1
|
Eto F, Nezu T, Sakahara H, Yamamoto Y, Aoki S, Shimomura M, Karakawa S, Maruyama H. Intravenous Thrombolysis for Pediatric Ischemic Stroke Secondary to Cancer Therapy-related Cardiac Dysfunction. Intern Med 2025; 64:287-291. [PMID: 38811218 DOI: 10.2169/internalmedicine.3755-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/31/2024] Open
Abstract
An 11-year-old boy developed cardioembolic stroke (CES) and cancer therapy-related cardiac dysfunction (CTRCD). He originally developed Ewing sarcoma and was treated with high-dose chemotherapy including doxorubicin. On admission, he had severe aphasia, and magnetic resonance imaging showed occlusion of the left middle cerebral artery M3 segment. Transthoracic echocardiography revealed severe left ventricular dysfunction and a mobile thrombus at the left ventricular apex. Intravenous thrombolysis was administered, and effective recanalization was achieved. The patient did not exhibit any neurological deficits during discharge. Reperfusion therapy for pediatric patients has not yet been established; however, it may be effective for CES secondary to CTRCD.
Collapse
Affiliation(s)
- Futoshi Eto
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Tomohisa Nezu
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Hideaki Sakahara
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Yumiko Yamamoto
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Shiro Aoki
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Maiko Shimomura
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Shuhei Karakawa
- Department of Pediatrics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| |
Collapse
|
2
|
Feng L, Wang X, Gao Z, Tong Y, Yuan X, Wu T, Xia D, Hu Y. Enhancing Chemotherapy Efficacy via an Autologous Erythrocyte-Anchoring Strategy with a Closed-System Drug-Transfer Device. ACS Biomater Sci Eng 2025; 11:429-441. [PMID: 39696880 DOI: 10.1021/acsbiomaterials.4c02128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Chemotherapeutic drugs often fail to localize efficiently to tumors when administered intravenously, causing off-target effects. This study proposes an autologous erythrocyte (ER)-anchoring strategy to improve chemotherapy efficacy and reduce side effects. Utilizing a modified hemodialysis instrument, a closed-system drug-transfer device was developed for autologous ER procurement and immunogenicity mitigation. Doxorubicin (DOX) and indocyanine green (ICG) were encapsulated in autologous ERs and then modified with DSPE-PEG-FA. The final product, DOX-ICG@ER-D, was reintroduced into circulation to enhance chemotherapy. These obtained DOX-ICG@ER-D showed good stability, minimal cardiotoxicity, and extended circulation time. Compared to free DOX, DOX-ICG@ER-D had a higher accumulation of DOX in hepatocellular carcinoma and the release of DOX could be controlled by laser irradiation. Tumor-bearing rats treated by these DOX-ICG@ER-D demonstrated improved antitumor efficacy and reduced cardiotoxicity. Thus, this autologous ER-anchoring strategy offers a promising alternative to intravenous chemotherapy in the clinic.
Collapse
Affiliation(s)
- Lingzi Feng
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing 210033, China
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, Jiangsu 226019, P. R. China
| | - Xiangqian Wang
- Department of Radiotherapy, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong 226361, P. R. China
| | - Ziyi Gao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu 210029, China
| | - Yuqing Tong
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing 210033, China
| | - Xiaopeng Yuan
- Department of Radiotherapy, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong 226361, P. R. China
| | - Ting Wu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu 210029, China
| | - Donglin Xia
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, Jiangsu 226019, P. R. China
| | - Yong Hu
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing 210033, China
| |
Collapse
|
3
|
Zhang J, Ding W, Yin Z, Liu S, Zhao M, Xu Y, Liu J, Pan W, Peng S, Wei C, Zheng Z, Qin JJ, Wan J, Wang M. Interleukin-12p40 deficiency attenuates myocardial ferroptosis in doxorubicin-induced chronic cardiomyopathy by inhibiting Th17 differentiation and interleukin-17A production. Cardiovasc Res 2024; 120:2117-2133. [PMID: 39298642 DOI: 10.1093/cvr/cvae208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/07/2024] [Accepted: 08/06/2024] [Indexed: 09/22/2024] Open
Abstract
AIMS Interleukin (IL)-12p40 is a common subunit of the bioactive cytokines IL-12 and IL-23, and it also has its own intrinsic functional activity. However, its role in doxorubicin-induced chronic cardiomyopathy (DICCM) as well as the underlying mechanisms are still unknown. METHODS AND RESULTS In this study, we used IL-12p40-knockout mice, IL-23p19-knockout mice, Rag1-knockout mice, a ferroptosis inhibitor, recombinant IL-12 (rIL-12), rIL-23, rIL-12p40, rIL-12p80, and anti-IL17A to investigate the effects of IL-12p40 on DICCM and elucidate the underlying mechanisms. We found that myocardial ferroptosis were increased in DICCM and that the inhibition of ferroptosis protected against DICCM. The expression of IL-12p40 was upregulated, and IL-12p40 was predominantly expressed by CD4+ T cells in the hearts of mice with DICCM. IL-12p40 knockout attenuated cardiac dysfunction, fibrosis and ferroptosis in DICCM, and similar results were observed in the context of CD4+ T cell IL-12p40 deficiency in Rag1-/- mice. Treatment with rIL-23, but not rIL-12, rIL-12p40 monomer or rIL-12p80, abolished the protective effects of IL-12p40 knockout. Moreover, rIL-23 treatment and IL-23p19 knockout exacerbated and ameliorated DICCM, respectively. IL-12p40 knockout might protect against DICCM by inhibiting Th17 differentiation and IL-17A production but not Th1, Th2 and Treg differentiation. Neutralizing IL-17A with an antibody also attenuated cardiac dysfunction, fibrosis, and ferroptosis. The IL-12p40/Th17/IL-17A axis might promote cardiomyocyte ferroptosis by activating TNF receptor-associated factor 6 (TRAF6)/mitogen-activated protein kinase (MAPK)/P53 signalling in DICCM. CONCLUSION Interleukin-12p40 deficiency protects against DICCM by inhibiting Th17 differentiation and the production of IL-17A, which plays critical roles in cardiomyocyte ferroptosis in DICCM via activating TRAF6/MAPK/P53 signalling. Our study may provide novel insights for the identification of therapeutic targets for treating DICCM in the clinic.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
4
|
Wang D, Jin Y, Yang M, Xue Y, Zhang X, Guo Y, Li X, Ma K. Cardioprotective effect of Saussurea involucrata injection against Doxorubicin-induced cardiotoxicity by network pharmacology analysis and experimental verification. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39632659 DOI: 10.3724/abbs.2024170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Doxorubicin (Dox) is widely utilized in the clinical treatment of various cancers. Despite its efficacy, Dox induces numerous adverse effects in humans with significant cardiotoxicity, posing a major limitation to its use. Saussurea involucrata injection (SII), derived from Saussurea involucrata, exhibits notable anti-inflammatory and anti-oxidative stress properties. However, its potential protective effects against Dox-induced cardiotoxicity (DIC) remain unexplored. In this study, we investigate the ability of SII to mitigate DIC and elucidate the underlying mechanisms through experimental research and network pharmacology analysis. Results from both in vitro and in vivo experiments reveal that SII treatment significantly improves Dox-induced cardiac dysfunction, reducing pathological alterations and fibrosis in cardiomyocytes. Moreover, SII has cardioprotective effects by diminishing the inflammation, oxidative stress, and apoptosis triggered by Dox. Network pharmacological analysis further shows that SII downregulates P53 protein expression by activating the AKT/MDM2 signaling pathway, thus attenuating DIC. In conclusion, this study confirms that SII mitigates DIC through downregulation of the AKT/MDM2/P53 signaling pathway, suggesting a promising therapeutic strategy for alleviating DIC.
Collapse
Affiliation(s)
- Ding Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Yu Jin
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Mengyu Yang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Yajing Xue
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Xiaotong Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Yanli Guo
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Xinzhi Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| |
Collapse
|
5
|
Wesley CD, Sansonetti A, Neutel CHG, Krüger DN, De Meyer GRY, Martinet W, Guns PJ. Short-Term Proteasome Inhibition: Assessment of the Effects of Carfilzomib and Bortezomib on Cardiac Function, Arterial Stiffness, and Vascular Reactivity. BIOLOGY 2024; 13:844. [PMID: 39452152 PMCID: PMC11504385 DOI: 10.3390/biology13100844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024]
Abstract
Proteasome inhibitors such as bortezomib and carfilzomib induce apoptosis and are a cornerstone in the treatment of relapsed or refractory multiple myeloma. However, concerns have emerged concerning their link to cancer therapy-related cardiovascular dysfunction (CTRCD). Bortezomib, a reversible first-generation inhibitor, and carfilzomib, a second-generation irreversible inhibitor, are associated with hypertension, heart failure, and cardiac arrhythmias. The current study investigated the effects of bortezomib and carfilzomib on cardiac (left ventricular ejection fraction, LVEF) and vascular (arterial stiffness, vascular reactivity) function. Cardiac function assessment aimed to build upon existing evidence of proteasome inhibitors CTRCD, while arterial stiffness served as an early indicator of potential vascular remodeling. Groups of 12-week-old C57BL/6J male mice (n = 8 per group) were randomly assigned to receive vehicle, carfilzomib (8 mg/kg I.P.), or bortezomib (0.5 mg/kg I.P.). Additionally, proteasome inhibition was assessed in mice treated with L-NAME (0.5 mg/kg) to induce hypertension. Cardiac and vascular parameters were evaluated via echocardiography on days 0 and 3. On day 6, mice were sacrificed for ex vivo analysis of arterial stiffness and vascular reactivity. Overall, no changes in arterial stiffness were detected either in vivo or ex vivo at basal pressures. However, a steeper pressure-stiffness curve was observed for carfilzomib in normotensive (p < 0.01) and hypertensive (p < 0.0001) mice ex vivo. Additionally, in hypertensive mice, carfilzomib decreased LVEF (p = 0.06), with bortezomib exhibiting similar trends. Vascular reactivity remained largely unchanged, but proteasome inhibition tended to enhance endothelial-independent relaxations in both control and hypertensive mice. In conclusion, short-term treatment with carfilzomib and bortezomib is considered relatively safe for the protocols assessed in the study.
Collapse
Affiliation(s)
- Callan D. Wesley
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences University of Antwerp, Campus Drie Eiken, 2610 Antwerp, Belgium; (A.S.); (C.H.G.N.); (D.N.K.); (G.R.Y.D.M.); (W.M.); (P.-J.G.)
| | | | | | | | | | | | | |
Collapse
|
6
|
Krüger DN, Bosman M, Van Craenenbroeck EM, De Meyer GRY, Franssen C, Guns PJ. Dexrazoxane prevents vascular toxicity in doxorubicin-treated mice. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:65. [PMID: 39367508 PMCID: PMC11451066 DOI: 10.1186/s40959-024-00270-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Doxorubicin (DOX) is used for breast cancer and lymphoma, but can cause cardiotoxicity, arterial stiffness, and endothelial dysfunction. We recently reported SERPINA3N as biomarker of cardiovascular toxicity in patients and mice. Dexrazoxane (DEXRA) is an FDA-approved drug that prevents DOX-induced cardiac toxicity in high-risk patients. However, the effect of DEXRA on vascular dysfunction during DOX treatment has not been documented. Therefore, here we investigated whether DEXRA protects against DOX-induced arterial stiffness, endothelial dysfunction, and SERPINA3N upregulation in tissue and plasma from mice. METHODS Male C57BL6/J mice were treated with DOX (4 mg/kg), DEXRA (40 mg/kg), a combination (DEXRA + DOX), or VEHICLE (0.9% NaCl) weekly i.p. for 6 weeks (n = 8 per group). Cardiovascular function was measured in vivo by ultrasound imaging at baseline, weeks 2 and 6. Vascular reactivity was analyzed ex vivo in the thoracic aorta at week 6 and molecular analysis was performed. RESULTS DEXRA prevented left ventricular ejection fraction decline by DOX (DEXRA + DOX: 62 ± 2% vs DOX: 51 ± 2%). Moreover, DEXRA prevented the increase in pulse wave velocity by DOX (DEXRA + DOX: 2.1 ± 0.2 m/s vs DOX: 4.5 ± 0.3 m/s) and preserved endothelium-dependent relaxation (DEXRA + DOX: 82 ± 3% vs DOX: 62 ± 3%). In contrast to DOX-treated mice, SERPINA3N did not increase in the DEXRA + DOX group. CONCLUSION Our results not only confirm the cardioprotective effects of DEXRA against DOX-induced cardiotoxicity but also add preservation of vascular endothelial cell function as an important mechanism. Moreover, the study demonstrates the potential of SERPINA3N as a biomarker for monitoring cardiovascular complications of DOX in high-risk patients.
Collapse
Affiliation(s)
- Dustin N Krüger
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium.
| | - Matthias Bosman
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem, B-2650, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem, B-2650, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| |
Collapse
|
7
|
Zhang J, Sang X, Yuan Y, Shen J, Fang Y, Qin M, Zheng H, Zhu Z. 4-Deoxy- ε-Pyrromycinone: A Promising Drug/Lead Compound to Treat Tumors. Drug Des Devel Ther 2024; 18:2367-2379. [PMID: 38911033 PMCID: PMC11193465 DOI: 10.2147/dddt.s461594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024] Open
Abstract
Background Anthraquinone drugs are widely used in the treatment of tumors. However, multidrug resistance and severe cardiac toxicity limit its use, which have led to the discovery of new analogues. In this paper, 4-Deoxy-ε-pyrromycinone (4-Deo), belonging to anthraquinone compounds, was first been studied with the anti-tumor effects and the safety in vitro and in vivo as a new anti-tumor drug or lead compound. Methods The quantitative analysis of 4-Deo was established by UV methodology. The anti-cancer effect of 4-Deo in vitro was evaluated by cytotoxicity experiments of H22, HepG2 and Caco2, and the anti-cancer mechanism was explored by cell apoptosis and cycle. The tumor-bearing mouse model was established by subcutaneous inoculation of H22 cells to evaluate the anti-tumor effect of 4-Deo in vivo. The safety of 4-Deo was verified by the in vitro safety experiments of healthy cells and the in vivo safety experiments of H22 tumor-bearing mice. Tumor tissue sections were labeled with CRT, HMGB1, IL-6 and CD115 to explore the preliminary anti-cancer mechanism by immunohistochemistry. Results In vitro experiments demonstrated that 4-Deo could inhibit the growth of H22 by inducing cell necrosis and blocking cells in S phase, and 4-Deo has less damage to healthy cells. In vivo experiments showed that 4-Deo increased the positive area of CRT and HMGB1, which may inhibit tumor growth by triggering immunogenic cell death (ICD). In addition, 4-Deo reduced the positive area of CSF1R, and the anti-tumor effect may be achieved by blocking the transformation of tumor-associated macrophages (TAMs) to M2 phenotype. Conclusion In summary, this paper demonstrated the promise of 4-Deo for cancer treatment in vitro and in vivo. This paper lays the foundation for the study of 4-Deo, which is beneficial for the further development anti-tumor drugs based on the lead compound of 4-Deo.
Collapse
Affiliation(s)
- Jiping Zhang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Xianan Sang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Yichao Yuan
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Jiawei Shen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Yuanyuan Fang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Minjing Qin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Hangsheng Zheng
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| | - Zhihong Zhu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, 311402, People’s Republic of China
| |
Collapse
|
8
|
Shen H, Zhou W, ChunrongTu, Peng Y, Li X, Liu D, Wang X, Zhang X, Zeng X, Zhang J. Thoracic aorta injury detected by 4D flow MRI predicts subsequent main adverse cardiovascular events in breast cancer patients receiving anthracyclines: A longitudinal study. Magn Reson Imaging 2024; 109:67-73. [PMID: 38484947 DOI: 10.1016/j.mri.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/02/2024] [Accepted: 03/08/2024] [Indexed: 04/09/2024]
Abstract
PURPOSE To investigate longitudinal thoracic aorta injury using 3-dimensional phase-contrast magnetic resonance imaging (4D flow MRI) parameters and to evaluate their value for predicting the subsequent main adverse cardiovascular events (MACEs) in breast cancer patients receiving anthracyclines. METHODS Between July 2020 and July 2021, eighty-eight female participants with breast cancer scheduled to receive anthracyclines with or without trastuzumab prospectively enrolled. Each subjects underwent 4D flow MRI at baseline, 3 and 6 months in relation to baseline. The diameter, peak velocity (Vpeak), wall shear stress (WSS), pulse wave velocity (PWV), energy loss (EL) and pressure gradient (PG) of thoracic aorta were measured. The association between these parameters and subsequent MACEs was performed by Cox proportional hazard models. RESULTS Ten participants had subsequently MACEs. The Vpeak and PG gradually decreased and the WSS, PWV and EL progressively increased at 3 and 6 months compared with baseline. Adjusted multivariable analysis showed that the WSS of the proximal, mid- and distal ascending aorta [HR, 1.314 (95% confidence interval (CI): 1.003, 1.898)], [HR, 1.320 (95% CI: 1.002, 1.801)] and [HR, 1.322 (95% CI: 1.001, 1.805)] and PWV of ascending aorta [HR, 2.223 (95% CI: 1.010, 4.653)] at 3 months were associated with subsequent MACEs. Combined WSS and PWV of ascending aorta at 3 months yielded the highest AUC (0.912) for predicting subsequent MACEs. CONCLUSION Combined WSS and PWV of ascending aorta at 3 months is helpful for predicting the subsequent MACEs in breast cancer patients treated by anthracyclines.
Collapse
Affiliation(s)
- Hesong Shen
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Wenqi Zhou
- Department of Breast Cancer Center, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - ChunrongTu
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Yangling Peng
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Xiaoqin Li
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Daihong Liu
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Xiaoxia Wang
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Xiaoyong Zhang
- Clinical Science, Philips Healthcare, 1268 Tianfu Avenue, Hitech Zone, Chengdu, China
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China.
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China.
| |
Collapse
|
9
|
He J, Hou L, Liu Q, Zhou R. Irisin links Claudin-5 preservation and Mfn2-mediated mitochondrial dynamics to resist doxorubicin-induced cardiac endothelial damage. Biochem Biophys Res Commun 2024; 696:149501. [PMID: 38232667 DOI: 10.1016/j.bbrc.2024.149501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/19/2024]
Abstract
Irisin is protective in the cardiac microenvironment and can resist doxorubicin-induced cardiotoxicity. The purpose of this study was to investigate the connection between Irisin, endothelial cell integrity, and mitochondrial dynamics. Primary cardiac microvascular endothelial cells (CMECs) were used to explore the regulatory effects of Irisin on tight junction proteins, mitochondrial dynamics, β-catenin expression, and transcriptional activity. Results showed that Irisin can suppress doxorubicin-induced upregulation of MMP2 and MMP9, thereby reducing the degradation of tight junction proteins (ZO-1 and Claudin-5) and VE-cadherin. The preservation of Claudin-5 contributes to maintaining Mfn2 expression, which in turn supports mitochondrial fusion. Although Irisin restores doxorubicin-induced downregulation of β-catenin, it concurrently limits β-catenin transcriptional activity via Mfn2-mediated sulfenylation. Therefore, this study revealed a novel mechanism linking the protective effects of Irisin on the tight junction proteins and mitochondrial dynamics upon doxorubicin exposure.
Collapse
Affiliation(s)
- Jun He
- Department of Cardiology, Neijiang Second People's Hospital, Neijiang, 641000, China
| | - Lin Hou
- Department of Nursing, Neijiang Second People's Hospital, Neijiang, 641000, China
| | - Quanwei Liu
- Department of Cardiology, Neijiang Second People's Hospital, Neijiang, 641000, China
| | - Rui Zhou
- National Regional Children's Medical Center (Northwest); Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province; Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases; Xi'an Children's Hospital. Xi'an 710003, China.
| |
Collapse
|