1
|
Hong L, Cai X, Zhan Y, Liu S, Zou P, Chen Y, Shao L. TLR2 activates AP-1 to facilitate CTGF transcription and stimulate doxorubicin-induced myocardial injury. Br J Pharmacol 2025. [PMID: 40097259 DOI: 10.1111/bph.17423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/17/2024] [Accepted: 11/11/2024] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND AND PURPOSE Our study aimed to explore the mechanistic network of toll-like receptor 2 (TLR2)/activator protein-1 (AP-1) combined with SOX10 activation of the mitogen-activated protein kinase (MAPK) pathway via connective tissue growth factor (CTGF) in doxorubicin (Dox)-induced myocardial injury. EXPERIMENTAL APPROACH Rats with Dox-induced myocardial injury were treated with a TLR2 inhibitor or CTGF silencing lentiviral vector. H9c2 cells were treated with genetic vectors or MAPK pathway activators. Cardiac function was tested using echocardiography and serum markers. H&E, Sirius red and TUNEL staining were used to detect myocardial pathological changes, collagen accumulation and apoptosis. Western blot was used to detect proteins related to cardiac hypertrophy, fibrosis, apoptosis and the MAPK pathway. H9c2 cell injury was assessed by testing cell viability, lactate dehydrogenase (LDH) release and mitochondrial membrane potential. KEY RESULTS TLR2 and CTGF were highly expressed in patients with heart failure, and Dox treatment further increased their expression. Inhibiting TLR2 or silencing CTGF improved cardiac function and reduced myocardial fibrosis and apoptosis in Dox-treated rats. Silencing of TLR2 alleviated Dox-induced H9c2 cell injury, which was nullified by CTGF overexpression. TLR2 activated AP-1, which cooperated with SOX10 to promote CTGF transcription. MAPK activation aggravated H9c2 cells against Dox-induced injury. CONCLUSIONS AND IMPLICATIONS TLR2 activates AP-1 which cooperates with SOX10 to promote CTGF transcription and subsequently activate the MAPK pathway, thereby stimulating Dox-induced myocardial injury.
Collapse
Affiliation(s)
- Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xinyong Cai
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yuliang Zhan
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Songtao Liu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Pengtao Zou
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yanmei Chen
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
2
|
Shi X, Cao Y, Wang H, Zhao Q, Yan C, Li S, Jing L. Vaccarin Ameliorates Doxorubicin-Induced Cardiotoxicity via Inhibition of p38 MAPK Mediated Mitochondrial Dysfunction. J Cardiovasc Transl Res 2024; 17:1155-1171. [PMID: 38886316 PMCID: PMC11519163 DOI: 10.1007/s12265-024-10525-7] [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: 11/02/2023] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
Doxorubicin is a frequently used chemotherapeutic agent for treating various malignancies. However, it leads to severe cardiotoxic side effects, such as heart failure, and elevates the risk of sudden cardiac death among cancer patients. While oxidative stress has been identified as the primary cause of doxorubicin-induced cardiotoxicity, therapeutic antioxidant approaches have yielded unsatisfactory outcomes. The aim of this study is to explore the therapeutic potential of vaccarin, an active flavonoid glycoside extracted from traditional Chinese herbal agent Semen Vaccariae, in doxorubicin-induced cardiotoxicity. We observed that vaccarin significantly ameliorates doxorubicin-induced heart dysfunction in mouse model and suppresses oxidative stress mediated cell apoptosis via specifically inhibiting the activation of p38 MAPK pathway. In vitro, we observed that vaccarin alleviates doxorubicin-induced mitochondrial membrane depolarization and ROS generation in H9c2 cell, but the p38 MAPK agonist anisomycin reverses these effects. Our findings provide a promising natural antioxidant to protect against DOX-induced cardiotoxicity.
Collapse
MESH Headings
- Animals
- p38 Mitogen-Activated Protein Kinases/metabolism
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- Doxorubicin/toxicity
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Cardiotoxicity
- Oxidative Stress/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Apoptosis/drug effects
- Disease Models, Animal
- Cell Line
- Male
- Antioxidants/pharmacology
- Membrane Potential, Mitochondrial/drug effects
- Mice, Inbred C57BL
- Reactive Oxygen Species/metabolism
- Signal Transduction/drug effects
- Heart Diseases/chemically induced
- Heart Diseases/prevention & control
- Heart Diseases/pathology
- Heart Diseases/metabolism
- Heart Diseases/enzymology
- Rats
- Ventricular Function, Left/drug effects
- Glycosides/pharmacology
- Protein Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Xin Shi
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Yang Cao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Hongyu Wang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Qi Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Cong Yan
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Shengzhu Li
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China
| | - Ling Jing
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang Qu, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
3
|
Gao S, Qiu Y, Meng Y, Jia Y, Lang X, Zhao H, Sun H, Zhang J, Ding L. Blockage of PHLPP1 protects against myocardial ischemia/reperfusion injury in diabetic mice via activation of STAT3 signaling. J Bioenerg Biomembr 2023; 55:325-339. [PMID: 37584737 DOI: 10.1007/s10863-023-09977-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/14/2023] [Indexed: 08/17/2023]
Abstract
Diabetes can exacerbate myocardial ischemia/reperfusion (IR) injury. However, the sensitivity to IR injury and the underlying mechanisms in diabetic hearts remain unclear. Inhibition of PH domain leucine-rich repeating protein phosphatase (PHLPP1) could reduce myocardial IR injury, our previous study demonstrated that the expression of PHLPP1 was upregulated in diabetic myocardial IR model. Thus, this study aimed to investigate the mechanism of PHLPP1 in diabetic myocardial IR injury. Nondiabetic and diabetic C57BL/6 mice underwent 45 min of coronary artery occlusion followed by 2 h of reperfusion. Male C57BL/6 mice were injected with streptozotocin for five consecutive days to establish a diabetes model. H9c2 cells were exposed to normal or high glucose and subjected to 4 h of hypoxia followed by 4 h of reoxygenation. Diabetes or hyperglycemia increased postischemic infarct size, cellular injury, release of creatine kinase-MB, apoptosis, and oxidative stress, while exacerbating mitochondrial dysfunction. This was accompanied by enhanced expression of PHLPP1 and decreased levels of p-STAT3 and p-Akt. These effects were counteracted by PHLPP1 knockdown. Moreover, PHLPP1 knockdown resulted in an increase in mitochondrial translocation of p-STAT3 Ser727 and nuclear translocation of p-STAT3 Tyr705 and p-STAT3 Ser727. However, the effect of PHLPP1 knockdown in reducing posthypoxic cellular damage was nullified by either Stattic or LY294002. Additionally, a co-immunoprecipitation assay indicated a direct interaction between PHLPP1 and p-STAT3 Ser727, but not p-STAT3 Tyr705. The abnormal expression of PHLPP1 plays a significant role in exacerbating myocardial IR injury in diabetic mice. Knockdown of PHLPP1 to activate the STAT3 signaling pathway may represent a novel strategy for alleviating myocardial IR injury in diabetes.
Collapse
Affiliation(s)
- Sumin Gao
- Department of Emergency Medicine, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yun Qiu
- Department of Emergency Medicine, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yuming Meng
- Department of Emergency Medicine, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yajuan Jia
- Department of Emergency Medicine, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Xuemei Lang
- Department of Emergency Medicine, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Hongmei Zhao
- Department of Emergency Medicine, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Hong Sun
- Department of Emergency Medicine, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jinsong Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Lianshu Ding
- Department of Neurosurgery, The Affiliated Huaian NO.1 People's Hospital of Nanjing Medical University, Huai'an, China.
| |
Collapse
|
4
|
Xu X, Mao C, Zhang C, Zhang M, Gong J, Wang X. Salvianolic Acid B Inhibits Ferroptosis and Apoptosis during Myocardial Ischemia/Reperfusion Injury via Decreasing the Ubiquitin-Proteasome Degradation of GPX4 and the ROS-JNK/MAPK Pathways. Molecules 2023; 28:molecules28104117. [PMID: 37241859 DOI: 10.3390/molecules28104117] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Myocardial ischemia/reperfusion injury (MIRI) is related to ferroptosis and apoptosis elicited by reactive oxygen species (ROS). In this research, we investigated the protective effect of salvianolic acid B (SAB) as a natural antioxidant on ferroptosis and apoptosis in the MIRI process, and discussed the protective mechanism inhibiting ubiquitin-proteasome degradation of glutathione peroxidase 4 (GPX4) and the c-Jun N-terminal kinases (JNK) apoptosis signal pathway. We observed that ferroptosis and apoptosis occurred in the MIRI rat model in vivo and the H9c2 cardiomyocyte hypoxia/reoxygenation (H/R) damage model in vitro. SAB can alleviate tissue damage related to ROS, ferroptosis and apoptosis. Ubiquitin-proteasome degradation of GPX4 occurred in H/R models, and SAB reduced the ubiquitin-proteasome degradation of GPX4. SAB downregulates JNK phosphorylation and the expression of BCL2-Associated X (Bax)/B-cell lymphoma-2 (Bcl-2) and Caspase-3 to inhibit apoptosis. The role of GPX4 in the cardioprotection of SAB was further verified by the elimination effect of the GPX4 inhibitor RAS-selective lethal 3 (RSL3). This research shows that SAB may be used as a myocardial protective agent against oxidative stress, ferroptosis and apoptosis, and has potential clinical application prospects.
Collapse
Affiliation(s)
- Xiaojin Xu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Chenhan Mao
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Chengbo Zhang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Meng Zhang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianbin Gong
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xindong Wang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
5
|
Di Gregorio E, Romiti C, Di Lorenzo A, Cavallo F, Ferrauto G, Conti L. RGD_PLGA Nanoparticles with Docetaxel: A Route for Improving Drug Efficiency and Reducing Toxicity in Breast Cancer Treatment. Cancers (Basel) 2022; 15:cancers15010008. [PMID: 36612006 PMCID: PMC9817983 DOI: 10.3390/cancers15010008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer is the leading cause of cancer-related death in women. Although many therapeutic approaches are available, systemic chemotherapy remains the primary choice, especially for triple-negative and advanced breast cancers. Unfortunately, systemic chemotherapy causes serious side effects and requires high doses to achieve an effective concentration in the tumor. Thus, the use of nanosystems for drug delivery may overcome these limitations. Herein, we formulated Poly (lactic-co-glycolic acid) nanoparticles (PLGA-NPs) containing Docetaxel, a fluorescent probe, and a magnetic resonance imaging (MRI) probe. The cyclic RGD tripeptide was linked to the PLGA surface to actively target αvβ3 integrins, which are overexpressed in breast cancer. PLGA-NPs were characterized using dynamic light scattering, fast field-cycling 1H-relaxometry, and 1H-nuclear magnetic resonance. Their therapeutic effects were assessed both in vitro in triple-negative and HER2+ breast cancer cells, and in vivo in murine models. In vivo MRI and inductively coupled plasma mass spectrometry of excised tumors revealed a stronger accumulation of PLGA-NPs in the RGD_PLGA group. Targeted PLGAs have improved therapeutic efficacy and strongly reduced cardiac side effects compared to free Docetaxel. In conclusion, RGD-PLGA is a promising system for breast cancer treatment, with positive outcome in terms of therapeutic efficiency and reduction in side effects.
Collapse
Affiliation(s)
- Enza Di Gregorio
- Correspondence: (E.D.G.); (A.D.L.); Tel.: +39-011-6708459 (E.D.G.); +39-011-6706458 (A.D.L.)
| | | | - Antonino Di Lorenzo
- Correspondence: (E.D.G.); (A.D.L.); Tel.: +39-011-6708459 (E.D.G.); +39-011-6706458 (A.D.L.)
| | | | | | | |
Collapse
|
6
|
Hum NR, Sebastian A, Martin KA, Rios-Arce ND, Gilmore SF, Gravano DM, Wheeler EK, Coleman MA, Loots GG. IL-17A Increases Doxorubicin Efficacy in Triple Negative Breast Cancer. Front Oncol 2022; 12:928474. [PMID: 35924165 PMCID: PMC9340269 DOI: 10.3389/fonc.2022.928474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
Due to lack of targetable receptors and intertumoral heterogeneity, triple negative breast cancer (TNBC) remains particularly difficult to treat. Doxorubicin (DOX) is typically used as nonselective neoadjuvant chemotherapy, but the diversity of treatment efficacy remains unclear. Comparable to variability in clinical response, an experimental model of TNBC using a 4T1 syngeneic mouse model was found to elicit a differential response to a seven-day treatment regimen of DOX. Single-cell RNA sequencing identified an increase in T cells in tumors that responded to DOX treatment compared to tumors that continued to grow uninhibited. Additionally, compared to resistant tumors, DOX sensitive tumors contained significantly more CD4 T helper cells (339%), γδ T cells (727%), Naïve T cells (278%), and activated CD8 T cells (130%). Furthermore, transcriptional profiles of tumor infiltrated T cells in DOX responsive tumors revealed decreased exhaustion, increased chemokine/cytokine expression, and increased activation and cytotoxic activity. γδ T cell derived IL-17A was identified to be highly abundant in the sensitive tumor microenvironment. IL-17A was also found to directly increase sensitivity of TNBC cells in combination with DOX treatment. In TNBC tumors sensitive to DOX, increased IL-17A levels lead to a direct effect on cancer cell responsiveness and chronic stimulation of tumor infiltrated T cells leading to improved chemotherapeutic efficacy. IL-17A’s role as a chemosensitive cytokine in TNBC may offer new opportunities for treating chemoresistant breast tumors and other cancer types.
Collapse
Affiliation(s)
- Nicholas R. Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- Stem Cell Instrumentation Foundry, University of California Merced, Merced, CA, United States
| | - Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Kelly A. Martin
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Naiomy D. Rios-Arce
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Sean F. Gilmore
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - David M. Gravano
- Stem Cell Instrumentation Foundry, University of California Merced, Merced, CA, United States
| | - Elizabeth K. Wheeler
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Matthew A. Coleman
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- Department of Radiation Oncology, University of California Davis, Sacramento, CA, United States
| | - Gabriela G. Loots
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- Stem Cell Instrumentation Foundry, University of California Merced, Merced, CA, United States
- *Correspondence: Gabriela G. Loots,
| |
Collapse
|
7
|
Paeonol protects against doxorubicin-induced cardiotoxicity by promoting Mfn2-mediated mitochondrial fusion through activating the PKCε-Stat3 pathway. J Adv Res 2022; 47:151-162. [PMID: 35842187 PMCID: PMC10173194 DOI: 10.1016/j.jare.2022.07.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/13/2022] [Accepted: 07/10/2022] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION The anti-cancer medication doxorubicin (Dox) is largely restricted in clinical usage due to its significant cardiotoxicity. The only medication approved by the FDA for Dox-induced cardiotoxicity is dexrazoxane, while it may reduce the sensitivity of cancer cells to chemotherapy and is restricted for use. There is an urgent need for the development of safe and effective medicines to alleviate Dox-induced cardiotoxicity. OBJECTIVES The objective of this study was to determine whether Paeonol (Pae) has the ability to protect against Dox-induced cardiotoxicity and if so, what are the underlying mechanisms involved. METHODS Sprague-Dawley rats and primary cardiomyocytes were used to create Dox-induced cardiotoxicity models. Pae's effects on myocardial damage, mitochondrial function, mitochondrial dynamics and signaling pathways were studied using a range of experimental methods. RESULTS Pae enhanced Mfn2-mediated mitochondrial fusion, restored mitochondrial function and cardiac performance both in vivo and in vitro under the Dox conditions. The protective properties of Pae were blunted when Mfn2 was knocked down or knocked out in Dox-induced cardiomyocytes and hearts respectively. Mechanistically, Pae promoted Mfn2-mediated mitochondria fusion by activating the transcription factor Stat3, which bound to the Mfn2 promoter in a direct manner and up-regulated its transcriptional expression. Furthermore, molecular docking, surface plasmon resonance and co-immunoprecipitation studies showed that Pae's direct target was PKCε, which interacted with Stat3 and enabled its phosphorylation and activation. Pae-induced Stat3 phosphorylation and Mfn2-mediated mitochondrial fusion were inhibited when PKCε was knocked down. Furthermore, Pae did not interfere with Dox's antitumor efficacy in several tumor cells. CONCLUSION Pae protects the heart against Dox-induced damage by stimulating mitochondrial fusion via the PKCε-Stat3-Mfn2 pathway, indicating that Pae might be a promising therapeutic therapy for Dox-induced cardiotoxicity while maintaining Dox's anticancer activity.
Collapse
|
8
|
Lérida-Viso A, Estepa-Fernández A, Morellá-Aucejo Á, Lozano-Torres B, Alfonso M, Blandez JF, Bisbal V, Sepúlveda P, García-Fernández A, Orzáez M, Martínez-Máñez R. Pharmacological senolysis reduces doxorubicin-induced cardiotoxicity and improves cardiac function in mice. Pharmacol Res 2022; 183:106356. [PMID: 35843569 DOI: 10.1016/j.phrs.2022.106356] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 01/10/2023]
Abstract
Many anticancer agents used in clinics induce premature senescence in healthy tissues generating accelerated aging processes and adverse side-effects in patients. Cardiotoxicity is a well-known limiting factor of anticancer treatment with doxorubicin (DOX), a very effective anthracycline widely used as antitumoral therapy in clinical practice, that leads to long-term morbidity and mortality. DOX exposure severely affects the population of cardiac cells in both mice and human hearts by inducing premature senescence, which may represent the molecular basis of DOX-induced cardiomyopathy. Here, we demonstrate that senescence induction in the heart contributes to impaired cardiac function in mice upon DOX treatment. Concomitant elimination of senescent cells with the senolytic Navitoclax in different formulations produces a significant decrease in senescence and cardiotoxicity markers together with the restoration of the cardiac function in mice followed by echocardiography. These results evidence the potential clinical use of senolytic therapies to alleviate cardiotoxicities induced in chemotherapy-treated patients.
Collapse
Affiliation(s)
- Araceli Lérida-Viso
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València (UPV), Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Valencia 46026, Spain; Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Alejandra Estepa-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Ángela Morellá-Aucejo
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Beatriz Lozano-Torres
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - María Alfonso
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain
| | - Juan F Blandez
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València (UPV), Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Valencia 46026, Spain; Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Viviana Bisbal
- Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Av. Fernando Abril Martorell 106, Valencia 46026, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Alba García-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Mar Orzáez
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain.
| | - Ramón Martínez-Máñez
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València (UPV), Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Valencia 46026, Spain; Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
9
|
RRM2 Alleviates Doxorubicin-Induced Cardiotoxicity through the AKT/mTOR Signaling Pathway. Biomolecules 2022; 12:biom12020299. [PMID: 35204799 PMCID: PMC8869767 DOI: 10.3390/biom12020299] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic agent that plays an unparalleled role in cancer treatment. However, its serious dose-dependent cardiotoxicity, which eventually contributes to irreversible heart failure, has greatly limited the widespread clinical application of DOX. A previous study has demonstrated that the ribonucleotide reductase M2 subunit (RRM2) exerts salutary effects on promoting proliferation and inhibiting apoptosis and autophagy. However, the specific function of RRM2 in DOX-induced cardiotoxicity is yet to be determined. This study aimed to elucidate the role and potential mechanism of RRM2 on DOX-induced cardiotoxicity by investigating neonatal primary cardiomyocytes and mice treated with DOX. Subsequently, the results indicated that RRM2 expression was significantly reduced in mice hearts and primary cardiomyocytes. Apoptosis and autophagy-related proteins, such as cleaved-Caspase3 (C-Caspase3), LC3B, and beclin1, were distinctly upregulated. Additionally, RRM2 deficiency led to increased autophagy and apoptosis in cells. RRM2 overexpression, on the contrary, alleviated DOX-induced cardiotoxicity in vivo and in vitro. Consistently, DIDOX, an inhibitor of RRM2, attenuated the protective effect of RRM2. Mechanistically, we found that AKT/mTOR inhibitors could reverse the function of RRM2 overexpression on DOX-induced autophagy and apoptosis, which means that RRM2 could have regulated DOX-induced cardiotoxicity through the AKT/mTOR signaling pathway. In conclusion, our experiment established that RRM2 could be a potential treatment in reversing DOX-induced cardiac dysfunction.
Collapse
|
10
|
Evaluation of the Effect of Crocin on Doxorubicin-Induced Cardiotoxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1328:143-153. [PMID: 34981476 DOI: 10.1007/978-3-030-73234-9_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite newer advances in cancer treatment, chemotherapy is still one of the most widely used treatment strategies in this field. However, this treatment strategy faces major challenges. Doxorubicin (Dox) is an effective chemotherapeutic agent used to treat various cancers. However, several studies have shown that the use of Dox in therapeutic concentrations is associated with serious side effects, such as cardiac toxicity. The use of natural products in combination with chemotherapeutic agents to reduce side effects is a novel approach, and several studies have shown promising results. In this regard, we examined the effect of Crocin on doxorubicin-induced cardiotoxicity in rat and H9c2 cell line. The in vitro model on H9C2 cells and the in vivo models on rats were treated with doxorubicin. Cell viability, DNA damage, and apoptosis were measured in H9C2 cell line in the presence and absence of Crocin. Oxidative stress and various inflammatory parameters, as well as cardiac function tests, also were assessed in doxorubicin-induced cardiotoxicity animal model in the presence and absence of Crocin. Our results showed that Crocin can significantly decrease apoptosis in H9C2 cell line through a reduction in ROS production and DNA damages. Moreover, evaluation of the effect of Crocin on doxorubicin-induced cardiotoxicity animal model showed that Crocin also can significantly reduce oxidative stress and inflammatory parameters in the serum of the animals. Assessment of cardiac function revealed that Crocin has a significant protective effect against doxorubicin-induced cardiotoxicity in the animal model. Our data indicate that Crocin significantly attenuated doxorubicin-induced cardiotoxicity. Hence, Crocin could be potentially used as an adjuvant treatment in combination with Dox to reduce cardiotoxicity.
Collapse
|
11
|
Desole C, Gallo S, Vitacolonna A, Vigna E, Basilico C, Montarolo F, Zuppini F, Casanova E, Miggiano R, Ferraris DM, Bertolotto A, Comoglio PM, Crepaldi T. Engineering, Characterization, and Biological Evaluation of an Antibody Targeting the HGF Receptor. Front Immunol 2021; 12:775151. [PMID: 34925346 PMCID: PMC8679783 DOI: 10.3389/fimmu.2021.775151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The Hepatocyte growth factor (HGF) and its receptor (MET) promote several physiological activities such as tissue regeneration and protection from cell injury of epithelial, endothelial, neuronal and muscle cells. The therapeutic potential of MET activation has been scrutinized in the treatment of acute tissue injury, chronic inflammation, such as renal fibrosis and multiple sclerosis (MS), cardiovascular and neurodegenerative diseases. On the other hand, the HGF-MET signaling pathway may be caught by cancer cells and turned to work for invasion, metastasis, and drug resistance in the tumor microenvironment. Here, we engineered a recombinant antibody (RDO24) and two derived fragments, binding the extracellular domain (ECD) of the MET protein. The antibody binds with high affinity (8 nM) to MET ECD and does not cross-react with the closely related receptors RON nor with Semaphorin 4D. Deletion mapping studies and computational modeling show that RDO24 binds to the structure bent on the Plexin-Semaphorin-Integrin (PSI) domain, implicating the PSI domain in its binding to MET. The intact RDO24 antibody and the bivalent Fab2, but not the monovalent Fab induce MET auto-phosphorylation, mimicking the mechanism of action of HGF that activates the receptor by dimerization. Accordingly, the bivalent recombinant molecules induce HGF biological responses, such as cell migration and wound healing, behaving as MET agonists of therapeutic interest in regenerative medicine. In vivo administration of RDO24 in the murine model of MS, represented by experimental autoimmune encephalomyelitis (EAE), delays the EAE onset, mitigates the early clinical symptoms, and reduces inflammatory infiltrates. Altogether, these results suggest that engineered RDO24 antibody may be beneficial in multiple sclerosis and possibly other types of inflammatory disorders.
Collapse
Affiliation(s)
- Claudia Desole
- Department of Oncology, University of Turin, Candiolo, Italy
| | - Simona Gallo
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Annapia Vitacolonna
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Elisa Vigna
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Francesca Montarolo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | | | | | - Riccardo Miggiano
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy.,IXTAL srl, Novara, Italy
| | - Davide Maria Ferraris
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy.,IXTAL srl, Novara, Italy
| | | | | | - Tiziana Crepaldi
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| |
Collapse
|
12
|
Riaud M, Hilairet G, Sindji L, Perdomo L, Montero-Menei CN, Martinez MC. Pharmacology active microcarriers delivering HGF associated with extracellular vesicles for myocardial repair. Eur J Pharm Biopharm 2021; 169:268-279. [PMID: 34748934 DOI: 10.1016/j.ejpb.2021.10.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/19/2021] [Accepted: 10/27/2021] [Indexed: 01/20/2023]
Abstract
Despite the curative approaches developed against myocardial infarction, cardiac cell death causes dysfunctional heart contractions that depend on the extent of the ischemic area and the reperfusion period. Cardiac regeneration may allow neovascularization and limit the ventricular remodeling caused by the scar tissue. We have previously found that large extracellular vesicles, carrying Sonic Hedgehog (lEVs), displayed proangiogenic and antioxidant properties, and decreased myocardial infarction size when administrated by intravenous injection. We propose to associate lEVs with pharmacology active microcarriers (PAMs) to obtain a combined cardioprotective and regenerative action when administrated by intracardiac injection. PAMs made of poly-D,L-lactic-coglycolic acid-poloxamer 188-poly-D,L-lactic-coglycolic acid and covered by fibronectin/poly-D-lysine provided a biodegradable and biocompatible 3D biomimetic support for the lEVs. When compared with lEVs alone, lEVs-PAMs constructs possessed an enhanced in vitro pro-angiogenic ability. PAMs were designed to continuously release encapsulated hepatocyte growth factor (PAMsHGF) and thus, locally increase the activity of the lEVs by the combined anti-fibrotic properties and regenerative properties. Intracardiac administration of either lEVs alone or lEVs-PAMsHGF improved cardiac function in a similar manner, in a rat model of ischemia-reperfusion. Moreover, lEVs alone or the IEVs-PAMsHGF induced arteriogenesis, but only the latter reduced tissue fibrosis. Taken together, these results highlight a promising approach for lEVs-PAMsHGF in regenerative medicine for myocardial infarction.
Collapse
Affiliation(s)
- Melody Riaud
- SOPAM, U1063, INSERM, UNIV Angers, SFR ICAT, Angers, France; CRCINA, UMR 1232, INSERM, Université de Nantes, Université d'Angers, F-49933 Angers, France
| | | | - Laurence Sindji
- CRCINA, UMR 1232, INSERM, Université de Nantes, Université d'Angers, F-49933 Angers, France
| | | | - Claudia N Montero-Menei
- CRCINA, UMR 1232, INSERM, Université de Nantes, Université d'Angers, F-49933 Angers, France.
| | | |
Collapse
|
13
|
Adhikari A, Asdaq SMB, Al Hawaj MA, Chakraborty M, Thapa G, Bhuyan NR, Imran M, Alshammari MK, Alshehri MM, Harshan AA, Alanazi A, Alhazmi BD, Sreeharsha N. Anticancer Drug-Induced Cardiotoxicity: Insights and Pharmacogenetics. Pharmaceuticals (Basel) 2021; 14:ph14100970. [PMID: 34681194 PMCID: PMC8539940 DOI: 10.3390/ph14100970] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022] Open
Abstract
The advancement in therapy has provided a dramatic improvement in the rate of recovery among cancer patients. However, this improved survival is also associated with enhanced risks for cardiovascular manifestations, including hypertension, arrhythmias, and heart failure. The cardiotoxicity induced by chemotherapy is a life-threatening consequence that restricts the use of several chemotherapy drugs in clinical practice. This article addresses the prevalence of cardiotoxicity mediated by commonly used chemotherapeutic and immunotherapeutic agents. The role of susceptible genes and radiation therapy in the occurrence of cardiotoxicity is also reviewed. This review also emphasizes the protective role of antioxidants and future perspectives in anticancer drug-induced cardiotoxicities.
Collapse
Affiliation(s)
- Archana Adhikari
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Syed Mohammed Basheeruddin Asdaq
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Dariyah, Riyadh 13713, Saudi Arabia
- Correspondence: (S.M.B.A.); (M.C.)
| | - Maitham A. Al Hawaj
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Hofuf 31982, Saudi Arabia;
| | - Manodeep Chakraborty
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
- Correspondence: (S.M.B.A.); (M.C.)
| | - Gayatri Thapa
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Nihar Ranjan Bhuyan
- Department of Pharmaceutical Analysis, Himalayan Pharmacy Institute, Majhitar, Rangpo 737136, Sikkim, India;
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia;
| | | | - Mohammed M. Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh 11426, Saudi Arabia;
| | - Aishah Ali Harshan
- Department of Pharmaceutical Care, Northern Area Armed Forces Hospital, King Khalid Military City Hospital, Hafr Al-Batin 39745, Saudi Arabia;
| | - Abeer Alanazi
- Department of Pharmaceutical Care, First Health Cluster in Eastern Province, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia;
| | | | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa-31982, Saudi Arabia;
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bengaluru 560035, Karnataka, India
| |
Collapse
|
14
|
Regulatory role of endogenous and exogenous fibroblast growth factor 1 in the cardiovascular system and related diseases. Pharmacol Res 2021; 169:105596. [PMID: 33831565 DOI: 10.1016/j.phrs.2021.105596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/07/2021] [Accepted: 03/31/2021] [Indexed: 12/15/2022]
Abstract
Fibroblast growth factor 1 (FGF1) has a critical regulatory role in the development of the cardiovascular system (CVS) and is strongly associated with the progression or treatment of cardiovascular diseases (CVDs). However, the regulatory mechanisms of FGF1 in CVS and CVDs have not yet been fully elucidated. Therefore, this review article summarized the existing literature reports on the role of FGF1 in CVS under physiological and pathological conditions. First, the expression and physiological functions of endogenous FGF1 is fully demonstrated. Then, we analyzed the role of exogenous FGF1 in normal CVS and related pathological processes. Specifically, the potential signaling pathways might be mediated by FGF1 in CVDs treatment is discussed in detail. In addition, the barriers and feasible solutions for the application of FGF1 are further analyzed. Finally, we highlight therapeutic considerations of FGF1 for CVDs in the future. Thus, this article may be as a reference to provide some ideas for the follow-up research.
Collapse
|
15
|
Ren Y, Yuan B, Hou S, Sui Y, Yang T, Lv M, Zhou Y, Yu H, Li S, Peng H, Chang N, Liu Y. Delivery of RGD-modified liposome as a targeted colorectal carcinoma therapy and its autophagy mechanism. J Drug Target 2021; 29:863-874. [PMID: 33507113 DOI: 10.1080/1061186x.2021.1882469] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Liposomes are among the most extensively applied drug carriers due to their excellent biocompatibility, controllable size and ease of modification. In the present study, we prepared untargeted liposomes (LP) and targeting liposomes modified with Arg-Gly-Asp (RGD-LP), and Doxorubicin Hydrochloride (DOX) or fluorescent probe was loaded. RGD-LP/DOX was identified to be uniformly spherical in size 131.2 ± 2.7 nm. Based on flow cytometry analysis and the confocal laser scanning microscopy, RGD-LP had a higher uptake into HRT-18 colorectal cancer cells than LP. Further, in vivo imaging study further suggested that RGD-LP could significantly increase the liposome accumulation in the tumour tissues of the mice bearing subcutaneous tumours. By investigating the targeting mechanism of RGD-LP, we found that they entered the cell via macropinocytosis. When loaded with DOX, RGD-LP exerted stronger tumour growth inhibitory activity against tumours of colorectal carcinoma compared to LP. Moreover, RGD-LP induced autophagy. Therefore, RGD-LP have the potential to be applied as a targeted colorectal carcinoma therapy.
Collapse
Affiliation(s)
- Yachao Ren
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Bingchuan Yuan
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Shenghua Hou
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Yilei Sui
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Tinghui Yang
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Meilin Lv
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Yulong Zhou
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hui Yu
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Sen Li
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Haisheng Peng
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Naidan Chang
- Department for a affiliation.Department of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Yang Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Ye J, Wang Y, Xu Y, Wang Z, Liu L, Wang M, Ye D, Zhang J, Yang Z, Lin Y, Ji Q, Wan J. Interleukin-22 deficiency alleviates doxorubicin-induced oxidative stress and cardiac injury via the p38 MAPK/macrophage/Fizz3 axis in mice. Redox Biol 2020; 36:101636. [PMID: 32863209 PMCID: PMC7371904 DOI: 10.1016/j.redox.2020.101636] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/26/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023] Open
Abstract
Several interleukin (IL) family members have been demonstrated to be involved in doxorubicin (DOX)-induced cardiac injury. This study aimed to investigate the role of IL-22 in DOX-induced cardiac injury and explore its possible mechanisms. In this study, mice were given DOX, and the cardiac expression and sources of IL-22 were determined. Then, IL-22 was knocked out to observe the effects on DOX-induced cardiac injury in mice. In addition, the p38 mitogen-activated protein kinase (MAPK) pathway was inhibited, macrophages were depleted and adoptively transferred, and Fizz3 was up-regulated in mice to explore the mechanisms. The results showed that cardiac IL-22 expression was significantly increased by DOX treatment and was mostly derived from cardiac macrophages. IL-22 knockout significantly reduced cardiac vacuolization and the expression of cardiomyocyte injury markers in both serum and left ventricular tissue and improved cardiac function in DOX-treated mice. In addition, IL-22 knockout reversed DOX-induced cardiac M1 macrophage/M2 macrophage imbalance, reduced oxidative stress and protected against cardiomyocyte apoptosis. p38 MAPK pathway inhibition with SB203580 and macrophage depletion further alleviated the above effects in DOX-treated IL-22-knockout mice. The effects were stronger IL-22-knockout mice with adoptive transfer of WT macrophages than in those with adoptive transfer of IL-22-knockout macrophages. Furthermore, increasing the expression of Fizz3 reduced cardiomyocyte apoptosis and alleviated cardiac dysfunction. Our results may suggest that IL-22 knockout alleviate DOX-induced oxidative stress and cardiac injury by inhibiting macrophage differentiation and thereby increasing the expression of Fizz3. Reductions in IL-22 expression may be beneficial for clinical chemotherapy in tumor patients.
Collapse
Affiliation(s)
- Jing Ye
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China; Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Yuan Wang
- Department of Thyroid Breast Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Ling Liu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Zicong Yang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Yingzhong Lin
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530021, China.
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China.
| |
Collapse
|
17
|
The Long-Lasting Protective Effect of HGF in Cardiomyoblasts Exposed to Doxorubicin Requires a Positive Feed-Forward Loop Mediated by Erk1,2-Timp1-Stat3. Int J Mol Sci 2020; 21:ijms21155258. [PMID: 32722178 PMCID: PMC7432797 DOI: 10.3390/ijms21155258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/16/2022] Open
Abstract
Previous studies showed that the hepatocyte growth factor (HGF)–Met receptor axis plays long-lasting cardioprotection against doxorubicin anti-cancer therapy. Here, we explored the mechanism(s) underlying the HGF protective effect. DNA damage was monitored by histone H2AX phosphorylation and apoptosis by proteolytic cleavage of caspase 3. In doxorubicin-treated H9c2 cardiomyoblasts, the long-lasting cardioprotection is mediated by activation of the Ras/Raf/Mek/Erk (extracellular signal-regulated kinase 1,2) signaling pathway and requires Stat3 (signal transducer and activator of transcription 3) activation. The HGF protection was abrogated by the Erk1,2 inhibitor, PD98059. This translated into reduced Y705 phosphorylation and impaired nuclear translocation of Stat3, showing crosstalk between Erk1,2 and Stat3 signaling. An array of 29 cytokines, known to activate Stat3, was interrogated to identify the molecule(s) linking the two pathways. The analysis showed a selective increase in expression of the tissue inhibitor of metalloproteinases-1 (Timp1). Consistently, inhibition in cardiomyoblasts of Timp1 translation by siRNAs blunted both Stat3 activation and the cardioprotective effect of HGF. Thus, Timp1 is responsible for the generation of a feed-forward loop of Stat3 activation and helps cardiomyocytes to survive during the genotoxic stress induced by anthracyclines.
Collapse
|
18
|
Cristóbal I, Rubio J, Santos A, Luque M, Sanz-Alvarez M, Rojo F, García-Foncillas J. Therapeutic implications of the PP2A/MET signalling axis in doxorubicin-induced cardiotoxicity and antitumour properties. Br J Pharmacol 2020; 177:3850-3851. [PMID: 32535881 DOI: 10.1111/bph.15130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 04/30/2020] [Accepted: 05/09/2020] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain.,Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | - Jaime Rubio
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain.,Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | - Andrea Santos
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain.,Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | - Melani Luque
- Pathology Department, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | - Marta Sanz-Alvarez
- Pathology Department, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | - Federico Rojo
- Pathology Department, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| | - Jesús García-Foncillas
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, Madrid, Spain.,Translational Oncology Division, Oncohealth Institute, IIS-Fundacion Jimenez Diaz-UAM, University Hospital "Fundacion Jimenez Diaz", Madrid, Spain
| |
Collapse
|
19
|
Gallo S, Spilinga M, Albano R, Ferrauto G, Di Gregorio E, Casanova E, Balmativola D, Bonzano A, Boccaccio C, Sapino A, Comoglio PM, Crepaldi T. Activation of the MET receptor attenuates doxorubicin-induced cardiotoxicity in vivo and in vitro. Br J Pharmacol 2020; 177:3107-3122. [PMID: 32133617 PMCID: PMC7280013 DOI: 10.1111/bph.15039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Background and Purpose Doxorubicin anti‐cancer therapy is associated with cardiotoxicity, resulting from DNA damage response (DDR). Hepatocyte growth factor (HGF) protects cardiomyocytes from injury, but its effective use is compromised by low biodistribution. In this study, we have investigated whether the activation of the HGF receptor—encoded by the Met gene—by an agonist monoclonal antibody (mAb) could protect against doxorubicin‐induced cardiotoxicity. Experimental Approach The mAb (5 mg·kg−1) was injected in vivo into C57BL/6J mice, before doxorubicin (three doses of 7 mg·kg−1). Cardiac functions were evaluated through MRI after treatment termination. Heart histological staining and mRNA levels of genes associated with heart failure (Acta1 and Nppa), inflammation (IL‐6), and fibrosis (Ctgf, Col1a2, Timp1, and Mmp9) were assessed. MAb (100 nM) was administered in vitro to H9c2 cardiomyoblasts before addition of doxorubicin (25 μM). DDR and apoptosis markers were evaluated by quantitative western blotting, flow cytometry, and immunofluorescence. Stattic was used for pharmacological inactivation of STAT3. Key Results In vivo, administration of the mAb alleviated doxorubicin‐induced cardiac dysfunction and fibrosis. In vitro, mAb mimicked the response to HGF by (a) inhibiting histone H2AX phosphorylation at S139, (b) quenching the expression of the DNA repair enzyme PARP1, and (c) reducing the proteolytic activation of caspase 3. The MET‐driven cardioprotection involved, at least in vitro, the phosphorylation of STAT3. Conclusion and Implications The MET agonist mAb provides a new tool for cardioprotection against anthracycline cardiotoxicity.
Collapse
Affiliation(s)
- Simona Gallo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Martina Spilinga
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| | | | - Giuseppe Ferrauto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Enza Di Gregorio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Elena Casanova
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | | | | | - Carla Boccaccio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Tiziana Crepaldi
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|