1
|
Yuan X, Yu T, Zhang Z, Li S. Non-invasive assessment of proarrhythmic risks associated with isoprenaline and the dietary supplement ingredient synephrine using human induced pluripotent stem cell-derived cardiomyocytes. Front Cardiovasc Med 2024; 11:1407138. [PMID: 38911513 PMCID: PMC11190318 DOI: 10.3389/fcvm.2024.1407138] [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: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
Background There have been conflicting reports about the proarrhythmic risk of p-synephrine (SYN). To address this, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) combined with the microelectrode array (MEA) system have been utilized to assess arrhythmia risks, particularly in the context of adrenomimetic drugs. Aim This study aims to determine whether MEA recordings from hiPSC-CMs could predict the proarrhythmic risk of adrenomimetic drugs and to investigate the cardiovascular effects and mechanisms of SYN. Materials and methods We employed MEA recordings to assess the electrophysiological properties of hiPSC-CMs and conducted concentration-response analyses to evaluate the effects of SYN and Isoprenaline (ISO) on beating rate and contractility. A risk scoring system for proarrhythmic risks was established based on hiPSC-CMs in this study. ISO, a classic beta-adrenergic drug, was also evaluated. Furthermore, the study evaluated the risk of SYN and recorded the concentration-response of beating rate, contractility and the change in the presence or absence of selective β1, β2 and β3 adrenergic blockers. Results Our results suggested that ISO carries a high risk of inducing arrhythmias, aligning with existing literature. SYN caused a 30% prolongation of the field potential duration (FPD) at a concentration of 206.326 μM, a change significantly different from baseline measurements and control treatments. The half maximal effective concentration (EC50) of SYN (3.31 μM) to affect hiPSC-CM beating rate is much higher than that of ISO (18.00 nM). The effect of SYN at an EC50 of 3.31 μM is about ten times more potent in hiPSC-CMs compared to neonatal rat cardiomyocytes (34.12 μM). SYN increased the contractility of cardiomyocytes by 29.97 ± 11.65%, compared to ISO's increase of 50.56 ± 24.15%. β1 receptor blockers almost eliminated the beating rate increase induced by both ISO and SYN, while neither β2 nor β3 blockers had a complete inhibitory effect. Conclusion The MEA and hiPSC-CM system could effectively predict the risk of adrenomimetic drugs. The study concludes that the proarrhythmia risk of SYN at conventional doses is low. SYN is more sensitive in increasing beating rate and contractility in human cardiomyocytes compared to rats, primarily activating β1 receptor.
Collapse
Affiliation(s)
| | | | | | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Ogonowski N, Rukavina Mikusic NL, Kouyoumdzian NM, Choi MR, Fellet A, Balaszczuk AM, Celuch SM. Cardiotoxic Effects of the Antineoplastic Doxorubicin in a Model of Metabolic Syndrome: Oxidative Stress and Transporter Expression in the Heart. J Cardiovasc Pharmacol 2021; 78:784-791. [PMID: 34524257 DOI: 10.1097/fjc.0000000000001137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/18/2021] [Indexed: 12/22/2022]
Abstract
ABSTRACT The aim of the present work was to examine whether metabolic syndrome-like conditions in rats with fructose (F) overload modify the cardiotoxic effects induced by doxorubicin (DOX) and whether the treatment altered the expression of P-gp, breast cancer resistance protein, and organic cation/carnitine transporters in the heart. Male Sprague-Dawley rats received either tap water (control group [C]; n = 16) or water with F 10% wt/vol (n = 16) during 8 weeks. Three days before being killed, the animals received a single dose of DOX (6 mg/kg, ip, md) (C-DOX and F-DOX groups) or vehicle (VEH; ISS 1 mL/kg BW; ip) (C-VEH and F-VEH groups) (n = 8 per group). F overload enhanced thiobarbituric acid-reactive substance levels in the left ventricle, and DOX injection further increased those values. DOX did not alter thiobarbituric acid-reactive substance production in C animals. DOX caused a decrease of 30% in the ejection fraction and a nearly 40% reduction in the fractional shortening in F animals, but not in C rats. Cardiac tissue levels of P-gp decreased by about 30% in F rats compared with the C groups. DOX did not modify cardiac P-gp expression. Breast cancer resistance protein and organic cation/carnitine transporter (OCTN 1/2/3) protein levels did not change with either F or DOX. It is suggested that DOX could cause greater cardiotoxicity in rats receiving F, probably due to enhanced cardiac lipid peroxidation and lower expression of cardiac P-gp. These results support the hypothesis that the cardiotoxicity of DOX could be increased under metabolic syndrome-like conditions or in other health disorders that involve cardiovascular risk factors.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Animals
- Antibiotics, Antineoplastic
- Cardiotoxicity
- Disease Models, Animal
- Doxorubicin
- Heart Diseases/chemically induced
- Heart Diseases/metabolism
- Heart Diseases/pathology
- Heart Diseases/physiopathology
- Lipid Peroxidation
- Male
- Metabolic Syndrome/complications
- Metabolic Syndrome/metabolism
- Myocardium/metabolism
- Myocardium/pathology
- Organic Cation Transport Proteins/genetics
- Organic Cation Transport Proteins/metabolism
- Oxidative Stress
- Rats, Sprague-Dawley
- Ventricular Function, Left/drug effects
- Rats
Collapse
Affiliation(s)
- Natalia Ogonowski
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Fisiología, Instituto de Química y Metabolismo del Fármaco, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Natalia Lucía Rukavina Mikusic
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Ciudad Autónoma de Buenos Aires, Argentina
| | - Nicolás Martín Kouyoumdzian
- Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET, Ciudad Autónoma de Buenos Aires, Argentina; and
| | - Marcelo Roberto Choi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET), CONICET, Ciudad Autónoma de Buenos Aires, Argentina; and
| | - Andrea Fellet
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Fisiología, Instituto de Química y Metabolismo del Fármaco, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ana María Balaszczuk
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Fisiología, Instituto de Química y Metabolismo del Fármaco, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Stella Maris Celuch
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas, CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
3
|
Iacopo F, Branch M, Cardinale D, Middeldorp M, Sanders P, Cohen JB, Achirica MC, Jaiswal S, Brown SA. Preventive Cardio-Oncology: Cardiovascular Disease Prevention in Cancer Patients and Survivors. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2021. [DOI: 10.1007/s11936-020-00883-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
4
|
Sasserath T, Rumsey JW, McAleer CW, Bridges LR, Long CJ, Elbrecht D, Schuler F, Roth A, Bertinetti‐LaPatki C, Shuler ML, Hickman JJ. Differential Monocyte Actuation in a Three-Organ Functional Innate Immune System-on-a-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000323. [PMID: 32670763 PMCID: PMC7341107 DOI: 10.1002/advs.202000323] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/09/2020] [Indexed: 05/24/2023]
Abstract
A functional, human, multiorgan, pumpless, immune system-on-a-chip featuring recirculating THP-1 immune cells with cardiomyocytes, skeletal muscle, and liver in separate compartments in a serum-free medium is developed. This in vitro platform can emulate both a targeted immune response to tissue-specific damage, and holistic proinflammatory immune response to proinflammatory compound exposure. The targeted response features fluorescently labeled THP-1 monocytes selectively infiltrating into an amiodarone-damaged cardiac module and changes in contractile force measurements without immune-activated damage to the other organ modules. In contrast to the targeted immune response, general proinflammatory treatment of immune human-on-a-chip systems with lipopolysaccharide (LPS) and interferon-γ (IFN-γ) causes nonselective damage to cells in all three-organ compartments. Biomarker analysis indicates upregulation of the proinflammation cytokines TNF-α, IL-6, IL-10, MIP-1, MCP-1, and RANTES in response to LPS + IFN-γ treatment indicative of the M1 macrophage phenotype, whereas amiodarone treatment only leads to an increase in the restorative cytokine IL-6 which is a marker for the M2 phenotype. This system can be used as an alternative to humanized animal models to determine direct immunological effects of biological therapeutics including monoclonal antibodies, vaccines, and gene therapies, and the indirect effects caused by cytokine release from target tissues in response to a drug's pharmacokinetics (PK)/pharmacodynamics (PD) profile.
Collapse
Affiliation(s)
- Trevor Sasserath
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
| | - John W. Rumsey
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
| | | | | | | | - Daniel Elbrecht
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
| | - Franz Schuler
- Hoffmann‐La RochePharmaceuticals DivisionBldg 73, Rm 117bBasel4070Switzerland
| | - Adrian Roth
- Hoffmann‐La RochePharmaceuticals DivisionBldg 73, Rm 117bBasel4070Switzerland
| | | | | | - James J. Hickman
- Hesperos, Inc.12501 Research Parkway, Suite 100OrlandoFL32826USA
- NanoScience Technology Center, University of Central Florida12424 Research Parkway, Suite 400OrlandoFL32826USA
| |
Collapse
|
5
|
Villanueva S, Zhang W, Zecchinati F, Mottino A, Vore M. ABC Transporters in Extrahepatic Tissues: Pharmacological Regulation in Heart and Intestine. Curr Med Chem 2019; 26:1155-1184. [PMID: 29589524 DOI: 10.2174/0929867325666180327092639] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 02/26/2018] [Accepted: 03/09/2018] [Indexed: 12/17/2022]
Abstract
ATP binding cassette (ABC) transporters are transmembrane proteins expressed in secretory epithelia like the liver, kidneys and intestine, in the epithelia exhibiting barrier function such as the blood-brain barrier and placenta, and to a much lesser extent, in tissues like reproductive organs, lungs, heart and pancreas, among others. They regulate internal distribution of endogenous metabolites and xenobiotics including drugs of therapeutic use and also participate in their elimination from the body. We here describe the function and regulation of ABC transporters in the heart and small intestine, as examples of extrahepatic tissues, in which ABC proteins play clearly different roles. In the heart, they are involved in tissue pathogenesis as well as in protecting this organ against toxic compounds and druginduced oxidative stress. The small intestine is highly exposed to therapeutic drugs taken orally and, consequently, ABC transporters localized on its surface strongly influence drug absorption and pharmacokinetics. Examples of the ABC proteins currently described are Multidrug Resistance-associated Proteins 1 and 2 (MRP1 and 2) for heart and small intestine, respectively, and P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) for both organs.
Collapse
Affiliation(s)
- Silvina Villanueva
- Instituto de Fisiologia Experimental, Facultad de Ciencias Bioquimicas y Farmaceuticas, CONICET-UNR. 2000 Rosario, Argentina
| | - Wei Zhang
- Department of Toxicology & Cancer Biology, University of Kentucky, Lexington, KY 40536-0305, United States
| | - Felipe Zecchinati
- Instituto de Fisiologia Experimental, Facultad de Ciencias Bioquimicas y Farmaceuticas, CONICET-UNR. 2000 Rosario, Argentina
| | - Aldo Mottino
- Instituto de Fisiologia Experimental, Facultad de Ciencias Bioquimicas y Farmaceuticas, CONICET-UNR. 2000 Rosario, Argentina
| | - Mary Vore
- Department of Toxicology & Cancer Biology, University of Kentucky, Lexington, KY 40536-0305, United States
| |
Collapse
|
6
|
Alemany R, Moura DS, Redondo A, Martinez-Trufero J, Calabuig S, Saus C, Obrador-Hevia A, Ramos R, Villar VH, Valverde C, Vaz MA, Medina J, Felipe-Abrio I, Hindi N, Taron M, Martin-Broto J. Nilotinib as Coadjuvant Treatment with Doxorubicin in Patients with Sarcomas: A Phase I Trial of the Spanish Group for Research on Sarcoma. Clin Cancer Res 2018; 24:5239-5249. [PMID: 30037815 DOI: 10.1158/1078-0432.ccr-18-0851] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/16/2018] [Accepted: 07/17/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Nilotinib plus doxorubicin showed to be synergistic regarding apoptosis in several sarcoma cell lines. A phase I/II trial was thus designed to explore the feasibility of nilotinib as coadjuvant of doxorubicin by inhibiting MRP-1/P-gp efflux activity. The phase I part of the study is presented here.Patients and Methods: Nilotinib 400 mg/12 hours was administered in fixed dose from day 1 to 6, and doxorubicin on day 5 of each cycle. Three dose escalation levels for doxorubicin at 60, 65, and 75 mg/m2 were planned. Cycles were repeated every 3 weeks for a total of 4 cycles. Eligible subtypes were retroperitoneal liposarcoma, leiomyosarcoma, and unresectable/metastatic high-grade chondrosarcoma.Results: Thirteen patients were enrolled: 7 chondrosarcoma, 4 liposarcoma, and 2 leiomyosarcoma. In 46 cycles administered, the most relevant grade 3/4 adverse effects per patient were neutropenia 54%, febrile neutropenia 15%, and asthenia 8%. No cardiac toxicity was observed. Only one dose-limiting toxicity (febrile neutropenia) was reported in the third dose level. With regard to efficacy, 1 partial response (1 liposarcoma), 9 stable diseases (5 chondrosarcoma, 2 liposarcoma, 1 leiomyosarcoma), and 3 progressive diseases (2 chondrosarcoma and 1 leiomyosarcoma) were present. ABCB1 and ABCC1 RNA expression levels decreased by 58.47-fold and 1.47-fold, respectively, on day 5 of the cycle.Conclusions: Combination of MRP-1/P-gp inhibitor, nilotinib, as coadjuvant with doxorubicin is feasible; it appears not to add substantial toxicity compared with doxorubicin alone. Pharmacodynamic study supports this concept. The recommended dose for the phase II part for doxorubicin was 75 mg/m2 Clin Cancer Res; 24(21); 5239-49. ©2018 AACR.
Collapse
Affiliation(s)
- Regina Alemany
- Department of Biology, Balearic Islands University, Palma de Mallorca, Spain.,Group of Advanced Therapies and Biomarkers in Clinical Oncology, Institut d'Investigació Sanitària de les Illes Balears (IdISBa-IUNICS), Palma de Mallorca, Spain
| | - David S Moura
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain
| | - Andres Redondo
- Medical Oncology Department, University Hospital La Paz, Madrid, Spain
| | | | - Silvia Calabuig
- Molecular Oncology Laboratory, Fundación Investigación, Hospital General Universitario de Valencia, Valencia, (Spain). Centro de Investigación Biomédica en Red de Cáncer (CIBEROnc), Madrid, Spain. Department of Pathology, Universitat de València, Valencia, Spain
| | - Carlos Saus
- Pathology Department, University Hospital Son Espases, Palma de Mallorca, Spain
| | - Antonia Obrador-Hevia
- Group of Advanced Therapies and Biomarkers in Clinical Oncology, Institut d'Investigació Sanitària de les Illes Balears (IdISBa-IUNICS), Palma de Mallorca, Spain.,Sequencing Unit, University Hospital Son Espases, Palma de Mallorca, Spain
| | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, Palma de Mallorca, Spain
| | - Victor H Villar
- Department of Biology, Balearic Islands University, Palma de Mallorca, Spain
| | - Claudia Valverde
- Medical Oncology Department, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Maria Angeles Vaz
- Medical Oncology Department, University Hospital Ramon y Cajal, Madrid, Spain
| | - Javier Medina
- Medical Oncology Department, Hospital Virgen de la Salud, Toledo, Spain
| | - Irene Felipe-Abrio
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain.,Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Nadia Hindi
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain.,Medical Oncology Department, University Hospital Virgen del Rocio, Sevilla, Spain
| | - Miguel Taron
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain
| | - Javier Martin-Broto
- Instituto de Biomedicina de Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Lab.215, Sevilla, Spain. .,Medical Oncology Department, University Hospital Virgen del Rocio, Sevilla, Spain
| |
Collapse
|
7
|
Kobuszewska A, Tomecka E, Zukowski K, Jastrzebska E, Chudy M, Dybko A, Renaud P, Brzozka Z. Heart-on-a-Chip: An Investigation of the Influence of Static and Perfusion Conditions on Cardiac (H9C2) Cell Proliferation, Morphology, and Alignment. SLAS Technol 2017; 22:536-546. [PMID: 28430559 DOI: 10.1177/2472630317705610] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lab-on-a-chip systems are increasingly used as tools for cultures and investigation of cardiac cells. In this article, we present how the geometry of microsystems and microenvironmental conditions (static and perfusion) influence the proliferation, morphology, and alignment of cardiac cells (rat cardiomyoblasts-H9C2). Additionally, studies of cell growth after incubation with verapamil hydrochloride were performed. For this purpose, poly(dimethylsiloxane) (PDMS)/glass microfluidic systems with three different geometries of microchambers (a circular chamber, a longitudinal channel, and three parallel microchannels separated by two rows of micropillars) were prepared. It was found that static conditions did not enhance the growth of H9C2 cells in the microsystems. On the contrary, perfusion conditions had an influence on division, morphology, and the arrangement of the cells. The highest number of cells, their parallel orientation, and their elongated morphology were obtained in the longitudinal microchannel. It showed that this kind of microsystem can be used to understand processes in heart tissue in detail and to test newly developed compounds applied in the treatment of cardiac diseases.
Collapse
Affiliation(s)
- Anna Kobuszewska
- 1 Institute of Biotechnology, Department of Microbioanalytics, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Ewelina Tomecka
- 1 Institute of Biotechnology, Department of Microbioanalytics, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Kamil Zukowski
- 1 Institute of Biotechnology, Department of Microbioanalytics, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Elzbieta Jastrzebska
- 1 Institute of Biotechnology, Department of Microbioanalytics, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Michal Chudy
- 1 Institute of Biotechnology, Department of Microbioanalytics, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Artur Dybko
- 1 Institute of Biotechnology, Department of Microbioanalytics, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Philippe Renaud
- 2 Microsystems Laboratory (LMIS4), École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Zbigniew Brzozka
- 1 Institute of Biotechnology, Department of Microbioanalytics, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| |
Collapse
|
8
|
Cellular Models and In Vitro Assays for the Screening of modulators of P-gp, MRP1 and BCRP. Molecules 2017; 22:molecules22040600. [PMID: 28397762 PMCID: PMC6153761 DOI: 10.3390/molecules22040600] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/24/2017] [Accepted: 03/28/2017] [Indexed: 12/12/2022] Open
Abstract
Adenosine triphosphate (ATP)-binding cassette (ABC) transporters are highly expressed in tumor cells, as well as in organs involved in absorption and secretion processes, mediating the ATP-dependent efflux of compounds, both endogenous substances and xenobiotics, including drugs. Their expression and activity levels are modulated by the presence of inhibitors, inducers and/or activators. In vitro, ex vivo and in vivo studies with both known and newly synthesized P-glycoprotein (P-gp) inducers and/or activators have shown the usefulness of these transport mechanisms in reducing the systemic exposure and specific tissue access of potentially harmful compounds. This article focuses on the main ABC transporters involved in multidrug resistance [P-gp, multidrug resistance-associated protein 1 (MRP1) and breast cancer resistance protein (BCRP)] expressed in tissues of toxicological relevance, such as the blood-brain barrier, cardiovascular system, liver, kidney and intestine. Moreover, it provides a review of the available cellular models, in vitro and ex vivo assays for the screening and selection of safe and specific inducers and activators of these membrane transporters. The available cellular models and in vitro assays have been proposed as high throughput and low-cost alternatives to excessive animal testing, allowing the evaluation of a large number of compounds.
Collapse
|
9
|
Wang S, Zhai C, Zhang Y, Yu Y, Zhang Y, Ma L, Li S, Qiao Y. Cardamonin, a Novel Antagonist of hTRPA1 Cation Channel, Reveals Therapeutic Mechanism of Pathological Pain. Molecules 2016; 21:molecules21091145. [PMID: 27589700 PMCID: PMC6274095 DOI: 10.3390/molecules21091145] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 12/04/2022] Open
Abstract
The increasing demand for safe and effective treatments of chronic pain has promoted the investigation of novel analgesic drugs. Some herbals have been known to be able to relieve pain, while the chemical basis and target involved in this process remained to be clarified. The current study aimed to find anti-nociceptive candidates targeting transient receptor potential ankyrin 1 (TRPA1), a receptor that implicates in hyperalgesia and neurogenic inflammation. In the current study, 156 chemicals were tested for blocking HEK293/TRPA1 ion channel by calcium-influx assay. Docking study was conducted to predict the binding modes of hit compound with TRPA1 using Discovery Studio. Cytotoxicity in HEK293 was conducted by Cell Titer-Glo assay. Additionally, cardiotoxicity was assessed via xCELLigence RTCA system. We uncovered that cardamonin selectively blocked TRPA1 activation while did not interact with TRPV1 nor TRPV4 channel. A concentration-dependent inhibitory effect was observed with IC50 of 454 nM. Docking analysis of cardamonin demonstrated a compatible interaction with A-967079-binding site of TRPA1. Meanwhile, cardamonin did not significantly reduce HEK293 cell viability, nor did it impair cardiomyocyte constriction. Our data suggest that cardamonin is a selective TRPA1 antagonist, providing novel insight into the target of its anti-nociceptive activity.
Collapse
Affiliation(s)
- Shifeng Wang
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wangjing Zhonghuan South Road, Chaoyang District, Beijing 100102, China.
| | - Chenxi Zhai
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wangjing Zhonghuan South Road, Chaoyang District, Beijing 100102, China.
| | - Yanling Zhang
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wangjing Zhonghuan South Road, Chaoyang District, Beijing 100102, China.
| | - Yangyang Yu
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wangjing Zhonghuan South Road, Chaoyang District, Beijing 100102, China.
| | - Yuxin Zhang
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wangjing Zhonghuan South Road, Chaoyang District, Beijing 100102, China.
| | - Lianghui Ma
- HD Biosciences, Co., Ltd., 590 Ruiqing Road, Zhangjiang Hi-Tech Park East Campus, Pudong New Area, Shanghai 201201, China.
| | - Shiyou Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing 100101, China.
| | - Yanjiang Qiao
- Key Laboratory of TCM-Information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 6 Wangjing Zhonghuan South Road, Chaoyang District, Beijing 100102, China.
| |
Collapse
|
10
|
Zhou ZY, Wan LL, Yang QJ, Han YL, Li D, Lu J, Guo C. Nilotinib reverses ABCB1/P-glycoprotein-mediated multidrug resistance but increases cardiotoxicity of doxorubicin in a MDR xenograft model. Toxicol Lett 2016; 259:124-132. [PMID: 27491883 DOI: 10.1016/j.toxlet.2016.07.710] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/06/2016] [Accepted: 07/30/2016] [Indexed: 11/30/2022]
Abstract
The BCR-Abl tyrosine kinase inhibitor (TKI), nilotinib, was developed to surmount resistance or intolerance to imatinib in patients with Philadelphia-positive chronic myelogenous leukemia. Recent studies have shown that nilotinib induces potent sensitization to anticancer agents by blocking the functions of ABCB1/P-glycoprotein (P-gp) in multidrug resistance (MDR). However, changes in P-gp expression or function affect the cardiac disposition and prolong the presence of both doxorubicin (DOX) and doxorubicinol (DOXol) in cardiac tissue, thus, enhancing the risk of cardiotoxicity. In this study, we used a MDR xenograft model to evaluate the antitumor activity, tissue distribution and cardiotoxicity of DOX when co-administered with nilotinib. This information will provide more insight into the pharmacological role of nilotinib in MDR reversal and the risk of DOX cardiotoxicity. Our results showed that nilotinib significantly enhanced DOX cytotoxicity and increased intracellular rhodamine 123 accumulation in MG63/DOX cells in vitro and strongly enhanced DOX inhibition of growth of P-gp-overexpressing MG63/DOX cell xenografts in nude mice. Additionally, nilotinib significantly increased DOX and DOXol accumulation in serum, heart, liver and tumor tissues. Importantly, nilotinib induced a disproportionate increase in DOXol in cardiac tissue. In the co-administration group, CBR1 and AKR1A1 protein levels were significantly increased in cardiac tissue, with more severe necrosis and vacuole formation. These results indicate that nilotinib reverses P-gp- mediated MDR by blocking the efflux function and potentiates DOX-induced cardiotoxicity. These findings represent a guide for the design of future clinical trials and studies of pharmacokinetic interactions and may be useful in guiding the use of nilotinib in combination therapy of cancer in clinical practice.
Collapse
Affiliation(s)
- Zhi-Yong Zhou
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, China; College of Medical Science, Three Gorges University, 443002 Yichang, Hubei, China
| | - Li-Li Wan
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, China
| | - Quan-Jun Yang
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, China
| | - Yong-Long Han
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, China
| | - Dan Li
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, China
| | - Jin Lu
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, China
| | - Cheng Guo
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233 Shanghai, China.
| |
Collapse
|
11
|
Brown SA, Sandhu N, Herrmann J. Systems biology approaches to adverse drug effects: the example of cardio-oncology. Nat Rev Clin Oncol 2015; 12:718-31. [PMID: 26462128 DOI: 10.1038/nrclinonc.2015.168] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Increased awareness of the cardiovascular toxic effects of chemotherapy has led to the emergence of cardio-oncology (or onco-cardiology), which focuses on screening, monitoring and treatment of patients with cardiovascular dysfunctions resulting from chemotherapy. Anthracyclines, such as doxorubicin, and HER2 inhibitors, such as trastuzumab, both have cardiotoxic effects. The biological rationale, mechanisms of action and cardiotoxicity profiles of these two classes of drugs, however, are completely different, suggesting that cardiotoxic effects can occur in a range of different ways. Advances in genomics and proteomics have implicated several genomic variants and biological pathways that can influence the susceptibility to cardiotoxicity from these, and other drugs. Established pathways include multidrug resistance proteins, energy utilization pathways, oxidative stress, cytoskeletal regulation and apoptosis. Gene-expression profiles that have revealed perturbed pathways have vastly increased our knowledge of the complex processes involved in crosstalk between tumours and cardiac function. Utilization of mathematical and computational modelling can complement pharmacogenomics and improve individual patient outcomes. Such endeavours should enable identification of variations in cardiotoxicity, particularly in those patients who are at risk of not recovering, even with the institution of cardioprotective therapy. The application of systems biology holds substantial potential to advance our understanding of chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Sherry-Ann Brown
- Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Nicole Sandhu
- Division of General Internal Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Joerg Herrmann
- Division of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
12
|
Hsieh A, Feric NT, Radisic M. Combined hypoxia and sodium nitrite pretreatment for cardiomyocyte protection in vitro. Biotechnol Prog 2015; 31:482-92. [PMID: 25582867 DOI: 10.1002/btpr.2039] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/24/2014] [Indexed: 11/08/2022]
Abstract
Methods that increase cardiomyocyte survival upon exposure to ischemia, hypoxia and reoxygenation injuries are required to improve the efficacy of cardiac cell therapy and enhance the viability and function of engineered tissues. We investigated the effect of combined hypoxia/NaNO2 pretreatment on rat neonatal cardiomyocyte (CM), cardiac fibroblast, and human embryonic stem cell-derived CM (hESC-CM) survival upon exposure to hypoxia/reoxygenation (H/R) injury in vitro. Cells were pretreated with and without hypoxia and/or various concentrations of NaNO2 for 20 min, then incubated for 2 h under hypoxic conditions, followed by 2 h in normoxia. The control cells were maintained under normoxia for 4 h. Pretreatment with either hypoxia or NaNO2 significantly increased CM viability but had no effect on cardiac fibroblast viability. Combined hypoxia/NaNO2 pretreatment significantly increased CM viability but significantly decreased cardiac fibroblast viability. In rat neonatal CMs, cell death, as determined by lactate dehydrogenase (LDH) activity, was significantly reduced with hypoxia/NaNO2 pretreatment; and in hESC-CMs, hypoxia/NaNO2 pretreatment increased the BCL-2/BAX gene expression ratio, suggesting that hypoxia/NaNO2 pretreatment promotes cell viability by downregulating apoptosis. Additionally, we found a correlation between the prosurvival effect of hypoxia/NaNO2 pretreatment and the myoglobin content of the cells by comparing neonatal rat ventricular and atrial CMs, which express high and low myoglobin respectively. Functionally, hypoxia/NaNO2 pretreatment significantly improved the excitation threshold upon H/R injury to the level observed for uninjured cells, whereas pretreatment did not affect the maximum capture rate. Hence, hypoxia/NaNO2 pretreatment may serve as a strategy to increase CM survival in cardiac regenerative therapy applications and tissue engineering.
Collapse
Affiliation(s)
- Anne Hsieh
- Dept. of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|
13
|
Zhou ZY, Wan LL, Yang QJ, Han YL, Li Y, Yu Q, Guo C, Li X. Evaluation of the pharmacokinetics and cardiotoxicity of doxorubicin in rat receiving nilotinib. Toxicol Appl Pharmacol 2013; 272:238-44. [DOI: 10.1016/j.taap.2013.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 05/15/2013] [Accepted: 06/05/2013] [Indexed: 11/16/2022]
|
14
|
McCaffrey TA, Tziros C, Lewis J, Katz R, Siegel R, Weglicki W, Kramer J, Mak IT, Toma I, Chen L, Benas E, Lowitt A, Rao S, Witkin L, Lian Y, Lai Y, Yang Z, Fu SW. Genomic profiling reveals the potential role of TCL1A and MDR1 deficiency in chemotherapy-induced cardiotoxicity. Int J Biol Sci 2013; 9:350-60. [PMID: 23630447 PMCID: PMC3638290 DOI: 10.7150/ijbs.6058] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 02/27/2013] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Anthracyclines, such as doxorubicin (Adriamycin), are highly effective chemotherapeutic agents, but are well known to cause myocardial dysfunction and life-threatening congestive heart failure (CHF) in some patients. METHODS To generate new hypotheses about its etiology, genome-wide transcript analysis was performed on whole blood RNA from women that received doxorubicin-based chemotherapy and either did, or did not develop CHF, as defined by ejection fractions (EF)≤40%. Women with non-ischemic cardiomyopathy unrelated to chemotherapy were compared to breast cancer patients prior to chemo with normal EF to identify heart failure-related transcripts in women not receiving chemotherapy. Byproducts of oxidative stress in plasma were measured in a subset of patients. RESULTS The results indicate that patients treated with doxorubicin showed sustained elevations in oxidative byproducts in plasma. At the RNA level, women who exhibited low EFs after chemotherapy had 260 transcripts that differed >2-fold (p<0.05) compared to women who received chemo but maintained normal EFs. Most of these transcripts (201) were not altered in non-chemotherapy patients with low EFs. Pathway analysis of the differentially expressed genes indicated enrichment in apoptosis-related transcripts. Notably, women with chemo-induced low EFs had a 4.8-fold decrease in T-cell leukemia/lymphoma 1A (TCL1A) transcripts. TCL1A is expressed in both cardiac and skeletal muscle, and is a known co-activator for AKT, one of the major pro-survival factors for cardiomyocytes. Further, women who developed low EFs had a 2-fold lower level of ABCB1 transcript, encoding the multidrug resistance protein 1 (MDR1), which is an efflux pump for doxorubicin, potentially leading to higher cardiac levels of drug. In vitro studies confirmed that inhibition of MDR1 by verapamil in rat H9C2 cardiomyocytes increased their susceptibility to doxorubicin-induced toxicity. CONCLUSIONS It is proposed that chemo-induced cardiomyopathy may be due to a reduction in TCL1A levels, thereby causing increased apoptotic sensitivity, and leading to reduced cardiac MDR1 levels, causing higher cardiac levels of doxorubicin and intracellular free radicals. If so, screening for TCL1A and MDR1 SNPs or expression level in blood, might identify women at greatest risk of chemo-induced heart failure.
Collapse
Affiliation(s)
- Timothy A McCaffrey
- Department of Medicine, Division of Genomic Medicine, George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Wang J, Nachtigal MW, Kardami E, Cattini PA. FGF-2 protects cardiomyocytes from doxorubicin damage via protein kinase C-dependent effects on efflux transporters. Cardiovasc Res 2013; 98:56-63. [PMID: 23341575 DOI: 10.1093/cvr/cvt011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIMS The anti-cancer anthracycline doxorubicin (DOX) increases the risk of cardiac damage, indicating a need to protect the heart and still allow the benefits of drug treatment. Fibroblast growth factor-2 (FGF-2) is cardioprotective against ischaemia-reperfusion injury. Our aim is to investigate: (i) the ability of FGF-2 to protect against DOX-induced cardiomyocyte damage and (ii) the contribution of efflux drug transport to any increase in injury-resistance. METHODS AND RESULTS Neonatal rat cardiomyocyte damage was assessed by measuring cell death markers and lactate dehydrogenase (LDH) activity in the culture medium. LDH activity was increased significantly after incubation with 0.5 μM DOX for 24 h in the absence but not presence of 10 nM FGF-2; this beneficial effect of FGF-2 was blocked by tyrosine kinase (FGF) receptor inhibition. An increase in efflux drug transporter RNA levels was also detected after FGF-2 treatment in the presence of DOX. The beneficial effect of FGF-2 against cell damage and increased transporter RNA levels were blunted with protein kinase C (PKC) inhibition. Finally, FGF-2 stimulated efflux transport of calcein and DOX, and treatment with efflux transporter inhibitors significantly attenuated the protective effect of FGF-2 from DOX-induced injury. CONCLUSION Administered FGF-2 increases resistance to DOX-induced cardiomyocyte damage, by a mechanism dependent on PKC as well as regulation of efflux transporter production and/or function.
Collapse
Affiliation(s)
- Jie Wang
- Department of Physiology, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 3J7
| | | | | | | |
Collapse
|
16
|
Hasinoff BB, Patel D, Wu X. The Dual-Targeted HER1/HER2 Tyrosine Kinase Inhibitor Lapatinib Strongly Potentiates the Cardiac Myocyte-Damaging Effects of Doxorubicin. Cardiovasc Toxicol 2012; 13:33-47. [DOI: 10.1007/s12012-012-9183-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
17
|
Budde T, Haney J, Bien S, Schwebe M, Riad A, Tschöpe C, Staudt A, Jedlitschky G, Felix SB, Kroemer HK, Grube M. Acute exposure to doxorubicin results in increased cardiac P-glycoprotein expression. J Pharm Sci 2011; 100:3951-8. [PMID: 21590773 DOI: 10.1002/jps.22622] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 04/14/2011] [Accepted: 04/26/2011] [Indexed: 01/02/2023]
Abstract
Doxorubicin is a frequently used anticancer drug, but its use is restricted due to the occurrence of severe side effects, namely strong cardiotoxicity. It is known from cancer cells that doxorubicin enhanced the expression of its efflux pump P-glycoprotein (P-gp), which may modulate local drug concentrations. We therefore studied the cardiac expression of P-gp in doxorubicin-treated mice. Mice were treated with doxorubicin, and P-gp expression was studied after 1, 3, and 5 days. Thereby, we could show a significant upregulation of abcb1a (162 ± 15% of control) and abcb1b (418 ± 110% of control) mRNA transcripts after 3 days. On protein level, western blot analysis and immunofluorescence staining revealed a similar finding 5 days after doxorubicin administration. In addition, these results could be confirmed by in vitro models using primary rat cardiomyocytes and the murine cardiomyocyte-like HL-1 cells. Besides an enhanced mRNA and protein expression, doxorubicin-treated HL-1 cells also demonstrated an enhanced P-gp function as assessed by a daunorubicin accumulation assay. Our in vivo and in vitro results demonstrate a cardiac upregulation of P-gp in doxorubicin-treated mice on expression and functional level. This finding may be relevant for cardiac tissue concentrations of P-gp substrates and may represent a mechanism in cardiac self-protection against xenobiotics.
Collapse
Affiliation(s)
- Thomas Budde
- Department of Pharmacology, Research Center of Pharmacology and Experimental Therapeutics, Ernst-Moritz-Arndt University, Greifswald, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Protective effect of tetrahydroxystilbene glucoside on cardiotoxicity induced by doxorubicin in vitro and in vivo. Acta Pharmacol Sin 2009; 30:1479-87. [PMID: 19890356 DOI: 10.1038/aps.2009.144] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM To test the effect of 2,3,5,4'-tetrahydroxystilbene-2-O-beta-D-glucoside (THSG) on doxorubicin (DOX)-induced cardiotoxicity. METHODS We used neonate rat cardiomyocytes and an acute mouse model of DOX-induced cardiotoxicity to examine the protective effect of THSG. RESULTS In the mouse model, administration of THSG significantly reduced DOX-induced cardiotoxicity, including animal mortality, histopathological changes, and levels of serum creatine kinase (CK) and lactate dehydrogenase (LDH). Moreover, THSG was able to attenuate the increased malondialdehyde (MDA) and decreased reduced glutathione (GSH) caused by DOX. In in vitro studies, THSG 10-300 micromol/L ameliorated DOX-induced cardiomyocyte apoptosis in a concentration-dependent manner. Further studies showed that THSG inhibited reactive oxygen species (ROS) generation and prevented DOX-induced loss of mitochondrial membrane potential, caspase-3 activation and upregulation of Bax protein expression. We observed a protective response against damage after DOX treatment. The level of Bcl-2 protein was increased. Additionally, THSG inhibited a DOX-induced [Ca(2+)] increase. CONCLUSION These results showed that THSG protected against DOX-induced cardiotoxicity by decreasing ROS generation and intracellular [Ca(2+)] and by inhibiting apoptotic signaling pathways.
Collapse
|
19
|
Farokhpour M, Karbalaie K, Tanhaei S, Nematollahi M, Etebari M, Sadeghi HM, Nasr-Esfahani MH, Baharvand H. Embryonic stem cell-derived cardiomyocytes as a model system to study cardioprotective effects of dexamethasone in doxorubicin cardiotoxicity. Toxicol In Vitro 2009; 23:1422-8. [PMID: 19596060 DOI: 10.1016/j.tiv.2009.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 06/24/2009] [Accepted: 07/07/2009] [Indexed: 12/15/2022]
Abstract
Embryonic stem cell (ESC)-derived beating cardiomyocytes may be considered as a suitable model for in vitro assessment of pharmacological and toxicological studies. In this model, laboratory animals are not required. In addition, physiological functions, such as heart beat, are assessed rather than single parameters such as cell viability. Here we report that doxorubicin (DOX) cardiotoxicity on mouse ESC-derived beating cardiomyocytes can be ameliorated by treatment with dexamethasone (DEX) when DEX is administrated only before DOX and not in combination with DOX. DEX effect appears to be mediated via glucocorticoid receptor and increases cardiomyocyte-specific gene expression. Cardiotoxicity of DOX can be augmented by calcium channel blocker, verapamil (VER) which also decreases the expression of cardiac gene markers. This model provides us with a clinical suggestion which proposes that the beneficial effect of DEX is obtained when DEX was added before DOX administration.
Collapse
Affiliation(s)
- Mahboubeh Farokhpour
- Department of Cell and Molecular Biology, Royan Institute for Animal Biotechnology, Esfahan, Iran
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Costa VM, Ferreira LM, Branco PS, Carvalho F, Bastos ML, Carvalho RA, Carvalho M, Remião F. Cross-Functioning between the Extraneuronal Monoamine Transporter and Multidrug Resistance Protein 1 in the Uptake of Adrenaline and Export of 5-(Glutathion-S-yl)adrenaline in Rat Cardiomyocytes. Chem Res Toxicol 2008; 22:129-135. [DOI: 10.1021/tx8002577] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Vera Marisa Costa
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Department, Faculty of Pharmacy, University of Porto, Porto, Portugal, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal, Neurosciences Center of Coimbra, Department of Biochemistry, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal, and CEBIMED, Faculty of Health Sciences, University Fernando Pessoa, Porto, Portugal
| | - Luísa Maria Ferreira
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Department, Faculty of Pharmacy, University of Porto, Porto, Portugal, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal, Neurosciences Center of Coimbra, Department of Biochemistry, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal, and CEBIMED, Faculty of Health Sciences, University Fernando Pessoa, Porto, Portugal
| | - Paula Sério Branco
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Department, Faculty of Pharmacy, University of Porto, Porto, Portugal, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal, Neurosciences Center of Coimbra, Department of Biochemistry, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal, and CEBIMED, Faculty of Health Sciences, University Fernando Pessoa, Porto, Portugal
| | - Félix Carvalho
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Department, Faculty of Pharmacy, University of Porto, Porto, Portugal, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal, Neurosciences Center of Coimbra, Department of Biochemistry, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal, and CEBIMED, Faculty of Health Sciences, University Fernando Pessoa, Porto, Portugal
| | - Maria Lourdes Bastos
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Department, Faculty of Pharmacy, University of Porto, Porto, Portugal, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal, Neurosciences Center of Coimbra, Department of Biochemistry, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal, and CEBIMED, Faculty of Health Sciences, University Fernando Pessoa, Porto, Portugal
| | - Rui Albuquerque Carvalho
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Department, Faculty of Pharmacy, University of Porto, Porto, Portugal, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal, Neurosciences Center of Coimbra, Department of Biochemistry, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal, and CEBIMED, Faculty of Health Sciences, University Fernando Pessoa, Porto, Portugal
| | - Márcia Carvalho
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Department, Faculty of Pharmacy, University of Porto, Porto, Portugal, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal, Neurosciences Center of Coimbra, Department of Biochemistry, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal, and CEBIMED, Faculty of Health Sciences, University Fernando Pessoa, Porto, Portugal
| | - Fernando Remião
- REQUIMTE (Rede de Química e Tecnologia), Toxicology Department, Faculty of Pharmacy, University of Porto, Porto, Portugal, REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal, Neurosciences Center of Coimbra, Department of Biochemistry, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal, and CEBIMED, Faculty of Health Sciences, University Fernando Pessoa, Porto, Portugal
| |
Collapse
|
21
|
Theodossiou TA, Galanou MC, Paleos CM. Novel amiodarone-doxorubicin cocktail liposomes enhance doxorubicin retention and cytotoxicity in DU145 human prostate carcinoma cells. J Med Chem 2008; 51:6067-74. [PMID: 18783209 DOI: 10.1021/jm800493j] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have developed novel cocktail liposomes bearing doxorubicin in their hydrophilic cores, and amiodarone, a potent multidrug resistance inhibitor, in their lipid bilayers. The efficacy of these liposomes was studied in DU145 human prostate carcinoma cells. Intracellular calcein retention, which is inversely proportional to multidrug resistance activity, significantly increased following cell incubation with amiodarone loaded liposomes. Fluorescence confocal microscopy on cells incubated with the cocktail liposomes revealed enhanced intranuclear doxorubicin accumulation. Two liposomal drug concentration combinations were employed to assess the differential cytotoxicity of the cocktail liposomes, doxorubicin (1.4 microM)-amiodarone (15 microM) and doxorubicin 3 (microM)-amiodarone (45 microM), and two incubation times, 5 and 19 h. Cell toxicity was determined by XTT assays at 24, 48, and 72 h following incubation and was significantly enhanced for incubation with the cocktail liposomes. On the whole, we believe that these liposomes will greatly contribute to the cancer chemotherapy arena.
Collapse
|
22
|
Outomuro D, Grana DR, Azzato F, Milei J. Adriamycin-induced myocardial toxicity: New solutions for an old problem? Int J Cardiol 2007; 117:6-15. [PMID: 16863672 DOI: 10.1016/j.ijcard.2006.05.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Revised: 03/28/2006] [Accepted: 05/18/2006] [Indexed: 11/19/2022]
Abstract
Adriamycin is a potent and broad-spectrum antineoplastic agent that plays a major role in cancer chemotherapy. Unfortunately, its use has been hampered by conventional toxicities and cardiotoxicity manifested by congestive cardiomyopathy. Adriamycin is particularly toxic to heart tissue and constitutes a major cause of morbidity and mortality due to its complex pathogenesis. In this review, the different forms of cardiotoxicity produced by adriamycin as well as the biochemical changes induced by this drug are summarized. Secondly, the current hypotheses proposed to explain adriamycin-induced myocardial damage (the iron and free-radical hypothesis, the metabolic hypothesis, the "unifying hypothesis" and apoptosis) and the attempts to reduce adriamycin-induced myocardial toxicity are discussed (e.g. dose limitation, close cardiac monitoring, alteration of dosage schedules, development of new anthracycline analogs, and the administration of protective agents and liposomal encapsulation). Finally, we summarized our own experimental and clinical experience in ameliorating and or preventing adriamycin-induced cardiotoxicity and the latest attempts to prevent and/or monitor cardiac function. According to this, a combination of usual doses of calcium antagonist drugs plus vitamins A and E seems advisable.
Collapse
Affiliation(s)
- Delia Outomuro
- Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires-Conicet, Argentina
| | | | | | | |
Collapse
|
23
|
Couture L, Nash JA, Turgeon J. The ATP-binding cassette transporters and their implication in drug disposition: a special look at the heart. Pharmacol Rev 2006; 58:244-58. [PMID: 16714487 DOI: 10.1124/pr.58.2.7] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The passage of drugs across cell membranes dictates their absorption, distribution, metabolism, and excretion. This process is determined by several factors including the molecular weight of the compounds, their shape, degree of ionization, and binding to proteins. Accumulation of xenobiotics into tissues does not depend only on their ability to enter cells, but also on their ability to leave them. For instance, the role of efflux transporters such as ATP-binding cassette (ABC) proteins in the disposition of drugs is now well recognized. Actually, ABC transporters act in synergy with drug-metabolizing enzymes to protect the organism from toxic compounds. The most studied transporter from the ABC transporter superfamily, P-glycoprotein, was found to be overexpressed in tumor cells and associated with an acquired resistance to several anticancer drugs. P-glycoprotein, thought at first to be confined to tumor cells, was subsequently recognized to be expressed in normal tissues such as the liver, kidney, intestine, and heart. Even though information remains rather limited on the functional role of ABC transporters in the myocardium, it is hypothesized that they may modulate efficacy and toxicity of cardioactive agents. This review addresses recent progress on knowledge about the ABC transporters in drug disposition and more precisely their role in drug distribution to the heart.
Collapse
Affiliation(s)
- Lucie Couture
- Faculté de Pharmacie, Université de Montréal, C.P. 6128, Succursale Centre-Ville, Montreal, Quebec, Canada, H3C 3J7
| | | | | |
Collapse
|
24
|
Partridge CR, Johnson CD, Ramos KS. In vitro models to evaluate acute and chronic injury to the heart and vascular systems. Toxicol In Vitro 2005; 19:631-44. [PMID: 15893448 DOI: 10.1016/j.tiv.2005.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2004] [Accepted: 03/16/2005] [Indexed: 01/27/2023]
Abstract
Multiple in vitro model systems are currently available to evaluate structure and function relationships in the cardiovascular system as well as the system's response to injury. As the level of molecular sophistication continues to advance, so does the level of complexity of the analysis. One of the most daunting tasks faced by researchers interested in studying cardiovascular function and injury is the selection of the system or systems best suited to answer the particular question at hand. In order to successfully apply any given model system, the researcher must recognize the advantages and limitations in the system of choice. This review provides a listing of the historical and modern techniques used to study cardiovascular function and chemically-induced toxicity. With the growing number of new pharmaceuticals discovered each year, it is imperative to use experimental model systems that allow for identification of targets that participate in or mediate adverse outcomes. Clearly, in vitro analysis cannot replace in vivo experimentation, but the methods currently available allow for a reduction in the number of animals used for experimentation and a better understanding of the complexity associated with the injury response.
Collapse
Affiliation(s)
- Charles R Partridge
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, 580 S Preston, Louisville, KY 40292, USA
| | | | | |
Collapse
|
25
|
Darvari R, Boroujerdi M. Investigation of the influence of modulation of P-glycoprotein by a multiple dosing regimen of tamoxifen on the pharmacokinetics and toxicodynamics of doxorubicin. Cancer Chemother Pharmacol 2005; 56:497-509. [PMID: 15937726 DOI: 10.1007/s00280-005-1001-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Accepted: 02/07/2005] [Indexed: 10/25/2022]
Abstract
PURPOSE The in vivo effect of modulators of P-glycoprotein (Pgp) on organ accumulation of substrates of Pgp has not been fully investigated. We investigated the influence of a Pgp modulator (tamoxifen, TAM) on the pharmacokinetics and toxicodynamics of a Pgp substrate (doxorubicin, DOX) in rats. METHODS TAM was administered daily for 11 days before the administration of DOX in male Sprague-Dawley rats, with all doses being clinically relevant. The experimental design of the project consisted of two different protocols. One was to investigate the effect of DOX on the time course of Pgp-ATPase activity, sarcoplasmic reticulum Ca(2+) -ATPase (SERCA) activity, and DOX concentration in the heart, liver, and kidneys of TAM-pretreated animals; the other protocol was to study the effect of TAM pretreatment on the disposition of DOX in the body by investigating its time course in plasma, urine and bile. RESULTS The simultaneous curve fitting of plasma data with urine and bile data with the help of the related pharmacokinetic equations provided the calculated parameters and constants. The first-order rate constants between the central and the myocardial compartments (k(1H) and k(H1)) were decreased in the TAM-treated group. The treatment also significantly reduced the k(1H)/k(H1) ratio in comparison to that of the control group. The first-order biliary elimination rate constant (k(b)) was significantly decreased (29%) in the TAM-treated group. The reduction was estimated in comparison with that of the control group. This reduction could be attributed to the inhibitory effect of TAM on Pgp located on biliary canicular membranes. The initial reduction of Pgp activity in TAM-treated group was at 60% of the basal level. The activity declined and reached a plateau at 20% of the basal activity after 6 h and remained at that level for 24 h. The area under the curves of Pgp-ATPase activity time (AUC(Activity 0-24)) following DOX administration in TAM-treated group was significantly lower than that of the control group, indicating an overall inhibitory effect of TAM on Pgp-ATPase activity under the protocol of this study. The area under the curves of the SERCA activity-time curve following DOX administration in TAM-treated group demonstrated a 15% reduction in AUC(Activity 0-24) in comparison with that of the control group, an indication of increased toxicity. The amount of myocardial Pgp in the 24-h period following DOX administration was comparable to the control group and showed no significant deviation from the basal levels of the protein. CONCLUSIONS The effect of TAM on DOX accumulation in the myocardial tissue and the increase in cardiotoxicity can be related to the net inhibitory effect of TAM on the efflux activity of Pgp in the heart. The results of the present study supported the hypothesis of the project that multiple regimen pretreatment with Pgp modulator TAM increases the DOX accumulation in the heart and promotes DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ramin Darvari
- Epic Therapeutics Inc., a Subsidiary of Baxter Healthcare Corporation, Norwood, MA 02062, USA
| | | |
Collapse
|
26
|
Büyükokuroğlu ME, Taysi S, Buyukavci M, Bakan E. Prevention of acute adriamycin cardiotoxicity by dantrolene in rats. Hum Exp Toxicol 2004; 23:251-6. [PMID: 15222403 DOI: 10.1191/0960327104ht443oa] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Possible preventive effect of dantrolene against the peroxidative damage in rat heart which was induced by the administration of an acute dose of adriamycin (ADR, 20 mg/kg, i.p.) has been examined. Forty-eight hours after ADR administration, biochemical changes including the activities of serum creatine kinase isoenzyme (CK-MB), lactate dehydrogenase (LDH) and aspartate aminotransferase (AST) and the levels of malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in heart tissue were measured. Pretreatment of rats with dantrolene, given i.p. 30 min prior to ADR injection, substantially reduced the peroxidative damage in the myocardium, and markedly lowered the serum CK-MB, LDH and AST. The protective effects obtained by dantrolene administration, however, were not complete and did not reach those of the control group. Dantrolene, at 5 mg/kg, was useful to obtain significant protective effects, while the protector effect of higher dantrolene dosing level (10 mg/kg) was weak or absent. These results suggest that, at least in part, due to antioxidative properties, dantrolene may provide a significant protective effect against acute ADR-induced cardiotoxicity.
Collapse
|
27
|
Kitano T, Iizasa H, Terasaki T, Asashima T, Matsunaga N, Utoguchi N, Watanabe Y, Obinata M, Ueda M, Nakashima E. Polarized glucose transporters and mRNA expression properties in newly developed rat syncytiotrophoblast cell lines, TR-TBTs. J Cell Physiol 2002; 193:208-18. [PMID: 12384998 DOI: 10.1002/jcp.10165] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In this study, we have established new syncytiotrophoblast cell lines (TR-TBTs) from the recently developed transgenic rat harboring temperature-sensitive simian virus 40 large T-antigen gene (Tg-rat). Four conditionally immortalized syncytiotrophoblast cell lines (TR-TBT 18d-1 approximately 4) were obtained from pregnant Tg-rats at gestational day 18. These cell lines had a syncytium-like morphology, could be prepared as monolayers, expressed cytokeratins and rat syncytiotrophoblast markers, and exhibited apical or basal GLUT1 localizations and apical GLUT3 localizations. TR-TBTs express large T-antigen and grow well at 33 degrees C with a doubling time of about 30 h. TR-TBTs have processes for the uptake of dehydroepiandrosteron-3-sulfate (DHEAS) and these are predominantly located on the basal side, and this is the first report of an in vitro model of blood placental barrier (BPB) able to incorporate DHEAS. Therefore, TR-TBTs are an appropriate in vitro model for investigating carrier-mediated transport functions at the BPB. Moreover, TR-TBTs express betaine/GABA transporter (GAT-2/BGT-1), concentrative nucleoside transporter 2 (CNT2), equilibrative nucleoside transporter 1 (ENT1), and ENT2 and the expression of these transporters has been reported in blood-brain barrier (BBB). Thus, the expression patterns of nucleoside and neurotransmitter transporters examined are quite similar in both the BPB and BBB.
Collapse
Affiliation(s)
- Tomohide Kitano
- Department of Pharmaceutics, Kyoritsu College of Pharmacy, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Weiss M, Kang W. P-glycoprotein inhibitors enhance saturable uptake of idarubicin in rat heart: pharmacokinetic/pharmacodynamic modeling. J Pharmacol Exp Ther 2002; 300:688-94. [PMID: 11805234 DOI: 10.1124/jpet.300.2.688] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Little is known about cardiac uptake kinetics of idarubicin, including a possible protective role of P-glycoprotein (Pgp)-mediated transport. This study therefore investigated uptake and negative inotropic action of idarubicin in the single-pass isolated perfused rat heart by using a pharmacokinetic/pharmacodynamic modeling approach. Idarubicin was administered as a 10-min constant infusion of 0.5 mg followed by a 70-min washout period in the absence and presence of the Pgp antagonists verapamil or amiodarone. Outflow concentration and left ventricular developed pressure were measured and the model parameters were estimated by simultaneous nonlinear regression. The results indicate the existence of a saturable, Michaelis-Menten type uptake process into the heart (K(m) = 3.06 microM, V(max) = 46.0 microM/min). Verapamil and amiodarone significantly enhanced the influx rate (V(max) increased 1.8-fold), suggesting that idarubicin is transported by Pgp directly out of the membrane before it gets into the cell. Verapamil and amiodarone attenuated the negative inotropic action of idarubicin, which was linked to the intracellular concentration of idarubicin.
Collapse
Affiliation(s)
- Michael Weiss
- Section of Pharmacokinetics, Department of Pharmacology, Martin Luther University Halle-Wittenberg, Halle, Germany.
| | | |
Collapse
|
29
|
Chlopčíková Š, Psotová J, Miketová P. NEONATAL RAT CARDIOMYOCYTES - A MODEL FOR THE STUDY OF MORPHOLOGICAL, BIOCHEMICAL AND ELECTROPHYSIOLOGICAL CHARACTERISTICS OF THE HEART. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2001. [DOI: 10.5507/bp.2001.011] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|