1
|
Kim SK, Kim JH, Moon I, Min J, Park J, Kim MG. Cardioprotective effects of PARP Inhibitors: A meta-analysis of animal studies. Transl Res 2025; 280:29-40. [PMID: 40316011 DOI: 10.1016/j.trsl.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
Poly(adenosine diphosphate [ADP] ribose) polymerase (PARP) inhibitors are expected to provide benefits to the cardiovascular system. However, the cardioprotective effect of PARP inhibitors has not been systematically reviewed or quantitatively analyzed. This study aimed to assess the cardioprotective effects of PARP inhibitors through a meta-analysis of animal studies. Three databases PubMed, Web of Sciences, and Embase were searched until September 1, 2023. The risk of bias was assessed using SYRCLE's Risk of Bias. A total of 74 animal studies that investigated the cardiac function of PARP inhibitors compared to placebo or vehicle, were included. Outcome measures were hemodynamic indexes, cardiac contractility, and biomarkers of myocardial injury. Pooled effect size was estimated using a random-effects model with RevMan 5.4. PARP inhibitors were associated with enhanced hemodynamic indexes, including cardiac output (standardized mean difference, 0.86 [95 % CI, 0.54 to 1.17]; p < 0.00001) and stroke volume (0.42 [0.07 to 0.76]; p = 0.02). PARP inhibitors were associated with increased cardiac contractility, including ejection fraction (0.71 [0.42 to 1.01]; p < 0.00001) and fractional shortening (0.96 [0.62 to 1.31]; p < 0.00001). PARP inhibitors were associated with decreased troponin І (-1.42 [-2.16 to -0.68]; p = 0.0002), plasma B-type natriuretic peptide (-0.95 [-1.56 to -0.33]; p = 0.003), creatine kinase (-1.81 [-2.63 to -0.99]; p < 0.0001), and infarct size (-1.58 [-2.01 to -1.14]; p < 0.00001). PARP inhibitors improve cardiac functions and attenuate myocardial injury in animals, which indicate the cardioprotective effects. Further human studies are necessary.
Collapse
Affiliation(s)
- Seong Kyung Kim
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Jae Hyun Kim
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju, South Korea
| | - Inyeong Moon
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Jiwon Min
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Jieun Park
- College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Myeong Gyu Kim
- College of Pharmacy, Ewha Womans University, Seoul, South Korea; Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
2
|
Su S, Chen Z, Ke Q, Kocher O, Krieger M, Kang PM. Nanoparticle-Directed Antioxidant Therapy Can Ameliorate Disease Progression in a Novel, Diet-Inducible Model of Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2024; 44:2476-2488. [PMID: 39417229 PMCID: PMC11602363 DOI: 10.1161/atvbaha.124.321030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Oxidative stress plays a crucial role in the pathogenesis of coronary artery disease. In cardiovascular research using murine models, the generation and maintenance of models with robust coronary arterial atherosclerosis has been challenging. METHODS We characterized a new mouse model in which the last 3 amino acids of the carboxyl terminus of the HDL (high-density lipoprotein) receptor (SR-B1 [scavenger receptor, class B, type 1]) were deleted in a low-density lipoprotein receptor knockout (LDLR-/-) mouse model (SR-B1ΔCT/LDLR-/-) fed an atherogenic diet. We also tested the therapeutic effects of an oxidative stress-targeted nanoparticle in atherogenic diet-fed SR-B1ΔCT/LDLR-/- mice. RESULTS The SR-B1ΔCT/LDLR-/- mice fed an atherogenic diet had occlusive coronary artery atherosclerosis, impaired cardiac function, and a dramatically lower survival rate, compared with LDLR-/- mice fed the same diet. As SR-B1ΔCT/LDLR-/- mice do not exhibit female infertility or low pup yield, they are far easier and less costly to use than the previously described SR-B1-based models of coronary artery disease. We found that treatment with the targeted nanoparticles improved the cardiac functions and corrected hematologic abnormalities caused by the atherogenic diet in SR-B1ΔCT/LDLR-/- mice but did not alter the distinctive plasma lipid levels. CONCLUSIONS The SR-B1ΔCT/LDLR-/- mice developed diet-inducible, fatal atherosclerotic coronary artery disease, which could be ameliorated by targeted nanoparticle therapy. Our study provides new tools for the development of cardiovascular therapies.
Collapse
Affiliation(s)
- Shi Su
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zhifen Chen
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Qingen Ke
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Olivier Kocher
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peter M. Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Varkoly K, Beladi R, Hamada M, McFadden G, Irving J, Lucas AR. Viral SERPINS-A Family of Highly Potent Immune-Modulating Therapeutic Proteins. Biomolecules 2023; 13:1393. [PMID: 37759793 PMCID: PMC10526531 DOI: 10.3390/biom13091393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/03/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Serine protease inhibitors, SERPINS, are a highly conserved family of proteins that regulate serine proteases in the central coagulation and immune pathways, representing 2-10% of circulating proteins in the blood. Serine proteases form cascades of sequentially activated enzymes that direct thrombosis (clot formation) and thrombolysis (clot dissolution), complement activation in immune responses and also programmed cell death (apoptosis). Virus-derived serpins have co-evolved with mammalian proteases and serpins, developing into highly effective inhibitors of mammalian proteolytic pathways. Through interacting with extracellular and intracellular serine and cysteine proteases, viral serpins provide a new class of highly active virus-derived coagulation-, immune-, and apoptosis-modulating drug candidates. Viral serpins have unique characteristics: (1) function at micrograms per kilogram doses; (2) selectivity in targeting sites of protease activation; (3) minimal side effects at active concentrations; and (4) the demonstrated capacity to be modified, or fine-tuned, for altered protease targeting. To date, the virus-derived serpin class of biologics has proven effective in a wide range of animal models and in one clinical trial in patients with unstable coronary disease. Here, we outline the known viral serpins and review prior studies with viral serpins, considering their potential for application as new sources for immune-, coagulation-, and apoptosis-modulating therapeutics.
Collapse
Affiliation(s)
- Kyle Varkoly
- Department of Internal Medicine, McLaren Macomb Hospital, Michigan State University College of Human Medicine, 1000 Harrington St., Mt Clemens, MI 48043, USA;
| | - Roxana Beladi
- Department of Neurological Surgery, Ascension Providence Hospital, Michigan State University College of Human Medicine, 16001 W Nine Mile Rd., Southfield, MI 48075, USA;
| | - Mostafa Hamada
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA;
- Center for Immunotherapy Vaccines and Virotherapy, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| | - Grant McFadden
- Center for Immunotherapy Vaccines and Virotherapy, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| | - James Irving
- UCL Respiratory and the Institute of Structural and Molecular Biology, University College London, 5 University Street, London WC1E 6JF, UK
| | - Alexandra R. Lucas
- Center for Immunotherapy Vaccines and Virotherapy, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA
| |
Collapse
|
4
|
Han HH, Wang HM, Jangili P, Li M, Wu L, Zang Y, Sedgwick AC, Li J, He XP, James TD, Kim JS. The design of small-molecule prodrugs and activatable phototherapeutics for cancer therapy. Chem Soc Rev 2023; 52:879-920. [PMID: 36637396 DOI: 10.1039/d2cs00673a] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Cancer remains as one of the most significant health problems, with approximately 19 million people diagnosed worldwide each year. Chemotherapy is a routinely used method to treat cancer patients. However, current treatment options lack the appropriate selectivity for cancer cells, are prone to resistance mechanisms, and are plagued with dose-limiting toxicities. As such, researchers have devoted their attention to developing prodrug-based strategies that have the potential to overcome these limitations. This tutorial review highlights recently developed prodrug strategies for cancer therapy. Prodrug examples that provide an integrated diagnostic (fluorescent, photoacoustic, and magnetic resonance imaging) response, which are referred to as theranostics, are also discussed. Owing to the non-invasive nature of light (and X-rays), we have discussed external excitation prodrug strategies as well as examples of activatable photosensitizers that enhance the precision of photodynamic therapy/photothermal therapy. Activatable photosensitizers/photothermal agents can be seen as analogous to prodrugs, with their phototherapeutic properties at a specific wavelength activated in the presence of disease-related biomarkers. We discuss each design strategy and illustrate the importance of targeting biomarkers specific to the tumour microenvironment and biomarkers that are known to be overexpressed within cancer cells. Moreover, we discuss the advantages of each approach and highlight their inherent limitations. We hope in doing so, the reader will appreciate the current challenges and available opportunities in the field and inspire subsequent generations to pursue this crucial area of cancer research.
Collapse
Affiliation(s)
- Hai-Hao Han
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, P. R. China. .,State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China.,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, P. R. China
| | - Han-Min Wang
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Paramesh Jangili
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| | - Mingle Li
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| | - Luling Wu
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK.
| | - Yi Zang
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,Lingang laboratory, Shanghai 201203, China
| | - Adam C Sedgwick
- Chemistry Research Laboratory, University of Oxford, Mansfield Road, OX1 3TA, UK.
| | - Jia Li
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China.,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, P. R. China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, P. R. China. .,The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China.,National Center for Liver Cancer, Shanghai 200438, China
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK. .,School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
5
|
Fan X, Zhang Z, Zheng L, Wei W, Chen Z. Long non-coding RNAs in the pathogenesis of heart failure: A literature review. Front Cardiovasc Med 2022; 9:950284. [PMID: 35990951 PMCID: PMC9381960 DOI: 10.3389/fcvm.2022.950284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
Heart failure (HF) is a common cardiovascular disorder and a major cause of mortality and morbidity in older people. The mechanisms underlying HF are still not fully understood, restricting novel therapeutic target discovery and drug development. Besides, few drugs have been shown to improve the survival of HF patients. Increasing evidence suggests that long non-coding RNAs (lncRNAs) serve as a critical regulator of cardiac physiological and pathological processes, regarded as a new target of treatment for HF. lncRNAs are versatile players in the pathogenesis of HF. They can interact with chromatin, protein, RNA, or DNA, thereby modulating chromatin accessibility, gene expressions, and signaling transduction. In this review, we summarized the current knowledge on how lncRNAs involve in HF and categorized them into four aspects based on their biological functions, namely, cardiomyocyte contractility, cardiac hypertrophy, cardiac apoptosis, and myocardial fibrosis. Along with the extensive laboratory data, RNA-based therapeutics achieved great advances in recent years. These indicate that targeting lncRNAs in the treatment of HF may provide new strategies and address the unmet clinical needs.
Collapse
Affiliation(s)
- Xiaoyan Fan
- Postdoctoral Mobile Station of Shandong University of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Cardiovascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenwei Zhang
- Department of Urinary Surgery, No.3 People's Hospital, Jinan, China
| | - Liang Zheng
- Department of Cardiovascular Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Wei
- Postdoctoral Mobile Station of Wangjing Hospital, Wangjing Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
- *Correspondence: Wei Wei
| | - Zetao Chen
- Section of Integrated Chinese and Western Medicine, Shandong university of Traditional Chinese Medicine, Jinan, China
- Department of Geriatrics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Zetao Chen
| |
Collapse
|
6
|
Hu Y, Liang Y, Tian H, Xu C, Yu D, Zhang P, Ye H, Li M. Microplitis bicoloratus bracovirus regulates cyclophilin A-apoptosis-inducing factor interaction to induce cell apoptosis in the insect immunosuppressive process. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 110:e21877. [PMID: 35218062 PMCID: PMC9285338 DOI: 10.1002/arch.21877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 05/27/2023]
Abstract
Microplitis bicoloratus bracovirus (MbBV) induces apoptosis in hemocytes of the host (Spodoptera litura) via the cyclophilin A (CypA)-mediated signaling pathway. However, the mechanisms underlying CypA-mediated signaling during apoptosis remain largely unknown. Therefore, in this study, we investigated how CypA and apoptosis-inducing factor (AIF) interact during MbBV-mediated apoptosis. Our findings showed that MbBV induces apoptosis through the CypA-AIF axis of insect immune suppression. In MbBV-infected Spli221 cells, both the expression of the cypa gene and the release of AIF from the mitochondria increased the number of apoptotic cells. CypA and AIF underwent concurrent cytoplasm-nuclear translocation. Conversely, blocking of AIF release from mitochondria not only inhibited the CypA-AIF interaction but also inhibited the cytoplasmic-nuclear translocation of AIF and CypA. Importantly, the survival of the apoptotic phenotype was significantly rescued in MbBV-infected Spli221 cells. In addition, we found that the cyclosporine A-mediated inhibition of CypA did not prevent the formation of the CypA and AIF complex; rather, this only suppressed genomic DNA fragmentation. In vitro experiments revealed direct molecular interactions between recombinant CypA and AIF. Taken together, our results demonstrate that the CypA-AIF interaction plays an important role in MbBV-induced innate immune suppression. This study will help to clarify aspects of insect immunological mechanisms and will be relevant to biological pest control.
Collapse
Affiliation(s)
- Yan Hu
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Ya‐Ping Liang
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Hang‐Yu Tian
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Cui‐Xian Xu
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Dan Yu
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Pan Zhang
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Hui Ye
- School of Life SciencesYunnan UniversityKunmingChina
- School of AgricultureYunnan UniversityKunmingChina
| | - Ming Li
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| |
Collapse
|
7
|
The Effect of Thymus vulgaris on Hepatic Enzymes Activity and Apoptosis-Related Gene Expression in Streptozotocin-Induced Diabetic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2948966. [PMID: 35368767 PMCID: PMC8967521 DOI: 10.1155/2022/2948966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/04/2021] [Accepted: 03/03/2022] [Indexed: 11/17/2022]
Abstract
Many diseases, including diabetes, are involved in the development of liver disorders through changes in the expression of genes such as apoptosis-related genes. In the present study, the effect of Thymus vulgaris (T. vulgaris) on hepatic enzyme activity and apoptosis-related gene expression in streptozotocin (STZ)-induced diabetic rats was examined. In this study, 50 adult male Wistar rats weighing approximately 200–220 g were divided into five groups. Diabetes was induced by an intraperitoneal injection of STZ (60 mg/kg). Following 18 days, all the animals in different groups were weighed and blood samples were taken from their cardiac veins. Gas chromatography-mass spectrometry (GC-MS) analysis revealed 45 different compounds in the T. vulgaris, including thymol (39.1%), p-cymene (20.63%), and γ-terpinene (14.85%). The results showed a significant increase in liver enzymes (aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP)) in diabetic or STZ mice compared to the control group (healthy mice) (P < 0.0001). The levels of AST, ALT, and ALP in rats treated with 200 mg/kg and 400 mg/kg of T. vulgaris extract showed a significant decrease in these enzymes in comparison with diabetic rats (P < 0.0001). The expression of caspase 3 and 9 genes in the groups treated with thyme significantly decreased compared to diabetic mice (P < 0.0001), and the expression of B-cell lymphoma-2 (Bcl-2) in the group receiving 400 mg/kg of thyme significantly increased compared to diabetic mice (P < 0.0001). Due to its antioxidant compounds, thyme improves the liver tissue cells in STZ-induced diabetic mice by reducing caspases 3 and 9 as well as increasing Bcl-2.
Collapse
|
8
|
Adini A, Adini I, Grad E, Tal Y, Danenberg HD, Kang PM, Matthews BD, D’Amato RJ. The Prominin-1-Derived Peptide Improves Cardiac Function Following Ischemia. Int J Mol Sci 2021; 22:5169. [PMID: 34068392 PMCID: PMC8153573 DOI: 10.3390/ijms22105169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022] Open
Abstract
Myocardial infarction (MI) remains the leading cause of death in the western world. Despite advancements in interventional revascularization technologies, many patients are not candidates for them due to comorbidities or lack of local resources. Non-invasive approaches to accelerate revascularization within ischemic tissues through angiogenesis by providing Vascular Endothelial Growth Factor (VEGF) in protein or gene form has been effective in animal models but not in humans likely due to its short half-life and systemic toxicity. Here, we tested the hypothesis that PR1P, a small VEGF binding peptide that we developed, which stabilizes and upregulates endogenous VEGF, could be used to improve outcome from MI in rodents. To test this hypothesis, we induced MI in mice and rats via left coronary artery ligation and then treated animals with every other day intraperitoneal PR1P or scrambled peptide for 14 days. Hemodynamic monitoring and echocardiography in mice and echocardiography in rats at 14 days showed PR1P significantly improved multiple functional markers of heart function, including stroke volume and cardiac output. Furthermore, molecular biology and histological analyses of tissue samples showed that systemic PR1P targeted, stabilized and upregulated endogenous VEGF within ischemic myocardium. We conclude that PR1P is a potential non-invasive candidate therapeutic for MI.
Collapse
Affiliation(s)
- Avner Adini
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.D.M.); (R.J.D.)
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Irit Adini
- Department of Surgery, Harvard Medical School, The Center for Engineering in Medicine, Mass General Hospital, Shriners Hospitals for Children Boston, Boston, MA 02114, USA;
| | - Etty Grad
- Interventional Cardiology, Heart Institute, Hadassah Hebrew University Medical Center, Jerusalem 91200, Israel; (E.G.); (H.D.D.)
| | - Yuval Tal
- Allergy and Clinical Immunology Unit and Department of Medicine, Hadassah University Medical Center, Jerusalem 91200, Israel;
| | - Haim D. Danenberg
- Interventional Cardiology, Heart Institute, Hadassah Hebrew University Medical Center, Jerusalem 91200, Israel; (E.G.); (H.D.D.)
| | - Peter M. Kang
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Benjamin D. Matthews
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.D.M.); (R.J.D.)
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert J. D’Amato
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.D.M.); (R.J.D.)
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
9
|
Hassanpour M, Aghamohamadzade N, Cheraghi O, Heidarzadeh M, Nouri M. Current status of cardiac regenerative medicine; An update on point of view to cell therapy application. J Cardiovasc Thorac Res 2021; 12:256-268. [PMID: 33510874 PMCID: PMC7828760 DOI: 10.34172/jcvtr.2020.44] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 09/19/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally. Because of the economic and social burden of acute myocardial infarction and its chronic consequences in surviving patients, understanding the pathophysiology of myocardial infarction injury is a major priority for cardiovascular research. MI is defined as cardiomyocytes death caused by an ischemic that resulted from the apoptosis, necrosis, necroptosis, and autophagy. The phases of normal repair following MI including inflammatory, proliferation, and maturation. Normal repair is slow and inefficient generally so that other treatments are required. Because of difficulties, outcomes, and backwashes of traditional therapies including coronary artery bypass grafting, balloon angioplasty, heart transplantation, and artificial heart operations, the novel strategy in the treatment of MI, cell therapy, was newly emerged. In cell therapy, a new population of cells has created that substitute with damaged cells. Different types of stem cell and progenitor cells have been shown to improve cardiac function through various mechanisms, including the formation of new myocytes, endothelial cells, and vascular smooth muscle cells. Bone marrow- and/or adipose tissue-derived mesenchymal stem cells, embryonic stem cells, autologous skeletal myoblasts, induced pluripotent stem cells, endothelial progenitor cells, cardiac progenitor cells and cardiac pericytes considered as a source for cell therapy. In this study, we focused on the point of view of the cell sources.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Omid Cheraghi
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | | | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Yaron JR, Zhang L, Guo Q, Burgin M, Schutz LN, Awo E, Wise L, Krause KL, Ildefonso CJ, Kwiecien JM, Juby M, Rahman MM, Chen H, Moyer RW, Alcami A, McFadden G, Lucas AR. Deriving Immune Modulating Drugs from Viruses-A New Class of Biologics. J Clin Med 2020; 9:E972. [PMID: 32244484 PMCID: PMC7230489 DOI: 10.3390/jcm9040972] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Viruses are widely used as a platform for the production of therapeutics. Vaccines containing live, dead and components of viruses, gene therapy vectors and oncolytic viruses are key examples of clinically-approved therapeutic uses for viruses. Despite this, the use of virus-derived proteins as natural sources for immune modulators remains in the early stages of development. Viruses have evolved complex, highly effective approaches for immune evasion. Originally developed for protection against host immune responses, viral immune-modulating proteins are extraordinarily potent, often functioning at picomolar concentrations. These complex viral intracellular parasites have "performed the R&D", developing highly effective immune evasive strategies over millions of years. These proteins provide a new and natural source for immune-modulating therapeutics, similar in many ways to penicillin being developed from mold or streptokinase from bacteria. Virus-derived serine proteinase inhibitors (serpins), chemokine modulating proteins, complement control, inflammasome inhibition, growth factors (e.g., viral vascular endothelial growth factor) and cytokine mimics (e.g., viral interleukin 10) and/or inhibitors (e.g., tumor necrosis factor) have now been identified that target central immunological response pathways. We review here current development of virus-derived immune-modulating biologics with efficacy demonstrated in pre-clinical or clinical studies, focusing on pox and herpesviruses-derived immune-modulating therapeutics.
Collapse
Affiliation(s)
- Jordan R. Yaron
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Liqiang Zhang
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Qiuyun Guo
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Michelle Burgin
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Lauren N. Schutz
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Enkidia Awo
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Lyn Wise
- University of Otago, Dunedin 9054, New Zealand; (L.W.); (K.L.K.)
| | - Kurt L. Krause
- University of Otago, Dunedin 9054, New Zealand; (L.W.); (K.L.K.)
| | | | - Jacek M. Kwiecien
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S4L8, Canada
| | - Michael Juby
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Masmudur M. Rahman
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Hao Chen
- The Department of Tumor Surgery, Second Hospital of Lanzhou University, Lanzhou 730030, China;
| | - Richard W. Moyer
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA;
| | - Antonio Alcami
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Cantoblanco, 28049 Madrid, Spain;
| | - Grant McFadden
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA; (J.R.Y.); (L.Z.); (Q.G.); (M.B.); (L.N.S.); (E.A.); (M.J.)
- Center for Immunotherapy, Vaccines and Virotherapy, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA (G.M.)
- St Joseph Hospital, Dignity Health, Creighton University, Phoenix, AZ 85013, USA
| |
Collapse
|
11
|
Abstract
Anthracycline-based chemotherapy can result in the development of a cumulative and progressively developing cardiomyopathy. Doxorubicin is one of the most highly prescribed anthracyclines in the United States due to its broad spectrum of therapeutic efficacy. Interference with different mitochondrial processes is chief among the molecular and cellular determinants of doxorubicin cardiotoxicity, contributing to the development of cardiomyopathy. The present review provides the basis for the involvement of mitochondrial toxicity in the different functional hallmarks of anthracycline toxicity. Our objective is to understand the molecular determinants of a progressive deterioration of functional integrity of mitochondria that establishes a historic record of past drug treatments (mitochondrial memory) and renders the cancer patient susceptible to subsequent regimens of drug therapy. We focus on the involvement of doxorubicin-induced mitochondrial oxidative stress, disruption of mitochondrial oxidative phosphorylation, and permeability transition, contributing to altered metabolic and redox circuits in cardiac cells, ultimately culminating in disturbances of autophagy/mitophagy fluxes and increased apoptosis. We also suggest some possible pharmacological and nonpharmacological interventions that can reduce mitochondrial damage. Understanding the key role of mitochondria in doxorubicin-induced cardiomyopathy is essential to reduce the barriers that so dramatically limit the clinical success of this essential anticancer chemotherapy.
Collapse
Affiliation(s)
- Kendall B Wallace
- From the Department of Biomedical Sciences, University of Minnesota Medical School, Duluth (K.B.W.)
| | - Vilma A Sardão
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal (V.A.S., P.J.O.)
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal (V.A.S., P.J.O.)
| |
Collapse
|
12
|
Xu Z, Chen L, Xiao Z, Zhu Y, Jiang H, Jin Y, Gu C, Wu Y, Wang L, Zhang W, Zuo J, Zhou D, Luan J, Shen J. Potentiation of the anticancer effect of doxorubicinin drug-resistant gastric cancer cells by tanshinone IIA. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 51:58-67. [PMID: 30466628 DOI: 10.1016/j.phymed.2018.05.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/28/2018] [Accepted: 05/18/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Gastric cancer is the fifth commonest cancer and the third cause of cancer-related deaths all over the world. The effectiveness of chemotherapy is still limited by drug resistance in gastric cancer. Tanshinones, abietane diterpenes isolated from the traditional Chinese medicine Danshen (Salvia miltiorrhiza), have exhibited versatile anticancer activities in particular the ability to overcome drug resistance in different cancers. PURPOSE The current study aimed to explore the capacity of tanshinone IIA, the most abundant tanshinone found in the plant Danshen, to overcome drug resistance of gastric cancer cells to a commonly used anticancer drug doxorubicin. STUDY DESIGN Sensitivity of cell lines to doxorubicin was determined by MTT assay. Doxorubicin resistant gastric cancer cell lines was established by step selection with increasing concentrations of doxorubicin. Cell cycle arrest, apoptosis and doxorubicin efflux were analyzed by flow cytometry. The expression of MRP1 was determined by realtime PCR and western-blot. RESULTS Based on the IC50 values of doxorubicin, we identified the doxorubicin-sensitive gastric cancer cell lines SNU-719 and SNU-610 as well as the cell lines relatively resistant to doxorubicin including SNU-638, SNU-668, SNU-216 and SNU-620. We also established two drug-resistant cell lines SNU-719R and SNU-610R. Despite the fact that tanshinone IIA alone showed no cytotoxicity on these gastric cells, we found the potentiation of the anticancer effect of doxorubicin in drug-resistant gastric cancer cells by tanshinone IIA. Furthermore, using doxorubicin-sensitive cell line SNU-719 and doxorubicin-resistant cell lines SNU-719R and SNU-620, we revealed the pivotal roles of MRP1. Its overexpression impaired cell cycle arrest and suppressed apoptosis in the development of both intrinsic and acquired drug resistance in gastric cancer cells to doxorubicin. Importantly, inhibition of MRP1 function enhanced cell cycle arrest, increased apoptosis and induced autophagic cell death which contributed to the capability of tanshinone IIA to potentiate the anticancer effect of doxorubicin in drug-resistant gastric cancer cells. CONCLUSION Tanshinone IIA is an interesting agent with potential to treat drug-resistant gastric cancer in combination therapy.
Collapse
Affiliation(s)
- Zhenyu Xu
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Lu Chen
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yanhong Zhu
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Hui Jiang
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Yan Jin
- Department of Gastrointestinal Surgery, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Cheng Gu
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Yilai Wu
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Lin Wang
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Wen Zhang
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jian Zuo
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Dexi Zhou
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jiajie Luan
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China.
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
13
|
Kankeu C, Clarke K, Passante E, Huber HJ. Doxorubicin-induced chronic dilated cardiomyopathy-the apoptosis hypothesis revisited. J Mol Med (Berl) 2016; 95:239-248. [PMID: 27933370 DOI: 10.1007/s00109-016-1494-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/17/2016] [Accepted: 11/25/2016] [Indexed: 01/08/2023]
Abstract
The chemotherapeutic agent doxorubicin (DOX) has significantly increased survival rates of pediatric and adult cancer patients. However, 10% of pediatric cancer survivors will 10-20 years later develop severe dilated cardiomyopathy (DCM), whereby the exact molecular mechanisms of disease progression after this long latency time remain puzzling. We here revisit the hypothesis that elevated apoptosis signaling or its increased likelihood after DOX exposure can lead to an impairment of cardiac function and cause a cardiac dilation. Based on recent literature evidence, we first argue why a dilated phenotype can occur when little apoptosis is detected. We then review findings suggesting that mature cardiomyocytes are protected against DOX-induced apoptosis downstream, but not upstream of mitochondrial outer membrane permeabilisation (MOMP). This lack of MOMP induction is proposed to alter the metabolic phenotype, induce hypertrophic remodeling, and lead to functional cardiac impairment even in the absence of cardiomyocyte apoptosis. We discuss findings that DOX exposure can lead to increased sensitivity to further cardiomyocyte apoptosis, which may cause a gradual loss in cardiomyocytes over time and a compensatory hypertrophic remodeling after treatment, potentially explaining the long lag time in disease onset. We finally note similarities between DOX-exposed cardiomyocytes and apoptosis-primed cancer cells and propose computational system biology as a tool to predict patient individual DOX doses. In conclusion, combining recent findings in rodent hearts and cardiomyocytes exposed to DOX with insights from apoptosis signal transduction allowed us to obtain a molecularly deeper insight in this delayed and still enigmatic pathology of DCM.
Collapse
Affiliation(s)
- Cynthia Kankeu
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Kylie Clarke
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Egle Passante
- School of Pharmacy and Biomedical Sciences, Univ. of Central Lancashire, Preston, UK
| | - Heinrich J Huber
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium. .,Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| |
Collapse
|
14
|
Pal R, Ke Q, Pihan GA, Yesilaltay A, Penman ML, Wang L, Chitraju C, Kang PM, Krieger M, Kocher O. Carboxy-terminal deletion of the HDL receptor reduces receptor levels in liver and steroidogenic tissues, induces hypercholesterolemia, and causes fatal heart disease. Am J Physiol Heart Circ Physiol 2016; 311:H1392-H1408. [PMID: 27694217 DOI: 10.1152/ajpheart.00463.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/13/2016] [Indexed: 01/15/2023]
Abstract
The HDL receptor SR-BI mediates the transfer of cholesteryl esters from HDL to cells and controls HDL abundance and structure. Depending on the genetic background, loss of SR-BI causes hypercholesterolemia, anemia, reticulocytosis, splenomegaly, thrombocytopenia, female infertility, and fatal coronary heart disease (CHD). The carboxy terminus of SR-BI (505QEAKL509) must bind to the cytoplasmic adaptor PDZK1 for normal hepatic-but not steroidogenic cell-expression of SR-BI protein. To determine whether SR-BI's carboxy terminus is also required for normal protein levels in steroidogenic cells, we introduced into SR-BI's gene a 507Ala/STOP mutation that produces a truncated receptor (SR-BIΔCT). As expected, the dramatic reduction of hepatic receptor protein in SR-BIΔCT mice was similar to that in PDZK1 knockout (KO) mice. Unlike SR-BI KO females, SR-BIΔCT females were fertile. The severity of SR-BIΔCT mice's hypercholesterolemia was intermediate between those of SR-BI KO and PDZK1 KO mice. Substantially reduced levels of the receptor in adrenal cortical cells, ovarian cells, and testicular Leydig cells in SR-BIΔCT mice suggested that steroidogenic cells have an adaptor(s) functionally analogous to hepatic PDZK1. When SR-BIΔCT mice were crossed with apolipoprotein E KO mice (SR-BIΔCT/apoE KO), pathologies including hypercholesterolemia, macrocytic anemia, hepatic and splenic extramedullary hematopoiesis, massive splenomegaly, reticulocytosis, thrombocytopenia, and rapid-onset and fatal occlusive coronary arterial atherosclerosis and CHD (median age of death: 9 wk) were observed. These results provide new insights into the control of SR-BI in steroidogenic cells and establish SR-BIΔCT/apoE KO mice as a new animal model for the study of CHD.
Collapse
Affiliation(s)
- Rinku Pal
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Qingen Ke
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - German A Pihan
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Ayce Yesilaltay
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Marsha L Penman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Li Wang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Chandramohan Chitraju
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Peter M Kang
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Monty Krieger
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; and
| | - Olivier Kocher
- Department of Pathology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
15
|
Lee D, Park S, Bae S, Jeong D, Park M, Kang C, Yoo W, Samad MA, Ke Q, Khang G, Kang PM. Hydrogen peroxide-activatable antioxidant prodrug as a targeted therapeutic agent for ischemia-reperfusion injury. Sci Rep 2015; 5:16592. [PMID: 26563741 PMCID: PMC4643254 DOI: 10.1038/srep16592] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 10/16/2015] [Indexed: 12/14/2022] Open
Abstract
Overproduction of hydrogen peroxide (H2O2) causes oxidative stress and is the main culprit in the pathogenesis of ischemia/reperfusion (I/R) injury. Suppression of oxidative stress is therefore critical in the treatment of I/R injury. Here, we report H2O2-activatable antioxidant prodrug (BRAP) that is capable of specifically targeting the site of oxidative stress and exerting anti-inflammatory and anti-apoptotic activities. BRAP with a self-immolative boronic ester protecting group was designed to scavenge H2O2 and release HBA (p-hydroxybenzyl alcohol) with antioxidant and anti-inflammatory activities. BRAP exerted potent antioxidant and anti-inflammatory activity in lipopolysaccharide (LPS)- and H2O2-stimulated cells by suppressing the generation of ROS and pro-inflammatory cytokines. In mouse models of hepatic I/R and cardiac I/R, BRAP exerted potent antioxidant, anti-inflammatory and anti-apoptotic activities due to the synergistic effects of H2O2-scavenging boronic esters and therapeutic HBA. In addition, administration of high doses of BRAP daily for 7 days showed no renal or hepatic function abnormalities. Therefore BRAP has tremendous therapeutic potential as H2O2-activatable antioxidant prodrug for the treatment of I/R injuries.
Collapse
Affiliation(s)
- Dongwon Lee
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea.,Polymer Fusion Research Center, Department of Polymer·Nano Science and Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Seunggyu Park
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Soochan Bae
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, United States
| | - Dahee Jeong
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Minhyung Park
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Changsun Kang
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Wooyoung Yoo
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Mohammed A Samad
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea.,Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, United States
| | - Qingen Ke
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, United States
| | - Gilson Khang
- Department of BIN Convergence Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea.,Polymer Fusion Research Center, Department of Polymer·Nano Science and Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, Republic of Korea
| | - Peter M Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, United States
| |
Collapse
|
16
|
Moreira AC, Branco AF, Sampaio SF, Cunha-Oliveira T, Martins TR, Holy J, Oliveira PJ, Sardão VA. Mitochondrial apoptosis-inducing factor is involved in doxorubicin-induced toxicity on H9c2 cardiomyoblasts. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2468-78. [DOI: 10.1016/j.bbadis.2014.09.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 09/19/2014] [Accepted: 09/26/2014] [Indexed: 01/22/2023]
|
17
|
Choudhury S, Bae S, Ke Q, Lee JY, Singh SS, St-Arnaud R, del Monte F, Kang PM. Abnormal calcium handling and exaggerated cardiac dysfunction in mice with defective vitamin d signaling. PLoS One 2014; 9:e108382. [PMID: 25268137 PMCID: PMC4182450 DOI: 10.1371/journal.pone.0108382] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/20/2014] [Indexed: 12/31/2022] Open
Abstract
Aim Altered vitamin D signaling is associated with cardiac dysfunction, but the pathogenic mechanism is not clearly understood. We examine the mechanism and the role of vitamin D signaling in the development of cardiac dysfunction. Methods and Results We analyzed 1α-hydroxylase (1α-OHase) knockout (1α-OHase−/−) mice, which lack 1α-OH enzymes that convert the inactive form to hormonally active form of vitamin D. 1α-OHase−/− mice showed modest cardiac hypertrophy at baseline. Induction of pressure overload by transverse aortic constriction (TAC) demonstrated exaggerated cardiac dysfunction in 1α-OHase−/− mice compared to their WT littermates with a significant increase in fibrosis and expression of inflammatory cytokines. Analysis of calcium (Ca2+) transient demonstrated profound Ca2+ handling abnormalities in 1α-OHase−/− mouse cardiomyocytes (CMs), and treatment with paricalcitol (PC), an activated vitamin D3 analog, significantly attenuated defective Ca2+ handling in 1α-OHase−/− CMs. We further delineated the effect of vitamin D deficiency condition to TAC by first correcting the vitamin D deficiency in 1α-OHase−/− mice, followed then by either a daily maintenance dose of vitamin D or vehicle (to achieve vitamin D deficiency) at the time of sham or TAC. In mice treated with vitamin D, there was a significant attenuation of TAC-induced cardiac hypertrophy, interstitial fibrosis, inflammatory markers, Ca2+ handling abnormalities and cardiac function compared to the vehicle treated animals. Conclusions Our results provide insight into the mechanism of cardiac dysfunction, which is associated with severely defective Ca2+ handling and defective vitamin D signaling in 1α-OHase−/− mice.
Collapse
Affiliation(s)
- Sangita Choudhury
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Soochan Bae
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Qingen Ke
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ji Yoo Lee
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sylvia S. Singh
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - René St-Arnaud
- Shriners Hospital and Departments of Surgery and Human Genetics, McGill University, Montreal, Canada
| | - Federica del Monte
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter M. Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
18
|
Kang JJ, Samad MA, Kim KS, Bae S. Comparative Anti-inflammatory Effects of Anti-arthritic Herbal Medicines and Ibuprofen. Nat Prod Commun 2014. [DOI: 10.1177/1934578x1400900932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDS), such as ibuprofen, are widely used over-the-counter drugs to treat arthritis, but they are often associated with side effects. Herbal medicines have been used to treat various diseases such as arthritis, but the scientific profiles are not well understood. In this study, we examined, in comparison with ibuprofen, the inhibitory effects on various inflammatory markers of the most commonly used herbal medicines to treat arthritis, boswellia (Boswellia sapindales), licorice (Glycyrrhiza glabra), guggul (Commiphora wightii), and neem (Azadirachta indica). To elicit inflammatory response, we exposed mouse myoblast C2C12 cells to lipopolysaccharide (LPS). Tumor necrosis factor-alpha (TNF-α) and monocyte chemotactic protein-1 (MCP-1), which are cytokines activated during an inflammatory response, were determined. The optimal non-toxic concentration was determined by exposing different concentrations of drugs (from 0.01 to 10 mg/mL). Cell death measurement revealed that the drug concentrations lower than 0.05 mg/mL were non-toxic concentrations for each drug, and these doses were used for the main experiments. We found that neem and licorice showed robust anti-inflammatory responses compared with ibuprofen. However, boswellia and guggul did not demonstrate significant anti-inflammatory responses. We concluded that neem and licorice are more effective than ibuprofen in suppressing LPS-induced inflammation in C2C12 cells.
Collapse
Affiliation(s)
- Joshua J. Kang
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Mohammed A. Samad
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
- Department of BIN Fusion Technology, Chonbuk National University, Dukjin, Jeonju, 561–756, South Korea
| | - Kye S. Kim
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| | - Soochan Bae
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Park S, Yoon J, Bae S, Park M, Kang C, Ke Q, Lee D, Kang PM. Therapeutic use of H2O2-responsive anti-oxidant polymer nanoparticles for doxorubicin-induced cardiomyopathy. Biomaterials 2014; 35:5944-53. [PMID: 24767791 DOI: 10.1016/j.biomaterials.2014.03.084] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 03/28/2014] [Indexed: 11/15/2022]
Abstract
Doxorubicin (DOX) is a commonly used anti-neoplastic agent but its clinical use is limited due to serious hepatic and cardiac side effects. DOX-induced toxicity is mainly associated with overproduction of reactive species oxygen (ROS) such as hydrogen peroxide (H2O2). We have recently developed H2O2-responsive anti-oxidant polymer, polyoxalate containing vanillyl alcohol (PVAX), which is designed to rapidly scavenge H2O2 and release vanillyl alcohol with anti-oxidant, anti-inflammatory and anti-apoptotic properties. In this study, we report that PVAX nanoparticles are novel therapeutic agents for treating DOX-induced cardiac and hepatic toxicity. Intraperitoneal injection of PVAX nanoparticles (4 mg/kg/day) resulted in significant inhibition in apoptosis in liver and heart of DOX-treated mice by suppressing the activation of poly (ADP ribose) polymerase 1 (PARP-1) and caspase-3. PVAX treatment also prevented DOX-induced cardiac dysfunction. Furthermore, survival rate (vehicle = 35% vs. PVAX = 75%; p < 0.05) was significantly improved in a PVAX nanoparticles-treated group compared with vehicle treated groups. Taken together, we anticipate that PVAX nanoparticles could be a highly specific and potent treatment modality in DOX-induced cardiac and hepatic toxicity.
Collapse
Affiliation(s)
- Seunggyu Park
- Department of BIN Fusion Technology, Chonbuk National University, Dukjin 664-14, Jeonju, Chonbuk 561-756, South Korea
| | - Jooheung Yoon
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, Boston, MA 02215, USA
| | - Soochan Bae
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, Boston, MA 02215, USA
| | - Minhyung Park
- Department of BIN Fusion Technology, Chonbuk National University, Dukjin 664-14, Jeonju, Chonbuk 561-756, South Korea
| | - Changsun Kang
- Department of BIN Fusion Technology, Chonbuk National University, Dukjin 664-14, Jeonju, Chonbuk 561-756, South Korea
| | - Qingen Ke
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, Boston, MA 02215, USA
| | - Dongwon Lee
- Department of BIN Fusion Technology, Chonbuk National University, Dukjin 664-14, Jeonju, Chonbuk 561-756, South Korea; Polymer Fusion Research Center, Department of Polymer⋅Nano Science and Technology, Chonbuk National University, Dukjin 664-14, Jeonju, Chonbuk 561-756, South Korea.
| | - Peter M Kang
- Department of BIN Fusion Technology, Chonbuk National University, Dukjin 664-14, Jeonju, Chonbuk 561-756, South Korea; Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, Boston, MA 02215, USA.
| |
Collapse
|
20
|
Lee D, Bae S, Ke Q, Lee J, Song B, Karumanchi SA, Khang G, Choi HS, Kang PM. Hydrogen peroxide-responsive copolyoxalate nanoparticles for detection and therapy of ischemia-reperfusion injury. J Control Release 2013; 172:1102-10. [PMID: 24096013 DOI: 10.1016/j.jconrel.2013.09.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 09/02/2013] [Accepted: 09/24/2013] [Indexed: 01/01/2023]
Abstract
The main culprit in the pathogenesis of ischemia/reperfusion (I/R) injury is the generation of high level of hydrogen peroxide (H2O2). In this study, we report a novel diagnostic and therapeutic strategy for I/R injury based on H2O2-activatable copolyoxalate nanoparticles using a murine model of hind limb I/R injury. The nanoparticles are composed of hydroxybenzyl alcohol (HBA)-incorporating copolyoxalate (HPOX) that, in the presence of H2O2, degrades completely into three known and safe compounds, cyclohexanedimethanol, HBA and CO2. HPOX effectively scavenges H2O2 in a dose-dependent manner and hydrolyzes to release HBA which exerts intrinsic antioxidant and anti-inflammatory activities both in vitro and in vivo models of hind limb I/R. HPOX nanoparticles loaded with fluorophore effectively and robustly image H2O2 generated in hind limb I/R injury, demonstrating their potential for bioimaging of H2O2-associated diseases. Furthermore, HPOX nanoparticles loaded with anti-apoptotic drug effectively release the drug payload after I/R injury, exhibiting their effectiveness for a targeted drug delivery system for I/R injury. We anticipate that multifunctional HPOX nanoparticles have great potential as H2O2 imaging agents, therapeutics and drug delivery systems for H2O2-associated diseases.
Collapse
Affiliation(s)
- Dongwon Lee
- Department of BIN Fusion Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, South Korea; Department of Polymer Nano Science and Technology, Chonbuk National University, Jeonju, Chonbuk 561-756, South Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bae S, Singh SS, Yu H, Lee JY, Cho BR, Kang PM. Vitamin D signaling pathway plays an important role in the development of heart failure after myocardial infarction. J Appl Physiol (1985) 2013; 114:979-87. [PMID: 23429874 DOI: 10.1152/japplphysiol.01506.2012] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Accumulating evidence suggests that vitamin D deficiency plays a crucial role in heart failure. However, whether vitamin D signaling itself plays an important role in cardioprotection is poorly understood. In this study, we examined the mechanism of modulating vitamin D signaling on progression to heart failure after myocardial infarction (MI) in mice. Vitamin D signaling was activated by administration of paricalcitol (PC), an activated vitamin D analog. Wild-type (WT) mice underwent sham or MI surgery and then were treated with either vehicle or PC. Compared with vehicle group, PC attenuated development of heart failure after MI associated with decreases in biomarkers, apoptosis, inflammation, and fibrosis. There was also improvement of cardiac function with PC treatment after MI. Furthermore, vitamin D receptor (VDR) mRNA and protein levels were restored by PC treatment. Next, to explore whether defective vitamin D signaling exhibited deleterious responses after MI, WT and VDR knockout (KO) mice underwent sham or MI surgery and were analyzed 4 wk after MI. VDR KO mice displayed a significant decline in survival rate and cardiac function compared with WT mice after MI. VDR KO mice also demonstrated a significant increase in heart failure biomarkers, apoptosis, inflammation, and fibrosis. Vitamin D signaling promotes cardioprotection after MI through anti-inflammatory, antifibrotic and antiapoptotic mechanisms.
Collapse
Affiliation(s)
- Soochan Bae
- Cardiovascular Division, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
22
|
Yang Z, Huang B, Zhang Z, Kang Y, Fu M, Liao W. Effects of IL-1 receptor-associated kinase-4 gene silencing on human osteoblast-like cells. Connect Tissue Res 2012; 53:498-507. [PMID: 22606974 DOI: 10.3109/03008207.2012.694927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The aim of this study is to identify the effects of interleukin-1 receptor-associated kinase-4 (IRAK-4) gene silencing on human osteoblast-like cells. The siRNA sequences of the target gene, IRAK-4, were constructed and transferred into MG63 cells (control group = MG63 cells; SC group = MG63 cells transfected with scrambled IRAK-4 siRNA; KD group = MG63 cells transfected with 75 nM IRAK-4 siRNA). The morphological changes, cell growth, cell-cycle progression, apoptosis, and the expression of various cytokines and proteins were compared. Compared with the control and SC groups, IRAK-4 gene silencing in MG63 cells caused morphological changes, inhibited growth, altered the cell-cycle distribution, increased apoptosis (p < 0.05), decreased bone alkaline phosphatase and osteocalcin levels (p < 0.05), and decreased protein expression of Bcl-2/Bax and Bcl-2, p-JNK1/2, p-ERK1/2, and p-p38MAPK (p < 0.05). The results indicated that IRAK-4 gene silencing in MG63 cells inhibited cell proliferation and function and increase apoptosis, which may be related to the decreased Bcl-2/Bax ratio and inhibition of the protein expression of various components of the mitogen-activated protein kinase pathways. The results of this study may help improve the understanding of the relationship between IRAK-4 and osteoblast-like cells and the interactions between various cytokines in the periprosthetic inflammatory environment.
Collapse
Affiliation(s)
- Zibo Yang
- Department of Orthopedic and Joint Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | | | | | | | | | | |
Collapse
|
23
|
Shi J, Zhang L, Zhang YW, Surma M, Mark Payne R, Wei L. Downregulation of doxorubicin-induced myocardial apoptosis accompanies postnatal heart maturation. Am J Physiol Heart Circ Physiol 2012; 302:H1603-13. [PMID: 22328080 DOI: 10.1152/ajpheart.00844.2011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Doxorubicin is a highly effective chemotherapeutic agent used for treating a wide spectrum of tumors, but its usage is limited because of its dose-dependent cardiotoxicity, especially in pediatric patients. Accumulating evidence indicates that caspase-dependent apoptosis contributes to the cardiotoxicity of doxorubicin. However, less attention has been paid to the effects of age on doxorubicin-induced apoptosis signaling in myocardium. This study focused on investigating differential apoptotic sensitivity between neonatal and adult myocardium, in particular, between neonatal and adult cardiomyocytes in vivo. Our results show that caspase-3 activity in normal mouse hearts decreased by ≥ 20-fold within the first 3 wk after birth, associated with a rapid downregulation in the expression of key proapoptotic proteins in intrinsic and extrinsic pathways. This rapid downregulation of caspase-3 activity was confirmed by immunostaining for cleaved caspase-3 and terminal deoxynucleotidyl transferase dUTP-mediated nick-end label staining. Doxorubicin treatment induced a dose-dependent increase in caspase-3 activity and apoptosis in neonatal mouse hearts, and both caspase-8 and caspase-9 activations were involved. Using transgenic mice with a nuclear localized LacZ reporter gene to label cardiomyocytes in vivo, we observed a fourfold higher level of doxorubicin-induced cardiomyocyte apoptosis in 1-wk-old mice compared with that in 3-wk-old mice. This study points to a major difference in apoptotic signaling in doxorubicin cardiotoxicity between neonatal and adult mouse hearts and reveals a critical transition from high to low susceptibility to doxorubicin-induced apoptosis during postnatal heart maturation.
Collapse
Affiliation(s)
- Jianjian Shi
- Riley Heart Research Center, Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana 46202-5225, USA
| | | | | | | | | | | |
Collapse
|
24
|
Chiong M, Wang ZV, Pedrozo Z, Cao DJ, Troncoso R, Ibacache M, Criollo A, Nemchenko A, Hill JA, Lavandero S. Cardiomyocyte death: mechanisms and translational implications. Cell Death Dis 2011; 2:e244. [PMID: 22190003 PMCID: PMC3252742 DOI: 10.1038/cddis.2011.130] [Citation(s) in RCA: 350] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality worldwide. Although treatments have improved, development of novel therapies for patients with CVD remains a major research goal. Apoptosis, necrosis, and autophagy occur in cardiac myocytes, and both gradual and acute cell death are hallmarks of cardiac pathology, including heart failure, myocardial infarction, and ischemia/reperfusion. Pharmacological and genetic inhibition of autophagy, apoptosis, or necrosis diminishes infarct size and improves cardiac function in these disorders. Here, we review recent progress in the fields of autophagy, apoptosis, and necrosis. In addition, we highlight the involvement of these mechanisms in cardiac pathology and discuss potential translational implications.
Collapse
Affiliation(s)
- M Chiong
- Centro Estudios Moleculares de la Celula, Departamento de Bioquimica y Biología Molecular, Facultad de Ciencias Quimicas y Farmaceuticas and Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Apoptosis is a tightly regulated physiologic process of programmed cell death that occurs in both normal and pathologic tissues. Numerous in vitro or in vivo studies have indicated that cardiomyocyte death through apoptosis and necrosis is a primary contributor to the progression of anthracycline-induced cardiomyopathy. There are now several pieces of evidence to suggest that activation of intrinsic and extrinsic apoptotic pathways contribute to anthracycline-induced apoptosis in the heart. Novel strategies were developed to address a wide variety of cardiotoxic mechanisms and apoptotic pathways by which anthracycline influences cardiac structure and function. Anthracycline-induced apoptosis provides a very valid representation of cardiotoxicity in the heart, an argument which has implications for the most appropriate animal models of damaged heart plus diverse pharmacological effects. In this review we describe various aspects of the current understanding of apoptotic cell death triggered by anthracycline. Differences in the sensitivity to anthracycline-induced apoptosis between young and adult hearts are also discussed.
Collapse
Affiliation(s)
- Jianjian Shi
- Riley Heart Research Center, Wells Center for Pediatric Research, Department of Pediatrics Indiana University, School of Medicine, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
26
|
New Targets to Treat the Structural Remodeling of the Myocardium. J Am Coll Cardiol 2011; 58:1833-43. [DOI: 10.1016/j.jacc.2011.06.058] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 06/21/2011] [Indexed: 11/20/2022]
|
27
|
Choudhury S, Bae S, Ke Q, Lee JY, Kim J, Kang PM. Mitochondria to nucleus translocation of AIF in mice lacking Hsp70 during ischemia/reperfusion. Basic Res Cardiol 2011; 106:397-407. [PMID: 21387140 PMCID: PMC3205442 DOI: 10.1007/s00395-011-0164-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 02/02/2011] [Accepted: 02/21/2011] [Indexed: 11/25/2022]
Abstract
Heat shock protein 70 (Hsp70) has been shown to have an anti-apoptotic function, but its mechanism is not clear in heart. In this study, we examined the effect of Hsp70 deletion on AIF-induced apoptosis during ischemia/reperfusion (I/R) in vivo. Although Hsp70 KO and WT mice demonstrated similar amounts of AIF released from mitochondria after I/R surgery, Hsp70 KO mice showed a significantly greater increase in apoptosis, larger infarct size, and decreased cardiac output. There was also a significant fourfold increase in the nuclear accumulation of AIF in Hsp70 KO mice compared with WT mice. Treatment with 4-AN (4-amino-1,8-napthalimide, 3 mg/kg), a potent inhibitor of PARP-1, which is a critical regulator of AIF-induced apoptosis, significantly blocked the release of AIF from mitochondria and the translocation of AIF into the nuclei after I/R in both WT and Hsp70 KO mice. In addition, 4-AN treatment resulted in a significant inhibition of apoptosis, a reduction of infarct size, and attenuated cardiac dysfunction in both WT and Hsp70 KO mice after I/R. The anti-apoptotic function of Hsp70 occurs through the inhibition of AIF-induced apoptosis by blocking the mitochondria to nucleus translocation of AIF. PARP-1 inhibition improves cardiac function by blocking AIF-induced apoptosis.
Collapse
Affiliation(s)
- Sangita Choudhury
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Soochan Bae
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Qingen Ke
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Ji Yoo Lee
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jacob Kim
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Peter M. Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA, Department of BIN Fusion Technology, Chonbuk National University, Jeonju, South Korea, Cardiovascular Institute, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, CLS-910, Boston, MA 02215, USA,
| |
Collapse
|