51
|
Rocca C, Pasqua T, Cerra MC, Angelone T. Cardiac Damage in Anthracyclines Therapy: Focus on Oxidative Stress and Inflammation. Antioxid Redox Signal 2020; 32:1081-1097. [PMID: 31928066 DOI: 10.1089/ars.2020.8016] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Despite their serious side effects, anthracyclines (ANTs) are the most prescribed chemotherapeutic drugs because of their strong efficacy in both solid and hematological tumors. A major limitation to ANTs clinical application is the severe cardiotoxicity observed both acutely and chronically. The mechanism underlying cardiac dysfunction under chemotherapy is mainly dependent on the generation of oxidative stress and systemic inflammation, both of which lead to progressive cardiomyopathy and heart failure. Recent Advances: Over the years, the iatrogenic ANTs-induced cardiotoxicity was believed to be simply given by iron metabolism and reactive oxygen species production; however, several experimental data indicate that ANTs may use alternative damaging mechanisms, such as topoisomerase 2β inhibition, inflammation, pyroptosis, immunometabolism, and autophagy. Critical Issues: In this review, we aimed at discussing ANTs-induced cardiac injury from different points of view, updating and focusing on oxidative stress and inflammation, since these pathways are not exclusive or independent from each other but they together importantly contribute to the complexity of ANTs-induced multifactorial cardiotoxicity. Future Directions: A deeper understanding of the mechanistic signaling leading to ANTs side effects could reveal crucial targeting molecules, thus representing strategic knowledge to promote better therapeutic efficacy and lower cardiotoxicity during clinical application.
Collapse
Affiliation(s)
- Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Maria Carmela Cerra
- Laboratory of Organ and System Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| |
Collapse
|
52
|
Tong Y, Wang K, Sheng S, Cui J. Polydatin ameliorates chemotherapy-induced cognitive impairment (chemobrain) by inhibiting oxidative stress, inflammatory response, and apoptosis in rats. Biosci Biotechnol Biochem 2020; 84:1201-1210. [PMID: 31992173 DOI: 10.1080/09168451.2020.1722057] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Most breast cancer survivors receiving chemotherapy have severe cognitive impairment, often referred to as "chemobrain." Polydatin (PLD) is known to have many biological activities. Thus, this study aimed to determine whether symptoms of chemobrain can be prevented or relieved by PLD. The chemobrain models were established by intraperitoneal injection of doxorubicin (DOX, 2 mg/kg) in rats once a week for 4 weeks (DOX group and DOX+PLD group). In the PLD group and DOX+PLD group, PLD (50 mg/kg) was administered orally to rats every day. We found that PLD treatment significantly protected against DOX-induced learning and memory impairment, restored hippocampal histopathological architecture. Furthermore, PLD suppressed DOX-induced oxidative stress through up-regulating Nrf2, inhibited inflammatory response by activating the NF-κB pathway, and reduced hippocampal apoptosis. Therefore, the present study indicated that PLD offered neuroprotection against DOX-induced chemobrain. PLD may assist in preventing chemobrain after chemotherapy in patients with cancers.
Collapse
Affiliation(s)
- Yifan Tong
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Department of Breast Cancer Surgery, Tangshan Gongren Hospital, Tangshan, Hebei, People's Republic of China
| | - Kaijie Wang
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, Hebei, People's Republic of China
| | - Shuhai Sheng
- Department of Breast Cancer Surgery, Tangshan Gongren Hospital, Tangshan, Hebei, People's Republic of China
| | - Jianzhong Cui
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, Hebei, People's Republic of China
| |
Collapse
|
53
|
Hall SE, Smuder AJ, Hayward R. Effects of Calorie Restriction and Voluntary Exercise on Doxorubicin-Induced Cardiotoxicity. Integr Cancer Ther 2019; 18:1534735419843999. [PMID: 30999765 PMCID: PMC6475835 DOI: 10.1177/1534735419843999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Introduction: Doxorubicin (DOX) is a widely used chemotherapeutic agent with known cardiotoxic properties, while calorie restriction (CR) and exercise have well-documented cardioprotective effects. No studies have investigated the effects of CR alone or the combined effects of CR and exercise on DOX cardiotoxicity. Methods: Rats were divided into 4 groups based on their food intake (ad libitum or CR) and activity (sedentary or voluntary wheel running [WR]). After completing a 16-week treatment, animals received either DOX (15 mg/kg) or saline (SAL) and cardiac function was measured 5 days after treatment. Chromatography was used to quantify left ventricular DOX accumulation. Results: Left ventricular developed pressure (LVDP), end systolic pressure (ESP), and left ventricular maximal rate of pressure development (dP/dtmax) were significantly higher in the CR + DOX group when compared with DOX. Fractional shortening, LVDP, ESP, dP/dtmax, and dP/dtmin were significantly higher in the CR + WR + DOX group compared with the DOX group. In addition, the CR + WR + DOX group showed significantly higher LVDP and ESP compared with the WR + DOX group. DOX accumulation in the heart was 5-fold lower (P < .05) in the CR + WR + DOX group compared with the DOX group. Conclusion: This is the first study to demonstrate that CR can reduce cardiac DOX accumulation, and confirms the protective role of CR against DOX-induced cardiac dysfunction. Our data also show that combining a known cardioprotective intervention, exercise training, with CR results in additive benefits in the protection against DOX cardiotoxicity.
Collapse
Affiliation(s)
| | | | - Reid Hayward
- 3 University of Northern Colorado, Greeley, CO, USA
| |
Collapse
|
54
|
Liao D, Zhang C, Liu N, Cao L, Wang C, Feng Q, Yao D, Long M, Jiang P. Involvement of neurotrophic signaling in doxorubicin-induced cardiotoxicity. Exp Ther Med 2019; 19:1129-1135. [PMID: 32010279 DOI: 10.3892/etm.2019.8276] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/08/2019] [Indexed: 12/22/2022] Open
Abstract
Dose dependent cardiotoxicity is the primary side effect of doxorubicin (DOX), but the underlying molecular mechanisms remain unclear. An increasing amount of evidence has demonstrated that neurotrophic signaling plays a pivotal role in both neurons and the heart, but the biological association between neurotrophic signaling and DOX-induced cardiotoxicity remains unknown. The present study determined the level of neurotrophins and their receptors in the heart of rats following DOX administration. DOX was administered 7 times at a dose of 2.5 mg/kg once every 2 days via intraperitoneal injection. The present study revealed that cardiac injury parameters, such as creatine kinase (CK), creatine kinase-myocardial bound, lactate dehydrogenase, troponin T and aspartate transaminase in serum were significantly increased in the DOX group. Both the gene and protein expression of brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) in the heart were markedly decreased following DOX treatment. Notably, the protein level of BDNF in the serum was inhibited in DOX-treated rats, whereas DOX induced a significant increase in the protein level of NGF in the serum. DOX induced a significant decrease in the level of tropomyosin-associated kinase A (TrkA) and the ratio of pTrkA/TrkA and pTrkB/TrkB. Furthermore, the administration of DOX suppressed downstream protein kinase B and extracellular signal regulated kinase phosphorylation. The present study first demonstrated that BDNF/TrkB signaling and NGF/TrkA signaling were altered by DOX, which indicated that neurotrophic signaling was involved in DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Dehua Liao
- Department of Pharmacy, Hunan Cancer Hospital, Changsha, Hunan 410013, P.R. China
| | - Chen Zhang
- Department of Cardiology, Jining First People's Hospital, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Ni Liu
- Department of Pharmacy, Hunan Cancer Hospital, Changsha, Hunan 410013, P.R. China
| | - Lizhi Cao
- Department of Pharmacy, Hunan Cancer Hospital, Changsha, Hunan 410013, P.R. China
| | - Changshui Wang
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Qingyan Feng
- Department of Neurology, Jining First People's Hospital, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Dunwu Yao
- Department of Pharmacy, Hunan Cancer Hospital, Changsha, Hunan 410013, P.R. China
| | - Minghui Long
- Department of Pharmacy, Hunan Cancer Hospital, Changsha, Hunan 410013, P.R. China
| | - Pei Jiang
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, Shandong 272000, P.R. China
| |
Collapse
|
55
|
Jafarinezhad Z, Rafati A, Ketabchi F, Noorafshan A, Karbalay‐Doust S. Cardioprotective effects of curcumin and carvacrol in doxorubicin-treated rats: Stereological study. Food Sci Nutr 2019; 7:3581-3588. [PMID: 31763008 PMCID: PMC6848807 DOI: 10.1002/fsn3.1210] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 08/12/2019] [Indexed: 12/02/2022] Open
Abstract
Doxorubicin (DOX) is a cardiotoxic drug. To reduce the harmful effects of DOX, two plant-derived components, including curcumin (CUR) and carvacrol (CAR), were considered. This study aimed to assess the protective effects of CUR and CAR on DOX-induced cardiotoxicity using physiological and stereological evaluations. Male rats were randomly allocated to six groups. Group's I-VI received phosphate-buffered saline (PBS), CUR (100 mg kg-1 day-1), CAR (50 mg kg-1 day-1), DOX (4 mg kg-1 week-1), DOX-CUR, and DOX-CAR, respectively. On day 24, plasma troponin I and ECG were analyzed and the left ventricle underwent stereological assessment. The results showed a fivefold increase in troponin I in the DOX-treated animals compared to the PBS ones. Additionally, heart rate and QRS amplitude, respectively, reduced by 18% and 31% and QT interval and QRS duration, respectively, increased by 41% and 24% in the DOX group in comparison with the PBS rats (p < .05). The total volume of the myocardium and vessels and the number of cardiomyocyte nuclei also, respectively, decreased by 30%, 45%, and 43% in the DOX group compared to the PBS animals (atrophy of the ventricular tissues, p < .01). Besides, the mean volumes of the connective tissue and cardiomyocytes, respectively, increased by 46% and 52% in the DOX group (p < .01). In the DOX-CUR and DOX-CAR groups, the changes were prevented extensively in comparison with the DOX group (p < .01). Co-administration of CUR or CAR and doxorubicin for 24 days could improve the heart function and structural changes.
Collapse
Affiliation(s)
- Zahra Jafarinezhad
- Histomorphometry and Stereology Research CenterShiraz University of Medical SciencesShirazIran
- Department of PhysiologyShiraz University of Medical SciencesShirazIran
| | - Ali Rafati
- Histomorphometry and Stereology Research CenterShiraz University of Medical SciencesShirazIran
- Department of PhysiologyShiraz University of Medical SciencesShirazIran
| | - Farzaneh Ketabchi
- Department of PhysiologyShiraz University of Medical SciencesShirazIran
| | - Ali Noorafshan
- Histomorphometry and Stereology Research CenterShiraz University of Medical SciencesShirazIran
- Department of AnatomyShiraz University of Medical SciencesShirazIran
| | - Saied Karbalay‐Doust
- Histomorphometry and Stereology Research CenterShiraz University of Medical SciencesShirazIran
- Department of AnatomyShiraz University of Medical SciencesShirazIran
| |
Collapse
|
56
|
Al-malky HS, Al Harthi SE, Osman AMM. Major obstacles to doxorubicin therapy: Cardiotoxicity and drug resistance. J Oncol Pharm Pract 2019; 26:434-444. [DOI: 10.1177/1078155219877931] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BackgroundDoxorubicin is one of the most commonly prescribed and time-tested anticancer drugs. Although being considered as a first line drug in different types of cancers, the two main obstacles to doxorubicin therapy are drug-induced cardiotoxicity and drug resistance.MethodThe study utilizes systemic reviews on publications of previous studies obtained from scholarly journal databases including PubMed, Medline, Ebsco Host, Google Scholar, and Cochrane. The study utilizes secondary information obtained from health organizations using filters and keywords to sustain information relevancy. The study utilizes information retrieved from studies captured in the peer-reviewed journals on “doxorubicin-induced cardiotoxicity” and “doxorubicin resistance.”Discussion and resultsThe exact mechanisms of cardiotoxicity are not known; various hypotheses are studied. Doxorubicin can lead to free radical generation in various ways. The commonly proposed underlying mechanisms promoting doxorubicin resistance are the expression of multidrug resistance proteins as well as other causes.ConclusionIn this review, we have described the major obstacles to doxorubicin therapy, doxorubicin-induced cardiotoxicity as well as the mechanisms of cancer drug resistance and in following the treatment failures.
Collapse
Affiliation(s)
- Hamdan S Al-malky
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sameer E Al Harthi
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdel-Moneim M Osman
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
57
|
Zhang Y, Knight W, Chen S, Mohan A, Yan C. Multiprotein Complex With TRPC (Transient Receptor Potential-Canonical) Channel, PDE1C (Phosphodiesterase 1C), and A2R (Adenosine A2 Receptor) Plays a Critical Role in Regulating Cardiomyocyte cAMP and Survival. Circulation 2019; 138:1988-2002. [PMID: 29871977 DOI: 10.1161/circulationaha.118.034189] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND cAMP plays a critical role in regulating cardiomyocyte survival. Various cAMP signaling pathways behave distinctly or in opposition. We have previously reported that activation of cAMP hydrolysis by cyclic nucleotide phosphodiesterase 1C (PDE1C) promotes cardiomyocytes death/apoptosis, yet the underlying molecular mechanism remains unknown. In this study, we aimed to identify the specific cAMP signaling pathway modulated by PDE1C and determine the mechanism by which Ca2+/calmodulin-stimulated PDE1C is activated. METHODS To study cardiomyocyte death/apoptosis, we used both isolated mouse adult cardiomyocytes in vitro and doxorubicin-induced cardiotoxicity in vivo. We used a variety of pharmacological activators and inhibitors as well as genetically engineered molecular tools to manipulate the expression and activity of proteins of interest. RESULTS We found that the protective effect of PDE1C inhibition/deficiency on Ang II or doxorubicin-induced cardiomyocyte death/apoptosis is dependent on cAMP-generating adenosine A2 receptors (A2Rs), suggesting that PDE1C's cAMP-hydrolyzing activity selectively modulates A2R-cAMP signaling in cardiomyocytes. In addition, we found that the effects of PDE1C activation on Ang II-mediated cAMP reduction and cardiomyocyte death are dependent on transient receptor potential-canonical (TRPC) channels, in particular TRPC3. We also observed synergistic protective effects on cardiomyocyte survival from the combination of A2R stimulation together with PDE1 or TRPC inhibition. Coimmunostaining and coimmunoprecipitation studies showed that PDE1C is localized in proximity with A2R and TRPC3 in the plasma membrane and perhaps T tubules. It is important to note that we found that doxorubicin-induced cardiac toxicity and dysfunction in mice are attenuated by the PDE1 inhibitor IC86340 or in PDE1C knockout mice, and this protective effect is significantly diminished by A2R antagonism. CONCLUSIONS We have characterized a novel multiprotein complex comprised of A2R, PDE1C, and TRPC3, in which PDE1C is activated by TRPC3-derived Ca2+, thereby antagonizing A2R-cAMP signaling and promoting cardiomyocyte death/apoptosis. Targeting these molecules individually or in combination may represent a compelling therapeutic strategy for potentiating cardiomyocyte survival.
Collapse
Affiliation(s)
- Yishuai Zhang
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Walter Knight
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY.,Department of Pharmacology and Physiology (W.K., S.C.), University of Rochester School of Medicine and Dentistry, NY
| | - Si Chen
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY.,Department of Pharmacology and Physiology (W.K., S.C.), University of Rochester School of Medicine and Dentistry, NY
| | - Amy Mohan
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| | - Chen Yan
- Aab Cardiovascular Research Institute, Department of Medicine (Y.Z., W.K., S.C., A.M., C.Y.), University of Rochester School of Medicine and Dentistry, NY
| |
Collapse
|
58
|
Chang D, Li H, Qian C, Wang Y. DiOHF Protects Against Doxorubicin-Induced Cardiotoxicity Through ERK1 Signaling Pathway. Front Pharmacol 2019; 10:1081. [PMID: 31611788 PMCID: PMC6777440 DOI: 10.3389/fphar.2019.01081] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 08/26/2019] [Indexed: 11/13/2022] Open
Abstract
Doxorubicin (DOX) is an effective anticancer agent. Its clinical use is, however, limited due to its detrimental side effects, especially the cardiotoxicity caused by ROS, mitochondrial dysfunction and apoptosis. 3’,4’-dihydroxyflavonol (DiOHF) is a recently developed potent synthetic flavonoid which has been reported to exert anti-oxidative activity in myocardial ischemia–reperfusion injury and maintain the normal mitochondrial function. The aim of this study was to explore the protective effects of DiOHF on the DOX-induced cardiotoxicity. We established DOX-induced cardiotoxicity in H9C2 cells by incubation with 1 μM DOX and in BALB/c mice by DOX injection. DiOHF effectively prevented and reversed the DOX-induced cardiotoxicity, including ROS production, mitochondrial dysfunction, and apoptosis. The DOX-induced cardiotoxicity was accompanied by ERK1/2 activation and abolished by the silence of ERK1, rather than ERK2. Furthermore, DOX treatment in mice induced an increase in serum CK-MB level and myocardial fibrosis with a reduction in left ventricular (LV) function. These detrimental effects were blunted by DiOHF administration. Conclusion: DiOHF suppresses and reverses the DOX-induced cardiotoxicity by inhibiting ROS release, stabilizing mitochondrial function and reducing apoptosis through activation of the ERK1 signaling.
Collapse
Affiliation(s)
- Danqi Chang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hang Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Cheng Qian
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yanggan Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.,Medical Research Institute of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
59
|
Curdione Ameliorated Doxorubicin-Induced Cardiotoxicity Through Suppressing Oxidative Stress and Activating Nrf2/HO-1 Pathway. J Cardiovasc Pharmacol 2019; 74:118-127. [DOI: 10.1097/fjc.0000000000000692] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
60
|
Sun Y, Nemec-Bakk AS, Mallik AU, Bagchi AK, Singal PK, Khaper N. Blueberry extract attenuates doxorubicin-induced damage in H9c2 cardiac cells 1. Can J Physiol Pharmacol 2019; 97:880-884. [PMID: 31365282 DOI: 10.1139/cjpp-2019-0031] [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] [Indexed: 12/21/2022]
Abstract
The objective of this study was to analyze the cardioprotective roles of 3 wild blueberry genotypes and one commercial blueberry genotype by measuring markers of oxidative stress and cell death in H9c2 cardiac cells exposed to doxorubicin. Ripe berries of the 3 wild blueberry genotypes were collected from a 10-year-old clearcut forest near Nipigon, Ontario, Canada (49°1'39″N, 87°52'21″W), whereas the commercial blueberries were purchased from a local grocery store. H9c2 cardiac cells were incubated with 15 μg gallic acid equivalent/mL blueberry extract for 4 h followed by 5 μM doxorubicin for 4 h, and oxidative stress and active caspase 3/7 were analyzed. The surface area as well as total phenolic content was significantly higher in all 3 wild blueberry genotypes compared with the commercial species. Increase in oxidative stress due to doxorubicin exposure was attenuated by pre-treatment with all 3 types of wild blueberries but not by commercial berries. Furthermore, increase in caspase 3/7 activity was also attenuated by all 3 wild genotypes as well. These data demonstrate that wild blueberry extracts can attenuate doxorubicin-induced damage to H9c2 cardiomyocytes through reduction in oxidative stress and apoptosis, whereas the commercial blueberry had little effect.
Collapse
Affiliation(s)
- Yue Sun
- Department of Biology, Northern Ontario School of Medicine, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| | | | - Azim U Mallik
- Department of Biology, Northern Ontario School of Medicine, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| | - Ashim K Bagchi
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Pawan K Singal
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Neelam Khaper
- Department of Biology, Northern Ontario School of Medicine, Lakehead University, Thunder Bay, ON P7B 5E1, Canada.,Medical Sciences Division, Northern Ontario School of Medicine, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| |
Collapse
|
61
|
Astragaloside IV alleviates doxorubicin induced cardiomyopathy by inhibiting NADPH oxidase derived oxidative stress. Eur J Pharmacol 2019; 859:172490. [PMID: 31229536 DOI: 10.1016/j.ejphar.2019.172490] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 01/26/2023]
Abstract
Doxorubicin (DOX) is a classic anti-tumor chemotherapeutic used to treat a wide range of tumors. One major downfall of DOX treatment is it can induce fatal cardiotoxicity. Astragaloside IV (AS-IV) is one of the primary active ingredients that can be isolated from the traditional Chinese herbal medicine, Astragalus membranaceus. This study uses both in vitro and in vivo tools to investigate whether AS-IV alleviates DOX induced cardiomyopathy. We found that AS-IV supplementation alleviates body weight loss, myocardial injury, apoptosis of cardiomyocytes, cardiac fibrosis and cardiac dysfunction in DOX-treated mice. Also, DOX-induced cardiomyocyte injury and apoptosis were effectively improved by AS-IV treatment in vitro. NADPH oxidase (NOX) plays an important role in the progress of the oxidative signal transduction and DOX-induced cardiomyopathy. In this study, we found that AS-IV treatment relieves DOX-induced NOX2 and NOX4 expression and oxidative stress in cardiomyocytes. In conclusion, AS-IV, an antioxidant, attenuates DOX-induced cardiomyopathy through the suppression of NOX2 and NOX4.
Collapse
|
62
|
Bai T, Liu Y, Li B. LncRNA LOXL1‐AS1/miR‐let‐7a‐5p/
EGFR
‐related pathway regulates the doxorubicin resistance of prostate cancer DU‐145 cells. IUBMB Life 2019; 71:1537-1551. [PMID: 31188543 DOI: 10.1002/iub.2075] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/09/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Tianliang Bai
- Department of Gastrointestinal SurgeryAffiliated Hospital of Hebei University Baoding Hebei P.R.China
- Department of General SurgeryFourth Hospital of Hebei Medical University (Tumor Hospital of Hebei Province) Shijiiazhuang Hebei China
| | - Yabin Liu
- Department of Gastrointestinal SurgeryAffiliated Hospital of Hebei University Baoding Hebei P.R.China
- Department of General SurgeryFourth Hospital of Hebei Medical University (Tumor Hospital of Hebei Province) Shijiiazhuang Hebei China
| | - Binghui Li
- Department of Gastrointestinal SurgeryAffiliated Hospital of Hebei University Baoding Hebei P.R.China
- Department of General SurgeryFourth Hospital of Hebei Medical University (Tumor Hospital of Hebei Province) Shijiiazhuang Hebei China
| |
Collapse
|
63
|
Zheng WF, Zhang SY, Ma HF, Chang XW, Wang H. C1qTNF-related protein-6 protects against doxorubicin-induced cardiac injury. J Cell Biochem 2019; 120:10748-10755. [PMID: 30719766 DOI: 10.1002/jcb.28366] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 11/29/2018] [Indexed: 12/11/2022]
Abstract
The clinical use of doxorubicin (DOX) is limited by its toxic effect. However, there is no specific drug that can prevent DOX-related cardiac injury. C1qTNF-related protein-6 (CTRP6) is a newly identified adiponectin paralog with many protective functions on metabolism and cardiovascular diseases. However, little is known about the effect of CTRP6 on DOX-induced cardiac injury. The present study aimed to investigate whether CTRP6 could protect against DOX-related cardiotoxicity. To induce acute cardiotoxicity, the mice were intraperitoneally injected with a single dose of DOX (15 mg/kg). Cardiomyocyte-specific CTRP6 overexpression was achieved using an adenoassociated virus system at 4 weeks before DOX injection. The data in our study demonstrated that CTRP6 messenger RNA and protein expression were decreased in DOX-treated hearts. CTRP6 attenuated cardiac atrophy induced by DOX injection and inhibited cardiac apoptosis and improved cardiac function in vivo. CTRP6 also promoted the activation of protein kinase B (AKT/PKB) signaling pathway in DOX-treated mice. CTRP6 prevented cardiomyocytes from DOX-induced apoptosis and activated the AKT pathway in vitro. CTRP6 lost its protection against DOX-induced cardiac injury in mice with AKT inhibition. In conclusion, CTRP6 protected the heart from DOX-cardiotoxicity and improves cardiac function via activation of the AKT signaling pathway.
Collapse
Affiliation(s)
- Wei-Feng Zheng
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Shou-Yan Zhang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Hui-Fang Ma
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Xue-Wei Chang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Hao Wang
- Department of Cardiology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| |
Collapse
|
64
|
Kuzu M, Yıldırım S, Kandemir FM, Küçükler S, Çağlayan C, Türk E, Dörtbudak MB. Protective effect of morin on doxorubicin-induced hepatorenal toxicity in rats. Chem Biol Interact 2019; 308:89-100. [PMID: 31100273 DOI: 10.1016/j.cbi.2019.05.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/19/2019] [Accepted: 05/13/2019] [Indexed: 12/26/2022]
Abstract
Although Doxorubicin (DOX) is a widespread drug used in the treatment of cancer, its clinical use is restricted due to its common side effects. In addition, administrating DOX with an antioxidant has recently become a new strategy in preventing the side effects of DOX. The protective effects of morin, a natural flavonoid, against DOX-induced liver and kidney damage in rats were investigated biochemically, immunohistochemically and histopathologically in this study. The experimental procedure was planned as 10 days, and 5 groups consisting of seven rats were formed. Morin was given orally to rats at a dose of 50 and 100 mg/kg for 10 days and DOX was given a single dose of 40 mg/kg intraperitoneally on day 8. In order to determine the protective effect of morin against oxidative stress caused by DOX, reduced glutathione (GSH) and malondialdehyde (MDA) levels and superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) enzyme activities were measured in liver and kidney tissues. Liver and kidney tissue damage were determined both histopathologically and by serum alanine transaminase (ALT), aspartate transaminase (AST), urea and creatinine analysis. In order to determine the effect of DOX-induced inflammation and against the effect of morin, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and nuclear factor kappa B (NF-κB) levels were determined in both tissues. Liver and kidney B-cell lymphoma-2 (Bcl-2) levels were determined biochemically. In addition, Bax expression in liver tissue and aquaporin-2 (AQP-2) and nephrin expression in renal tissue were determined immunohistochemically. It was determined that oxidative damage caused by DOX decreased and improvement of liver and kidney function markers were observed in the groups that were treated with morin. In addition, pre-treatment of morin showed a regulatory effect on TNF-α, IL-1β and NF-κB levels. It prevented the increase in DOX-induced Bax expression and decrease in Bcl-2 level, AQP-2 and nephrin expression. Histopathological examination revealed that it prevented tissue damage in liver and kidney tissues.
Collapse
Affiliation(s)
- Müslüm Kuzu
- Department of Biochemistry, Faculty of Science and Letter, İbrahim Çeçen University of Ağrı, Ağrı, Turkey.
| | - Serkan Yıldırım
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Fatih Mehmet Kandemir
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Sefa Küçükler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Cüneyt Çağlayan
- Department of Biochemistry, Faculty of Veterinary Medicine, Bingöl University, Bingöl, Turkey
| | - Erdinç Türk
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Hatay, Turkey
| | | |
Collapse
|
65
|
Korga A, Ostrowska M, Jozefczyk A, Iwan M, Wojcik R, Zgorka G, Herbet M, Vilarrubla GG, Dudka J. Apigenin and hesperidin augment the toxic effect of doxorubicin against HepG2 cells. BMC Pharmacol Toxicol 2019; 20:22. [PMID: 31053173 PMCID: PMC6499973 DOI: 10.1186/s40360-019-0301-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 04/11/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common malignancies, with an increasing incidence. Despite the fact that systematic chemotherapy with a doxorubicin provides only marginal improvements in survival of the HCC patients, the doxorubicin is being used in transarterial therapies or combined with the target drug - sorafenib. The aim of the study was to evaluate the effect of natural flavonoids on the cytotoxicity of the doxorubicin against human hepatocellular carcinoma cell line HepG2. METHODS The effect of apigenin and its glycosides - cosmosiin, rhoifolin; baicalein and its glycosides - baicalin as well as hesperetin and its glycosides - hesperidin on glycolytic genes expression of HepG2 cell line, morphology and cells' viability at the presence of doxorubicin have been tested. In an attempt to elucidate the mechanism of observed results, the fluorogenic probe for reactive oxygen species (ROS), the DNA oxidative damage, the lipid peroxidation and the double strand breaks were evaluated. To assess impact on the glycolysis pathway, the mRNA expression for a hexokinase 2 (HK2) and a lactate dehydrogenase A (LDHA) enzymes were measured. The results were analysed statistically with the one-way analysis of variance (ANOVA) and post hoc multiple comparisons. RESULTS The apigenin and the hesperidin revealed the strongest effect on the toxicity of doxorubicin. Both flavonoids simultaneously changed the expression of the glycolytic pathway genes - HK2 and LDHA, which play a key role in the Warburg effect. Although separate treatment with doxorubicin, apigenin and hesperidin led to a significant oxidative DNA damage and double strand breaks, simultaneous administration of doxorubicin and apigenin or hesperidin abolished these damage with the simultaneous increase in the doxorubicin toxicity. CONCLUSION The obtained results indicate the existence of a very effective cytotoxic mechanism in the HepG2 cells of the combined effect of doxorubicin and apigenin (or hesperidin), not related to the oxidative stress. To explain this synergy mechanism, further research is needed, The observed intensification of the cytotoxic effect of doxorubicin by this flavonoids may be a promising direction of the research on the therapy of hepatocellular carcinoma, especially in a chemoembolization.
Collapse
Affiliation(s)
- Agnieszka Korga
- Independent Medical Biology Unit, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Marta Ostrowska
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Aleksandra Jozefczyk
- Department of Pharmacognosy with Medicinal Plant Laboratory, Medical University of Lublin, 1 Chodzko Street, 20-093 Lublin, Poland
| | - Magdalena Iwan
- Independent Medical Biology Unit, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Rafal Wojcik
- Department of Human Anatomy, Medical University of Lublin, 4 Jaczewski Street, 20-090 Lublin, Poland
| | - Grazyna Zgorka
- Department of Pharmacognosy with Medicinal Plant Laboratory, Medical University of Lublin, 1 Chodzko Street, 20-093 Lublin, Poland
| | - Mariola Herbet
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Gemma Gomez Vilarrubla
- Independent Medical Biology Unit, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Jaroslaw Dudka
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| |
Collapse
|
66
|
Ibrahim MA, Bakhaat GA, Tammam HG, Mohamed RM, El-Naggar SA. Cardioprotective effect of green tea extract and vitamin E on Cisplatin-induced cardiotoxicity in mice: Toxicological, histological and immunohistochemical studies. Biomed Pharmacother 2019; 113:108731. [DOI: 10.1016/j.biopha.2019.108731] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/16/2019] [Accepted: 02/22/2019] [Indexed: 02/01/2023] Open
|
67
|
Korga A, Ostrowska M, Iwan M, Herbet M, Dudka J. Inhibition of glycolysis disrupts cellular antioxidant defense and sensitizes HepG2 cells to doxorubicin treatment. FEBS Open Bio 2019; 9:959-972. [PMID: 30973680 PMCID: PMC6487699 DOI: 10.1002/2211-5463.12628] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/04/2019] [Accepted: 03/14/2019] [Indexed: 11/27/2022] Open
Abstract
Increased glucose consumption is a known hallmark of cancer cells. Increased glycolysis provides ATP, reducing agents and substrates for macromolecular synthesis in intensely dividing cells. Therefore, inhibition of glycolysis is one strategy in anticancer therapy as well as in improved efficacy of conventional anticancer chemotherapeutic agents. One such agent is doxorubicin (DOX), but the mechanism of sensitization of tumor cells to DOX by inhibition of glycolysis has not been fully elucidated. As oxidative stress is an important phenomenon accompanying DOX action and antioxidant defense is closely related to energy metabolism, the aim of the study was the evaluation of oxidative stress markers and antioxidant abilities of cancer cells treated with DOX while glycolysis is inhibited. HepG2 cells were treated with DOX and one of three glycolysis inhibitors: 2-deoxyglucose, dichloroacetate or 3-promopyruvate. To evaluate the possible interaction mechanisms, we assessed mRNA expression of selected genes related to energy metabolism and antioxidant defense; oxidative stress markers; and reduced glutathione (GSH) and NADPH levels. Additionally, glutamine consumption was measured. It was demonstrated that the chemotherapeutic agent and glycolysis inhibitors induced oxidative stress and associated damage in HepG2 cells. However, simultaneous treatment with both agents resulted in even greater lipid peroxidation and a significant reduction in GSH and NADPH levels. Moreover, in the presence of the drug and an inhibitor, HepG2 cells had a reduced ability to take up glutamine. These results indicated that cells treated with DOX while glycolysis was inhibited had significantly reduced ability to produce NADPH and antioxidant defenses.
Collapse
Affiliation(s)
- Agnieszka Korga
- Independent Medical Biology UnitMedical University of LublinPoland
| | | | - Magdalena Iwan
- Independent Medical Biology UnitMedical University of LublinPoland
| | - Mariola Herbet
- Department of ToxicologyMedical University of LublinPoland
| | - Jaroslaw Dudka
- Department of ToxicologyMedical University of LublinPoland
| |
Collapse
|
68
|
Zaki SM, Algaleel WA, Imam RA, Abdelmoaty MM. Mesenchymal stem cells pretreated with platelet-rich plasma modulate doxorubicin-induced cardiotoxicity. Hum Exp Toxicol 2019; 38:857-874. [PMID: 30991846 DOI: 10.1177/0960327119842613] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The cardiotoxic adverse effect of doxorubicin (DOX) is the major factor limiting its use. Recently, mesenchymal stem cells (MSCs) have been implicated in the preclinical studies of treatment of DOX-induced cardiotoxicity. The question is MSCs pretreated with platelet-rich plasma (PRP) have a better influence on DOX-induced cardiotoxicity compared to the influence of MSCs alone. Twenty-four Wistar rats were categorized into control, DOX-treated, MSC-treated, and PRP/MSC-treated groups. DOX was injected for two consecutive weeks. Light microscopic, biochemical markers (interleukin 10 (IL-10), tumor necrosis factor alpha (TNF-α), and creatine kinase-MB (CK-MB)), immunohistochemical (Bax, Bcl2, vascular endothelial growth factor (VEGF), and cardiac troponin-I (CT-I)), and oxidative/antioxidative markers (malondialdehyde (MDA)/superoxide dismutase (SOD)) were measured. Degenerative cardiac changes were detected in the DOX-treated group with complete loss of the architecture and coagulative necrosis. These changes were accompanied with the elevation of the serum level of CK-MB and loss of CT-I immunoreactivity. The major factors in the DOX-induced cardiotoxicity were the oxidative stress (elevated MDA/decreased SOD), inflammation (elevated TNF-α/decreased IL-10), and cardiac apoptosis (lower Bcl2, higher Bax, and lower Bcl2/Bax ratio). MSCs and PRP/MSCs attenuate DOX-induced cardiotoxicity. Better attenuation was observed in the PRP/MSC-treated group. PRP/MSC combination reduced greatly the MDA and TNF-α and increased IL-10, Bcl2/Bax ratio, and VEGF. PRP had no significant influence over the Bcl2, Bax, and SOD. In conclusion, DOX in its toxic dose induced myocardial injury. This destructive effect is related to oxidative stress, inflammation, and cardiac apoptosis. PRP/MSC possesses a better attenuation over the DOX-induced toxicity compared to MSC alone.
Collapse
Affiliation(s)
- S M Zaki
- 1 Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt.,2 Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Wa Abd Algaleel
- 1 Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt.,2 Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - R A Imam
- 1 Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - M M Abdelmoaty
- 1 Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
69
|
Rocca C, Scavello F, Colombo B, Gasparri AM, Dallatomasina A, Granieri MC, Amelio D, Pasqua T, Cerra MC, Tota B, Corti A, Angelone T. Physiological levels of chromogranin A prevent doxorubicin-induced cardiotoxicity without impairing its anticancer activity. FASEB J 2019; 33:7734-7747. [PMID: 30973759 DOI: 10.1096/fj.201802707r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The clinical use of doxorubicin (Doxo), a widely used anticancer chemotherapeutic drug, is limited by dose-dependent cardiotoxicity. We have investigated whether chromogranin A (CgA), a cardioregulatory protein released in the blood by the neuroendocrine system and by the heart itself, may contribute to regulation of the cardiotoxic and antitumor activities of Doxo. The effects of a physiologic dose of full-length recombinant CgA on Doxo-induced cardiotoxicity and antitumor activity were investigated in rats using in vivo and ex vivo models and in murine models of melanoma, fibrosarcoma, lymphoma, and lung cancer, respectively. The effect of Doxo on circulating levels of CgA was also investigated. In vivo and ex vivo mechanistic studies showed that CgA can prevent Doxo-induced heart inflammation, oxidative stress, apoptosis, fibrosis, and ischemic injury. On the other hand, CgA did not impair the anticancer activity of Doxo in all the murine models investigated. Furthermore, we observed that Doxo can reduce the intracardiac expression and release of CgA in the blood (i.e., an important cardioprotective agent). These findings suggest that administration of low-dose CgA to patients with low levels of endogenous CgA might represent a novel approach to prevent Doxo-induced adverse events without impairing antitumor effects.-Rocca, C., Scavello, F., Colombo, B., Gasparri, A. M., Dallatomasina, A., Granieri, M. C., Amelio, D., Pasqua, T., Cerra, M. C., Tota, B., Corti, A., Angelone, T. Physiological levels of chromogranin A prevent doxorubicin-induced cardiotoxicity without impairing its anticancer activity.
Collapse
Affiliation(s)
- Carmine Rocca
- Laboratory of Cellular and Molecular Cardiac Pathophysiology, Department of Biology, Ecology, and Earth Science, University of Calabria, Rende (Cosenza), Italy
| | - Francesco Scavello
- Laboratory of Cellular and Molecular Cardiac Pathophysiology, Department of Biology, Ecology, and Earth Science, University of Calabria, Rende (Cosenza), Italy
| | - Barbara Colombo
- Division of Experimental Oncology, Vita-Salute San Raffaele University-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Anna Maria Gasparri
- Division of Experimental Oncology, Vita-Salute San Raffaele University-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Alice Dallatomasina
- Division of Experimental Oncology, Vita-Salute San Raffaele University-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Maria Concetta Granieri
- Laboratory of Cellular and Molecular Cardiac Pathophysiology, Department of Biology, Ecology, and Earth Science, University of Calabria, Rende (Cosenza), Italy
| | - Daniela Amelio
- Laboratory of Cellular and Molecular Cardiac Pathophysiology, Department of Biology, Ecology, and Earth Science, University of Calabria, Rende (Cosenza), Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiac Pathophysiology, Department of Biology, Ecology, and Earth Science, University of Calabria, Rende (Cosenza), Italy
| | - Maria Carmela Cerra
- Laboratory of Cellular and Molecular Cardiac Pathophysiology, Department of Biology, Ecology, and Earth Science, University of Calabria, Rende (Cosenza), Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| | - Bruno Tota
- Laboratory of Cellular and Molecular Cardiac Pathophysiology, Department of Biology, Ecology, and Earth Science, University of Calabria, Rende (Cosenza), Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| | - Angelo Corti
- Division of Experimental Oncology, Vita-Salute San Raffaele University-Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiac Pathophysiology, Department of Biology, Ecology, and Earth Science, University of Calabria, Rende (Cosenza), Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| |
Collapse
|
70
|
Pan J, Rostamizadeh K, Filipczak N, Torchilin VP. Polymeric Co-Delivery Systems in Cancer Treatment: An Overview on Component Drugs' Dosage Ratio Effect. Molecules 2019; 24:E1035. [PMID: 30875934 PMCID: PMC6471357 DOI: 10.3390/molecules24061035] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
Multiple factors are involved in the development of cancers and their effects on survival rate. Many are related to chemo-resistance of tumor cells. Thus, treatment with a single therapeutic agent is often inadequate for successful cancer therapy. Ideally, combination therapy inhibits tumor growth through multiple pathways by enhancing the performance of each individual therapy, often resulting in a synergistic effect. Polymeric nanoparticles prepared from block co-polymers have been a popular platform for co-delivery of combinations of drugs associated with the multiple functional compartments within such nanoparticles. Various polymeric nanoparticles have been applied to achieve enhanced therapeutic efficacy in cancer therapy. However, reported drug ratios used in such systems often vary widely. Thus, the same combination of drugs may result in very different therapeutic outcomes. In this review, we investigated polymeric co-delivery systems used in cancer treatment and the drug combinations used in these systems for synergistic anti-cancer effect. Development of polymeric co-delivery systems for a maximized therapeutic effect requires a deeper understanding of the optimal ratio among therapeutic agents and the natural heterogenicity of tumors.
Collapse
Affiliation(s)
- Jiayi Pan
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA.
| | - Kobra Rostamizadeh
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA.
- Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan 4513956184, Iran.
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA.
- Laboratory of Lipids and Liposomes, Department of Biotechnology, University of Wroclaw, 50-383 Wroclaw, Poland.
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
71
|
Doxorubicin-induced cardiomyopathy associated with inhibition of autophagic degradation process and defects in mitochondrial respiration. Sci Rep 2019; 9:2002. [PMID: 30765730 PMCID: PMC6376057 DOI: 10.1038/s41598-018-37862-3] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/15/2018] [Indexed: 12/21/2022] Open
Abstract
Doxorubicin (Dox) is a highly effective anticancer drug but cause acute ventricular dysfunction, and also induce late-onset cardiomyopathy and heart failure. Despite extensive studies, the pathogenic sequelae leading to the progression of Dox-associated cardiomyopathy remains unknown. We assessed temporal changes in autophagy, mitochondrial dynamics, and bioenergetics in mouse models of acute and chronic Dox-cardiomyopathy. Time course study of acute Dox-treatment showed accumulation of LC3B II in heart lysates. Autophagy flux assays confirmed that the Dox-induced accumulation of autophagosomes occurs due to blockage of the lysosomal degradation process. Dox-induced autophagosomes and autolysosome accumulation were confirmed in vivo by using GFP-LC3 and mRFP-GFP-LC3 transgenic (Tg) mice. Mitochondria isolated from acute Dox-treated hearts showed significant suppression of oxygen consumption rate (OCR). Chronic Dox-cardiotoxicity also exhibited time-dependent accumulation of LC3B II levels and increased accumulation of green puncta in GFP-LC3 Tg hearts. Mitochondria isolated from chronic Dox-treated hearts also showed significant suppression of mitochondrial OCR. The in vivo impairment of autophagic degradation process and mitochondrial dysfunction data were confirmed in vitro using cultured neonatal cardiomyocytes. Both acute and chronic Dox-associated cardiomyopathy involves a multifocal disease process resulting from autophagosomes and autolysosomes accumulation, altered expression of mitochondrial dynamics and oxidative phosphorylation regulatory proteins, and mitochondrial respiratory dysfunction.
Collapse
|
72
|
Khajavi Rad A, Mohebbati R. Zataria multiflora extract and carvacrol affect cardiotoxicity induced by Adriamycin in rat. J Basic Clin Physiol Pharmacol 2018; 30:73-79. [PMID: 30110251 DOI: 10.1515/jbcpp-2018-0008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/25/2018] [Indexed: 06/08/2023]
Abstract
Background Because of the antioxidant effects of Zataria multiflora (ZM) and carvacrol (CAR) and also the role of oxidative stress in the induction of cardiotoxicity induced by Adriamycin (ADR), the aim of this study was to investigate the improvement effects of ZM extract and CAR on cardiotoxicity induced by ADR in rats. Methods Twenty-eight male rats were randomly assigned to four groups including (1) the control group; (2) the ADR group, which received ADR intravenously at the beginning of the study and the (3) ZM+ADR and (4) CAR+ADR groups, which received ZM and CAR by gavage for 28 consecutive days and ADR as single dose. Blood samples were collected on days 0 and 28 to determine serum glutamic oxaloacetic transaminase (SGOT), serum glutamic pyruvic transaminase (SGPT) and lactate dehydrogenase (LDH). Also, cardiac tissue was removed for redox marker evaluation. Results In the ADR group, malondialdehyde (MDA) significantly increased and superoxide dismutase (SOD) activity and total thiol contents significantly reduced, as compared with the control group, while CAR administration significantly improved this condition. Treatment with ZM significantly increased the SOD activity and total thiol content, as compared with the ADR group. The level of LDH significantly increased on day 28 in the ADR group compared to the control group, and administration of ZM and CAR significantly decreased it. The SGPT and SGOT levels in the ADR group significantly increased, and CAR administration significantly reduced them. Conclusion The results indicate that the administration of ZM hydroalcoholic extracts and its active ingredient, CAR, could reduce the oxidative stress damage through promotion of the cardiac and systemic antioxidant system. Also, CAR administration demonstrated better improvement in cardiotoxicity with ADR in rats.
Collapse
Affiliation(s)
- Abolfazl Khajavi Rad
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Mohebbati
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran, Phone: +985138828565, Fax: +985138828564, E-mail:
| |
Collapse
|
73
|
Ray S, Li Z, Hsu CH, Hwang LP, Lin YC, Chou PT, Lin YY. Dendrimer- and copolymer-based nanoparticles for magnetic resonance cancer theranostics. Theranostics 2018; 8:6322-6349. [PMID: 30613300 PMCID: PMC6299700 DOI: 10.7150/thno.27828] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/20/2018] [Indexed: 01/06/2023] Open
Abstract
Cancer theranostics is one of the most important approaches for detecting and treating patients at an early stage. To develop such a technique, accurate detection, specific targeting, and controlled delivery are the key components. Various kinds of nanoparticles have been proposed and demonstrated as potential nanovehicles for cancer theranostics. Among them, polymer-like dendrimers and copolymer-based core-shell nanoparticles could potentially be the best possible choices. At present, magnetic resonance imaging (MRI) is widely used for clinical purposes and is generally considered the most convenient and noninvasive imaging modality. Superparamagnetic iron oxide (SPIO) and gadolinium (Gd)-based dendrimers are the major nanostructures that are currently being investigated as nanovehicles for cancer theranostics using MRI. These structures are capable of specific targeting of tumors as well as controlled drug or gene delivery to tumor sites using pH, temperature, or alternating magnetic field (AMF)-controlled mechanisms. Recently, Gd-based pseudo-porous polymer-dendrimer supramolecular nanoparticles have shown 4-fold higher T1 relaxivity along with highly efficient AMF-guided drug release properties. Core-shell copolymer-based nanovehicles are an equally attractive alternative for designing contrast agents and for delivering anti-cancer drugs. Various copolymer materials could be used as core and shell components to provide biostability, modifiable surface properties, and even adjustable imaging contrast enhancement. Recent advances and challenges in MRI cancer theranostics using dendrimer- and copolymer-based nanovehicles have been summarized in this review article, along with new unpublished research results from our laboratories.
Collapse
Affiliation(s)
- Sayoni Ray
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Zhao Li
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Chao-Hsiung Hsu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Lian-Pin Hwang
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Ying-Chih Lin
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Pi-Tai Chou
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Yung-Ya Lin
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
74
|
Husain M, Nolan TS, Foy K, Reinbolt R, Grenade C, Lustberg M. An overview of the unique challenges facing African-American breast cancer survivors. Support Care Cancer 2018; 27:729-743. [PMID: 30460398 DOI: 10.1007/s00520-018-4545-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 11/07/2018] [Indexed: 01/08/2023]
Abstract
PURPOSE The existence of cancer disparities is well known. Focus on alleviating such disparities centers on diagnosis, treatment, and mortality. This review surveyed current knowledge of health disparities that exist in the acute survivorship period (immediately following diagnosis and treatment) and their contributors, particularly for African-American breast cancer survivors (AA-BCS). METHODS Utilizing the ASCO four components of survivorship care, we explore disparities in surveillance and effects of cancer and therapies that AA-BCS face within the acute survivorship period (the years immediately following diagnosis). A literature review of PUBMED, Scopus, and Cochrane databases was conducted to identify articles related to AA-BCS acute survivorship. The search yielded 97 articles. Of the 97 articles, 38 articles met inclusion criteria. RESULTS AA-BCS experience disparate survivorship care, which negatively impacts quality of life and health outcomes. Challenges exist in surveillance, interventions for late effects (e.g., quality-of-life outcomes, cardiotoxicity, and cognitive changes), preventing recurrence with promotion of healthy living, and coordinating care among the healthcare team. CONCLUSIONS This overview identified current knowledge on the challenges in survivorship among AA-BCS. Barriers to optimal survivorship care inhibit progress in eliminating breast cancer disparities. Research addressing best practices for survivorship care is needed for this population. Implementation of culturally tailored care may reduce breast cancer disparities among AA-BCS.
Collapse
Affiliation(s)
- Marium Husain
- The Ohio State University, 320 W. 10th Ave, Suite A455, Columbus, OH, 43210, USA.
| | - Timiya S Nolan
- The Ohio State University, 320 W. 10th Ave, Suite A455, Columbus, OH, 43210, USA
| | - Kevin Foy
- The Ohio State University, 320 W. 10th Ave, Suite A455, Columbus, OH, 43210, USA
| | - Raquel Reinbolt
- Medical Oncology, The Ohio State University, 320 W. 10th Ave, Suite A455, Columbus, OH, 43210, USA
| | - Cassandra Grenade
- Medical Oncology, The Ohio State University, 320 W. 10th Ave, Suite A455, Columbus, OH, 43210, USA
| | - Maryam Lustberg
- Medical Oncology, The Ohio State University, 320 W. 10th Ave, Suite A455, Columbus, OH, 43210, USA
| |
Collapse
|
75
|
Khadka D, Kim HJ, Oh GS, Shen A, Lee S, Lee SB, Sharma S, Kim SY, Pandit A, Choe SK, Kwak TH, Yang SH, Sim H, Eom GH, Park R, So HS. Augmentation of NAD + levels by enzymatic action of NAD(P)H quinone oxidoreductase 1 attenuates adriamycin-induced cardiac dysfunction in mice. J Mol Cell Cardiol 2018; 124:45-57. [PMID: 30291911 DOI: 10.1016/j.yjmcc.2018.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 09/18/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Adriamycin (ADR) is a powerful chemotherapeutic agent extensively used to treat various human neoplasms. However, its clinical utility is hampered due to severe adverse side effects i.e. cardiotoxicity and heart failure. ADR-induced cardiomyopathy (AIC) has been reported to be caused by myocardial damage and dysfunction through oxidative stress, DNA damage, and inflammatory responses. Nonetheless, the remedies for AIC are even not established. Therefore, we illustrate the role of NAD+/NADH modulation by NAD(P)H quinone oxidoreductase 1 (NQO1) enzymatic action on AIC. METHODS AND RESULTS AIC was established by intraperitoneal injection of ADR in C57BL/6 wild-type (WT) and NQO1 knockout (NQO1-/-) mice. All Mice were orally administered dunnione (named NQO1 substrate) before and after exposure to ADR. Cardiac biomarker levels in the plasma, cardiac dysfunction, oxidative biomarkers, and mRNA and protein levels of pro-inflammatory mediators were determined compared the cardiac toxicity of each experimental group. All biomarkers of Cardiac damage and oxidative stress, and mRNA levels of pro-inflammatory cytokines including cardiac dysfunction were increased in ADR-treated both WT and NQO1-/- mice. However, this increase was significantly reduced by dunnione in WT, but not in NQO1-/- mice. In addition, a decrease in SIRT1 activity due to a reduction in the NAD+/NADH ratio by PARP-1 hyperactivation was associated with AIC through increased nuclear factor (NF)-κB p65 and p53 acetylation in both WT and NQO1-/- mice. While an elevation in NAD+/NADH ratio via NQO1 enzymatic action using dunnione recovered SIRT1 activity and subsequently deacetylated NF-κB p65 and p53, however not in NQO1-/- mice, thereby attenuating AIC. CONCLUSION Thus, modulation of NAD+/NADH by NQO1 may be a novel therapeutic approach to prevent chemotherapy-associated heart failure, including AIC.
Collapse
Affiliation(s)
- Dipendra Khadka
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Hyung-Jin Kim
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Gi-Su Oh
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - AiHua Shen
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - SeungHoon Lee
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Su-Bin Lee
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Subham Sharma
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Seon Young Kim
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Arpana Pandit
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Seong-Kyu Choe
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Tae Hwan Kwak
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea
| | - Sei-Hoon Yang
- Internal Medicine, School of Medicine Wonkwang, University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Hyuk Sim
- Internal Medicine, School of Medicine Wonkwang, University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Gwang Hyeon Eom
- Department of Pharmacology, Medical Research Center for Gene Regulation Chonnam, National University Medical School, Hwasungun Jeollanam-do 58128, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science & Engineering, Institute of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hong-Seob So
- Center for Metabolic Function Regulation, & Department of Microbiology, Republic of Korea..
| |
Collapse
|
76
|
Chen H, Gao L, Huang Z, Liu Y, Guo S, Xing J, Meng Z, Liang C, Li Y, Yao R, Li L, Zhang Y, Gu H, Liu Y. C1qTNF-related protein 1 attenuates doxorubicin-induced cardiac injury via activation of AKT. Life Sci 2018; 207:492-498. [DOI: 10.1016/j.lfs.2018.06.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/27/2018] [Accepted: 06/28/2018] [Indexed: 11/15/2022]
|
77
|
Men Y, Peng S, Yang P, Jiang Q, Zhang Y, Shen B, Dong P, Pang Z, Yang W. Biodegradable Zwitterionic Nanogels with Long Circulation for Antitumor Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:23509-23521. [PMID: 29947223 DOI: 10.1021/acsami.8b03943] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Zwitterionic nanocarriers have emerged as a new class of biocompatible nanomaterials with outstanding stealth capability in blood circulation. In this work, a novel biodegradable zwitterionic nanogel based on poly(sulfobetaine methacrylate) (PSBMA) was developed for reduction-responsive drug delivery to tumors. PSBMA nanogels were facilely fabricated by one-step reflux precipitation polymerization with the advantage of being surfactant-free and time-saving. The disulfide bond not only endowed the nanogels degradability in a reduction environment but also be modified with a fluorescent group after partial reduction. In vitro release experiments disclosed that doxorubicin (DOX)-loaded PSBMA nanogels could hold the drugs firmly in physiological conditions (only 7% release in 24 h) and release the drugs rapidly and sufficiently in 10 mM glutathione (85% in 8 h). More interestingly, PSBMA nanogels displayed long circulation in blood after intravenous injection, and small change was found in half-life of nanogels between the first (34.1 h) and the second injection (30.5 h), indicating that there was no accelerated blood clearance phenomenon for these nanogels. Meanwhile, no obvious immunogenic response was detected after PSBMA nanogels were injected into BALB/c mice. Furthermore, PSBMA nanogels showed a high accumulation of 9.5 and 10.7% of injected dose per gram of tissue in tumors at 24 and 48 h post intravenous injection, respectively. With outstanding long circulation time, high tumor accumulation, and sufficient drug release in a reduction environment, DOX-loaded PSBMA nanogels demonstrated the strongest tumor growth inhibition effect among all of the treatment groups in human hypopharyngeal carcinoma-bearing mouse models. Therefore, our study provided a facile drug delivery platform based on biodegradable zwitterionic nanogels and may have great potential in tumor drug delivery.
Collapse
Affiliation(s)
- Yongzhi Men
- Shanghai General Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200080 , P. R. China
| | - Shaojun Peng
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science , Fudan University , Shanghai 200433 , P. R. China
| | - Peng Yang
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science , Fudan University , Shanghai 200433 , P. R. China
| | - Qin Jiang
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science , Fudan University , Shanghai 200433 , P. R. China
| | - Yanhui Zhang
- Shanghai General Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200080 , P. R. China
| | - Bin Shen
- Shanghai General Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200080 , P. R. China
| | - Pin Dong
- Shanghai General Hospital , Shanghai Jiao Tong University School of Medicine , Shanghai 200080 , P. R. China
| | - Zhiqing Pang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai 201203 , P. R. China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers & Department of Macromolecular Science , Fudan University , Shanghai 200433 , P. R. China
| |
Collapse
|
78
|
Cochera F, Dinca D, Bordejevic DA, Citu IM, Mavrea AM, Andor M, Trofenciuc M, Tomescu MC. Nebivolol effect on doxorubicin-induced cardiotoxicity in breast cancer. Cancer Manag Res 2018; 10:2071-2081. [PMID: 30038521 PMCID: PMC6053261 DOI: 10.2147/cmar.s166481] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Purpose The aim of this study was to assess whether nebivolol treatment could have beneficial effects in the prevention of anthracyclines-induced cardiotoxicity. Patients and methods Our prospective study included 60 women, mean age 52.6±13 years, with HER2 negative breast cancer, scheduled to undergo treatment with doxorubicin. The patients were randomly divided into two groups: the treatment group (n=30) which received nebivolol 5 mg once daily for the duration of chemotherapy and the control group (n=30) without treatment with nebivolol. Cytostatic treatment was performed with doxorubicin 70 mg/m2 administered intravenously every 21 days for six cycles. The average cumulative dose of doxorubicin was 520±8 mg/m2. Echocardiography was performed immediately before and after six cycles of doxorubicin therapy. Results We found no significant differences between the two groups regarding baseline clinical and echocardiographic parameters. The two groups reached a similar cumulative dose of doxorubicin. No patient died during the study. None of the patients withdrew from chemotherapy. After six cycles of doxorubicin therapy, the left ventricular (LV) ejection fraction, shortening fraction, and LV diameters changed, but not significantly. Tissue Doppler imaging (TDI) detected in the control group a significant decrease of myocardial velocities, indicating a LV diastolic dysfunction. In the same group, speckle tracking imaging (STI) revealed a statistically significant alteration of the ventricular deformation, which means a decrease in LV systolic function. In the nebivolol treatment group, no significant alterations in the LV systolic and diastolic function were observed. Conclusion The results of this study show the benefit of new echocardiographic imaging methods such as TDI and STI in the screening of early cardiac dysfunction induced by cytostatic treatment. Nebivolol treatment prevented the occurrence of anthracyclines-induced cardiomyopathy in the short term. In order to confirm these preliminary results, larger studies with a longer follow-up period are required.
Collapse
Affiliation(s)
- Flavia Cochera
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania,
| | - Daniel Dinca
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania,
| | - Diana Aurora Bordejevic
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania,
| | - Ioana Mihaela Citu
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania,
| | - Adelina Marioara Mavrea
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania,
| | - Minodora Andor
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania,
| | - Mihai Trofenciuc
- Cardiology Department, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania,
| | | |
Collapse
|
79
|
Silva RC, Britto DMC, de Fátima Pereira W, Brito-Melo GEA, Machado CT, Pedreira MM. Effect of short- and medium-term toxicity of doxorubicin on spermatogenesis in adult Wistar rats. Reprod Biol 2018; 18:169-176. [DOI: 10.1016/j.repbio.2018.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/24/2018] [Accepted: 03/13/2018] [Indexed: 10/17/2022]
|
80
|
Sangomla S, Saifi MA, Khurana A, Godugu C. Nanoceria ameliorates doxorubicin induced cardiotoxicity: Possible mitigation via reduction of oxidative stress and inflammation. J Trace Elem Med Biol 2018; 47:53-62. [PMID: 29544808 DOI: 10.1016/j.jtemb.2018.01.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 01/26/2018] [Accepted: 01/30/2018] [Indexed: 12/20/2022]
Abstract
Doxorubicin (DOX) is one of the most commonly used anticancer drugs but its use has been limited due to constraints of cardiotoxic side effects. The precise mechanism underlying cardiotoxicity is not yet fully understood but oxidative stress has been found to be a primary mechanism behind this. In addition, DOX induced cardiotoxicity also shows involvement of proinflammatory cytokines such as IL-6 and TNF-α. Since oxidative stress plays major role in DOX induced cardiotoxicity, different antioxidants have been tried to prevent cardiotoxicity of DOX. Nanoparticles have risen up as a promising material with a wide variety of actions, and cerium oxide nanoparticles or nanoceria (NC) is one of such kind with great antioxidant potential. NC has emerged as a promising antioxidant in different pathological conditions. The present study was aimed to investigate possible protective effects of NC in DOX induced cardiotoxicity. Cardiotoxicity was induced in Swiss mice by DOX administration through i.p. route at a dose level of 15 mg/kg in two divided doses on alternate days. In our study, NC was found to mitigate cardiotoxic potential of DOX and prevented weight loss. NC restored the levels of cardiac injury markers lactate dehydrogenase (LDH) and creatinine kinase MB (CK-MB). Moreover, NC reduced malondialdehyde (MDA) levels and increased endogenous antioxidants such as reduced glutathione (GSH) and catalase levels. In addition, NC decreased proinflammatory cytokine levels and also prevented the alteration in normal structure of heart samples. Our study showed protective effects of NC in DOX induced cardiotoxicity which can become a potential therapeutic intervention against DOX induced cardiotoxicity.
Collapse
Affiliation(s)
- Swetha Sangomla
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Mohd Aslam Saifi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Amit Khurana
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
81
|
Angiotensin-converting enzyme 2 overexpression protects against doxorubicin-induced cardiomyopathy by multiple mechanisms in rats. Oncotarget 2018; 8:24548-24563. [PMID: 28445944 PMCID: PMC5421869 DOI: 10.18632/oncotarget.15595] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/13/2017] [Indexed: 11/25/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is considered a potential therapeutic target of the renin-angiotensin system (RAS) for the treatment of cardiovascular diseases. We aimed to explore the effects of ACE2 overexpression on doxorubicin-induced cardiomyopathy in rats. Rats were randomly divided into treatment and control groups. The rats of treatment group were injected intraperitoneally with 6 doses of doxorubicin (2.5 mg/kg) within a period of two weeks. Two weeks after the initial injection of doxorubicin, these rats were randomly divided into Mock, Ad-EGFP, Ad-ACE2, and Cilazapril groups. The rats of Ad-EGFP and Ad-ACE2 groups received intramyocardial injection of Ad-EGFP and Ad-ACE2, respectively. The rats of Cilazapril group received cilazapril (10 mg/kg/day) via intragastric intubation. Apoptosis, inflammation, oxidative stress, cardiac function, the extent of myocardial fibrosis, and levels of ACE2, ACE, angiotensin II (AngII), and angiotensin (1–7) were evaluated. Four weeks after ACE2 gene transfer, the Ad-ACE2 group showed not only reduced apoptosis, inflammatory response, oxidative stress, left ventricular (LV) volume, extent of myocardial fibrosis and mortality of rats, but also increased LV ejection fraction and ACE2 expression level compared with the Mock and Ad-EGFP groups. ACE2 overexpression was superior to cilazapril in improving doxorubicin-induced cardiomyopathy. The putative mechanisms may involve activation of the AMPK and PI3K-AKT pathways, inhibition of the ERK pathway, decrease of TGF-β1 expression, and interactions of shifting RAS components, such as decreased myocardium AngII levels, increased myocardium Ang (1–7) levels, and reduced ACE expression. Thus, ACE2 may be a novel therapeutic approach to prevent and treat doxorubicin-induced cardiomyopathy.
Collapse
|
82
|
Pecoraro M, Ciccarelli M, Fiordelisi A, Iaccarino G, Pinto A, Popolo A. Diazoxide Improves Mitochondrial Connexin 43 Expression in a Mouse Model of Doxorubicin-Induced Cardiotoxicity. Int J Mol Sci 2018. [PMID: 29518932 PMCID: PMC5877618 DOI: 10.3390/ijms19030757] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Doxorubicin (DOXO) administration induces alterations in Connexin 43 (Cx43) expression and localization, thus, inducing alterations in chemical and electrical signal transmission between cardiomyocytes and in intracellular calcium homeostasis even evident after a single administration. This study was designed to evaluate if Diazoxide (DZX), a specific opener of mitochondrial KATP channels widely used for its cardioprotective effects, can fight DOXO-induced cardiotoxicity in a short-time mouse model. DZX (20 mg/kg i.p.) was administered 30 min before DOXO (10 mg/kg i.p.) in C57BL/6j female mice for 1–3 or seven days once every other day. A recovery of cardiac parameters, evaluated by Echocardiography, were observed in DZX+DOXO co-treated mice. Western blot analysis performed on heart lysates showed an increase in sarco/endoplasmic reticulum Ca2+-ATPase (SERCAII) and a reduction in phospholamban (PLB) amounts in DZX+DOXO co-treated mice. A contemporary recovery of intracellular Ca2+-signal, detected spectrofluorometrically by means of FURA-2AM, was observed in these mice. Cx43 expression and localization, analyzed by Western blot and confirmed by immunofluorescence analysis, showed that DZX co-treatement increases Cx43 amount both on sarcoplasmic membrane and on mitochondria. In conclusion, our data demonstrate that, in a short-time mouse model of DOXO-induced cardiotoxicity, DZX exerts its cardioprotective effects also by enhancing the amount Cx43.
Collapse
Affiliation(s)
- Michela Pecoraro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy.
| | - Michele Ciccarelli
- Department of Medicine and Surgery, University of Salerno, 84084 Baronissi, Italy.
| | - Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, Federico II University, 80138 Naples, Italy.
| | - Guido Iaccarino
- Department of Medicine and Surgery, University of Salerno, 84084 Baronissi, Italy.
| | - Aldo Pinto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy.
| | - Ada Popolo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, Italy.
| |
Collapse
|
83
|
Mohajeri M, Sahebkar A. Protective effects of curcumin against doxorubicin-induced toxicity and resistance: A review. Crit Rev Oncol Hematol 2018; 122:30-51. [PMID: 29458788 DOI: 10.1016/j.critrevonc.2017.12.005] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/28/2017] [Accepted: 12/11/2017] [Indexed: 02/08/2023] Open
Abstract
Doxorubicin (DOX)-induced toxicity and resistance are major obstacles in chemotherapeutic approaches. Despite effective in the treatment of numerous malignancies, some clinicians have voiced concern that DOX has the potential to cause debilitating consequences in organ tissues, especially the heart. The mechanisms of toxicity and resistance are respectively related to induction of reactive oxygen species (ROS) and up-regulation of ATP-binding cassette (ABC) transporter. Curcumin (CUR) with several biological and pharmacological properties is expected to restore DOX-mediated impairments to tissues. This review is intended to address the current knowledge on DOX adverse effects and CUR protective actions in the heart, kidneys, liver, brain, and reproductive organs. Coadministration of CUR and DOX is capable of ameliorating DOX toxicity pertained to antioxidant, apoptosis, autophagy, and mitochondrial permeability.
Collapse
Affiliation(s)
- Mohammad Mohajeri
- Department of Medical Biotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
84
|
Possible Mechanisms of the Prevention of Doxorubicin Toxicity by Cichoric Acid-Antioxidant Nutrient. Nutrients 2018; 10:nu10010044. [PMID: 29303987 PMCID: PMC5793272 DOI: 10.3390/nu10010044] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 12/13/2022] Open
Abstract
Skin is the largest organ in the human body, and which protects organism against unfavorable external factors e.g., chemicals, environment pollutants, allergens, microorganisms, and it plays a crucial role in maintaining general homeostasis. It is also an important target of oxidative stress due to the activity of oxygen reactive species (ROS), which are constantly generated in the fibroblasts in response to exogenous or endogenous prooxidant agents. An example of such compound with proved prooxidant activity is Doxorubicin (DOX), which is an effective anticancer agent belongs in anthracycline antibiotic group. Increasingly frequent implementation of various strategies to reduce undesirable DOX side effects was observed. Very promising results come from the combination of DOX with dietary antioxidants from the polyphenol group of compounds, such as cichoric acid (CA) in order to lower oxidative stress level. The aim of this work was to evaluate the influence of CA combined with DOX on the oxidative stress parameters in fibroblasts, which constitute the main cells in human skin. We also wanted to examine anti-apoptotic activity of CA in fibroblasts treated with selected concentrations of DOX. Results obtained from the combination of DOX with CA revealed that CA exhibits cytoprotective activity against DOX-induced damage by lowering oxidative stress level and by inhibiting apoptosis. The present finding may indicate that CA may serve as antioxidative and anti-apoptotic agent, active against DOX-induced damage.
Collapse
|
85
|
Cappetta D, Rossi F, Piegari E, Quaini F, Berrino L, Urbanek K, De Angelis A. Doxorubicin targets multiple players: A new view of an old problem. Pharmacol Res 2018; 127:4-14. [DOI: 10.1016/j.phrs.2017.03.016] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 01/22/2023]
|
86
|
Hosseini A, Sahebkar A. Reversal of Doxorubicin-induced Cardiotoxicity by Using Phytotherapy: A Review. J Pharmacopuncture 2017; 20:243-256. [PMID: 30151294 PMCID: PMC6104714 DOI: 10.3831/kpi.2017.20.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 12/13/2022] Open
Abstract
Doxorubicin as a chemotherapeutic drug is widely used for the treatment of patients with cancer. However, clinical use of this drug is hampered by its cardiotoxicity, which is manifested as electrocardiographic abnormalities, arrhythmias, irreversible degenerative cardiomyopathy and congestive heart failure. The precise mechanisms underlying the cardiotoxicity of doxorubicin are not clear, but impairment of calcium homeostasis, generation of iron complexes, production of oxygen radicals, mitochondrial dysfunction and cell membrane damage have been suggested as potential etiologic factors. Compounds that can neutralize the toxic effect of doxorubicin on cardiac cells without reducing the drug's antitumor activity are needed. In recent years, numerous studies have shown that herbal medicines and bioactive phytochemicals can serve as effective add-on therapies to reduce the cardiotoxic effects of doxorubicin. This review describes different phytochemicals and herbal products that have been shown to counterbalance doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Azar Hosseini
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad,
Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad,
Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad,
Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad,
Iran
| |
Collapse
|
87
|
Niccoli S, Boreham DR, Phenix CP, Lees SJ. Non-radioactive 2-deoxy-2-fluoro-D-glucose inhibits glucose uptake in xenograft tumours and sensitizes HeLa cells to doxorubicin in vitro. PLoS One 2017; 12:e0187584. [PMID: 29095925 PMCID: PMC5667878 DOI: 10.1371/journal.pone.0187584] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/23/2017] [Indexed: 12/23/2022] Open
Abstract
A glucose analog called 2-deoxy-D-glucose (2DG) has been successfully used to sensitize cancer cells to ROS-inducing cancer treatments such as ionizing radiation, through the inhibition of glycolysis. However, the use of 2DG can be limited by several factors such as availability, non-specific cytotoxicity, and chemoresistance under hypoxic conditions. The purpose of this study was to investigate the use of non-radioactive 2-deoxy-2-fluoro-D-glucose (19FDG), a drug that potentially addresses current limitations of 2DG. The effectiveness of using either 2DG or 19FDG in combination with doxorubicin (Dox) in HeLa cells was determined in both normoxia and hypoxia. We have also shown that under both oxygen conditions, 19FDG-treated cells produce less lactate than 2DG-treated cells, an important finding that suggests improved inhibition of glycolysis, the preferential pathway for cancerous cells. When used in combination with Dox, we have demonstrated a significant decrease in the number of viable cells, with the effect of 19FDG remaining stable across both normoxic and hypoxic conditions. Moreover, the assessment of apoptosis and necrosis revealed that 19FDG maintained its ability to sensitize HeLa cells to Dox in hypoxia, but 2DG was only effective under normoxic conditions. The retained effectiveness of 19FDG in combination with Dox under hypoxic conditions, suggests that 19FDG may be efficacious for sensitizing hypoxic regions of solid tumour masses. Importantly, the ability of 19FDG to inhibit glucose uptake in vivo was also confirmed using positron emission tomography (PET) of xenograft tumours. The results displayed here suggest 19FDG is a promising combination therapy, which may lead to decreased ROS scavenging via glycolysis, and enhanced treatment success.
Collapse
Affiliation(s)
- Sarah Niccoli
- Medical Sciences, Lakehead University Faculty of Medicine, Thunder Bay, Ontario, Canada
| | - Douglas R. Boreham
- Medical Sciences Division, Northern Ontario School of Medicine, Thunder Bay, Ontario, Canada
| | - Christopher P. Phenix
- Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Thunder Bay Regional Health Research Institute, Thunder Bay, Ontario, Canada
| | - Simon J. Lees
- Medical Sciences, Lakehead University Faculty of Medicine, Thunder Bay, Ontario, Canada
- Medical Sciences Division, Northern Ontario School of Medicine, Thunder Bay, Ontario, Canada
- Biology, Lakehead University, Thunder Bay, Ontario, Canada
- * E-mail:
| |
Collapse
|
88
|
Dhingra A, Jayas R, Afshar P, Guberman M, Maddaford G, Gerstein J, Lieberman B, Nepon H, Margulets V, Dhingra R, Kirshenbaum LA. Ellagic acid antagonizes Bnip3-mediated mitochondrial injury and necrotic cell death of cardiac myocytes. Free Radic Biol Med 2017; 112:411-422. [PMID: 28838842 DOI: 10.1016/j.freeradbiomed.2017.08.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/03/2017] [Accepted: 08/13/2017] [Indexed: 01/10/2023]
Abstract
The Bcl-2 protein Bnip3 is crucial for provoking oxidative injury to mitochondria following anthracycline treatment or ischemia-reperfusion injury. Herein, we investigate the effects of the polyphenolic compound ellagic acid (EA) on Bnip3 mediated mitochondrial injury and necrotic cell death in cardiac myocytes. In contrast to vehicle treated cardiomyocytes, Bnip3 was highly enriched in mitochondrial fractions of cardiac myocytes treated with the anthracycline doxorubicin or in cells subjected to hypoxia (HPX). Mitochondrial associated Bnip3 was accompanied by mPTP opening and loss of ∆Ψm. The dynamin related fission protein Drp-1 was phosphorylated (Drp1616) and coincided with excessive mitochondrial fragmentation, mitophagy and necrosis in cardiac myocytes treated with doxorubicin or subjected to hypoxia. Moreover, knock-down of Bnip3 was sufficient to prevent mitochondrial fission and doxorubicin-induced cell death supporting the involvement of Bnip3 in doxorubicin cardiotoxity. Interestingly, mitochondrial associated Bnip3 in cells treated with doxorubicin was markedly reduced by EA. This resulted in significantly less mitochondrial fission and cell death. Notably, EA similarly suppressed mitochondrial injury and cell death induced by hypoxia or Bnip3 over-expression. Herein, we identify a novel signaling axis that operationally links EA and Bnip3 for suppression of cardiac cell death. We provide compelling new evidence that EA suppresses mitochondrial injury and necrotic cell death of cardiac myocytes by functionally abrogating Bnip3 activity. Hence, by suppressing mitochondrial injury induced by Bnip3, EA may provide a therapeutic advantage in reducing oxidative injury and cardiac dysfunction in cancer patients undergoing anthracycline treatment or individuals with ischemic cardiac stress.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antibiotics, Antineoplastic/toxicity
- Apoptosis/drug effects
- Autophagy/drug effects
- Cell Hypoxia/drug effects
- Doxorubicin/antagonists & inhibitors
- Doxorubicin/toxicity
- Dynamins/genetics
- Dynamins/metabolism
- Ellagic Acid/pharmacology
- Membrane Potential, Mitochondrial/drug effects
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondrial Dynamics/drug effects
- Mitochondrial Dynamics/genetics
- Mitochondrial Membrane Transport Proteins/genetics
- Mitochondrial Membrane Transport Proteins/metabolism
- Mitochondrial Permeability Transition Pore
- Mitochondrial Proteins/antagonists & inhibitors
- Mitochondrial Proteins/genetics
- Mitochondrial Proteins/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Necrosis/genetics
- Necrosis/metabolism
- Necrosis/pathology
- Phosphorylation/drug effects
- Primary Cell Culture
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Abhinav Dhingra
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Rahul Jayas
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Pegah Afshar
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Matthew Guberman
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Graham Maddaford
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Johnathan Gerstein
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Brooke Lieberman
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Hilary Nepon
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Victoria Margulets
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Rimpy Dhingra
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Lorrie A Kirshenbaum
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6.
| |
Collapse
|
89
|
Renu K, Abilash V, Tirupathi Pichiah P, Syeda TA, Arunachalam S. Adriamycin-induced cardiomyopathy can serve as a model for diabetic cardiomyopathy – a hypothesis. Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2017.09.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
90
|
Jiang Y, Li M, Lu Z, Wang Y, Yu X, Sui D, Fu L. Ginsenoside Rg3 induces ginsenoside Rb1-comparable cardioprotective effects independent of reducing blood pressure in spontaneously hypertensive rats. Exp Ther Med 2017; 14:4977-4985. [PMID: 29201202 PMCID: PMC5704305 DOI: 10.3892/etm.2017.5198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 08/17/2017] [Indexed: 11/19/2022] Open
Abstract
Ginsenoside Rg3 (Rg3) is a rare type of ginsenoside used as an anti-tumor medicine in China. Ginsenoside Rb1 (Rb1), which exhibits protective effects on the cardiovascular system, is similar to Rg3 in chemical structure. In the present study, Rb1 and Rg3 were administered for 6 weeks to spontaneously hypertensive rats (SHR) and their cardioprotective effects were assessed. According to echocardiography and histopathological examinations, the decrease in cardiac function and ventricular remodeling that occurred in SHR rats were attenuated by Rb1 and Rg3. However, tail-cuff blood pressure measurements indicated that Rb1 and Rg3 did not reduce blood pressure in SHR rats. The cardioprotective effects of Rb1 and Rg3 occurred independently of blood pressure reduction. Furthermore, immunohistochemistry (IHC) revealed that renin angiotensin system (RAS) activity in the myocardium of SHR was significantly attenuated by Rb1 and Rg3, whereas ELISA identified no significant changes of RAS activity in the serum. The results of IHC and reverse transcription-quantitative polymerase chain reaction demonstrated that levels of transforming growth factor β1, tumor necrosis factor-α, interleukin-6, interleukin-1 and endothelian-1 in the myocardium of SHR rats were reduced following Rb1 and Rg3 treatment. This may be due to the attenuation of RAS activity in the myocardium and the mechanisms of the cardioprotective effects of Rb1 and Rg3.
Collapse
Affiliation(s)
- Yichuan Jiang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Min Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zeyuan Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuchen Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Li Fu
- Jilin Yatai Pharmaceutical Co., Ltd., Changchun, Jilin 130033, P.R. China
- Institute of Dalian Fusheng Natural Medicine, Dalian, Liaoning 116600, P.R. China
| |
Collapse
|
91
|
Molecular mechanism of doxorubicin-induced cardiomyopathy - An update. Eur J Pharmacol 2017; 818:241-253. [PMID: 29074412 DOI: 10.1016/j.ejphar.2017.10.043] [Citation(s) in RCA: 396] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/11/2017] [Accepted: 10/20/2017] [Indexed: 12/27/2022]
Abstract
Doxorubicin is utilized for anti-neoplastic treatment for several decades. The utility of this drug is limited due to its side effects. Generally, doxorubicin toxicity is originated from the myocardium and then other organs are also ruined. The mechanism of doxorubicin is intercalated with the DNA and inhibits topoisomerase 2. There are various signalling mechanisms involved in doxorubicin cardiotoxicity. First and foremost, the doxorubicin-induced cardiotoxicity is due to oxidative stress. Cardiac mitochondrial damage is supposed after few hours following the revelation of doxorubicin. This has led important new uses for the mechanism of doxorubicin-induced cardiotoxicity and novel avenues of investigation to determine better pharmacotherapies and interventions for the impediment of cardiotoxicity. The idea of this review is to bring up to date the recent findings of the mechanism of doxorubicin cardiomyopathies such as calcium dysregulation, endoplasmic reticulum stress, impairment of progenitor cells, activation of immune, ubiquitous system and some other parameters.
Collapse
|
92
|
Jian CY, Ouyang HB, Xiang XH, Chen JL, Li YX, Zhou X, Wang JY, Yang Y, Zhong EY, Huang WH, Zhang HW. Naringin protects myocardial cells from doxorubicin‑induced apoptosis partially by inhibiting the p38MAPK pathway. Mol Med Rep 2017; 16:9457-9463. [PMID: 29152646 PMCID: PMC5780003 DOI: 10.3892/mmr.2017.7823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 04/06/2017] [Indexed: 01/06/2023] Open
Abstract
Doxorubicin (DOX) has been widely used to treat cancers as a first-line antitumor drug. However, it causes severe, irreversible, dose-dependent cardiotoxicity. To evaluate the protective effects of naringin (NRG) on cardiotoxicity, the authors investigated the molecular mechanism of the p38MAPK signaling pathway. H9c2 cells were treated for 24 h by using 5 µmol/l DOX without or with being pretreated by 1 µM NRG for 150 min or by 3 µM SB203580 for 60 min. Cell viability was detected by cell counting kit-8 assay. Intracellular reactive oxygen species (ROS) levels were detected based on the oxidative conversion of 2′,7′-dichlorfluorescein-diacetate (cell-permeable) to dichlorofluorescein (fluorescent). The expression of p38MAPK was determined by western blotting. The expression level of p-p38MAPK in H9c2 cells, which was significantly increased by exposure to 5 µM DOX for 60 min (P<0.01), was significantly decreased by pretreatment with 1 µM NRG for 150 min beforehand (P<0.01). The viability of H9c2 cells pretreated for 150 min with 1 µM NRG was significantly enhanced compared with that using DOX directly (P<0.01). Intracellular ROS levels were significantly reduced by being pretreated with 1 µM NRG for 150 min or with 3 µM SB203580 for 60 min before the cells were exposed to 5 µM DOX. Collectively, NRG protected H9c2 cells against the cardiotoxicity induced by DOX through suppressing the expression and activity of the p38MAPK pathway. The findings provided valuable evidence for the possible use of NRG to relieve DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Chun-Yan Jian
- Department of Cardiology, Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong 524000, P.R. China
| | - Han-Bin Ouyang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xian-Hong Xiang
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jia-Li Chen
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yong-Xin Li
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xin Zhou
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jin-Yang Wang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yang Yang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - En-Yi Zhong
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wen-Hua Huang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hong-Wu Zhang
- Department of Anatomy, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
93
|
Hajra S, Basu A, Singha Roy S, Patra AR, Bhattacharya S. Attenuation of doxorubicin-induced cardiotoxicity and genotoxicity by an indole-based natural compound 3,3'-diindolylmethane (DIM) through activation of Nrf2/ARE signaling pathways and inhibiting apoptosis. Free Radic Res 2017; 51:812-827. [PMID: 28922986 DOI: 10.1080/10715762.2017.1381694] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The most crucial complication related to doxorubicin (DOX) therapy is nonspecific cytotoxic effect on healthy normal cells. The clinical use of this broad-spectrum chemotherapeutic agent is restricted due to development of severe form of cardiotoxicity, myelosuppression, and genotoxicity which interfere with therapeutic schedule, compromise treatment outcome and may lead to secondary malignancy. 3,3'-diindolylmethane (DIM) is a naturally occurring plant alkaloid formed by the hydrolysis of indolylmethyl glucosinolate (glucobrassicin). Therefore, the present study was undertaken to investigate the protective role of DIM against DOX-induced toxicity in mice. DOX was administered (5 mg/kg b.w., i.p.) and DIM was administered (25 mg/kg b.w., p.o.) in concomitant and 15 days pretreatment schedule. Results showed that DIM significantly attenuated DOX-induced oxidative stress in the cardiac tissues by reducing the levels of free radicals and lipid peroxidation, and by enhancing the level of glutathione (reduced) and the activity of antioxidant enzymes. The chemoprotective potential of DIM was confirmed by histopathological evaluation of heart and bone marrow niche. Moreover, DIM considerably mitigated DOX-induced clastogenicity, DNA damage, apoptosis, and myeloid hyperplasia in bone marrow niche. In addition, oral administration of DIM significantly (p < .05) stimulated the Nrf2-mediated activation of antioxidant response element (ARE) pathway and promoted expression of ARE-driven cytoprotective proteins, HO-1, NQO1, and glutathione-S-transferase (GST). In connection with that, DIM significantly attenuated DOX-induced apoptosis by upregulation of Bcl-2 expression and downregulation of Bax and caspase-3 expression. Thus, this study suggests that DIM has promising chemoprotective efficacy against DOX-induced toxicity and indicates its future use as an adjuvant in chemotherapy.
Collapse
Affiliation(s)
- Subhadip Hajra
- a Department of Cancer Chemoprevention , Chittaranjan National Cancer Institute , Kolkata , India
| | - Abhishek Basu
- a Department of Cancer Chemoprevention , Chittaranjan National Cancer Institute , Kolkata , India
| | - Somnath Singha Roy
- a Department of Cancer Chemoprevention , Chittaranjan National Cancer Institute , Kolkata , India.,b Centre of Biomedical Research, SGPGIMS Campus , Lucknow , India
| | - Arup Ranjan Patra
- a Department of Cancer Chemoprevention , Chittaranjan National Cancer Institute , Kolkata , India
| | - Sudin Bhattacharya
- a Department of Cancer Chemoprevention , Chittaranjan National Cancer Institute , Kolkata , India
| |
Collapse
|
94
|
Stachowiak P, Wojtarowicz A, Milchert-Leszczyńska M, Safranow K, Falco M, Kaliszczak R, Kornacewicz-Jach Z. The paradox of the first cycle of chemotherapy-transient improvement of contractility and diastolic function after the first cycle of anthracycline-based chemotherapy: a prospective clinical trial. Oncotarget 2017; 8:96442-96452. [PMID: 29221219 PMCID: PMC5707113 DOI: 10.18632/oncotarget.21279] [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: 07/26/2017] [Accepted: 09/08/2017] [Indexed: 01/20/2023] Open
Abstract
Aims Breast cancer is the most common cancer among women, and anthracyclines are the most commonly administered drugs for these patients. Cardiotoxicity is one of the complications, which limits the success of this therapy. Very few studies have evaluated anthracycline toxicities within the first few hours after the first infusion, and the majority of published studies were performed in animal models. The present study aimed to evaluate changes in echocardiographic parameters in women with breast cancer 24 hours after receiving the first dose of an anthracycline. Materials and Methods and Results The present study included 75 chemotherapy-naive female patients without heart failure, who were diagnosed with breast cancer and were scheduled to undergo anthracycline-based chemotherapy (epirubicin and doxorubicin). During their visits to the Heart Center, the patients underwent detail echocardiographic examination, including assessment of systolic and diastolic function and longitudinal strain. There were no differences in baseline echocardiographic parameters between patients with and those without cardiotoxicity. Cardiotoxicity was observed during follow-up in 14 patients (18.7%). Improvements in left ventricular ejection fraction and global longitudinal strain were observed at 24 hours after administration of the cytotoxic agent in the subgroup of patients without further cardiotoxicity. The changes were transient and the assessment of left ventricular ejection fraction after completion of chemotherapy revealed similar values to those before the treatment. Conclusions The findings of our study suggest that transient improvement in contractility and systolic and diastolic function might occur 24 hours after anthracycline administration, especially in patients who do not develop cardiotoxicity.
Collapse
Affiliation(s)
- Paweł Stachowiak
- Department of Cardiology, Pomeranian Medical University, Szczecin, Poland
| | | | - Marta Milchert-Leszczyńska
- Department of Radiotherapy, West Pomeranian Oncology Center, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Michał Falco
- Department of Radiotherapy, West Pomeranian Oncology Center, Pomeranian Medical University, Szczecin, Poland
| | - Robert Kaliszczak
- Department of Cardiology, Pomeranian Medical University, Szczecin, Poland
| | | |
Collapse
|
95
|
Maillet A, Tan KP, Brunham LR. Use of Human Pluripotent Stem Cell Derived‐Cardiomyocytes to Study Drug‐Induced Cardiotoxicity. ACTA ACUST UNITED AC 2017; 73:22.5.1-22.5.22. [DOI: 10.1002/cptx.30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Agnes Maillet
- Translational Laboratory in Genetic Medicine, Agency for Science Technology and Research Singapore
| | - Kim Peng Tan
- Translational Laboratory in Genetic Medicine, Agency for Science Technology and Research Singapore
| | - Liam R. Brunham
- Translational Laboratory in Genetic Medicine, Agency for Science Technology and Research Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore Singapore
- Department of Medicine, Centre for Heart Lung Innovation, University of British Columbia Vancouver Canada
| |
Collapse
|
96
|
Xu F, Li X, Liu L, Xiao X, Zhang L, Zhang S, Lin P, Wang X, Wang Y, Li Q. Attenuation of doxorubicin-induced cardiotoxicity by esculetin through modulation of Bmi-1 expression. Exp Ther Med 2017; 14:2216-2220. [PMID: 28962145 PMCID: PMC5609155 DOI: 10.3892/etm.2017.4763] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/04/2017] [Indexed: 01/27/2023] Open
Abstract
The protective effects and mechanisms of esculetin on doxorubicin (DOX)-induced injury of H9c2 cells were investigated. H9c2 cells were cultured and the logarithmic growth phase of the cells was divided into a control group, a DOX group and an esculetin + DOX group. Cell viability was detected by MTT assay. Annexin V-PI (AV-PI) double staining flow cytometry was carried out to detect cell apoptosis. Intracellular reactive oxygen species (ROS) were detected by flow cytometry. Transmission electron microscope (TEM) was used to evaluate cell ultrastructure. Cleaved caspase-3, cleaved PARP, Bcl-2, Bid and Bmi-1 proteins levels were investigated by western blot analysis. Bmi-1 siRNA was used to detect the role of Bmi-1 in the protective effects of esculetin against DOX-induced toxicity in H9c2 cells. The MTT and AV-PI double staining results showed that esculetin significantly increased H9c2 cell viability. Compared with the control group, the levels of cleaved caspase-3, cleaved PARP, Bid and ROS levels were significantly decreased, but the expression of Bcl-2 and Bmi-1 were significantly increased in the esculetin + DOX group. TEM showed that the cell structure of the mitochondria was protected by esculetin. The results of Bmi-1 siRNA showed that esculetin could protect DOX-induced cardiotoxicity by modulating Bmi-1 expression. Esculetin can protect DOX-induced cardiotoxicity and the effects may be attributable to modulation of Bmi-1 expression, provoking intracellular ROS accumulation, protecting the structure of mitochondria and reducing cell apoptosis.
Collapse
Affiliation(s)
- Fan Xu
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Xiao Li
- Department of Radiology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Lanfang Liu
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Xu Xiao
- Department of Pharmacy, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Li Zhang
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Shenglin Zhang
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Pingping Lin
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Xiaojie Wang
- Basic Medical Institute, Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Yongwei Wang
- Department of Anatomy, Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Qingshan Li
- Department of Oncology, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
97
|
Korga A, Józefczyk A, Zgórka G, Homa M, Ostrowska M, Burdan F, Dudka J. Evaluation of the phytochemical composition and protective activities of methanolic extracts of Centaurea borysthenica and Centaurea daghestanica (Lipsky) Wagenitz on cardiomyocytes treated with doxorubicin. Food Nutr Res 2017; 61:1344077. [PMID: 28747863 PMCID: PMC5510226 DOI: 10.1080/16546628.2017.1344077] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 06/05/2017] [Indexed: 12/13/2022] Open
Abstract
Centaurea L. is a genus of the family Asteraceae that comprises over 600 taxa. Representatives of the Centaurea genus were used as natural medications for many diseases. Methanolic-aqueous extracts from aerial parts of two Centaurea species: C. borysthenica Gruner and C. daghestanica (Lipsky) Wagenitz were studied for their polyphenolic composition and potential protective effect on cardiomyocytes treated with doxorubicin. Effectiveness of doxorubicin in cancer therapy is limited by a dose-dependent cardiotoxicity. Oxidative stress is a widely recognized mechanism of this phenomenon. One of the most important strategies has been an application of drug together with antioxidant agents. A cardioprotective effect of selected extracts of Centaurea species was suspected in this study. Cell viability, oxidative stress, and mitochondrial membrane potential analyses showed protective activity of the methanolic extract of C. borysthenica and C. daghestanica on rat cardiomyocytes treated with doxorubicin. Although C. borysthenica is more effective as a cardiomyocyte protective agent, in higher concentrations it weakened the drug activity. C. daghestanica extract did not change the doxorubicin efficacy in the evaluated experiment. Interestingly, both tested extracts were cytotoxic for myeloma cells. The detected antioxidant activity of the studied extracts can be used in the prevention of doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Agnieszka Korga
- Independent Medical Biology Unit, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Józefczyk
- Department of Pharmacognosy with Medicinal Plant Unit, Medical University of Lublin, Lublin, Poland
| | - Grażyna Zgórka
- Department of Pharmacognosy with Medicinal Plant Unit, Medical University of Lublin, Lublin, Poland
| | - Mateusz Homa
- Independent Medical Biology Unit, Medical University of Lublin, Lublin, Poland
| | - Marta Ostrowska
- Department of Toxicology, Medical University of Lublin, Lublin, Poland
| | - Franciszek Burdan
- Department of Human Anatomy, Medical University of Lublin, Lublin, Poland
| | - Jarosław Dudka
- Independent Medical Biology Unit, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
98
|
Khames A, Khalaf MM, Gad AM, Abd El-Raouf OM. Ameliorative effects of sildenafil and/or febuxostat on doxorubicin-induced nephrotoxicity in rats. Eur J Pharmacol 2017; 805:118-124. [PMID: 28257823 DOI: 10.1016/j.ejphar.2017.02.046] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 02/16/2017] [Accepted: 02/27/2017] [Indexed: 02/05/2023]
Abstract
Sildenafil and febuxostat protect against doxorubicin-induced nephrotoxicity; however the exact mechanism remains to be elucidated. The effect of sildenafil and febuxostat on doxorubicin-induced nephrotoxicity in rats was studied. Male rats were subdivided into nine groups. The 1st group served as normal control, the 2nd group received dimethylsulfoxide 50% (DMSO), the 3rd group received doxorubicin (3.5mg/kg, i.p.), twice weekly for 3 weeks. The next 3 groups received sildenafil (5mg/kg; p.o.), febuxostat (10mg/kg; p.o.) and their combination, respectively daily for 21 days. The last 3 groups received doxorubicin in combination with sildenafil, febuxostat or their combination. Nephrotoxicity was evaluated histopathologically by light microscopy and biochemically through measuring the following parameters, Kidney function biomarkers [serum levels of urea, creatinine and uric acid], oxidative stress biomarkers [kidney contents of glutathione reduced (GSH) and malondialdehyde (MDA)], The apoptotic marker namely; caspase-3 in kidney tissue and the inflammatory mediator tumor necrosis factor alpha (TNF-α). doxorubicin-induced a significant elevation in nephrotoxicity markers, expression of caspase-3 and caused induction of inflammation and oxidative stress. Histological changes in the kidney was tubular necrosis. Sildenafil and/or febuxostat administration with doxorubicin caused a significant decrease in nephrotoxicity markers and inflammatory mediators, restoration of normal values of oxidative stress biomarkers and hampering the expression of renal caspase-3. They also ameliorate histological changes induced by doxorubicin. sildenafil and febuxostat are promising protective agents against doxorubicin-nephrotoxicity through improving biochemical, inflammatory, histopathological and immunohistochemical alterations induced by doxorubicin.
Collapse
Affiliation(s)
- Ali Khames
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Marwa M Khalaf
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| | - Amany M Gad
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Ola M Abd El-Raouf
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| |
Collapse
|
99
|
Khames A, khalaf MM, Gad AM, Abd El-Raouf OM. Ameliorative effects of sildenafil and/or febuxostat on doxorubicin-induced nephrotoxicity in rats. Eur J Pharmacol 2017. [DOI: https://doi.org/10.1016/j.ejphar.2017.02.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
100
|
Marano F, Frairia R, Rinella L, Argenziano M, Bussolati B, Grange C, Mastrocola R, Castellano I, Berta L, Cavalli R, Catalano MG. Combining doxorubicin-nanobubbles and shockwaves for anaplastic thyroid cancer treatment: preclinical study in a xenograft mouse model. Endocr Relat Cancer 2017; 24:275-286. [PMID: 28487350 DOI: 10.1530/erc-17-0045] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/04/2017] [Indexed: 01/04/2023]
Abstract
Anaplastic thyroid cancer is one of the most lethal diseases, and a curative therapy does not exist. Doxorubicin, the only drug approved for anaplastic thyroid cancer treatment, has a very low response rate and causes numerous side effects among which cardiotoxicity is the most prominent. Thus, doxorubicin delivery to the tumor site could be an import goal aimed to improve the drug efficacy and to reduce its systemic side effects. We recently reported that, in human anaplastic thyroid cancer cell lines, combining doxorubicin-loaded nanobubbles with extracorporeal shock waves, acoustic waves used in lithotripsy and orthopedics without side effects, increased the intracellular drug content and in vitro cytotoxicity. In the present study, we tested the efficacy of this treatment on a human anaplastic thyroid cancer xenograft mouse model. After 21 days, the combined treatment determined the greatest drug accumulation in tumors with consequent reduction of tumor volume and weight, and an extension of the tumor doubling time. Mechanistically, the treatment induced tumor apoptosis and decreased cell proliferation. Finally, although doxorubicin caused the increase of fibrosis markers and oxidative stress in animal hearts, loading doxorubicin into nanobubbles avoided these effects preventing heart damage. The improvement of doxorubicin anti-tumor effects together with the prevention of heart damage suggests that the combination of doxorubicin-loaded nanobubbles with extracorporeal shock waves might be a promising drug delivery system for anaplastic thyroid cancer treatment.
Collapse
Affiliation(s)
| | - Roberto Frairia
- Department of Medical SciencesUniversity of Turin, Turin, Italy
| | - Letizia Rinella
- Department of Medical SciencesUniversity of Turin, Turin, Italy
| | - Monica Argenziano
- Department of Drug Science and TechnologyUniversity of Turin, Turin, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of Turin, Turin, Italy
| | - Cristina Grange
- Department of Medical SciencesUniversity of Turin, Turin, Italy
| | | | | | | | - Roberta Cavalli
- Department of Drug Science and TechnologyUniversity of Turin, Turin, Italy
| | | |
Collapse
|