1
|
Au Yeung VPW, Obrezanova O, Zhou J, Yang H, Bowen TJ, Ivanov D, Saffadi I, Carter AS, Subramanian V, Dillmann I, Hall A, Corrigan A, Viant MR, Pointon A. Computational approaches identify a transcriptomic fingerprint of drug-induced structural cardiotoxicity. Cell Biol Toxicol 2024; 40:50. [PMID: 38940987 PMCID: PMC11213733 DOI: 10.1007/s10565-024-09880-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/15/2024] [Indexed: 06/29/2024]
Abstract
Structural cardiotoxicity (SCT) presents a high-impact risk that is poorly tolerated in drug discovery unless significant benefit is anticipated. Therefore, we aimed to improve the mechanistic understanding of SCT. First, we combined machine learning methods with a modified calcium transient assay in human-induced pluripotent stem cell-derived cardiomyocytes to identify nine parameters that could predict SCT. Next, we applied transcriptomic profiling to human cardiac microtissues exposed to structural and non-structural cardiotoxins. Fifty-two genes expressed across the three main cell types in the heart (cardiomyocytes, endothelial cells, and fibroblasts) were prioritised in differential expression and network clustering analyses and could be linked to known mechanisms of SCT. This transcriptomic fingerprint may prove useful for generating strategies to mitigate SCT risk in early drug discovery.
Collapse
Affiliation(s)
- Victoria P W Au Yeung
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
- Phenomics, Data Sciences & Quantitative Biology, R&D AstraZeneca, Cambridge, UK.
| | - Olga Obrezanova
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Jiarui Zhou
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Hongbin Yang
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Tara J Bowen
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Delyan Ivanov
- High-Throughput Screening, R&D, AstraZeneca, Alderley Park, UK
| | - Izzy Saffadi
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Alfie S Carter
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Vigneshwari Subramanian
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Inken Dillmann
- Disease Molecular Profiling, Discovery Biology, R&D AstraZeneca, Gothenburg, Sweden
| | - Andrew Hall
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Adam Corrigan
- Phenomics, Data Sciences & Quantitative Biology, R&D AstraZeneca, Cambridge, UK
| | - Mark R Viant
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, UK
| | - Amy Pointon
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
2
|
Xie S, Sun Y, Zhao X, Xiao Y, Zhou F, Lin L, Wang W, Lin B, Wang Z, Fang Z, Wang L, Zhang Y. An update of the molecular mechanisms underlying anthracycline induced cardiotoxicity. Front Pharmacol 2024; 15:1406247. [PMID: 38989148 PMCID: PMC11234178 DOI: 10.3389/fphar.2024.1406247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Anthracycline drugs mainly include doxorubicin, epirubicin, pirarubicin, and aclamycin, which are widely used to treat a variety of malignant tumors, such as breast cancer, gastrointestinal tumors, lymphoma, etc. With the accumulation of anthracycline drugs in the body, they can induce serious heart damage, limiting their clinical application. The mechanism by which anthracycline drugs cause cardiotoxicity is not yet clear. This review provides an overview of the different types of cardiac damage induced by anthracycline-class drugs and delves into the molecular mechanisms behind these injuries. Cardiac damage primarily involves alterations in myocardial cell function and pathological cell death, encompassing mitochondrial dysfunction, topoisomerase inhibition, disruptions in iron ion metabolism, myofibril degradation, and oxidative stress. Mechanisms of uptake and transport in anthracycline-induced cardiotoxicity are emphasized, as well as the role and breakthroughs of iPSC in cardiotoxicity studies. Selected novel cardioprotective therapies and mechanisms are updated. Mechanisms and protective strategies associated with anthracycline cardiotoxicity in animal experiments are examined, and the definition of drug damage in humans and animal models is discussed. Understanding these molecular mechanisms is of paramount importance in mitigating anthracycline-induced cardiac toxicity and guiding the development of safer approaches in cancer treatment.
Collapse
Affiliation(s)
- Sicong Xie
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuwei Sun
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan Zhao
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiqun Xiao
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fei Zhou
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liang Lin
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Wang
- College of Electronic and Optical Engineering and College of Flexible Electronics, Future Technology, Nanjing University of Posts and Telecommunications, Nanjing, China
| | - Bin Lin
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Department of Pharmacy, Changxing People's Hospital, Huzhou, China
| | - Zun Wang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zixuan Fang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Wang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhang
- Department of Rehabilitation Medicine, School of Acupuncture-Moxibustion and Tuina and School of Health Preservation and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Department of Pharmacy, Changxing People's Hospital, Huzhou, China
| |
Collapse
|
3
|
Attanasio U, Di Sarro E, Tricarico L, Di Lisi D, Armentaro G, Miceli S, Fioretti F, Deidda M, Correale M, Novo G, Sciacqua A, Nodari S, Cadeddu C, Tocchetti CG, Palazzuoli A, Mercurio V. Cardiovascular Biomarkers in Cardio-Oncology: Antineoplastic Drug Cardiotoxicity and Beyond. Biomolecules 2024; 14:199. [PMID: 38397436 PMCID: PMC10887095 DOI: 10.3390/biom14020199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Serum biomarkers represent a reproducible, sensitive, minimally invasive and inexpensive method to explore possible adverse cardiovascular effects of antineoplastic treatments. They are useful tools in risk stratification, the early detection of cardiotoxicity and the follow-up and prognostic assessment of cancer patients. In this literature review, we aim at describing the current state of knowledge on the meaning and the usefulness of cardiovascular biomarkers in patients with cancer; analyzing the intricate relationship between cancer and cardiovascular disease (especially HF) and how this affects cardiovascular and tumor biomarkers; exploring the role of cardiovascular biomarkers in the risk stratification and in the identification of chemotherapy-induced cardiotoxicity; and providing a summary of the novel potential biomarkers in this clinical setting.
Collapse
Affiliation(s)
- Umberto Attanasio
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
| | - Elena Di Sarro
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
| | - Lucia Tricarico
- Cardiology Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (L.T.); (M.C.)
| | - Daniela Di Lisi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (D.D.L.); (G.N.)
- Division of Cardiology, University Hospital Paolo Giaccone, Via del Vespro 129, 90127 Palermo, Italy
| | - Giuseppe Armentaro
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy; (G.A.); (S.M.); (A.S.)
| | - Sofia Miceli
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy; (G.A.); (S.M.); (A.S.)
| | - Francesco Fioretti
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Spedali Civili Hospital and University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy; (F.F.); (S.N.)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy (C.C.)
| | - Michele Correale
- Cardiology Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (L.T.); (M.C.)
| | - Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (D.D.L.); (G.N.)
- Division of Cardiology, University Hospital Paolo Giaccone, Via del Vespro 129, 90127 Palermo, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy; (G.A.); (S.M.); (A.S.)
| | - Savina Nodari
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Spedali Civili Hospital and University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy; (F.F.); (S.N.)
| | - Christian Cadeddu
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy (C.C.)
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Alberto Palazzuoli
- Cardiovascular Diseases Unit, Cardio-thoracic and Vascular Department Le Scotte Hospital, University of Siena, Strada delle Scotte 14, 53100 Siena, Italy;
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
| |
Collapse
|
4
|
Dougherty BV, Moore CJ, Rawls KD, Jenior ML, Chun B, Nagdas S, Saucerman JJ, Kolling GL, Wallqvist A, Papin JA. Identifying metabolic adaptations characteristic of cardiotoxicity using paired transcriptomics and metabolomics data integrated with a computational model of heart metabolism. PLoS Comput Biol 2024; 20:e1011919. [PMID: 38422168 PMCID: PMC10931521 DOI: 10.1371/journal.pcbi.1011919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/12/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Improvements in the diagnosis and treatment of cancer have revealed long-term side effects of chemotherapeutics, particularly cardiotoxicity. Here, we present paired transcriptomics and metabolomics data characterizing in vitro cardiotoxicity to three compounds: 5-fluorouracil, acetaminophen, and doxorubicin. Standard gene enrichment and metabolomics approaches identify some commonly affected pathways and metabolites but are not able to readily identify metabolic adaptations in response to cardiotoxicity. The paired data was integrated with a genome-scale metabolic network reconstruction of the heart to identify shifted metabolic functions, unique metabolic reactions, and changes in flux in metabolic reactions in response to these compounds. Using this approach, we confirm previously seen changes in the p53 pathway by doxorubicin and RNA synthesis by 5-fluorouracil, we find evidence for an increase in phospholipid metabolism in response to acetaminophen, and we see a shift in central carbon metabolism suggesting an increase in metabolic demand after treatment with doxorubicin and 5-fluorouracil.
Collapse
Affiliation(s)
- Bonnie V. Dougherty
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Connor J. Moore
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Kristopher D. Rawls
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Matthew L. Jenior
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Bryan Chun
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sarbajeet Nagdas
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, Virginia, United States of America
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Glynis L. Kolling
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland, United States of America
| | - Jason A. Papin
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
5
|
Antoniadi K, Thomaidis N, Nihoyannopoulos P, Toutouzas K, Gikas E, Kelaidi C, Polychronopoulou S. Prognostic Factors for Cardiotoxicity among Children with Cancer: Definition, Causes, and Diagnosis with Omics Technologies. Diagnostics (Basel) 2023; 13:1864. [PMID: 37296716 PMCID: PMC10252297 DOI: 10.3390/diagnostics13111864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/03/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Improvements in the treatment of childhood cancer have considerably enhanced survival rates over the last decades to over 80% as of today. However, this great achievement has been accompanied by the occurrence of several early and long-term treatment-related complications major of which is cardiotoxicity. This article reviews the contemporary definition of cardiotoxicity, older and newer chemotherapeutic agents that are mainly involved in cardiotoxicity, routine process diagnoses, and methods using omics technology for early and preventive diagnosis. Chemotherapeutic agents and radiation therapies have been implicated as a cause of cardiotoxicity. In response, the area of cardio-oncology has developed into a crucial element of oncologic patient care, committed to the early diagnosis and treatment of adverse cardiac events. However, routine diagnosis and the monitoring of cardiotoxicity rely on electrocardiography and echocardiography. For the early detection of cardiotoxicity, in recent years, major studies have been conducted using biomarkers such as troponin, N-terminal pro b-natriuretic peptide, etc. Despite the refinements in diagnostics, severe limitations still exist due to the increase in the above-mentioned biomarkers only after significant cardiac damage has occurred. Lately, the research has expanded by introducing new technologies and finding new markers using the omics approach. These new markers could be used not only for early detection but also for the early prevention of cardiotoxicity. Omics science, which includes genomics, transcriptomics, proteomics, and metabolomics, offers new opportunities for biomarker discovery in cardiotoxicity and may provide an understanding of the mechanisms of cardiotoxicity beyond traditional technologies.
Collapse
Affiliation(s)
- Kondylia Antoniadi
- Department of Pediatric Hematology-Oncology (T.A.O.), “Aghia Sophia” Children’s Hospital, Goudi, 11527 Athens, Greece
| | - Nikolaos Thomaidis
- Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Petros Nihoyannopoulos
- First Department of Cardiology, University of Athens, Hippokration Hospital, 11527 Athens, Greece
| | - Konstantinos Toutouzas
- First Department of Cardiology, University of Athens, Hippokration Hospital, 11527 Athens, Greece
| | - Evangelos Gikas
- Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Charikleia Kelaidi
- Department of Pediatric Hematology-Oncology (T.A.O.), “Aghia Sophia” Children’s Hospital, Goudi, 11527 Athens, Greece
| | - Sophia Polychronopoulou
- Department of Pediatric Hematology-Oncology (T.A.O.), “Aghia Sophia” Children’s Hospital, Goudi, 11527 Athens, Greece
| |
Collapse
|
6
|
Metabolomic Profiles on Antiblastic Cardiotoxicity: New Perspectives for Early Diagnosis and Cardioprotection. J Clin Med 2022; 11:jcm11226745. [PMID: 36431222 PMCID: PMC9693331 DOI: 10.3390/jcm11226745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/19/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Antiblastic drugs-induced cardiomyopathy remains a relevant cause of morbidity and mortality, during and after chemotherapy, despite the progression in protective therapy against cardiovascular diseases and myocardial function. In the last few decades, many groups of researchers have focused their attention on studying the metabolic profile, first in animals, and, subsequently, in humans, looking for profiles which could be able to predict drug-induced cardiotoxicity and cardiovascular damage. In clinical practice, patients identified as being at risk of developing cardiotoxicity undergo a close follow-up and more tailored therapies. Injury to the heart can be a consequence of both new targeted therapies, such as tyrosine kinase inhibitors, and conventional chemotherapeutic agents, such as anthracyclines. This review aims to describe all of the studies carried on this topic of growing interest.
Collapse
|
7
|
S R, P G, P B, Hn Y, Ak D. Solid lipid nanoformulation of berberine attenuates Doxorubicin triggered in vitro inflammation in H9c2 rat cardiomyocytes. Comb Chem High Throughput Screen 2022; 25:1695-1706. [PMID: 35718970 DOI: 10.2174/1386207325666220617113744] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/06/2022] [Accepted: 04/08/2022] [Indexed: 11/22/2022]
Abstract
AIM To evaluate berberine solid lipid nanoparticles' efficacy against doxorubicin-induced cardiotoxicity. BACKGROUND Berberine (Ber) is cardioprotective, but its oral bioavailability is low and its effect in chemotherapy-induced cardiotoxicity has not been studied. OBJECTIVE Solid lipid nanoparticles (SLNs) of berberine chloride were prepared, characterized and evaluated in vitro against Doxorubicin induced cardiomyocyte injury. METHODOLOGY Berberine loaded SLNs (Ber-SLNs) were synthesized using water-in-oil microemulsion technique with tripalmitin, Tween 80 and poloxamer 407. Ber-SLNs were evaluated for preventive effect against toxicity of Doxorubicin in H9c2 cells. The culture was pre-treated (24 h) with Ber (10 µM) and Ber-SLNs (1 and 10 µM) and exposed to 1 µM of Doxorubicin (Dox) was added for 3 h. The cell viability (LDH (Lactate dehydrogenase) assay and MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide)), levels of Creatine kinase-MB (CK-MB), Nitrite, MDA (Malondialdehyde), ROS (Reactive oxygen species) generation and apoptotic DNA (Deoxyribonucleic acid) content were assessed. RESULTS Ber-SLNs had a mean particle size of 13.12±1.188 nm, zeta potential of -1.05 ± 0.08 mV, poly-dispersity index (PDI) of 0.317 ± 0.05 and entrapment efficiency of 50 ± 4.8%. Cell viability was 81 0.17% for Ber-SLNs (10 µM) and 73.22 0.83% for Ber (10 µM) treated cells in MTT assay. Percentage cytotoxicity calculated from LDH release was 58.91 0.54% after Dox, 40.3 1.3% with Ber (10 µM) and 40.7 1.3% with Ber-SLNs (1 µM) (p<0.001). Inflammation and oxidative stress markers were lower with Ber and Ber-SLNs. Attenuation of ROS generation and apoptosis of cardiomyocytes were noted on fluorescence microscopy. CONCLUSION Ber loaded SLNs effectively prevented Doxorubicin-induced inflammation and oxidative stress in rat cardiomyocytes. The results demonstrate that microemulsion is a simple, cost-effective technique to prepare Ber-SLNs and may be considered as a drug delivery vehicle for berberine.
Collapse
Affiliation(s)
- Rawal S
- Department of Pharmacology, AIIMS, New Delhi
| | - Gupta P
- Department of Pharmacology, AIIMS, New Delhi
| | - Bhatnagar P
- Department of Pharmacology, AIIMS, New Delhi
| | - Yadav Hn
- Department of Pharmacology, AIIMS, New Delhi
| | - Dinda Ak
- Department of Pathology, AIIMS, New Delhi
| |
Collapse
|
8
|
Gryshkova V, Lushbough I, Palmer J, Burrier R, Delaunois A, Donley E, Valentin JP. microRNAs signatures as potential biomarkers of structural cardiotoxicity in human-induced pluripotent stem-cell derived cardiomyocytes. Arch Toxicol 2022; 96:2033-2047. [PMID: 35488128 DOI: 10.1007/s00204-022-03280-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/14/2022] [Indexed: 12/11/2022]
Abstract
Identification of early biomarkers of heart injury and drug-induced cardiotoxicity is important to eliminate harmful drug candidates early in preclinical development and to prevent severe drug effects. The main objective of this study was to investigate the expression of microRNAs (miRNAs) in human-induced pluripotent stem cell cardiomyocytes (hiPSC-CM) in response to a broad range of cardiotoxic drugs. Next generation sequencing was applied to hiPSC-CM treated for 72 h with 40 drugs falling into the categories of functional (i.e., ion channel blockers), structural (changes in cardiomyocytes structure), and general (causing both functional and structural) cardiotoxicants as well as non-cardiotoxic drugs. The largest changes in miRNAs expression were observed after treatments with structural or general cardiotoxicants. The number of deregulated miRNAs was the highest for idarubicin, mitoxantrone, and bortezomib treatments. RT-qPCR validation confirmed upregulation of several miRNAs across multiple treatments at therapeutically relevant concentrations: hsa-miR-187-3p, hsa-miR-146b-5p, hsa-miR-182-5p (anthracyclines); hsa-miR-365a-5p, hsa-miR-185-3p, hsa-miR-184, hsa-miR-182-5p (kinase inhibitors); hsa-miR-182-5p, hsa-miR-126-3p and hsa-miR-96-5p (common some anthracyclines, kinase inhibitors and bortezomib). Further investigations showed that an upregulation of hsa-miR-187-3p and hsa-miR-182-5p could serve as a potential biomarker of structural cardiotoxicity and/or an additional endpoint to characterize cardiac injury in vitro.
Collapse
Affiliation(s)
| | - Isabel Lushbough
- UCB Biopharma SRL, Braine L'Alleud, Belgium
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | |
Collapse
|
9
|
Cadeddu Dessalvi C, Deidda M, Noto A, Madeddu C, Cugusi L, Santoro C, López-Fernández T, Galderisi M, Mercuro G. Antioxidant Approach as a Cardioprotective Strategy in Chemotherapy-Induced Cardiotoxicity. Antioxid Redox Signal 2021; 34:572-588. [PMID: 32151144 DOI: 10.1089/ars.2020.8055] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Chemotherapy-induced cardiotoxicity (CTX) has been associated with redox signaling imbalance. In fact, redox reactions are crucial for normal heart physiology, whereas excessive oxidative stress can cause cardiomyocyte structural damage. Recent Advances: An antioxidant approach as a cardioprotective strategy in this setting has shown encouraging results in preventing anticancer drug-induced CTX. Critical Issues: In fact, traditional heart failure drugs as well as many other compounds and nonpharmacological strategies, with a partial effect in reducing oxidative stress, have been shown to counterbalance chemotherapy-induced CTX in this setting to some extent. Future Directions: Given the various pathways of toxicity involved in different chemotherapeutic schemes, interactions with redox balance need to be fine-tuned and a personalized cardioprotective approach seems to be required.
Collapse
Affiliation(s)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Antonio Noto
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Lucia Cugusi
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Ciro Santoro
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Teresa López-Fernández
- Cardiology Service, Cardio-Oncology Unit, La Paz University Hospital, IdiPAz Research Institute, Ciber CV, Madrid, Spain
| | - Maurizio Galderisi
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
10
|
Russo M, Della Sala A, Tocchetti CG, Porporato PE, Ghigo A. Metabolic Aspects of Anthracycline Cardiotoxicity. Curr Treat Options Oncol 2021; 22:18. [PMID: 33547494 PMCID: PMC7864817 DOI: 10.1007/s11864-020-00812-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 12/13/2022]
Abstract
OPINION STATEMENT Heart failure (HF) is increasingly recognized as the major complication of chemotherapy regimens. Despite the development of modern targeted therapies such as monoclonal antibodies, doxorubicin (DOXO), one of the most cardiotoxic anticancer agents, still remains the treatment of choice for several solid and hematological tumors. The insurgence of cardiotoxicity represents the major limitation to the clinical use of this potent anticancer drug. At the molecular level, cardiac side effects of DOXO have been associated to mitochondrial dysfunction, DNA damage, impairment of iron metabolism, apoptosis, and autophagy dysregulation. On these bases, the antioxidant and iron chelator molecule, dexrazoxane, currently represents the unique FDA-approved cardioprotectant for patients treated with anthracyclines.A less explored area of research concerns the impact of DOXO on cardiac metabolism. Recent metabolomic studies highlight the possibility that cardiac metabolic alterations may critically contribute to the development of DOXO cardiotoxicity. Among these, the impairment of oxidative phosphorylation and the persistent activation of glycolysis, which are commonly observed in response to DOXO treatment, may undermine the ability of cardiomyocytes to meet the energy demand, eventually leading to energetic failure. Moreover, increasing evidence links DOXO cardiotoxicity to imbalanced insulin signaling and to cardiac insulin resistance. Although anti-diabetic drugs, such as empagliflozin and metformin, have shown interesting cardioprotective effects in vitro and in vivo in different models of heart failure, their mechanism of action is unclear, and their use for the treatment of DOXO cardiotoxicity is still unexplored.This review article aims at summarizing current evidence of the metabolic derangements induced by DOXO and at providing speculations on how key players of cardiac metabolism could be pharmacologically targeted to prevent or cure DOXO cardiomyopathy.
Collapse
Affiliation(s)
- Michele Russo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Via Nizza 52, 10126, Torino, Italy
| | - Angela Della Sala
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Via Nizza 52, 10126, Torino, Italy
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Paolo Ettore Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
11
|
Metabonomics Analysis of Myocardial Metabolic Dysfunction in Patients with Cardiac Natriuretic Peptide Resistance. Cardiol Res Pract 2020; 2020:1416945. [PMID: 33376601 PMCID: PMC7744244 DOI: 10.1155/2020/1416945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/29/2020] [Accepted: 10/31/2020] [Indexed: 12/31/2022] Open
Abstract
Brain natriuretic peptide (BNP) is an important biological marker and regulator of cardiac function. BNP resistance is characterized by high concentrations of less functionally effective BNP and common in heart failure (HF) patients. However, the roles and consequences of BNP resistance remain poorly understood. Investigate the effects of cardiac BNP resistance and identify potential metabolic biomarkers for screening and diagnosis. Thirty patients and thirty healthy subjects were enrolled in this study. Cardiac functions were evaluated by echocardiography. The plasma levels of cyclic guanosine monophosphate (cGMP) and BNP were measured by enzyme-linked immunosorbent assay (ELISA) and the cGMP/BNP ratio is calculated to determine cardiac natriuretic peptide resistance. Liquid chromatograph tandem mass spectrometry (LC-MS) based untargeted metabolomics analysis was applied to screen metabolic changes. The cGMP/BNP ratio was markedly lower in HF patients than controls. The cGMP/BNP ratio and ejection fraction (EF) were strongly correlated (R2 = 0.676, P < 0.05). Importantly, metabolic profiles were substantially different between HF patients and healthy controls. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis demonstrated that the differentially expressed metabolites are involved in signaling pathways that regulate cardiac functions. In HF patients, BNP resistance develops in association with a reduction in heart function and metabolic remodeling. It suggests possible functional roles of BNP resistance in the regulation of cardiac metabolism.
Collapse
|
12
|
A Comparative Study of Rat Urine 1H-NMR Metabolome Changes Presumably Arising from Isoproterenol-Induced Heart Necrosis Versus Clarithromycin-Induced QT Interval Prolongation. BIOLOGY 2020; 9:biology9050098. [PMID: 32414184 PMCID: PMC7284797 DOI: 10.3390/biology9050098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/07/2020] [Accepted: 05/10/2020] [Indexed: 12/18/2022]
Abstract
Cardiotoxicity remains a challenging concern both in drug development and in the management of various clinical situations. There are a lot of examples of drugs withdrawn from the market or stopped during clinical trials due to unpredicted cardiac adverse events. Obviously, current conventional methods for cardiotoxicity assessment suffer from a lack of predictivity and sensitivity. Therefore, there is a need for developing new tools to better identify and characterize any cardiotoxicity that can occur during the pre-clinical and clinical phases of drug development as well as after marketing in exposed patients. In this study, isoproterenol and clarithromycin were used as prototypical cardiotoxic agents in rats in order to evaluate potential biomarkers of heart toxicity at very early stages using 1H-NMR-based metabonomics. While isoproterenol is known to cause heart necrosis, clarithromycin may induce QT interval prolongation. Heart necrosis and QT prolongation were validated by histological analysis, serum measurement of lactate dehydrogenase/creatine phosphate kinase and QTc measurement by electrocardiogram (ECG). Urine samples were collected before and repeatedly during daily exposure to the drugs for 1H-NMR based-metabonomics investigations. Specific metabolic signatures, characteristic of each tested drug, were obtained from which potential predictive biomarkers for drug-induced heart necrosis and drug-induced QT prolongation were retrieved. Isoproterenol-induced heart necrosis was characterized by higher levels of taurine, creatine, glucose and by lower levels of Krebs cycle intermediates, creatinine, betaine/trimethylamine N-oxide (TMAO), dimethylamine (DMA)/sarcosine. Clarithromycin-induced QT prolongation was characterized by higher levels of creatinine, taurine, betaine/TMAO and DMA/sarcosine and by lower levels of Krebs cycle intermediates, glucose and hippurate.
Collapse
|
13
|
Metabolomic Analysis of Patients with Chronic Myeloid Leukemia and Cardiovascular Adverse Events after Treatment with Tyrosine Kinase Inhibitors. J Clin Med 2020; 9:jcm9041180. [PMID: 32326001 PMCID: PMC7231160 DOI: 10.3390/jcm9041180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Cardiovascular adverse events (CV-AEs) are considered critical complications in chronic myeloid leukemia (CML) patients treated with second- and third-generation tyrosine kinase inhibitors (TKIs). The aim of our study was to assess the correlation between metabolic profiles and CV-AEs in CML patients treated with TKIs. Methods: We investigated 39 adult CML patients in chronic-phase (mean age 49 years, range 24–70 years), with no comorbidities evidenced at baseline, who were consecutively identified with CML and treated with imatinib, nilotinib, dasatinib, and ponatinib. All patients performed Gas-Chromatography-Mass-Spectrometry-based metabolomic analysis and were divided into two groups (with and without CV-AEs). Results: Ten CV-AEs were documented. Seven CV-AEs were rated as 3 according to the Common Toxicity Criteria, and one patient died of a dissecting aneurysm of the aorta. The patients’ samples were clearly separated into two groups after analysis and the main discriminant metabolites were tyrosine, lysine, glutamic acid, ornithine, 2-piperdinecarboxylic acid, citric acid, proline, phenylalanine, threonine, mannitol, leucine, serine, creatine, alanine, and 4-hydroxyproline, which were more abundant in the CV-AE group. Conversely, myristic acid, oxalic acid, arabitol, 4-deoxy rithronic acid, ribose, and elaidic acid were less represented in the CV-AE group. Conclusions: CML patients with CV-AEs show a different metabolic profile, suggesting probable mechanisms of endothelial damage.
Collapse
|