1
|
Al-Maamari A, Sultan M, Wu S, Zhang T, Wang C, Han B, Duan Y, Ding SS, Chen N, Zhang H, Sun F, Chen X, Yu D, Su S. Activation of sigma 1 receptor attenuates doxorubicin-induced cardiotoxicity by alleviating oxidative stress, mitochondria dysfunction, ER stress-related apoptosis, and autophagy impairment. Int J Biol Macromol 2025; 310:143549. [PMID: 40294674 DOI: 10.1016/j.ijbiomac.2025.143549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
The cardiotoxic effects of doxorubicin (DOX) are a significant clinical challenge. This study explored the mechanisms underlying the cardioprotective effects of fluvoxamine (FLU) in DOX-induced cardiotoxicity. In vitro and in vivo models of DOX-induced cardiotoxicity were established using H9c2 cardiomyocytes and BALB/c mice, respectively. The cardioprotective effects of FLU were evaluated using echocardiography, electrocardiography, HE staining, myocardial injury indicators, immunohistochemical staining, immunofluorescence staining, and western blotting. Small interfering RNA (siRNA) targeting the sigma-1 receptor (Sig1R), the Sig1R inhibitor haloperidol (HALO), and the nuclear factor erythroid 2-related factor 2 inhibitor ML385 were used to verify the mechanism of FLU in H9c2 cells, and molecular docking was performed to compare the binding affinities of DOX and FLU to Sig1R. DOX treatment significantly decreased Sig1R expression, leading to cardiac dysfunction, endoplasmic reticulum stress, apoptosis, impaired autophagy, oxidative stress, and mitochondrial dysfunction. Treatment with FLU mitigated these effects. Molecular docking simulations revealed that FLU showed better binding affinity for Sig1R than DOX. Moreover, the beneficial effects of FLU in DOX-induced cardiotoxicity were reduced in the presence of HALO, ML385, or Sig1R siRNA. Our study indicates that FLU effectively reduces DOX-induced cardiotoxicity by activating Sig1R, highlighting its potential as a therapeutic agent against chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ahmed Al-Maamari
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Marwa Sultan
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Shang Wu
- Breast Cancer Center, The Fourth Hospital, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Tao Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Chuchu Wang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Boye Han
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Yuxin Duan
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Shan-Shan Ding
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Nannan Chen
- School of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Fangyi Sun
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Xueyan Chen
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Ding Yu
- Cardiovascular Intensive Care Unit, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050023, PR China
| | - Suwen Su
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China.
| |
Collapse
|
2
|
Wiese W, Galita G, Siwecka N, Rozpędek-Kamińska W, Slupianek A, Majsterek I. Endoplasmic Reticulum Stress in Acute Myeloid Leukemia: Pathogenesis, Prognostic Implications, and Therapeutic Strategies. Int J Mol Sci 2025; 26:3092. [PMID: 40243748 PMCID: PMC11988921 DOI: 10.3390/ijms26073092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy that poses a significant therapeutic challenge due to its high recurrence rate and demanding treatment regimens. Increasing evidence suggests that endoplasmic reticulum (ER) stress and downstream activation of the unfolded protein response (UPR) pathway play a key role in the pathogenesis of AML. ER stress is triggered by the accumulation of misfolded or unfolded proteins within the ER. This causes activation of the UPR to restore cellular homeostasis. However, the UPR can shift from promoting survival to inducing apoptosis under prolonged or excessive stress conditions. AML cells can manipulate the UPR pathway to evade apoptosis, promoting tumor progression and resistance against various therapeutic strategies. This review provides the current knowledge on ER stress in AML and its prognostic and therapeutic implications.
Collapse
MESH Headings
- Humans
- Endoplasmic Reticulum Stress
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/diagnosis
- Unfolded Protein Response
- Prognosis
- Apoptosis
- Animals
- Signal Transduction
Collapse
Affiliation(s)
- Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| | - Grzegorz Galita
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| | - Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| | - Artur Slupianek
- Office of the Vice President for Research, Temple University, Philadelphia, PA 19140, USA
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (W.W.); (G.G.); (N.S.); (W.R.-K.)
| |
Collapse
|
3
|
Xu W, Tang C, Zhao R, Wang Y, Jiao H, Ang H, Chen Y, Wang X, Liang Y. Sydnthiones are versatile bioorthogonal hydrogen sulfide donors. Nat Commun 2024; 15:10288. [PMID: 39604436 PMCID: PMC11603141 DOI: 10.1038/s41467-024-54765-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
Hydrogen sulfide (H2S) is an important endogenous gasotransmitter, but the bioorthogonal reaction triggered H2S donors are still rare. Here we show one type of bioorthogonal H2S donors, sydnthiones (1,2,3-oxadiazol-3-ium-5-thiolate derivatives), which was designed with the aid of density functional theory (DFT) calculations. The reactions between sydnthiones and strained alkynes provide a platform for controllable, tunable and mitochondria-targeted release of H2S. We investigate the reactivity of sydnthiones‒dibenzoazacyclooctyne (DIBAC) reactions and their orthogonality with two other bioorthogonal cycloaddition pairs: tetrazine‒norbornene (Nor) and tetrazine‒monohydroxylated cyclooctyne (MOHO). By taking advantage of these mutually orthogonal reactions, we can realize selective labeling or drug release. Furthermore, we explore the role of H2S, which is released from the sydnthione-DIBAC reaction, on doxorubicin-induced cytotoxicity. The results demonstrate that the viability of H9c2 cells can be significantly improved by pretreating with sydnthione 1b and DIBAC for 6 h prior to exposure to Dox.
Collapse
Affiliation(s)
- Wenyuan Xu
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Inter-disciplinary Research Center, Nanjing University, Nanjing, 210023, China
| | - Cheng Tang
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Inter-disciplinary Research Center, Nanjing University, Nanjing, 210023, China
| | - Ruohan Zhao
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Inter-disciplinary Research Center, Nanjing University, Nanjing, 210023, China
| | - Yajun Wang
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Inter-disciplinary Research Center, Nanjing University, Nanjing, 210023, China
| | - Hongyun Jiao
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Inter-disciplinary Research Center, Nanjing University, Nanjing, 210023, China
| | - Han Ang
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Inter-disciplinary Research Center, Nanjing University, Nanjing, 210023, China
| | - Yinghan Chen
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Inter-disciplinary Research Center, Nanjing University, Nanjing, 210023, China.
| | - Xin Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, 475004, China.
| | - Yong Liang
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Inter-disciplinary Research Center, Nanjing University, Nanjing, 210023, China.
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
4
|
Sun M, Zhang X, Tan B, Zhang Q, Zhao X, Dong D. Potential role of endoplasmic reticulum stress in doxorubicin-induced cardiotoxicity-an update. Front Pharmacol 2024; 15:1415108. [PMID: 39188945 PMCID: PMC11345228 DOI: 10.3389/fphar.2024.1415108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/01/2024] [Indexed: 08/28/2024] Open
Abstract
As a chemotherapy agent, doxorubicin is used to combat cancer. However, cardiotoxicity has limited its use. The existing strategies fail to eliminate doxorubicin-induced cardiotoxicity, and an in-depth exploration of its pathogenesis is in urgent need to address the issue. Endoplasmic reticulum stress (ERS) occurs when Endoplasmic Reticulum (ER) dysfunction results in the accumulation of unfolded or misfolded proteins. Adaptive ERS helps regulate protein synthesis to maintain cellular homeostasis, while prolonged ERS stimulation may induce cell apoptosis, leading to dysfunction and damage to tissue and organs. Numerous studies on doxorubicin-induced cardiotoxicity strongly link excessive activation of the ERS to mechanisms including oxidative stress, calcium imbalance, autophagy, ubiquitination, and apoptosis. The researchers also found several clinical drugs, chemical compounds, phytochemicals, and miRNAs inhibited doxorubicin-induced cardiotoxicity by targeting ERS. The present review aims to outline the interactions between ERS and other mechanisms in doxorubicin-induced cardiotoxicity and summarize ERS's role in this type of cardiotoxicity. Additionally, the review enumerates several clinical drugs, phytochemicals, chemical compounds, and miRNAs targeting ERS for considering therapeutic regimens that address doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Xin Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Boxuan Tan
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| | - Qingya Zhang
- Innovation Institute, China Medical University, Shenyang, Liaoning, China
| | - Xiaopeng Zhao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
5
|
Reis-Mendes A, Ferreira M, Padrão AI, Duarte JA, Duarte-Araújo M, Remião F, Carvalho F, Sousa E, Bastos ML, Costa VM. The Role of Nrf2 and Inflammation on the Dissimilar Cardiotoxicity of Doxorubicin in Two-Time Points: a Cardio-Oncology In Vivo Study Through Time. Inflammation 2024; 47:264-284. [PMID: 37833616 PMCID: PMC10799157 DOI: 10.1007/s10753-023-01908-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023]
Abstract
Doxorubicin (DOX) is a topoisomerase II inhibitor used in cancer therapy. Despite its efficacy, DOX causes serious adverse effects, such as short- and long-term cardiotoxicity. This work aimed to assess the short- and long-term cardiotoxicity of DOX and the role of inflammation and antioxidant defenses on that cardiotoxicity in a mice model. Adult CD-1 male mice received a cumulative dose of 9.0 mg/kg of DOX (2 biweekly intraperitoneal injections (ip), for 3 weeks). One week (1W) or 5 months (5M) after the last DOX administration, the heart was collected. One week after DOX, a significant increase in p62, tumor necrosis factor receptor (TNFR) 2, glutathione peroxidase 1, catalase, inducible nitric oxide synthase (iNOS) cardiac expression, and a trend towards an increase in interleukin (IL)-6, TNFR1, and B-cell lymphoma 2 associated X (Bax) expression was observed. Moreover, DOX induced a decrease on nuclear factor erythroid-2 related factor 2 (Nrf2) cardiac expression. In both 1W and 5M, DOX led to a high density of infiltrating M1 macrophages, but only the 1W-DOX group had a significantly higher number of nuclear factor κB (NF-κB) p65 immunopositive cells. As late effects (5M), an increase in Nrf2, myeloperoxidase, IL-33, tumor necrosis factor-α (TNF-α), superoxide dismutase 2 (SOD2) expression, and a trend towards increased catalase expression were observed. Moreover, B-cell lymphoma 2 (Bcl-2), cyclooxygenase-2 (COX-2), and carbonylated proteins expression decreased, and a trend towards decreased p38 mitogen-activated protein kinase (MAPK) expression were seen. Our study demonstrated that DOX induces adverse outcome pathways related to inflammation and oxidative stress, although activating different time-dependent response mechanisms.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Mariana Ferreira
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Ana Isabel Padrão
- Research Center in Physical Activity, Faculty of Sport, University of Porto, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, Porto, Portugal
| | - José Alberto Duarte
- Research Center in Physical Activity, Faculty of Sport, University of Porto, Porto, Portugal
- 1H-TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, University of Porto, Porto, Portugal
- Department of Immuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemistry Department, Faculty of Pharmacy, University of Porto, Porto, Portugal
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, Porto, Portugal
| | - Maria Lourdes Bastos
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
- UCIBIO - Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
6
|
Łoboda A, Dulak J. Cardioprotective Effects of Hydrogen Sulfide and Its Potential Therapeutic Implications in the Amelioration of Duchenne Muscular Dystrophy Cardiomyopathy. Cells 2024; 13:158. [PMID: 38247849 PMCID: PMC10814317 DOI: 10.3390/cells13020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Hydrogen sulfide (H2S) belongs to the family of gasotransmitters and can modulate a myriad of biological signaling pathways. Among others, its cardioprotective effects, through antioxidant, anti-inflammatory, anti-fibrotic, and proangiogenic activities, are well-documented in experimental studies. Cardiorespiratory failure, predominantly cardiomyopathy, is a life-threatening complication that is the number one cause of death in patients with Duchenne muscular dystrophy (DMD). Although recent data suggest the role of H2S in ameliorating muscle wasting in murine and Caenorhabditis elegans models of DMD, possible cardioprotective effects have not yet been addressed. In this review, we summarize the current understanding of the role of H2S in animal models of cardiac dysfunctions and cardiac cells. We highlight that DMD may be amenable to H2S supplementation, and we suggest H2S as a possible factor regulating DMD-associated cardiomyopathy.
Collapse
Affiliation(s)
- Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7 Street, 30-387 Kraków, Poland;
| | | |
Collapse
|
7
|
Chang WT, Wu CC, Liao IC, Lin YW, Chen YC, Ho CH, Lee WC, Lin YC, Chen ZC, Shih JY, Wu NC, Kan WC. Dapagliflozin protects against doxorubicin-induced nephrotoxicity associated with nitric oxide pathway-A translational study. Free Radic Biol Med 2023; 208:103-111. [PMID: 37549754 DOI: 10.1016/j.freeradbiomed.2023.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Doxorubicin (Dox) is a potent anticancer agent, but its associated organ toxicity, including nephrotoxicity, restricts clinical applications. Dapagliflozin (DAPA), a sodium-glucose cotransporter-2 inhibitor, has been shown to slow the progression of kidney disease in patients with and without diabetes. However, the effect of DAPA to counteract Dox-induced nephrotoxicity remains uncertain. Therefore, in this study, we aimed to elucidate the effects of DAPA in mitigating Dox-induced nephrotoxicity. We analyzed the Taiwan National Health Insurance Database to evaluate the incidence of renal failure among breast cancer patients receiving Dox treatment compared to those without. After adjusting for age and comorbidities, we found that the risk of renal failure was significantly higher in Dox-treated patients (incidence rate ratio, 2.45; confidence interval, 1.41-4.26; p = 0.0014). In a parallel study, we orally administered DAPA to Sprague-Dawley rats for 6 weeks, followed by Dox for 4 weeks. DAPA ameliorated Dox-induced glomerular atrophy, renal fibrosis, and dysfunction. Furthermore, DAPA effectively suppressed Dox-induced apoptosis and reactive oxygen species production. On a cellular level, DAPA in HK-2 cells mitigated Dox-mediated suppression of the endothelial NOS pathway and reduced Dox-induced activities of reactive oxygen species and apoptosis-associated proteins. DAPA improved Dox-induced apoptosis and renal dysfunction, suggesting its potential utility in preventing nephrotoxicity in patients with cancer undergoing Dox treatment.
Collapse
Affiliation(s)
- Wei-Ting Chang
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan; Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan; Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chia-Chun Wu
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - I-Chuang Liao
- Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yu-Wen Lin
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Yi-Chen Chen
- Department of Hospital and Health Care Administration, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chung-Han Ho
- Department of Hospital and Health Care Administration, Chi-Mei Medical Center, Tainan, Taiwan; Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Wei-Chieh Lee
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan; Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - You-Cheng Lin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan
| | - Zhih-Cherng Chen
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan; Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Jhih-Yuan Shih
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan; Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Nan-Chun Wu
- Division of Cardiovascular Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan; Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Tainan, Taiwan.
| | - Wei-Chih Kan
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan.
| |
Collapse
|
8
|
Pharoah BM, Zhang C, Khodade VS, Keceli G, McGinity C, Paolocci N, Toscano JP. Hydropersulfides (RSSH) attenuate doxorubicin-induced cardiotoxicity while boosting its anticancer action. Redox Biol 2023; 60:102625. [PMID: 36773545 PMCID: PMC9929489 DOI: 10.1016/j.redox.2023.102625] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/19/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Cardiotoxicity is a frequent and often lethal complication of doxorubicin (DOX)-based chemotherapy. Here, we report that hydropersulfides (RSSH) are the most effective reactive sulfur species in conferring protection against DOX-induced toxicity in H9c2 cardiac cells. Mechanistically, RSSH supplementation alleviates the DOX-evoked surge in reactive oxygen species (ROS), activating nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent pathways, thus boosting endogenous antioxidant defenses. Simultaneously, RSSH turns on peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a master regulator of mitochondrial function, while decreasing caspase-3 activity to inhibit apoptosis. Of note, we find that RSSH potentiate anticancer DOX effects in three different cancer cell lines, with evidence that suggests this occurs via induction of reductive stress. Indeed, cancer cells already exhibit much higher basal hydrogen sulfide (H2S), sulfane sulfur, and reducing equivalents compared to cardiac cells. Thus, RSSH may represent a new promising avenue to fend off DOX-induced cardiotoxicity while boosting its anticancer effects.
Collapse
Affiliation(s)
- Blaze M Pharoah
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Chengximeng Zhang
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Vinayak S Khodade
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Gizem Keceli
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Christopher McGinity
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Department of Biomedical Sciences, University of Padova, Padova, Italy.
| | - John P Toscano
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, United States.
| |
Collapse
|
9
|
Liao D, Shangguan D, Wu Y, Chen Y, Liu N, Tang J, Yao D, Shi Y. Curcumin protects against doxorubicin induced oxidative stress by regulating the Keap1-Nrf2-ARE and autophagy signaling pathways. Psychopharmacology (Berl) 2023; 240:1179-1190. [PMID: 36949340 PMCID: PMC10102057 DOI: 10.1007/s00213-023-06357-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/13/2023] [Indexed: 03/24/2023]
Abstract
BACKGROUND Doxorubicin (DOX)-induced neurotoxicity is widely reported in previous studies. Oxidative stress has been validated as a critical event involved in DOX-induced neurotoxicity. As a selective autophagy adaptor protein, p62 is reported to regulate Keap1-Nrf2-ARE antioxidant pathway in response to oxidative stress. Curcumin (CUR) relieves depressive-like state through the mitigation of oxidative stress and the activation of Nrf2-ARE signaling pathway. However, the exact mechanism of CUR in alleviating DOX-induced neurotoxicity is still unknown. MATERIALS AND METHODS The rats were randomly divided into three groups: control group, DOX group, and DOX + CUR group. At the end of 3 weeks, the behavior tests as sucrose preference test (SPT), forced swimming test (FST), and novelty-suppressed feeding test (NSFT) were performed to assess anxiety- and depression-like behaviors. The rats were sacrificed after behavior tests, and the brain tissues were collected for biochemical analysis. RESULTS It was observed that the administration of CUR could effectively reverse DOX-induced depressive-like behaviors. The exposure of DOX activated autophagy and increased oxidative stress levels, and the administration of CUR could significantly inhibit DOX-induced autophagy and suppress oxidative stress. More importantly, we also found that Keap1-Nrf2-ARE signaling pathway was involved in DOX-induced neurotoxicity and oxidative stress regulated by autophagy. CONCLUSION Our study demonstrated that CUR could effectively reverse DOX-induced neurotoxicity through suppressing autophagy and mitigating oxidative stress and endoplasmic reticulum (ER) stress.
Collapse
Affiliation(s)
- Dehua Liao
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, China
| | - Danggang Shangguan
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, China
| | - Yi Wu
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, China
| | - Yun Chen
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, China
| | - Ni Liu
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, China
| | - Jingyi Tang
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, China
| | - Dunwu Yao
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, China.
| | - Yingrui Shi
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
10
|
H2S Donors with Cytoprotective Effects in Models of MI/R Injury and Chemotherapy-Induced Cardiotoxicity. Antioxidants (Basel) 2023; 12:antiox12030650. [PMID: 36978898 PMCID: PMC10045576 DOI: 10.3390/antiox12030650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous signaling molecule that greatly influences several important (patho)physiological processes related to cardiovascular health and disease, including vasodilation, angiogenesis, inflammation, and cellular redox homeostasis. Consequently, H2S supplementation is an emerging area of interest, especially for the treatment of cardiovascular-related diseases. To fully unlock the medicinal properties of hydrogen sulfide, however, the development and refinement of H2S releasing compounds (or donors) are required to augment its bioavailability and to better mimic its natural enzymatic production. Categorizing donors by the biological stimulus that triggers their H2S release, this review highlights the fundamental chemistry and releasing mechanisms of a range of H2S donors that have exhibited promising protective effects in models of myocardial ischemia-reperfusion (MI/R) injury and cancer chemotherapy-induced cardiotoxicity, specifically. Thus, in addition to serving as important investigative tools that further advance our knowledge and understanding of H2S chemical biology, the compounds highlighted in this review have the potential to serve as vital therapeutic agents for the treatment (or prevention) of various cardiomyopathies.
Collapse
|
11
|
Mauro AG, Mezzaroma E, Toldo S, Melendez GC, Franco RL, Lesnefsky EJ, Abbate A, Hundley WG, Salloum FN. NLRP3-mediated inflammation in cardio-oncology: sterile yet harmful. Transl Res 2023; 252:9-20. [PMID: 35948198 PMCID: PMC9839540 DOI: 10.1016/j.trsl.2022.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 01/17/2023]
Abstract
Despite significant advances and the continuous development of novel, effective therapies to treat a variety of malignancies, cancer therapy-induced cardiotoxicity has been identified as a prominent cause of morbidity and mortality, closely competing with secondary malignancies. This unfortunate limitation has prompted the inception of the field of cardio-oncology with its purpose to provide the necessary knowledge and key information on mechanisms that support the use of the most efficacious cancer therapy with minimal or no interruption while paying close attention to preventing cardiovascular related morbidity and mortality. Several mechanisms that contribute to cancer therapy-induced cardiotoxicity have been proposed and studied. These mainly involve mitochondrial dysfunction and reactive oxygen species-induced oxidative stress, lysosomal damage, impaired autophagy, cell senescence, DNA damage, and sterile inflammation with the formation and activation of the NLRP3 inflammasome. In this review, we focus on describing the principal mechanisms for different classes of cancer therapies that lead to cardiotoxicity involving the NLRP3 inflammasome. We also summarize current evidence of cardio-protection with inflammasome inhibitors in the context of heart disease in general, and further highlight the potential application of this evidence for clinical translation in at risk patients for the purpose of preventing cancer therapy associated cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Adolfo G Mauro
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Eleonora Mezzaroma
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Stefano Toldo
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Giselle C Melendez
- Department of Internal Medicine, Sections on Cardiovascular Medicine, Department of Pathology, Section on Comparative Medicine, Wake Forest, School of Medicine, Winston-Salem, NC
| | - R Lee Franco
- College of Humanities and Sciences, Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA
| | - Edward J Lesnefsky
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA; Department of the Medical Service of the McGuire Veterans Affairs Medical Center, Richmond, VA
| | - Antonio Abbate
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - W Gregory Hundley
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Fadi N Salloum
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA.
| |
Collapse
|
12
|
Jia TT, Zhang Y, Hou JT, Niu H, Wang S. H 2S-based fluorescent imaging for pathophysiological processes. Front Chem 2023; 11:1126309. [PMID: 36778034 PMCID: PMC9911449 DOI: 10.3389/fchem.2023.1126309] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Hydrogen sulfide (H2S), as an important endogenous signaling molecule, plays a vital role in many physiological processes. The abnormal behaviors of hydrogen sulfide in organisms may lead to various pathophysiological processes. Monitoring the changes in hydrogen sulfide is helpful for pre-warning and treating these pathophysiological processes. Fluorescence imaging techniques can be used to observe changes in the concentration of analytes in organisms in real-time. Therefore, employing fluorescent probes imaging to investigate the behaviors of hydrogen sulfide in pathophysiological processes is vital. This paper reviews the design strategy and sensing mechanisms of hydrogen sulfide-based fluorescent probes, focusing on imaging applications in various pathophysiological processes, including neurodegenerative diseases, inflammation, apoptosis, oxidative stress, organ injury, and diabetes. This review not only demonstrates the specific value of hydrogen sulfide fluorescent probes in preclinical studies but also illuminates the potential application in clinical diagnostics.
Collapse
Affiliation(s)
- Tong-Tong Jia
- College of Chemistry and Chemical Engineering, Luoyang Normal University, Luoyang, China
| | - Yuanyuan Zhang
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Ji-Ting Hou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Huawei Niu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Shan Wang
- Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Tailoring carrier-free nanocombo of small-molecule prodrug for combinational cancer therapy. J Control Release 2022; 352:256-275. [PMID: 36272660 DOI: 10.1016/j.jconrel.2022.10.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
The outcomes of monotherapy could not satisfy clinical cancer treatment owing to the challenges of tumor heterogeneity, multi-drug resistance, tumor metastasis and relapse. In response, the significance of combinational cancer therapy has been highlighted. Traditional combinational schemes usually utilize "free" drug for multi drug administration, independently. The diverse pharmacokinetics and biodistribution greatly hinder the antitumor effects and cause systematic toxicity. To tackle the hinderance, various nanoparticulate drug delivery systems (Nano-DDSs) have been developed. However, conventional Nano-DDSs encapsulate drugs into carrier materials through noncovalent interactions, resulting in low drug loading, fixed multi drug encapsulation ratio, chemical instability and carrier-associated toxicity. Recently, carrier-free nanocombos based on self-assembling small-molecule prodrugs (SPNCs) have emerged as a versatile Nano-DDSs for multiple drug delivery. Benefited by the self-assembly capability, SPNCs could be facilely fabricated with distinct merits of ultra-high drug loading, adjustable drug ratio and negligible carrier-associated toxicity. Herein, we summarize the latest trends of SPNCs. First, a basic review on self-assembling small-molecule prodrugs is presented. Additionally, facile techniques to prepare SPNCs are introduced. Furthermore, advanced combinational therapies based on SPNCs are spotlighted with special emphasis on synergistic mechanisms. Finally, future prospects and challenges are discussed.
Collapse
|
14
|
Kim SW, Ahn BY, Tran TTV, Pyun JH, Kang JS, Leem YE. PRMT1 suppresses doxorubicin-induced cardiotoxicity by inhibiting endoplasmic reticulum stress. Cell Signal 2022; 98:110412. [PMID: 35863589 DOI: 10.1016/j.cellsig.2022.110412] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/24/2022] [Accepted: 07/15/2022] [Indexed: 11/03/2022]
Abstract
Doxorubicin (Dox) is a widely used anti-cancer drug that has a significant limitation, which is cardiotoxicity. Its cardiotoxic side effect is dose dependent and occurs through any age. Dox has been known to exert its toxic effect through oxidative stress, but an emerging mechanism is endoplasmic reticulum (ER) stress that activates proapoptotic pathway involving PERK/ATF4/CHOP axis. These stresses lead to dysfunction of myocardium associated with cell death. Although accumulating evidence support their involvement to Dox-induced cardiotoxicity, the mechanism is not well elucidated. Protein arginine methyltransferases 1 (PRMT1) has been known to play a role in cardiomyocyte cell survival through modulation of ER response. In this study, we demonstrate an important role of PRMT1 in Dox-induced cardiotoxicity via ER stress. Depletion of PRMT1 in H9c2 cardiomyocytes enhanced Dox-stimulated cell death, and increased reactive oxygen species (ROS) production and DNA damage by enhancing the levels of proapoptotic cleaved Caspase-3 and γH2AX in response to Dox. Consistently, overexpression of PRMT1 attenuated the apoptotic effect of Dox. In addition, the acute treatment of Dox induced a substantial increase in PRMT1 activity and the translocation of PRMT1 to ER. Overexpression of PRMT1 in cardiomyocyte diminished Dox-induced ER stress, and ATF4 methylation by PRMT1 was involved in the suppression of ER stress. Taken together, our data suggest that PRMT1 is a novel target molecule for protection from Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Su Woo Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Byeong-Yun Ahn
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Thi Thuy Vy Tran
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Jung-Hoon Pyun
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Young-Eun Leem
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Republic of Korea.
| |
Collapse
|
15
|
Chang WT, Shih JY, Lin YW, Chen ZC, Kan WC, Lin TH, Hong CS. Dapagliflozin protects against doxorubicin-induced cardiotoxicity by restoring STAT3. Arch Toxicol 2022; 96:2021-2032. [PMID: 35438302 DOI: 10.1007/s00204-022-03298-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/31/2022] [Indexed: 01/02/2023]
Abstract
Doxorubicin (Dox), an effective therapy in different types of cancer, is known to exhibit cardiotoxic effects. Despite previous studies indicating the benefits of dapagliflozin (DAPA) in patients experiencing heart failure, it remains uncertain whether DAPA exerts a protective effect on Dox-induced cardiac dysfunction. Signal transducer and activator of transcription 3 (STAT3) participates in various mechanisms of cardioprotection. Herein, we aimed to investigate the effects of DAPA on Dox-induced cardiotoxicity and the role of STAT3. Sprague-Dawley rats were pretreated with oral DAPA for 6 weeks followed by Dox for 4 weeks. Sequential echocardiography was applied to assess cardiac function. For in vitro analysis, cardiomyocytes were treated with 10 μM DAPA and subsequently exposed to 1 μM Dox. The expression of reactive oxygen species- and apoptosis-related proteins was measured. Using STAT3 siRNA, we further examined the effects of STAT3 effect on DAPA-associated protection against Dox-induced apoptosis. In rats treated with Dox, DAPA significantly reduced cardiac fibrosis and improved cardiac function and hemodynamics. Additionally, DAPA effectively inhibited Dox-induced apoptosis and reactive oxygen species (ROS) in cardiomyocytes. Mechanistically, we showed that DAPA decreased cardiac expression of Bax and cleaved caspase 3 but increased Bcl-2 expression. DAPA also significantly rescued Dox-suppressed STAT3 expression. Conversely, knocking down STAT3 in cardiomyocytes reversed the DAPA-related protective effects on Dox-induced cell apoptosis and ROS. Collectively, our findings indicate that DAPA could be useful for preventing Dox-induced cardiotoxicity by restoring STAT3.
Collapse
Affiliation(s)
- Wei-Ting Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Jhih-Yuan Shih
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Yu-Wen Lin
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Zhih-Cherng Chen
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Wei-Chih Kan
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, 901, Zhonghua Road, Yongkang District, Tainan, Taiwan ROC.
- Department of Biological Science and Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan.
| | - Tsung-Hsien Lin
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chon-Seng Hong
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
16
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
17
|
Xiang Y, Chen L, Liu C, Yi X, Li L, Huang Y. Redirecting Chemotherapeutics to the Endoplasmic Reticulum Increases Tumor Immunogenicity and Potentiates Anti-PD-L1 Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104591. [PMID: 34859582 DOI: 10.1002/smll.202104591] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/28/2021] [Indexed: 05/21/2023]
Abstract
The endoplasmic reticulum (ER) in cancer cells has been considered as a pharmacological target. Still, the effects of a ER-targeted system remain less investigated, due to the fact that most chemo-drugs take actions in the nucleus. Here, it is demonstrated that ER-targeted delivery of doxorubicin (DOX), a typically nucleus-tropic-and-acting agent, attenuates its original effect on cytotoxicity while generating new functions favorable for immune activation. First, a library of DOX derivatives with variable ER-targeting abilities is synthesized. The results reveal that higher ER-targeting efficiency correlates with greater ER stress. As compared with naïve drug, ER-targeted DOX considerably alters the mode of action from nuclear DNA damage-associated cytotoxicity to ER stress-mediated calreticulin exposure. Consequently, ER-targeted DOX decreases cytotoxicity but increases the capability to induce immunogenic cell death (ICD). Therefore, a platform combining naïve and ER-targeted DOX is constructed for in vivo application. Conventional polymer-DOX conjugate inhibits tumor growth by exerting a direct killing effect, and ER-targeted polymer-DOX conjugate suppresses residual tumors by eliciting ICD-associated immunity, together resulting in considerable tumor regression. In addition, simultaneous inhibition of adaptive PD-L1 enrichment (due to negative-feedback to ICD induction) further leads to greater therapeutic outcome. Collectively, ER-targeted therapy can enhance anticancer efficacy by promoting ICD-associated immunotherapy, and potentiating chemotherapy and checkpoint blockade therapy.
Collapse
Affiliation(s)
- Yucheng Xiang
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Liqiang Chen
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Chendong Liu
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Xiaoli Yi
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Lian Li
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| | - Yuan Huang
- Key laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu, 610041, P. R. China
| |
Collapse
|
18
|
Mu S, Zhang J, Gao H, Wang Y, Rizvi SFA, Ding N, Liu X, Wu L, Zhang H. Sequential detection of H 2S and HOBr with a novel lysosome-targetable fluorescent probe and its application in biological imaging. JOURNAL OF HAZARDOUS MATERIALS 2022; 422:126898. [PMID: 34416694 DOI: 10.1016/j.jhazmat.2021.126898] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 06/13/2023]
Abstract
Understanding the complex relationship between active small molecules is of great significance in various physiological processes. Herein, we present the design and synthesis of a sequential responsive Lysosome-Naphthalene imide-Azido (lyso-NP-N3) reporter for probing the H2S and HOBr within organelle (lysosome) in living cells. Probe lyso-NP-N3 exhibited high selectivity and sensitivity towards H2S (LOD = 23.5 nM) and HOBr (LOD = 254 nM). Additionally, lyso-NP-N3 possessed an excellent lysosome targeting ability and was utilized to visualize the exogenous/endogenous H2S and HOBr in RAW 264.7, Hela and HepG2 cells. Facilitated by this sequentially activated mechanism, the probe was successfully applied to confirm that the reported scavenger of HOBr, N-acetyl-L-cysteine (NAC) mainly relied on its metabolite H2S to eliminate excess HOBr, thereby playing the role of cell regulation and protection. These results establish the crosstalk between H2S and HOBr in lysosome and provide a promising tool to study metabolite interactions.
Collapse
Affiliation(s)
- Shuai Mu
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jinlong Zhang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Hong Gao
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Yaya Wang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Nana Ding
- College of Chemical Engineering, Northwest Minzu University, Lanzhou, Gansu 730030, China
| | - Xiaoyan Liu
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Lan Wu
- College of Chemical Engineering, Northwest Minzu University, Lanzhou, Gansu 730030, China.
| | - Haixia Zhang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metals Chemistry and Resources Utilization of Gansu Province and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
19
|
Yarmohammadi F, Hayes AW, Karimi G. The cardioprotective effects of hydrogen sulfide by targeting endoplasmic reticulum stress and the Nrf2 signaling pathway: A review. Biofactors 2021; 47:701-712. [PMID: 34161646 DOI: 10.1002/biof.1763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
Cardiac diseases are emerging due to lifestyle, urbanization, and the accelerated aging process. Oxidative stress has been associated with cardiac injury progression through interference with antioxidant strategies and endoplasmic reticulum (ER) function. Hydrogen sulfide (H2 S) is generated endogenously from l-cysteine in various tissues including heart tissue. Pharmacological evaluation of H2 S has suggested a potential role for H2 S against diabetic cardiomyopathy, ischemia/reperfusion injury, myocardial infarction, and cardiotoxicity. Nuclear factor E2-related factor 2 (Nrf2) activity is crucial for cell survival in response to oxidative stress. H2 S up-regulates Nrf2 expression and its related signaling pathway in myocytes. H2 S also suppresses the expression and activity of ER stress-related proteins. H2 S has been reported to improve various cardiac conditions through antioxidant and anti-ER stress-related activities.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, Florida, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Huang J, Lei Y, Lei S, Gong X. Cardioprotective effects of corilagin on doxorubicin induced cardiotoxicity via P13K/Akt and NF-κB signaling pathways in rats model. Toxicol Mech Methods 2021; 32:79-86. [PMID: 34369273 DOI: 10.1080/15376516.2021.1965274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Even though doxorubicin (DOX) is a potential chemotherapeutic drug, its usage is restricted due to its ability to induce cardiac damage. In order to prevent this damage, a potent cardioprotective agent should be associated with DOX treatment. Corilagin is a natural polyphenol tannic acid which unveils enormous pharmacological activities predominantly as an antitumor agent. Hence, the current work is designed to study the precise mechanisms of corilagin upon administration in doxorubicin induced cardiotoxicity in experimental rats. DOX treated rats showed diminished level of blood pressures and heart rate, whereas corilagin along with DOX treatment improved the status. Cardiotoxicity enzymes and biomarkers were found to be increased in the serum of DOX induced rats. Upon treatment, corilagin could reduce the cardiotoxicity enzymes and biomarkers in serum. Histopathological examination of cardiac tissue also revealed the anti-toxic effects of corilagin in contrast to DOX. Injection of DOX in rats showed inflammatory cells infiltration, necrosis and fragmented myofibrils. Corilagin treatment reverted the cardiac histology to near normal. Inflammatory mediators and P13K, Akt, and NF-κB were upregulated in DOX administered rats. Corilagin repressed the levels of P13K, Akt, and NF-κB in DOX induced rats. In the present investigations, corilagin improved cardiac function via reducing injury, inflammation and promoting apoptosis thereby suggesting that corilagin would be recommended for DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Jing Huang
- Department of Pharmacy, Wuxi No.2 People's Hospital, Wuxi, 214000, China
| | - Ying Lei
- Department of Cardiology, Ankang Hospital of Traditional Chinese Medicine, Ankang, 725000, China
| | - Shengping Lei
- Clinical Experimental Center, Xi'an International Medical Center Hospital, Xi'an, 710100, China
| | - Xinwen Gong
- Department of Cardiology, Ankang Hospital of Traditional Chinese Medicine, Ankang, 725000, China
| |
Collapse
|
21
|
Hu HJ, Qiu J, Zhang C, Tang ZH, Qu SL, Jiang ZS. Hydrogen sulfide improves ox‑LDL‑induced expression levels of Lp‑PLA 2 in THP‑1 monocytes via the p38MAPK pathway. Mol Med Rep 2021; 23:358. [PMID: 33760156 PMCID: PMC7974332 DOI: 10.3892/mmr.2021.11997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 01/22/2021] [Indexed: 12/22/2022] Open
Abstract
Hydrogen sulfide (H2S) exerts an anti‑atherosclerotic effect and decreases foam cell formation. Lipoprotein‑associated phospholipase A2 (Lp‑PLA2) is a key factor involved in foam cell formation. However, the association between H2S and Lp‑PLA2 expression levels with respect to foam cell formation has not yet been elucidated. The present study investigated whether H2S can affect foam cell formation and potential signalling pathways via regulation of the expression and activity of Lp‑PLA2. Using human monocytic THP‑1 cells as a model system, it was observed that oxidized low‑density lipoprotein (ox‑LDL) not only upregulates the expression level and activity of Lp‑PLA2, it also downregulates the expression level and activity of Cystathionine γ lyase. Exogenous supplementation of H2S decreased the expression and activity of Lp‑PLA2 induced by ox‑LDL. Moreover, ox‑LDL induced the expression level and activity of Lp‑PLA2 via activation of the p38MAPK signalling pathway. H2S blocked the expression levels and activity of Lp‑PLA2 induced by ox‑LDL via inhibition of the p38MAPK signalling pathway. Furthermore, H2S inhibited Lp‑PLA2 activity by blocking the p38MAPK signaling pathway and significantly decreased lipid accumulation in ox‑LDL‑induced macrophages, as detected by Oil Red O staining. The results of the present study indicated that H2S inhibited ox‑LDL‑induced Lp‑PLA2 expression levels and activity by blocking the p38MAPK signalling pathway, thereby improving foam cell formation. These findings may provide novel insights into the role of H2S intervention in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Heng-Jing Hu
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jie Qiu
- Department of Cardiology Laboratory, Huazhong University of Science and Technology Tongji Medical College First Clinical College, Wuhan, Hubei 430000, P.R. China
| | - Chi Zhang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Han Tang
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Sheng Jiang
- Department of Cardiology Laboratory, First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
22
|
Chang WT, Lin YW, Ho CH, Chen ZC, Liu PY, Shih JY. Dapagliflozin suppresses ER stress and protects doxorubicin-induced cardiotoxicity in breast cancer patients. Arch Toxicol 2021; 95:659-671. [PMID: 33211168 DOI: 10.1007/s00204-020-02951-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/05/2020] [Indexed: 01/12/2023]
Abstract
Cancer patients with diabetes have an increasing risk of Dox-induced cardiotoxicity. Despite previous studies reporting benefits of dapagliflozin on the cardiovascular system, it remains unknown whether dapagliflozin has a cardioprotective effect in cancer patients with diabetes. We aimed to investigate the potential of dapagliflozin for preventing doxorubicin (Dox)-induced cardiotoxicity. Using Taiwan National Health Insurance Database, the incidence of heart failure of cancer patients with or without diabetes was investigated. Streptozotocin (STZ)-induced diabetic rats were pretreated with oral dapagliflozin for 6 weeks followed by Dox for 4 weeks via intraperitoneal injection. Sequential echocardiography was applied to assess cardiac function. For in vitro analysis, cardiomyocytes cultured in high glucose were treated with dapagliflozin at 10 μM and subsequently exposed to Dox at 1 μM. Apoptosis and endoplasmic reticulum (ER) stress-related protein expression were measured. Among the studied patients, those with diabetes had a higher risk of major adverse cardiovascular events including the development of heart failure. In diabetic rats, dapagliflozin reduced cardiac fibrosis and significantly improved cardiac function. Dapagliflozin effectively inhibited Dox-induced apoptosis and reactive oxygen species in cardiomyocytes under high glucose. Mechanistically, we showed that dapagliflozin decreased the cardiac expression of Bax and cleaved caspase 3 but increased Bcl-2. Dapagliflozin also significantly reduced ER stress-associated proteins including GRP78, PERK, eIF-2α, ATF-4, and CHOP. Our study revealed for the first time that dapagliflozin mitigated Dox-induced cardiomyocyte apoptosis in diabetes. These results indicate that dapagliflozin could be useful for preventing cardiotoxicity in diabetic cancer patients receiving Dox treatment.
Collapse
Affiliation(s)
- Wei-Ting Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 901, Zhonghua Road, Yongkang District, Tainan, Taiwan R.O.C
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Yu-Wen Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 901, Zhonghua Road, Yongkang District, Tainan, Taiwan R.O.C
| | - Chung-Han Ho
- Department of Hospital and Health Care Administration, Chi-Mei Medical Center, Tainan, Taiwan
| | - Zhih-Cherng Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 901, Zhonghua Road, Yongkang District, Tainan, Taiwan R.O.C
| | - Ping-Yen Liu
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- Division of Cardiology, Internal Medicine, College of Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Jhih-Yuan Shih
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, 901, Zhonghua Road, Yongkang District, Tainan, Taiwan R.O.C..
| |
Collapse
|
23
|
Li Y, Liu M, Yi J, Song X, Zheng X, Liu D, Wang S, Chu C, Yang J. Exogenous hydrogen sulfide inhibits apoptosis by regulating endoplasmic reticulum stress-autophagy axis and improves myocardial reconstruction after acute myocardial infarction. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1325-1336. [PMID: 33210714 DOI: 10.1093/abbs/gmaa133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
During acute myocardial infarction, endoplasmic reticulum (ER) stress-induced autophagy and apoptosis have been shown as important pathogeneses of myocardial reconstruction. Importantly, hydrogen sulfide (H2S), as a third endogenous gas signaling molecule, exerts strong cytoprotective effect on anti-ER stress, autophagy regulation and antiapoptosis. Here, we showed that H2S treatment inhibits apoptosis by regulating ER stress-autophagy axis and improves myocardial reconstruction after acute myocardial infarction. We found that H2S intervention improved left ventricle function, reduced glycogen deposition in myocardial tissue mesenchyme, and inhibited apoptosis. Moreover, the expressions of fibrosis indicators (Col3a1 and Col1a2), ER stress-related proteins (CHOP and BIP/ERP78), autophagy-related proteins (Beclin and ATG5), apoptosis protein (Bax), as well as fibrosis protein Col4a3bp were all decreased after treatment with H2S. H2S administration also maintained MMP/TIMP balance. Mechanistically, H2S activated the PI3K/AKT signaling pathway. In addition, H2S treatment also reduced the expressions of ER stress-related proteins, autophagy-related proteins, and apoptins in in vitro experiments. Interestingly, activation of ER stress-autophagy axis could reverse the inhibitory effect of H2S on myocardial apoptosis. Altogether, these results suggested that exogenous H2S suppresses myocardial apoptosis by blocking ER stress-autophagy axis, which in turn reverses cardiac remodeling after myocardial infarction.
Collapse
Affiliation(s)
- Yaling Li
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Maojun Liu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Jiali Yi
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Xiong Song
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Xia Zheng
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Da Liu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Sen Wang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| |
Collapse
|
24
|
Yarmohammadi F, Rezaee R, Haye AW, Karimi G. Endoplasmic reticulum stress in doxorubicin-induced cardiotoxicity may be therapeutically targeted by natural and chemical compounds: A review. Pharmacol Res 2020; 164:105383. [PMID: 33348022 DOI: 10.1016/j.phrs.2020.105383] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022]
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent with marked, dose-dependent cardiotoxicity that leads to tachycardia, atrial and ventricular arrhythmia, and irreversible heart failure. Induction of the endoplasmic reticulum (ER) which plays a major role in protein folding and calcium homeostasis was reported as a key contributor to cardiac complications of DOX. This article reviews several chemical compounds that have been shown to regulate DOX-induced inflammation, apoptosis, and autophagy via inhibition of ER stress signaling pathways, such as the IRE1α/ASK1/JNK, IRE1α/JNK/Beclin-1, and CHOP pathways.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Rezaee
- Clinical Research Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Haye
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, United States
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
25
|
Mamipour M, Yousefi M, Dehnad A, Faridvand Y, Zarezadeh R, Khaksar M, Pouyafar A, Rahbarghazi R. Protective effect of bacterial lipase on lipopolysaccharide-induced toxicity in rat cardiomyocytes; H9C2 cell line. J Cardiovasc Thorac Res 2019; 12:35-42. [PMID: 32211136 PMCID: PMC7080329 DOI: 10.34172/jcvtr.2020.06] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022] Open
Abstract
Introduction: Cardiovascular system is highly sensitive to LPS-induced oxidative damage. This study aimed to show the inhibitory effect of bacterial Lipase on LPS-induced cardiomyoblasts toxicity. Methods: Rat cardiomyoblasts H9C2 were classified into Control, LPS (cells received 0.1, 1 and 10 μg/mL LPS) and LPS+ Lipase groups. In LPS+Lipase group, different concentrations of lipopolysaccharide were pre-incubated with 5 mg/mL bacterial lipase at 37˚C overnight prior to cell treatment. After 72 hours, cell viability was assessed by MTT assay. The expression of key genes related to toll-like receptor signaling pathways was assessed by real-time PCR assay. Percentage of fatty acids was evaluated in each group using gas chromatography assay. The levels of NO was also measured using the Griess reaction. Results: Data showed H9C2 cells viability was decreased after exposure to LPS in a dose-dependent manner (P < 0.05). Incubation of LPS with lipase increased cell survival rate and closed to near-to-control levels (P < 0.05). Lipase had the potential to blunt the increased expression of IRAK and NF-κB in cells after exposure to the LPS. Compared to the LPS group, lipase attenuated the increased level of NO-induced by LPS (P < 0.05). Gas chromatography analysis showed the reduction of saturated fatty acids in cells from LPS group while the activity of lipase prohibited impact of LPS on cell fatty acid composition. LPS decreased the ability of cardiomyoblasts to form colonies. Incubation of LPS with lipase enhanced clonogenic capacity. Conclusion: Reduction in lipopolysaccharide-induced cytotoxicity is possibly related to lipase activity and reduction of modified lipopolysaccharide with toll-like receptor.
Collapse
Affiliation(s)
- Mina Mamipour
- Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Yousefi
- Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Dehnad
- Biotechnology Department, East Azerbaijan Research and Education Center Agricultural and Natural Resources, AREEO, Tabriz, Iran.,Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran.,Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Faridvand
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Khaksar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayda Pouyafar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
26
|
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
|
27
|
Zhang S, You ZQ, Yang L, Li LL, Wu YP, Gu LQ, Xin YF. Protective effect of Shenmai injection on doxorubicin-induced cardiotoxicity via regulation of inflammatory mediators. Altern Ther Health Med 2019; 19:317. [PMID: 31744501 PMCID: PMC6862794 DOI: 10.1186/s12906-019-2686-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/16/2019] [Indexed: 12/05/2022]
Abstract
Background Doxorubicin (DOX) is a chemotherapy drug for malignant tumors. The clinical application of DOX is limited due to its dosage relative cardiotoxicity. Oxidative damage and cardiac inflammation appear to be involved in DOX-related cardiotoxicity. Shenmai injection (SMI), which mainly consists of Panax ginsengC.A.Mey.and Ophiopogon japonicus (Thunb.) Ker Gawl, is widely used for the treatment of atherosclerotic coronary heart disease and viral myocarditis in China. In this study, we investigated the protective effect of Shenmai injection on doxorubicin-induced acute cardiac injury via the regulation of inflammatory mediators. Methods Male ICR mice were randomly divided into seven groups: control, DOX (10 mg/kg), SMI (5 g/kg), DOX with pretreatment with SMI (0.5 g/kg, 1.5 g/kg or 5 g/kg) and DOX with post-treatment with SMI (5 g/kg). Forty-eight hours after the last DOX administration, all mice were anesthetized for ultrasound echocardiography. Then, serum was collected for biochemical and inflammatory cytokine detection, and heart tissue was collected for histological and Western blot detection. Results A cumulative dose of DOX (10 mg/kg) induced acute cardiotoxicity in mice manifested by altered echocardiographic outcome, and increased tumor necrosis factor, interleukin 6 (IL-6), monocyte chemotactic protein 1, interferon-γ, and serum AST and LDH levels, as well as cardiac cytoplasmic vacuolation and myofibrillar disarrangement. DOX also caused the increase in the expression of IKK-α and iNOS and produced a large amount of NO, resulting in the accumulation of nitrotyrosine in the heart tissue. Pretreatment with SMI elicited a dose-dependent cardioprotective effect in DOX-dosed mice as evidenced by the normalization of serum inflammatory mediators, as well as improve dcardiac function and myofibril disarrangement. Conclusions SMI could recover inflammatory cytokine levels and suppress the expression of IKK-α and iNOS in vivo, which was increased by DOX. Overall, there was evidence that SMI could ameliorate DOX-induced cardiotoxicity by inhibiting inflammation and recovering heart dysfunction.
Collapse
|
28
|
Maejima Y. The critical roles of protein quality control systems in the pathogenesis of heart failure. J Cardiol 2019; 75:219-227. [PMID: 31699567 DOI: 10.1016/j.jjcc.2019.09.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 01/30/2023]
Abstract
Heart failure is a refractory disease with a prevalence that has continuously increased around the world. Over the past decade, we have made remarkable progress in the treatment of heart failure, including drug therapies, device therapies, and regeneration therapies. However, as each of these heart failure therapies does not go much beyond symptomatic therapy, there is a compelling need to establish novel therapeutic strategies for heart failure in a fundamental way. As cardiomyocytes are terminally differentiated cells, protein quality control is critical for maintaining cellular homeostasis, optimal performance, and longevity. There are five evolutionarily conserved mechanisms for ensuring protein quality control in cells: the ubiquitin-proteasome system, autophagy, the unfolded protein response, SUMOylation, and NEDDylation. Recent research has clarified the molecular mechanism underlying how these processes degrade misfolded proteins and damaged organelles in cardiomyocytes. In addition, a growing body of evidence suggests that deviation from appropriate levels of protein quality control causes cellular dysfunction and death, which in turn leads to heart failure. We herein review recent advances in understanding the role of protein quality control systems in heart disease and discuss the therapeutic potential of modulating protein quality control systems in the human heart.
Collapse
Affiliation(s)
- Yasuhiro Maejima
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
29
|
Wang Y, Matson JB. Supramolecular nanostructures with tunable donor loading for controlled H 2S release. ACS APPLIED BIO MATERIALS 2019; 2:5093-5098. [PMID: 33283175 DOI: 10.1021/acsabm.9b00768] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Hydrogen sulfide (H2S), an endogenously generated and regulated signaling gas, plays a vital role in a variety of (patho)physiological processes. In the past few years, different kinds of H2S-releasing compounds (often referred to as H2S donors) have been developed for H2S delivery, but it is still challenging to make H2S donors with tunable payloads in a simple and efficient manner. Herein, a series of peptide-H2S donor conjugates (PHDCs) with tunable donor loadings are designed for controlled H2S release. The PHDCs self-assemble into nanoribbons with different geometries in aqueous solution. Upon addition of cysteine, these nanostructures release H2S, delivering their payload into H9C2 cells, as visualized using an H2S-selective fluorescent probe. Beyond imaging, in vitro studies show that the ability of PHDCs to mitigate doxorubicin-induced cardiotoxicity in H9C2 cardiomyocytes depends on their nanostructures and H2S release profiles. This strategy may enable the development of sophisticated H2S-releasing biomaterials for drug delivery and regenerative medicine.
Collapse
Affiliation(s)
- Yin Wang
- Department of Chemistry, Virginia Tech Center for Drug Discovery, and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| | - John B Matson
- Department of Chemistry, Virginia Tech Center for Drug Discovery, and Macromolecules Innovation Institute, Virginia Tech, Blacksburg, VA 24061, United States
| |
Collapse
|
30
|
Wang Y, Kaur K, Scannelli SJ, Bitton R, Matson JB. Self-Assembled Nanostructures Regulate H 2S Release from Constitutionally Isomeric Peptides. J Am Chem Soc 2018; 140:14945-14951. [PMID: 30369241 PMCID: PMC6225339 DOI: 10.1021/jacs.8b09320] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Indexed: 12/21/2022]
Abstract
We report here on three constitutionally isomeric peptides, each of which contains two glutamic acid residues and two lysine residues functionalized with S-aroylthiooximes (SATOs), termed peptide-H2S donor conjugates (PHDCs). SATOs decompose in the presence of cysteine to generate hydrogen sulfide (H2S), a biological signaling gas with therapeutic potential. The PHDCs self-assemble in aqueous solution into different morphologies, two into nanoribbons of different dimensions and one into a rigid nanocoil. The rate of H2S release from the PHDCs depends on the morphology, with the nanocoil-forming PHDC exhibiting a complex release profile driven by morphological changes promoted by SATO decomposition. The nanocoil-forming PHDC mitigated the cardiotoxicity of doxorubicin more effectively than its nanoribbon-forming constitutional isomers as well as common H2S donors. This strategy opens up new avenues to develop H2S-releasing biomaterials and highlights the interplay between structure and function from the molecular level to the nanoscale.
Collapse
Affiliation(s)
- Yin Wang
- Department
of Chemistry, Virginia Tech Center for Drug Discovery, and Macromolecules
Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Kuljeet Kaur
- Department
of Chemistry, Virginia Tech Center for Drug Discovery, and Macromolecules
Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Samantha J. Scannelli
- Department
of Chemistry, Virginia Tech Center for Drug Discovery, and Macromolecules
Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Ronit Bitton
- Department
of Chemical Engineering and the Ilze Kats Institute for Nanoscale
Science and Technology, Ben-Gurion University
of the Negev, Beer-Sheva 84105, Israel
| | - John B. Matson
- Department
of Chemistry, Virginia Tech Center for Drug Discovery, and Macromolecules
Innovation Institute, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
31
|
Jiao X, Xiao Y, Li Y, Liang M, Xie X, Wang X, Tang B. Evaluating Drug-Induced Liver Injury and Its Remission via Discrimination and Imaging of HClO and H 2S with a Two-Photon Fluorescent Probe. Anal Chem 2018; 90:7510-7516. [PMID: 29781282 DOI: 10.1021/acs.analchem.8b01106] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drug-induced liver injury (DILI) has aroused wide concern. Finding new markers or indicators as well as detoxification molecules for DILI is of great significance and good application prospect, which can help develop effective preclinical screening methodology and corresponding treatment protocols. Herein, in this article, DILI caused by antidepressant drugs of duloxetine and fluoxetine and its remission were evaluated by a two-photon fluorescent probe, RPC-1, through discriminating and imaging HClO and H2S simultaneously. By being applied both in vitro and in vivo, RPC-1 revealed slight up-regulation of HClO and negligible liver damage after administration of either of the two drugs. In contrast, an apparent up-regulation of HClO and obvious liver damage was observed after combined administration of the drugs. Meanwhile, the pretreatment of N-acetyl cysteine (NAC) resulted in the increasing of endogenous H2S level, which contributed to the remission of DILI. The histological analysis and serological test both gave good consistency with the imaging results. These findings demonstrate that HClO may be an appropriate indicator of DILI, and H2S plays an important role in the antidotal effect of NAC. We envision that RPC-1 can be used as a powerful tool to predict clinical DILI and study the effect of antidote, as well as explore the molecular mechanisms involved.
Collapse
Affiliation(s)
- Xiaoyun Jiao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| | - Yongsheng Xiao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| | - Yong Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| | - Muwen Liang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| | - Xilei Xie
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| | - Xu Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Molecular and Nano Science , Shandong Normal University , Jinan 250014 , P. R. China
| |
Collapse
|
32
|
New NO- and H2S-releasing doxorubicins as targeted therapy against chemoresistance in castration-resistant prostate cancer: in vitro and in vivo evaluations. Invest New Drugs 2018; 36:985-998. [PMID: 29607467 DOI: 10.1007/s10637-018-0590-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/15/2018] [Indexed: 12/12/2022]
Abstract
Chemotherapy for castration-resistant prostate cancer (CRPC) is only temporarily effective due to the onset of chemoresistance. We investigated the efficacy of NO- and H2S-releasing doxorubicins (NitDox and H2SDox) in overcoming drug resistance and evaluated their safety. New and innovative NO- and H2S-releasing doxorubicins (NitDox and H2SDox) showed a good intracellular accumulation and high cytotoxic activity in vitro in an androgen-independent and doxorubicin-resistant DU-145 prostate cancer cell line. Nude mice were subcutaneously injected with 4*106 DU-145 cells and treated once a week for 3 weeks with 5 mg/kg doxorubicin, NitDox, H2SDox or vehicle, i.p. Animal weight, tumor volume, intra-tumoral drug accumulation, apoptosis and the presence of nitrotyrosine and sulfhydryl (SH) groups within the tumor, were evaluated. Cardiotoxicity was assessed by measuring troponin plasma levels and the left ventricular wall thickness. In vivo, NitDox and H2SDox accumulated inside the tumors, significantly reduced tumor volumes by 60%, increased the percentage of apoptotic cells in both the inner and the outer parts of the tumors and the presence of nitrotyrosine and SH groups. Doxorubicin treatment was associated with reduced body weight and cardiotoxicity. On the contrary, NitDox and H2SDox were well tolerated and had a better safety profile. Combining efficacy with reduced cardiovascular side effects, NitDox and H2SDox are promising novel therapeutic agents for reversing chemoresistance in CRCP.
Collapse
|
33
|
Du S, Huang Y, Jin H, Wang T. Protective Mechanism of Hydrogen Sulfide against Chemotherapy-Induced Cardiotoxicity. Front Pharmacol 2018; 9:32. [PMID: 29434549 PMCID: PMC5790791 DOI: 10.3389/fphar.2018.00032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/11/2018] [Indexed: 12/12/2022] Open
Abstract
Over the past few decades, the number of long term survivors of childhood cancers has been increased exponentially. However, among these survivors, treatment-related toxicity, especially cardiotoxicity, is becoming the essential cause of morbidity and mortality. Thus, preventing the treatment-related adverse effects is important to increase the event free survival during the treatment of cancer in children and adolescents. Accumulating evidence has demonstrated that hydrogen sulfide (H2S) exerts a protective role on cardiomyocytes through a variety of mechanisms. Here, we mainly reviewed the cardioprotective role of H2S in the chemotherapy, and emphatically discussed the possible mechanisms.
Collapse
Affiliation(s)
- Shuxu Du
- Department of Pediatrics, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Tianyou Wang
- Hematology/Oncology Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
34
|
Lin J, Chen M, Liu D, Guo R, Lin K, Deng H, Zhi X, Zhang W, Feng J, Wu W. Exogenous hydrogen sulfide protects human umbilical vein endothelial cells against high glucose‑induced injury by inhibiting the necroptosis pathway. Int J Mol Med 2017; 41:1477-1486. [PMID: 29286079 PMCID: PMC5819925 DOI: 10.3892/ijmm.2017.3330] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 12/05/2017] [Indexed: 11/25/2022] Open
Abstract
Hyperglycemia is a key factor in the development of diabetic complications, including the processes of atherosclerosis. Receptor-interacting protein 3 (RIP3), a mediator of necroptosis, is implicated in atherosclerosis development. Additionally, hydrogen sulfide (H2S) protects the vascular endothelium against hyperglycemia-induced injury and attenuates atherosclerosis. On the basis of these findings, the present study aimed to confirm the hypothesis that necroptosis mediates high glucose (HG)-induced injury in human umbilical vein endothelial cells (HUVECs), and that the inhibition of necroptosis contributes to the protective effect of exogenous H2S against this injury. The results revealed that exposure of HUVECs to 40 mM HG markedly enhanced the expression level of RIP3, along with multiple injuries, including a decrease in cell viability, an increase in the number of apoptotic cells, an increase in the expression level of cleaved caspase-3, generation of reactive oxygen species (ROS), as well as dissipation of the mitochondrial membrane potential (MMP). Treatment of the cells with sodium hydrogen sulfide (NaHS; a donor of H2S) prior to exposure to HG significantly attenuated the increased RIP3 expression and the aforementioned injuries by HG. Notably, treatment of cells with necrostatin-1 (Nec-1), an inhibitor of necroptosis, prior to exposure to HG ameliorated the HG-induced injuries, leading to a decrease in ROS generation and a loss of MMP. However, pre-treatment of the cells with Nec-1 enhanced the HG-induced increase in the expression levels of cleaved caspases-3 and -9. By contrast, pre-treatment with Z-VAD-FMK, a pan-caspase inhibitor, promoted the increased expression of RIP3 by HG. Taken together, the findings of the present study have demonstrated, to the best of our knowledge for the first time, that exogenous H2S protects HUVECs against HG-induced injury through inhibiting necroptosis. The present study has also provided novel evidence that there is a negative interaction between necroptosis and apoptosis in the HG-treated HUVECs.
Collapse
Affiliation(s)
- Jiaqiong Lin
- Department of Endocrinology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatrics Institute, Guangzhou, Guangdong 510080, P.R. China
| | - Meiji Chen
- Department of Pediatrics, Huangpu Division of the First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510700, P.R. China
| | - Donghong Liu
- Department of Ultrasound, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou 510080, P.R. China
| | - Ruixian Guo
- Department of Physiology, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou 510080, P.R. China
| | - Kai Lin
- Department of Endocrinology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatrics Institute, Guangzhou, Guangdong 510080, P.R. China
| | - Haiou Deng
- Department of Endocrinology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatrics Institute, Guangzhou, Guangdong 510080, P.R. China
| | - Ximei Zhi
- Department of Endocrinology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatrics Institute, Guangzhou, Guangdong 510080, P.R. China
| | - Weijie Zhang
- Department of Endocrinology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatrics Institute, Guangzhou, Guangdong 510080, P.R. China
| | - Jianqiang Feng
- Department of Physiology, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou 510080, P.R. China
| | - Wen Wu
- Department of Endocrinology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangdong Geriatrics Institute, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
35
|
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
|
36
|
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: 392] [Impact Index Per Article: 49.0] [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
|
37
|
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
|
38
|
Yang CT, Chen L, Xu S, Day JJ, Li X, Xian M. Recent Development of Hydrogen Sulfide Releasing/Stimulating Reagents and Their Potential Applications in Cancer and Glycometabolic Disorders. Front Pharmacol 2017; 8:664. [PMID: 29018341 PMCID: PMC5623001 DOI: 10.3389/fphar.2017.00664] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/06/2017] [Indexed: 12/24/2022] Open
Abstract
As an important endogenous gaseous signaling molecule, hydrogen sulfide (H2S) exerts various effects in the body. A variety of pathological changes, such as cancer, glycometabolic disorders, and diabetes, are associated with altered endogenous levels of H2S, especially decreased. Therefore, the supplement of H2S is of great significance for the treatment of diseases containing the above pathological changes. At present, many efforts have been made to increase the in vivo levels of H2S by administration of gaseous H2S, simple inorganic sulfide salts, sophisticated synthetic slow-releasing controllable H2S donors or materials, and using H2S stimulating agents. In this article, we reviewed the recent development of H2S releasing/stimulating reagents and their potential applications in two common pathological processes including cancer and glycometabolic disorders.
Collapse
Affiliation(s)
- Chun-Tao Yang
- Affiliated Cancer Hospital and Institute, Key Laboratory of Protein Modification and Degradation in School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Department of Chemistry, Washington State University, Pullman, WA, United States
| | - Li Chen
- Affiliated Cancer Hospital and Institute, Key Laboratory of Protein Modification and Degradation in School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Shi Xu
- Department of Chemistry, Washington State University, Pullman, WA, United States
| | - Jacob J Day
- Department of Chemistry, Washington State University, Pullman, WA, United States
| | - Xiang Li
- Affiliated Cancer Hospital and Institute, Key Laboratory of Protein Modification and Degradation in School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, WA, United States
| |
Collapse
|
39
|
Inhibition of PTEN Attenuates Endoplasmic Reticulum Stress and Apoptosis via Activation of PI3K/AKT Pathway in Alzheimer’s Disease. Neurochem Res 2017; 42:3052-3060. [PMID: 28819903 DOI: 10.1007/s11064-017-2338-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 12/14/2022]
|
40
|
Wang CY, Zou W, Liang XY, Jiang ZS, Li X, Wei HJ, Tang YY, Zhang P, Tang XQ. Hydrogen sulfide prevents homocysteine‑induced endoplasmic reticulum stress in PC12 cells by upregulating SIRT‑1. Mol Med Rep 2017; 16:3587-3593. [PMID: 28713986 DOI: 10.3892/mmr.2017.7004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 05/15/2017] [Indexed: 11/06/2022] Open
Abstract
It was previously confirmed that hydrogen sulfide (H2S) has a neuroprotective effect, preventing homocysteine‑induced neurotoxicity. However, the exact molecular mechanisms underlying this protective effect remain to be fully elucidated. Endoplasmic reticulum (ER) stress contributes to homocysteine‑induced neurotoxicity. Silent mating type information regulator 2 homolog 1 (SIRT‑1) can attenuate ER stress, exerting its neuroprotective effect. Therefore, the present study aimed to investigate whether H2S protects PC12 cells against homocysteine‑induced ER stress and whether SIRT‑1 mediates this protective effect of H2S. Western blotting was used to detect the expression of SIRT‑1, glucose‑regulated protein 78 (GRP78), and cleaved caspase‑12 in PC12 cells. It was observed that sodium hydrosulfide (NaHS), an exogenous H2S donor, significantly attenuated the homocysteine‑induced ER stress responses, including increases in the protein expression levels of GRP78 and cleaved caspase‑12. Simultaneously, NaHS upregulated the expression of SIRT‑1 and reversed the homocysteine‑induced downregulation of SIRT‑1 in PC12 cells. Sirtinol, a specific inhibitor of SIRT‑1, eliminated the protective effects of H2S in homocysteine‑induced ER stress. These data indicated that H2S prevented homocysteine‑induced ER stress via enhancing the expression of SIRT‑1. These findings offer novel insight into the protective mechanisms of H2S against homocysteine‑induced neurotoxicity.
Collapse
Affiliation(s)
- Chun-Yan Wang
- The Institute of Cardiovascular Disease, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wei Zou
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiao-Yu Liang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhi-Sheng Jiang
- The Institute of Cardiovascular Disease, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiang Li
- Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hai-Jun Wei
- Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yi-Yun Tang
- Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ping Zhang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiao-Qing Tang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
41
|
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
|
42
|
Bauckneht M, Ferrarazzo G, Fiz F, Morbelli S, Sarocchi M, Pastorino F, Ghidella A, Pomposelli E, Miglino M, Ameri P, Emionite L, Ticconi F, Arboscello E, Buschiazzo A, Massimelli EA, Fiordoro S, Borra A, Cossu V, Bozzano A, Ibatici A, Ponzoni M, Spallarossa P, Gallamini A, Bruzzi P, Sambuceti G, Marini C. Doxorubicin Effect on Myocardial Metabolism as a Prerequisite for Subsequent Development of Cardiac Toxicity: A Translational 18F-FDG PET/CT Observation. J Nucl Med 2017. [PMID: 28646013 DOI: 10.2967/jnumed.117.191122] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The present translational study aimed to verify whether serial 18F-FDG PET/CT predicts doxorubicin cardiotoxicity. Methods: Fifteen athymic mice were treated intravenously with saline (n = 5) or with 5 or 7.5 mg of doxorubicin per kilogram (n = 5 each) and underwent dynamic small-animal PET beforehand and afterward to estimate left ventricular (LV) metabolic rate of glucose (MRGlu). Thereafter, we retrospectively identified 69 patients who had been successfully treated with a regimen of doxorubicin, bleomycin, vinblastine, and dacarbazine for Hodgkin disease (HD) and had undergone 4 consecutive 18F-FDG PET/CT scans. Volumes of interest were drawn on LV myocardium to quantify mean SUV. All patients were subsequently interviewed by telephone (median follow-up, 30 mo); 36 of them agreed to undergo electrocardiography and transthoracic echocardiography. Results: In mice, LV MRGlu was 17.9 ± 4.4 nmol × min-1 × g-1 at baseline. Doxorubicin selectively and dose-dependently increased this value in the standard-dose (27.9 ± 9 nmol × min-1 × g-1, P < 0.05 vs. controls) and high-dose subgroups (37.2 ± 7.8 nmol × min-1 × g-1, P < 0.01 vs. controls, P < 0.05 vs. standard-dose). In HD patients, LV SUV showed a progressive increase during doxorubicin treatment that persisted at follow-up. New-onset cardiac abnormalities appeared in 11 of 36 patients (31%). In these subjects, pretherapy LV SUV was markedly lower with respect to the remaining patients (1.53 ± 0.9 vs. 3.34 ± 2.54, respectively, P < 0.01). Multivariate analysis confirmed the predictive value of baseline LV SUV for subsequent cardiac abnormalities. Conclusion: Doxorubicin dose-dependently increases LV MRGlu, particularly in the presence of low baseline 18F-FDG uptake. These results imply that low myocardial 18F-FDG uptake before the initiation of doxorubicin chemotherapy in HD patients may predict the development of chemotherapy-induced cardiotoxicity, suggesting that prospective clinical trials are warranted to test this hypothesis.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Giulia Ferrarazzo
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Francesco Fiz
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy.,Nuclear Medicine Unit, Department of Radiology, Tübingen, Germany
| | - Silvia Morbelli
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Matteo Sarocchi
- Clinic of Cardiovascular Diseases, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Fabio Pastorino
- Unit of Experimental Therapy in Oncology, IRCCS Gaslini, Genoa, Italy
| | - Alberto Ghidella
- Clinic of Cardiovascular Diseases, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Elena Pomposelli
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Maurizio Miglino
- Haematology Clinic, University of Genoa, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Pietro Ameri
- Clinic of Cardiovascular Diseases, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Laura Emionite
- Animal Facility, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Flavia Ticconi
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | | | - Ambra Buschiazzo
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | | | - Salvatore Fiordoro
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Anna Borra
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Vanessa Cossu
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Annalisa Bozzano
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Adalberto Ibatici
- Haematology Clinic, University of Genoa, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Mirco Ponzoni
- Unit of Experimental Therapy in Oncology, IRCCS Gaslini, Genoa, Italy
| | - Paolo Spallarossa
- Clinic of Cardiovascular Diseases, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Andrea Gallamini
- Department of Research, Innovation, and Statistics, Lacassagne Cancer Centre, Nice, France
| | - Paolo Bruzzi
- Epidemiology Unit, IRCCS-AOU San Martino-IST, Genoa, Italy; and
| | - Gianmario Sambuceti
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Cecilia Marini
- CNR Institute of Bioimaging and Molecular Physiology, Section of Genoa, Milan, Italy
| |
Collapse
|
43
|
Liu SY, Li D, Zeng HY, Kan LY, Zou W, Zhang P, Gu HF, Tang XQ. Hydrogen Sulfide Inhibits Chronic Unpredictable Mild Stress-Induced Depressive-Like Behavior by Upregulation of Sirt-1: Involvement in Suppression of Hippocampal Endoplasmic Reticulum Stress. Int J Neuropsychopharmacol 2017; 20:867-876. [PMID: 28482013 PMCID: PMC5737807 DOI: 10.1093/ijnp/pyx030] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 05/03/2017] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Hydrogen sulfide (H2S) is a crucial signaling molecule with a wide range of physiological functions. Previously, we confirmed that stress-induced depression is accompanied with disturbance of H2S generation in hippocampus. The present work attempted to investigate the inhibitory effect of H2S on chronic unpredictable mild stress-induced depressive-like behaviors and the underlying mechanism. METHODS We established the rat model of chronic unpredictable mild stress to simulate depression. Open field test, forced swim test, and tail suspension test were used to assess depressive-like behaviors. The expression of Sirt-1 and three marked proteins related to endoplasmic reticulum stress (GRP-78, CHOP, and cleaved caspase-12) were detected by western blot. RESULTS We found that chronic unpredictable mild stress-exposed rats exhibit depression-like behavior responses, including significantly increased immobility time in the forced swim test and tail suspension test, and decreased climbing time and swimming time in the forced swim test. In parallel, chronic unpredictable mild stress-exposed rats showed elevated levels of hippocampal endoplasmic reticulum stress and reduced levels of Sirt-1. However, NaHS (a donor of H2S) not only alleviated chronic unpredictable mild stress-induced depressive-like behaviors and hippocampal endoplasmic reticulum stress, but it also increased the expression of hippocampal Sirt-1 in chronic unpredictable mild stress-exposed rats. Furthermore, Sirtinol, an inhibitor of Sirt-1, reversed the protective effects of H2S against chronic unpredictable mild stress-induced depression-like behaviors and hippocampal endoplasmic reticulum stress. CONCLUSION These results demonstrated that H2S has an antidepressant potential, and the underlying mechanism is involved in the inhibition of hippocampal endoplasmic reticulum stress by upregulation of Sirt-1 in hippocampus. These findings identify H2S as a novel therapeutic target for depression.
Collapse
Affiliation(s)
- Shu-Yun Liu
- Department of Neurology, Affiliated Center Hospital of Shenzhen Longhua New District, Guangdong Medical University, Shenzhen, Guangdong, P. R. China (Ms Liu and Ms Li); Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan, P. R. China (Ms Zeng, Ms Kan, Mr Zou, Mr Zhang, and Drs Gu and Tang); Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, P. R. China (Mr Zou); Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Dr Tang)
| | - Dan Li
- Department of Neurology, Affiliated Center Hospital of Shenzhen Longhua New District, Guangdong Medical University, Shenzhen, Guangdong, P. R. China (Ms Liu and Ms Li); Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan, P. R. China (Ms Zeng, Ms Kan, Mr Zou, Mr Zhang, and Drs Gu and Tang); Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, P. R. China (Mr Zou); Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Dr Tang)
| | - Hai-Ying Zeng
- Department of Neurology, Affiliated Center Hospital of Shenzhen Longhua New District, Guangdong Medical University, Shenzhen, Guangdong, P. R. China (Ms Liu and Ms Li); Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan, P. R. China (Ms Zeng, Ms Kan, Mr Zou, Mr Zhang, and Drs Gu and Tang); Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, P. R. China (Mr Zou); Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Dr Tang)
| | - Li-Yuan Kan
- Department of Neurology, Affiliated Center Hospital of Shenzhen Longhua New District, Guangdong Medical University, Shenzhen, Guangdong, P. R. China (Ms Liu and Ms Li); Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan, P. R. China (Ms Zeng, Ms Kan, Mr Zou, Mr Zhang, and Drs Gu and Tang); Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, P. R. China (Mr Zou); Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Dr Tang)
| | - Wei Zou
- Department of Neurology, Affiliated Center Hospital of Shenzhen Longhua New District, Guangdong Medical University, Shenzhen, Guangdong, P. R. China (Ms Liu and Ms Li); Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan, P. R. China (Ms Zeng, Ms Kan, Mr Zou, Mr Zhang, and Drs Gu and Tang); Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, P. R. China (Mr Zou); Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Dr Tang)
| | - Ping Zhang
- Department of Neurology, Affiliated Center Hospital of Shenzhen Longhua New District, Guangdong Medical University, Shenzhen, Guangdong, P. R. China (Ms Liu and Ms Li); Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan, P. R. China (Ms Zeng, Ms Kan, Mr Zou, Mr Zhang, and Drs Gu and Tang); Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, P. R. China (Mr Zou); Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Dr Tang)
| | - Hong-Feng Gu
- Department of Neurology, Affiliated Center Hospital of Shenzhen Longhua New District, Guangdong Medical University, Shenzhen, Guangdong, P. R. China (Ms Liu and Ms Li); Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan, P. R. China (Ms Zeng, Ms Kan, Mr Zou, Mr Zhang, and Drs Gu and Tang); Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, P. R. China (Mr Zou); Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Dr Tang)
| | - Xiao-Qing Tang
- Department of Neurology, Affiliated Center Hospital of Shenzhen Longhua New District, Guangdong Medical University, Shenzhen, Guangdong, P. R. China (Ms Liu and Ms Li); Institute of Neuroscience, Medical College, University of South China, Hengyang, Hunan, P. R. China (Ms Zeng, Ms Kan, Mr Zou, Mr Zhang, and Drs Gu and Tang); Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, P. R. China (Mr Zou); Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, Hunan, PR China (Dr Tang).,Correspondence: Xiao-Qing Tang, MD, PhD, Department of Physiology, Institute of Neuroscience, Medical College, University of South China, 28 West Changsheng Road, Hengyang 421001, Hunan Province, P. R. China ()
| |
Collapse
|
44
|
Hu HJ, Jiang ZS, Qiu J, Zhou SH, Liu QM. Protective effects of hydrogen sulfide against angiotensin II-induced endoplasmic reticulum stress in HUVECs. Mol Med Rep 2017; 15:2213-2222. [DOI: 10.3892/mmr.2017.6238] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/05/2016] [Indexed: 11/05/2022] Open
|
45
|
Ke X, Chen J, Peng L, Zhang W, Yang Y, Liao X, Mo L, Guo R, Feng J, Hu C, Nie R. Heat shock protein 90/Akt pathway participates in the cardioprotective effect of exogenous hydrogen sulfide against high glucose-induced injury to H9c2 cells. Int J Mol Med 2017; 39:1001-1010. [PMID: 28204829 DOI: 10.3892/ijmm.2017.2891] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 01/17/2017] [Indexed: 11/05/2022] Open
Abstract
It has been reported that exogenous hydrogen sulfide (H2S) protects against high glucose (HG)-induced cardiac injury and has a modulatory effect on heat shock protein (HSP) and Akt, which play a cardioprotective role. In this study, we examined whether the HSP90/Akt pathway contributes to the protective effects of exogenous H2S against HG-induced injury to H9c2 cardiac cells. Our results revealed that the exposure of H9c2 cardiac cells to 35 mM glucose (HG) for 1 to 24 h decreased the expression of HSP90 and markedly reduced the expression level of phosphorylated (p)-Akt in a time-dependent manner. Co-exposure of the cells to HG and geldanamycin (GA; an inhibitor of HSP90) aggravated the inhibition of the p-Akt expression level by HG. Of note, treatment of the cells with 400 µM NaHS (a donor of H2S) for 30 min prior to exposure to HG significantly attenuated the HG-induced decrease in the expression levels of both HSP90 and p-Akt, along with inhibition of HG-induced cell injury, as indicated by the increase in cell viability and superoxide dismutase (SOD) activity, and by a decrease in the number of apoptotic cells, reactive oxygen species (ROS) generation, as well as by the decreased dissipation of mitochondrial membrance potential (MMP). Importantly, treatment of the cells with GA or LY294002 (an inhibitor of Akt) prior to exposure to NaHS and HG considerably blocked the cardioprotective effects of NaHS against the HG-induced injury mentioned above. On the whole, the findings of this study demonstrate that the inhibition of the HSP90/Akt pathway may be an important mechanism responsible for HG-induced cardiomyocyte injury. We also provide novel evidence that exogenous H2S protects H9c2 cells against HG-induced injury by activating the HSP90/Akt pathway.
Collapse
Affiliation(s)
- Xiao Ke
- Department of Cardiology, Shenzhen Sun Yat-sen Cardiovascular Hospital, Shenzhen, Guangdong, P.R. China
| | - Jingfu Chen
- Department of Cardiovascular Medicine and Dongguan Cardiovascular Institute, The Third People's Hospital of Dongguan City, Dongguan, Guangdong, P.R. China
| | - Longyun Peng
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Wei Zhang
- Department of Cardiovasology and Cardiac Care Unit (CCU), Huangpu Division of The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Yiying Yang
- Department of Cardiovasology and Cardiac Care Unit (CCU), Huangpu Division of The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xinxue Liao
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Liqiu Mo
- Department of Anesthesiology, Huangpu Division of The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Ruixian Guo
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Jianqiang Feng
- Department of Anesthesiology, Huangpu Division of The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Chengheng Hu
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Ruqiong Nie
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen Universtity, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
46
|
Xu D, Jin H, Wen J, Chen J, Chen D, Cai N, Wang Y, Wang J, Chen Y, Zhang X, Wang X. Hydrogen sulfide protects against endoplasmic reticulum stress and mitochondrial injury in nucleus pulposus cells and ameliorates intervertebral disc degeneration. Pharmacol Res 2017; 117:357-369. [PMID: 28087442 DOI: 10.1016/j.phrs.2017.01.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/17/2016] [Accepted: 01/05/2017] [Indexed: 11/16/2022]
Abstract
It has been suggested that excessive apoptosis in intervertebral disc cells induced by inflammatory cytokines, such as interleukin (IL)-1β, is related to the process of intervertebral disc degeneration (IVDD). Hydrogen sulfide (H2S), a gaseous signaling molecule, has drawn attention for its anti-apoptosis role in various pathophysiological processes in degenerative diseases. To date, there has been no investigation of the correlation of H2S production and IVDD or of the effects of H2S on IL-1β-induced apoptosis in nucleus pulposus (NP) cells. Here, we found that the expression levels of cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE), two key enzymes in the generation of H2S, were significantly decreased in human degenerate NP tissues as well as in IL-1β-treated NP cells. NaHS (H2S donor) administration showed a protective effect by inhibiting the endoplasmic reticulum (ER) stress response and mitochondrial dysfunction induced by IL-1β stimulation in vitro, the effect was related to activation of the PI3K/Akt and ERK1/2 signaling pathways. Suppression of these pathways by specific inhibitors, LY294002 and PD98059, partially reduced the protective effect of NaHS. Moreover, in the percutaneous needle puncture disc degeneration rat tail model, disc degeneration was partially reversed by NaHS administration. Taken together, our results suggest that H2S plays a protective role in IVDD and the underlying mechanism involves PI3K/Akt and ERK1/2 signaling pathways-mediated suppression of ER stress and mitochondrial dysfunction in IL-1β-induced NP cells.
Collapse
Affiliation(s)
- Daoliang Xu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haiming Jin
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianxia Wen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiaoxiang Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Deheng Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ningyu Cai
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongli Wang
- Department of Orthopaedic Surgery, Huzhou Central Hospital, Huzhou, China
| | - Jianle Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yu Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaolei Zhang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang, China.
| | - Xiangyang Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
47
|
Alginate Oligosaccharide Prevents Acute Doxorubicin Cardiotoxicity by Suppressing Oxidative Stress and Endoplasmic Reticulum-Mediated Apoptosis. Mar Drugs 2016; 14:md14120231. [PMID: 27999379 PMCID: PMC5192468 DOI: 10.3390/md14120231] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 12/07/2016] [Accepted: 12/09/2016] [Indexed: 01/01/2023] Open
Abstract
Doxorubicin (DOX) is a highly potent chemotherapeutic agent, but its usage is limited by dose-dependent cardiotoxicity. DOX-induced cardiotoxicity involves increased oxidative stress and activated endoplasmic reticulum-mediated apoptosis. Alginate oligosaccharide (AOS) is a non-immunogenic, non-toxic and biodegradable polymer, with anti-oxidative, anti-inflammatory and anti-endoplasmic reticulum stress properties. The present study examined whether AOS pretreatment could protect against acute DOX cardiotoxicity, and the underlying mechanisms focused on oxidative stress and endoplasmic reticulum-mediated apoptosis. We found that AOS pretreatment markedly increased the survival rate of mice insulted with DOX, improved DOX-induced cardiac dysfunction and attenuated DOX-induced myocardial apoptosis. AOS pretreatment mitigated DOX-induced cardiac oxidative stress, as shown by the decreased expressions of gp91 (phox) and 4-hydroxynonenal (4-HNE). Moreover, AOS pretreatment significantly decreased the expression of Caspase-12, C/EBP homologous protein (CHOP) (markers for endoplasmic reticulum-mediated apoptosis) and Bax (a downstream molecule of CHOP), while up-regulating the expression of anti-apoptotic protein Bcl-2. Taken together, these findings identify AOS as a potent compound that prevents acute DOX cardiotoxicity, at least in part, by suppression of oxidative stress and endoplasmic reticulum-mediated apoptosis.
Collapse
|
48
|
Wu J, Guo W, Lin SZ, Wang ZJ, Kan JT, Chen SY, Zhu YZ. Gp130-mediated STAT3 activation by S-propargyl-cysteine, an endogenous hydrogen sulfide initiator, prevents doxorubicin-induced cardiotoxicity. Cell Death Dis 2016; 7:e2339. [PMID: 27537522 PMCID: PMC5108313 DOI: 10.1038/cddis.2016.209] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 12/20/2022]
Abstract
Doxorubicin (Dox) could trigger a large amount of apoptotic cells in the myocardium, which leads to dilated cardiomyopathy and heart failure. S-propargyl-cysteine (SPRC), a producing agent of endogenous hydrogen sulfide (H2S), possesses cardioprotective efficacy. However, the specific effect and mechanism of SPRC in Dox-induced cardiotoxicity remain elusive. Given gp130 with its main downstream signaling molecule, signal transducer and activator of transcription 3 (STAT3), is involved in cardiac myocyte survival and growth; the present study was performed to elucidate whether SPRC counteracts Dox-induced cardiotoxicity, and if so, whether the gp130/STAT3 pathway is involved in this cardioprotective activity. SPRC stimulated the activation of STAT3 via gp130-mediated transduction tunnel in vitro and in vivo. In Dox-stimulated cardiotoxicity, SPRC enhanced cell viability, restored expression of gp130/STAT3-regulated downstream genes, inhibited apoptosis and oxidative stress, and antagonized mitochondrial dysfunction and intracellular Ca(2+) overload. Intriguingly, blockade of gp130/STAT3 signaling abrogated all these beneficial capacities of SPRC. Our findings present the first piece of evidence for the therapeutic properties of SPRC in alleviating Dox cardiotoxicity, which could be attributed to the activation of gp130-mediated STAT3 signaling. This will offer a novel molecular basis and therapeutic strategy of H2S donor for the treatment of heart failure.
Collapse
Affiliation(s)
- J Wu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - W Guo
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - S-Z Lin
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Z-J Wang
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - J-T Kan
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - S-Y Chen
- Department of Cardiovascular Surgery, Guangdong General Hospital, Guangzhou, Guangdong, China
| | - Y-Z Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.,School of Pharmacy, Macau University of Science & Technology, Macau, China
| |
Collapse
|
49
|
Doxorubicin impairs cardiomyocyte viability by suppressing transcription factor EB expression and disrupting autophagy. Biochem J 2016; 473:3769-3789. [PMID: 27487838 DOI: 10.1042/bcj20160385] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/03/2016] [Indexed: 12/11/2022]
Abstract
Doxorubicin (DOX) is an effective anti-cancer agent. However, DOX treatment increases patient susceptibility to dilated cardiomyopathy. DOX predisposes cardiomyocytes to insult by suppressing mitochondrial energy metabolism, altering calcium flux, and disrupting proteolysis and proteostasis. Prior studies have assessed the role of macroautophagy in DOX cardiotoxicity; however, limited studies have examined whether DOX mediates cardiac injury through dysfunctions in inter- and/or intra-lysosomal signaling events. Lysosomal signaling and function is governed by transcription factor EB (TFEB). In the present study, we hypothesized that DOX caused myocyte injury by impairing lysosomal function and signaling through negative regulation of TFEB. Indeed, we found that DOX repressed cellular TFEB expression, which was associated with impaired cathepsin proteolytic activity across in vivo, ex vivo, and in vitro models of DOX cardiotoxicity. Furthermore, we observed that loss of TFEB was associated with reduction in macroautophagy protein expression, inhibition of autophagic flux, impairments in lysosomal cathepsin B activity, and activation of cell death. Restoration and/or activation of TFEB in DOX-treated cardiomyocytes prevented DOX-induced suppression of cathepsin B activity, reduced DOX-mediated reactive oxygen species (ROS) overproduction, attenuated activation of caspase-3, and improved cellular viability. Collectively, loss of TFEB inhibits lysosomal autophagy, rendering cardiomyocytes susceptible to DOX-induced proteotoxicity and injury. Our data reveal a novel mechanism wherein DOX primes cardiomyocytes for cell death by depleting cellular TFEB.
Collapse
|
50
|
Sheng CC, Amiri-Kordestani L, Palmby T, Force T, Hong CC, Wu JC, Croce K, Kim G, Moslehi J. 21st Century Cardio-Oncology: Identifying Cardiac Safety Signals in the Era of Personalized Medicine. JACC Basic Transl Sci 2016; 1:386-398. [PMID: 28713868 PMCID: PMC5508213 DOI: 10.1016/j.jacbts.2016.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 01/01/2023]
Abstract
Cardiotoxicity is a well-established complication of oncology therapies. Cardiomyopathy resulting from anthracyclines is a classic example. In the past decade, an explosion of novel cancer therapies, often targeted and more specific than traditional therapies, has revolutionized oncology therapy and dramatically changed cancer prognosis. However, some of these therapies have introduced an assortment of cardiovascular (CV) complications. At times, these devastating outcomes have only become apparent after drug approval and have limited the use of potent therapies. There is a growing need for better testing platforms, both for CV toxicity screening, as well as for elucidating mechanisms of cardiotoxicities of approved cancer therapies. This review discusses the utility of nonclinical models (in vitro, in vivo, & in silico) available and highlights recent advancements in modalities like human stem cell-derived cardiomyocytes for developing more comprehensive cardiotoxicity testing and new means of cardioprotection with targeted anticancer therapies.
Collapse
Affiliation(s)
- Calvin Chen Sheng
- Cardiovascular Division, Vanderbilt University School of Medicine, Nashville, Tennessee
- Cardio-Oncology Program, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Laleh Amiri-Kordestani
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Todd Palmby
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Thomas Force
- Cardiovascular Division, Vanderbilt University School of Medicine, Nashville, Tennessee
- Cardio-Oncology Program, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Charles C. Hong
- Cardiovascular Division, Vanderbilt University School of Medicine, Nashville, Tennessee
- Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
- Accelerating Drug Repurposing Incubator, Vanderbilt Institute for Clinical and Translational Research, Nashville, Tennessee
| | - Joseph C. Wu
- Cardiovascular Division, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
| | - Kevin Croce
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey Kim
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Javid Moslehi
- Cardiovascular Division, Vanderbilt University School of Medicine, Nashville, Tennessee
- Cardio-Oncology Program, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| |
Collapse
|