1
|
Yang BF, Li D, Liu CL, Luo Y, Shi J, Guo XQ, Fan HJ, Lv Q. Advances in rhabdomyolysis: A review of pathogenesis, diagnosis, and treatment. Chin J Traumatol 2025:S1008-1275(25)00010-0. [PMID: 40082140 DOI: 10.1016/j.cjtee.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/21/2024] [Accepted: 10/25/2024] [Indexed: 03/16/2025] Open
Abstract
Rhabdomyolysis (RM) is a multifactorial clinical syndrome characterized by the disintegration and necrosis of muscle tissue, leading to the release of cellular contents into the circulation. One of the most severe complications of RM is acute kidney injury, with a mortality rate of 20%-50%. Early and timely diagnosis is the key to improving the prognosis of patients with RM. The etiology of RM is complex and associated with various traumas, drugs, medications, and hereditary diseases, and the clinical symptoms are nonspecific. Therefore, its diagnosis highly relies on the doctor's experience and the level of medical equipment. However, RM often occurs in situations with limited medical resources, such as natural disasters, battlefields, and large-scale traffic accidents. In these scenarios, the varying levels of expertise among rescue personnel can lead to delays in diagnosis and treatment, thereby increasing the risk of mortality. This article provides a comprehensive review of the etiology, pathogenesis, complications, diagnostic, and treatment methods of RM. It also aims to offer new perspectives on the diagnosis and prognosis of RM by integrating machine learning and artificial intelligence. It is believed that this article can help pre-hospital rescuers and in-hospital doctors have a comprehensive understanding of RM to improve the patients' outcomes and overcome the challenges.
Collapse
Affiliation(s)
- Bo-Fan Yang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China
| | - Duo Li
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, Zhejiang Province, China
| | - Chun-Li Liu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, Zhejiang Province, China
| | - Yu Luo
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, Zhejiang Province, China
| | - Jie Shi
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, Zhejiang Province, China
| | - Xiao-Qin Guo
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, Zhejiang Province, China
| | - Hao-Jun Fan
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, Zhejiang Province, China
| | - Qi Lv
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin, 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
2
|
Bogomolova AP, Katrukha IA. Troponins and Skeletal Muscle Pathologies. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2083-2106. [PMID: 39865025 DOI: 10.1134/s0006297924120010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/19/2024] [Accepted: 12/01/2024] [Indexed: 01/28/2025]
Abstract
Skeletal muscles account for ~30-40% of the total weight of human body and are responsible for its most important functions, including movement, respiration, thermogenesis, and glucose and protein metabolism. Skeletal muscle damage negatively impacts the whole-body functioning, leading to deterioration of the quality of life and, in severe cases, death. Therefore, timely diagnosis and therapy for skeletal muscle dysfunction are important goals of modern medicine. In this review, we focused on the skeletal troponins that are proteins in the thin filaments of muscle fibers. Skeletal troponins play a key role in regulation of muscle contraction. Biochemical properties of these proteins and their use as biomarkers of skeletal muscle damage are described in this review. One of the most convenient and sensitive methods of protein biomarker measurement in biological liquids is immunochemical analysis; hence, we examined the factors that influence immunochemical detection of skeletal troponins and should be taken into account when developing diagnostic test systems. Also, we reviewed the available data on the skeletal troponin mutations that are considered to be associated with pathologies leading to the development of diseases and discussed utilization of troponins as drug targets for treatment of the skeletal muscle disorders.
Collapse
Affiliation(s)
- Agnessa P Bogomolova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Hytest Ltd., Turku, Finland
| | - Ivan A Katrukha
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
- Hytest Ltd., Turku, Finland
| |
Collapse
|
3
|
Lorenzoni R, Davies S, Cordenonsi LM, Roggia I, Viçosa JADS, Mezzomo NJ, de Oliveira AL, do Carmo GM, Vitalis G, Gomes P, Raffin RP, Alves OL, Vaucher RDA, Rech VC. Lipid-core nanocapsules containing simvastatin do not affect the biochemical and hematological indicators of toxicity in rats. Toxicol Res (Camb) 2024; 13:tfae189. [PMID: 39539252 PMCID: PMC11557222 DOI: 10.1093/toxres/tfae189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Our research group previously studied the effectiveness of lipid-core nanocapsules (LNC) containing simvastatin (SV-LNC) in treating cognitive impairment in rats. While our results were promising, we needed to evaluate the potential toxicity of the nanoparticles themselves. This study aimed to compare the biochemical and hematological parameters of adult Wistar rats receiving LNC or SV-LNC to those receiving low doses of simvastatin crystals dispersed in a saline solution over 45 days. We discovered that LNC and SV-LNC, which are both nanometers in size with low polydispersity index, negative zeta potential, and high SV encapsulation efficacy, were not more toxic than SV crystals based on various biochemical markers of hepatic, pancreatic, renal, mineral, bony, alkaline phosphatase, glucose, and uric acid damage. Furthermore, LNC exhibited no toxicity for hematological parameters, including red and white blood cell counts. Based on this animal model of toxicological study, our findings suggest that long-term administration of LNC is a safe and promising nanocarrier.
Collapse
Affiliation(s)
- Ricardo Lorenzoni
- Franciscan University, Postgraduate Program in Nanosciences, Rua dos Andradas, 1614 CEP: 97010-032 Santa Maria, RS, Brazil
| | - Samuel Davies
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Brazil, Avenida Ipiranga 2752, Porto Alegre, RS, Brazil
| | - Leticia Malgarim Cordenonsi
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Sul, Brazil, Avenida Ipiranga 2752, Porto Alegre, RS, Brazil
| | - Isabel Roggia
- Franciscan University, Postgraduate Program in Nanosciences, Rua dos Andradas, 1614 CEP: 97010-032 Santa Maria, RS, Brazil
| | - José Alcides da Silva Viçosa
- Franciscan University, Postgraduate Program in Nanosciences, Rua dos Andradas, 1614 CEP: 97010-032 Santa Maria, RS, Brazil
| | - Nathana Jamille Mezzomo
- Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Amanda Lima de Oliveira
- Franciscan University, Postgraduate Program in Nanosciences, Rua dos Andradas, 1614 CEP: 97010-032 Santa Maria, RS, Brazil
| | - Guilherme Machado do Carmo
- Franciscan University, Postgraduate Program in Nanosciences, Rua dos Andradas, 1614 CEP: 97010-032 Santa Maria, RS, Brazil
| | - Graciela Vitalis
- Franciscan University, Postgraduate Program in Nanosciences, Rua dos Andradas, 1614 CEP: 97010-032 Santa Maria, RS, Brazil
| | - Patrícia Gomes
- Franciscan University, Postgraduate Program in Nanosciences, Rua dos Andradas, 1614 CEP: 97010-032 Santa Maria, RS, Brazil
| | - Renata Platcheck Raffin
- Franciscan University, Postgraduate Program in Nanosciences, Rua dos Andradas, 1614 CEP: 97010-032 Santa Maria, RS, Brazil
| | - Oswaldo Luiz Alves
- Solid State Chemistry Laboratory, Institute of Chemistry, University of Campinas, PO Box 6154, 13083-970 Campinas, SP Brazil
| | - Rodrigo De Almeida Vaucher
- Postgraduate Program in Biochemistry and Bioprospecting, Federal University of Pelotas, Capão do Leão, CEP 96010-900, RS, Brazil
| | - Virginia Cielo Rech
- Franciscan University, Postgraduate Program in Nanosciences, Rua dos Andradas, 1614 CEP: 97010-032 Santa Maria, RS, Brazil
| |
Collapse
|
4
|
Favere K, Van Hecke M, Eens S, Bosman M, Stobbelaar K, Hotterbeekx A, Kumar-Singh S, L Delputte P, Fransen E, De Sutter J, Guns PJ, Roskams T, Heidbuchel H. The natural history of CVB3 myocarditis in C57BL/6J mice: an extended in-depth characterization. Cardiovasc Pathol 2024; 72:107652. [PMID: 38750778 DOI: 10.1016/j.carpath.2024.107652] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND AND AIMS Viral infections are the leading cause of myocarditis. Besides acute cardiac complications, late-stage sequelae such as myocardial fibrosis may develop, importantly impacting the prognosis. Coxsackievirus B3 (CVB)-induced myocarditis in mice is the most commonly used translational model to study viral myocarditis and has provided the majority of our current understanding of the disease pathophysiology. Nevertheless, the late stages of disease, encompassing fibrogenesis and arrhythmogenesis, have been underappreciated in viral myocarditis research to date. The present study investigated the natural history of CVB-induced myocarditis in C57BL/6J mice, expanding the focus beyond the acute phase of disease. In addition, we studied the impact of sex and inoculation dose on the disease course. METHODS AND RESULTS C57BL/6J mice (12 weeks old; n=154) received a single intraperitoneal injection with CVB to induce viral myocarditis, or vehicle (PBS) as control. Male mice (n=92) were injected with 5 × 105 (regular dose) (RD) or 5 × 106 (high dose) (HD) plaque-forming units of CVB, whereas female mice received the RD only. Animals were sacrificed 1, 2, 4, 8, and 11 weeks after CVB or PBS injection. Virally inoculated mice developed viral disease with a temporary decline in general condition and weight loss, which was less pronounced in female animals (P<.001). In male CVB mice, premature mortality occurred between days 8 and 23 after inoculation (RD: 21%, HD: 20%), whereas all female animals survived. Over the course of disease, cardiac inflammation progressively subsided, with faster resolution in female mice. There were no substantial group differences in the composition of the inflammatory cell infiltrates: predominance of cytotoxic T cells at day 7 and 14, and a switch from arginase1-reactive macrophages to iNOS-reactive macrophages from day 7 to 14 were the main findings. There was concomitant development and maturation of different patterns of myocardial fibrosis, with enhanced fibrogenesis in male mice. Virus was almost completely cleared from the heart by day 14. Serum biomarkers of cardiac damage and cardiac expression of remodeling genes were temporarily elevated during the acute phase of disease. Cardiac CTGF gene upregulation was less prolonged in female CVB animals. In vivo electrophysiology studies at weeks 8 and 11 demonstrated that under baseline conditions (i.e. in the absence of proarrhythmogenic drugs), ventricular arrhythmias could only be induced in CVB animals. The cumulative arrhythmia burden throughout the entire stimulation protocol was not significantly different between CVB and control groups. CONCLUSION CVB inoculation in C57BL/6J mice represents a model of acute self-limiting viral myocarditis, with progression to different patterns of myocardial fibrosis. Sex, but not inoculation dose, seems to modulate the course of disease.
Collapse
Affiliation(s)
- Kasper Favere
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; Department of Cardiology, Antwerp University Hospital, 2650 Antwerp, Belgium; Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium.
| | - Manon Van Hecke
- Translational Cell & Tissue Research, Department of Imaging & Pathology, University of Leuven, 3000 Leuven, Belgium
| | - Sander Eens
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium
| | - Matthias Bosman
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium
| | - Kim Stobbelaar
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, 2610 Antwerp, Belgium
| | - An Hotterbeekx
- Molecular Pathology Group, FGGW-Laboratory of Cell Biology and Histology, University of Antwerp, 2610 Antwerp, Belgium
| | - Samir Kumar-Singh
- Molecular Pathology Group, FGGW-Laboratory of Cell Biology and Histology, University of Antwerp, 2610 Antwerp, Belgium
| | - Peter L Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, 2610 Antwerp, Belgium
| | - Erik Fransen
- Centre for Medical Genetics, University of Antwerp, 2610 Antwerp, Belgium
| | - Johan De Sutter
- Department of Internal Medicine, Ghent University, 9000 Ghent, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, GENCOR, University of Antwerp, 2610 Antwerp, Belgium
| | - Tania Roskams
- Translational Cell & Tissue Research, Department of Imaging & Pathology, University of Leuven, 3000 Leuven, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, 2610 Antwerp, Belgium; Department of Cardiology, Antwerp University Hospital, 2650 Antwerp, Belgium
| |
Collapse
|
5
|
Reinal I, Ontoria-Oviedo I, Selva M, Casini M, Peiró-Molina E, Fambuena-Santos C, Climent AM, Balaguer J, Cañete A, Mora J, Raya Á, Sepúlveda P. Modeling Cardiotoxicity in Pediatric Oncology Patients Using Patient-Specific iPSC-Derived Cardiomyocytes Reveals Downregulation of Cardioprotective microRNAs. Antioxidants (Basel) 2023; 12:1378. [PMID: 37507917 PMCID: PMC10376252 DOI: 10.3390/antiox12071378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/25/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Anthracyclines are widely used in the treatment of many solid cancers, but their efficacy is limited by cardiotoxicity. As the number of pediatric cancer survivors continues to rise, there has been a concomitant increase in people living with anthracycline-induced cardiotoxicity. Accordingly, there is an ongoing need for new models to better understand the pathophysiological mechanisms of anthracycline-induced cardiac damage. Here we generated induced pluripotent stem cells (iPSCs) from two pediatric oncology patients with acute cardiotoxicity induced by anthracyclines and differentiated them to ventricular cardiomyocytes (hiPSC-CMs). Comparative analysis of these cells (CTX hiPSC-CMs) and control hiPSC-CMs revealed that the former were significantly more sensitive to cell injury and death from the anthracycline doxorubicin (DOX), as measured by viability analysis, cleaved caspase 3 expression, oxidative stress, genomic and mitochondrial damage and sarcomeric disorganization. The expression of several mRNAs involved in structural integrity and inflammatory response were also differentially affected by DOX. Functionally, optical mapping analysis revealed higher arrythmia complexity after DOX treatment in CTX iPSC-CMs. Finally, using a panel of previously identified microRNAs associated with cardioprotection, we identified lower levels of miR-22-3p, miR-30b-5p, miR-90b-3p and miR-4732-3p in CTX iPSC-CMs under basal conditions. Our study provides valuable phenotype information for cellular models of cardiotoxicity and highlights the significance of using patient-derived cardiomyocytes for studying the associated pathogenic mechanisms.
Collapse
Affiliation(s)
- Ignacio Reinal
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Imelda Ontoria-Oviedo
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Marta Selva
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Marilù Casini
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
| | - Esteban Peiró-Molina
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | | | - Andreu M Climent
- ITACA Institute, Universitat Politècnica de València, 46026 Valencia, Spain
| | - Julia Balaguer
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Transtational Research in Cancer Unit-Pediatric Oncology, Health Research Institute Hospital La Fe, 46026 Valencia, Spain
| | - Adela Cañete
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Transtational Research in Cancer Unit-Pediatric Oncology, Health Research Institute Hospital La Fe, 46026 Valencia, Spain
- Department of Pediatrics, University of Valencia, 46010 Valencia, Spain
| | - Jaume Mora
- Oncology Service, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Ángel Raya
- Regenerative Medicine Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain
- Program for Clinical Translation of Regenerative Medicine in Catalonia-P-[CMRC], L'Hospitalet de Llobregat, 08908 Barcelona, Spain
- Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Carlos III Institute of Health, 28029 Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
- Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Carlos III Institute of Health, 28029 Madrid, Spain
- Department of Pathology, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
6
|
Kim-Muller JY, Song L, LaCarubba Paulhus B, Pashos E, Li X, Rinaldi A, Joaquim S, Stansfield JC, Zhang J, Robertson A, Pang J, Opsahl A, Boucher M, Breen D, Hales K, Sheikh A, Wu Z, Zhang BB. GDF15 neutralization restores muscle function and physical performance in a mouse model of cancer cachexia. Cell Rep 2023; 42:111947. [PMID: 36640326 DOI: 10.1016/j.celrep.2022.111947] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/06/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Cancer cachexia is a disorder characterized by involuntary weight loss and impaired physical performance. Decline in physical performance of patients with cachexia is associated with poor quality of life, and currently there are no effective pharmacological interventions that restore physical performance. Here we examine the effect of GDF15 neutralization in a mouse model of cancer-induced cachexia (TOV21G) that manifests weight loss and muscle function impairments. With comprehensive assessments, our results demonstrate that cachectic mice treated with the anti-GDF15 antibody mAB2 exhibit body weight gain with near-complete restoration of muscle mass and markedly improved muscle function and physical performance. Mechanistically, the improvements induced by GDF15 neutralization are primarily attributed to increased caloric intake, while altered gene expression in cachectic muscles is restored in caloric-intake-dependent and -independent manners. The findings indicate potential of GDF15 neutralization as an effective therapy to enhance physical performance of patients with cachexia.
Collapse
Affiliation(s)
- Ja Young Kim-Muller
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - LouJin Song
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Brianna LaCarubba Paulhus
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Evanthia Pashos
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Xiangping Li
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Anthony Rinaldi
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Stephanie Joaquim
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - John C Stansfield
- Biostatistics, Early Clinical Development, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Jiangwei Zhang
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 10777 Science Center Dr., San Diego, CA, USA
| | - Andrew Robertson
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Jincheng Pang
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Alan Opsahl
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Magalie Boucher
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Danna Breen
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Katherine Hales
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Abdul Sheikh
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Zhidan Wu
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Bei B Zhang
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA.
| |
Collapse
|
7
|
Tonry C, Russel-Hallinan A, McCune C, Collier P, Harbinson M, Dixon L, Watson CJ. Circulating biomarkers for management of cancer therapeutics related cardiac dysfunction. Cardiovasc Res 2022; 119:710-728. [PMID: 35640873 PMCID: PMC10153425 DOI: 10.1093/cvr/cvac087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 05/09/2022] [Accepted: 05/15/2022] [Indexed: 11/14/2022] Open
Abstract
Cancer therapeutics related cardiac dysfunction (CTRCD) has emerged as a major cause of morbidity and mortality in cancer survivors. Effective clinical management of CTRCD is impeded by a lack of sensitive diagnostic and prognostic strategies. Circulating molecular markers could potentially address this need as they are often indicative of cardiac stress before cardiac damage can be detected clinically. A growing understanding of the underlying physiological mechanisms for CTRCD has inspired research efforts to identify novel pathophysiologically-relevant biomarkers that may also guide development of cardio-protective therapeutic approaches. The purpose of this review is to evaluate current circulating biomarkers of cardiac stress and their potential role in diagnosis and management of CTRCD. We also discuss some emerging avenues for CTRCD-focused biomarker investigations.
Collapse
Affiliation(s)
- Claire Tonry
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Adam Russel-Hallinan
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Claire McCune
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | | | | | | | - Chris J Watson
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
8
|
Schofield AL, Brown JP, Brown J, Wilczynska A, Bell C, Glaab WE, Hackl M, Howell L, Lee S, Dear JW, Remes M, Reeves P, Zhang E, Allmer J, Norris A, Falciani F, Takeshita LY, Seyed Forootan S, Sutton R, Park BK, Goldring C. Systems analysis of miRNA biomarkers to inform drug safety. Arch Toxicol 2021; 95:3475-3495. [PMID: 34510227 PMCID: PMC8492583 DOI: 10.1007/s00204-021-03150-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023]
Abstract
microRNAs (miRNAs or miRs) are short non-coding RNA molecules which have been shown to be dysregulated and released into the extracellular milieu as a result of many drug and non-drug-induced pathologies in different organ systems. Consequently, circulating miRs have been proposed as useful biomarkers of many disease states, including drug-induced tissue injury. miRs have shown potential to support or even replace the existing traditional biomarkers of drug-induced toxicity in terms of sensitivity and specificity, and there is some evidence for their improved diagnostic and prognostic value. However, several pre-analytical and analytical challenges, mainly associated with assay standardization, require solutions before circulating miRs can be successfully translated into the clinic. This review will consider the value and potential for the use of circulating miRs in drug-safety assessment and describe a systems approach to the analysis of the miRNAome in the discovery setting, as well as highlighting standardization issues that at this stage prevent their clinical use as biomarkers. Highlighting these challenges will hopefully drive future research into finding appropriate solutions, and eventually circulating miRs may be translated to the clinic where their undoubted biomarker potential can be used to benefit patients in rapid, easy to use, point-of-care test systems.
Collapse
Affiliation(s)
- Amy L Schofield
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Joseph P Brown
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Jack Brown
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Ania Wilczynska
- bit.bio, Babraham Research Campus, The Dorothy Hodgkin Building, Cambridge, CB22 3FH, UK
| | - Catherine Bell
- CVRM Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Warren E Glaab
- Merck & Co., Inc, 770 Sumneytown Pike, West Point, PA, 19486, USA
| | | | - Lawrence Howell
- GlaxoSmithKline (GSK), Stevenage, Greater Cambridge Area, UK
| | - Stephen Lee
- ABHI, 1 Duchess St, 4th Floor, Suite 2, London, W1W 6AN, UK
| | - James W Dear
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Mika Remes
- Genomics EMEA, QIAGEN Aarhus, Prismet, Silkeborgvej 2, 8000, Aarhus C, Denmark
| | - Paul Reeves
- Arcis Biotechnology Limited, Suite S07, Techspace One, Sci-tech Daresbury, Keckwick Lane, Daresbury, Warrington, WA4 4AB, UK
| | - Eunice Zhang
- Wolfson Centre for Personalised Medicine, Department of Pharmacology and Therapeutics, University of Liverpool, Crown Street, Liverpool, L69 3BX, UK
| | - Jens Allmer
- Applied Bioinformatics, Bioscience, Wageningen University and Research, Droevendaalsesteeg 4, 6708 PB, Wageningen, The Netherlands
| | - Alan Norris
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Francesco Falciani
- Computational Biology Facility, MerseyBio, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Louise Y Takeshita
- Computational Biology Facility, MerseyBio, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Shiva Seyed Forootan
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Robert Sutton
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7BE, UK
| | - B Kevin Park
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK
| | - Chris Goldring
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool, L69 3GE, UK.
| |
Collapse
|
9
|
Rankovic M, Draginic N, Jeremic J, Samanovic AM, Stojkov S, Mitrovic S, Jeremic N, Radonjic T, Srejovic I, Bolevich S, Svistunov A, Jakovljevic V, Turnic TN. Protective Role of Vitamin B 1 in Doxorubicin-Induced Cardiotoxicity in Rats: Focus on Hemodynamic, Redox, and Apoptotic Markers in Heart. Front Physiol 2021; 12:690619. [PMID: 34630136 PMCID: PMC8494423 DOI: 10.3389/fphys.2021.690619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/18/2021] [Indexed: 11/13/2022] Open
Abstract
Up until now, the specific mechanisms involved in doxorubicin (DOX)-induced cardiotoxicity have not been fully elucidated. Since thiamine deficiency is associated with myocardial dysfunction and it may lead to cardiomyopathy, we aimed to investigate whether thiamine (Vitamin B1) treatment provides cardioprotection and modulates DOX mediated subchronic cardiotoxicity as well as to determine possible mechanisms of its effects. The study involved 48 Wistar albino rats divided into four groups: healthy non-treated rats and healthy rats treated with thiamine and DOX rats without treatment and DOX rats treated with thiamine. DOX was applied as a single i.p.injection (15mg/kg), while thiamine treatment lasted 7days (25mg/kg/dayi.p.). Before and after the treatment hemodynamic changes were monitored in vivo by echocardiography. When the protocol was completed, animals were sacrificed and rat hearts were isolated in order to evaluate parameters of cardiac oxidative stress [superoxide anion radical-O2 -, hydrogen peroxide-H2O2, nitric oxide-NO-, index of lipid peroxidation-thiobarbituric acid (TBA) reactive substances (TBARS), superoxide dismutase - SOD, catalase (CAT), and reduced glutathione-GSH] and apoptosis (Bax, Bcl-2, caspases). DOX treatment significantly reduced the ejection fraction, while thiamine treatment led to its minor increase in the DOX-treated group. In that sense, heart oxidative stress markers were significantly increased in DOX-treated rats, while therapeutic dose of thiamine decreased the levels of free radicals. Our study demonstrated the promising ameliorative effects of thiamine against DOX-induced cardiotoxicity through modulation of oxidative stress, suppression of apoptosis, and possibility to improve myocardial performance and morphometric structure of rats` hearts.
Collapse
Affiliation(s)
- Marina Rankovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nevena Draginic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Department of Human Pathology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Jovana Jeremic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Svetlana Stojkov
- Department of Pharmacy, Novi Sad University Business Academy, College of Vocational Studies for the Education of Preschool Teachers and Sports Trainers, Subotica, Serbia
| | - Slobodanka Mitrovic
- Department of Pathology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nevena Jeremic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Ivan Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.,Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sergey Bolevich
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Andrey Svistunov
- Research Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir Jakovljevic
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Tamara Nikolic Turnic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
10
|
Yuan M, Zang L, Xu A, Gong M, Liu Q, Huo B, Wang J, Fu H, Tse G, Roever L, Li G, Wang H, Liu T. Dynamic Changes of Serum Heart Type-Fatty Acid Binding Protein in Cancer Patients Treated With Immune Checkpoint Inhibitors. Front Pharmacol 2021; 12:748677. [PMID: 34658887 PMCID: PMC8517171 DOI: 10.3389/fphar.2021.748677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
Objective: Immune checkpoint inhibitors (ICIs) are effective anti-cancer drugs that can improve survival in cancer patients, but their use may be associated with adverse cardiovascular side effects. Therefore, there is a clinical unmet need to identify non-invasive biomarker to detect subclinical cardiac toxicity after ICI treatment. The aim of this study is to examine the plasma levels of biomarkers in cancer survivors who were treated with ICIs. Patients and Methods: In a cohort of 19 cancer patients, biomarkers were evaluated at baseline, 1 month, 3 and 6 months after ICI administration. These biomarkers, hypothesized to be mechanistically relevant to cardiotoxicity, included cardiac troponin I (cTnI), high-sensitivity C-reactive protein (Hs-CRP), N-terminal pro-B-type natriuretic peptide (NT-pro BNP), CK (creatine kinase), CK-MB (creatine kinase-MB), Pentraxin-related protein 3 (PTX3), growth differentiation factor 15 (GDF-15), heart type-fatty acid binding protein (H-FABP) and galectin 3 (Gal-3). Results: H-FABP, but not other biomarkers, were increased at 3 months, which persisted at 6 months (529.28 ± 312.83 vs. 752.33 ± 283.65 vs. 808.00 ± 289.69 pg/ml, p = 0.031 and p = 0.013). Left ventricular ejection fraction (63.00 ± 4.15% vs. 63.74 ± 4.07%, p > 0.05) was not significantly reduced at this time point. Conclusions: H-FABP, but not other biomarkers, were increased in patients who were treated using ICIs. H-FABP might be a more sensitive biomarker to detect ICI-related subclinical myocardial damage than traditional cardiac biomarkers.
Collapse
Affiliation(s)
- Ming Yuan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
- Department of Cardiology, Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Zang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Aiqing Xu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Mengqi Gong
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Qing Liu
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bin Huo
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
- Central Laboratory/Tianjin Research Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jinhuan Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Huaying Fu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Leonardo Roever
- Department of Clinical Research, Federal University of Uberlândia, Uberlândia, Brazil
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haitao Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin, China
- Central Laboratory/Tianjin Research Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
11
|
Wagner KR, Guglieri M, Ramaiah SK, Charnas L, Marraffino S, Binks M, Vaidya VS, Palmer J, Goldstein R, Muntoni F. Safety and disease monitoring biomarkers in Duchenne muscular dystrophy: results from a Phase II trial. Biomark Med 2021; 15:1389-1396. [PMID: 34533053 DOI: 10.2217/bmm-2021-0222] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Evaluate the utility of glutamate dehydrogenase (GLDH) and cardiac troponin I as safety biomarkers, and creatine kinase and muscle injury panel as muscle health biomarkers in Duchenne muscular dystrophy. Patients & methods: Data were collected during a Phase II trial of domagrozumab. Results: GLDH was a more specific biomarker for liver injury than alanine aminotransferase. Cardiac troponin I elevations were variable and not sustained, limiting its applicability as a biomarker. Muscle injury panel biomarkers were no more informative than creatine kinase as a muscle health biomarker. Conclusion: Results support the use of GLDH as a specific biomarker for liver injury in patients with Duchenne muscular dystrophy. Clinical trial registration: ClinicalTrials.gov, NCT02310763.
Collapse
Affiliation(s)
- Kathryn R Wagner
- Center for Genetic Muscle Disorders, Kennedy Krieger Institute, Departments of Neurology & Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Michaela Guglieri
- The John Walton Muscular Dystrophy Research Centre, Newcastle University & Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | - Francesco Muntoni
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, London, UK
| |
Collapse
|
12
|
Bamberg K, Mehtälä L, Arola O, Laitinen S, Nordling P, Strandberg M, Strandberg N, Paltta J, Mali M, Espinosa-Ortega F, Pirilä L, Lundberg IE, Savukoski T, Pettersson K. Evaluation of a New Skeletal Troponin I Assay in Patients with Idiopathic Inflammatory Myopathies. J Appl Lab Med 2021; 5:320-331. [PMID: 32445386 DOI: 10.1093/jalm/jfz016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/01/2019] [Indexed: 11/13/2022]
Abstract
BACKGROUND The current biomarkers for diagnosis and monitoring of injured and diseased skeletal muscles, such as creatine kinase (CK), have limited tissue specificity and incapability to differentiate between pathological and physiological changes. Thus, new biomarkers with improved diagnostic accuracy are needed. Our aim was to develop and validate a novel assay for skeletal troponin I (skTnI), and to assess its clinical performance in patients with idiopathic inflammatory myopathies (IIM). METHODS A two-step fluoroimmunoassay was used to analyze samples from healthy reference individuals (n = 140), patients with trauma (n = 151), and patients with IIM (n = 61). RESULTS The limit of detection was 1.2 ng/mL, and the upper reference limit (90th percentile) was 5.2 ng/mL. The median skTnI concentrations were <limit of detection (LoD), 2.7 ng/mL, and 8.6 ng/mL in reference, trauma, and IIM cohorts, respectively. Differences in measured skTnI levels were statistically significant between all three study cohorts (Kruskal-Wallis P < 0.001; Mann-Whitney P < 0.001 for all). skTnI and CK had a strong positive correlation (Spearman's r = 0.771, P < 0.001), and the longitudinal changes in skTnI mirrored those observed with CK. CONCLUSIONS With the skTnI assay, patients with IIM were identified from healthy individuals and from patients with traumatic muscular injuries. When compared to CK, skTnI appeared to be more accurate in managing patients with low-grade IIM disease activities. The developed assay serves as a reliable analytical tool for the assessment of diagnostic accuracy of skTnI in the diagnosis and monitoring of myopathies.
Collapse
Affiliation(s)
- Katriina Bamberg
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Laura Mehtälä
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Olli Arola
- Intensive Care Medicine and Pain Management, Turku University Hospital, Turku, Finland
| | | | | | | | - Niko Strandberg
- Department of Orthopaedic Surgery, Turku University Hospital, Turku, Finland
| | - Johanna Paltta
- Department of Rheumatology, Turku University Hospital, Turku, Finland
| | - Markku Mali
- Department of Rheumatology, Turku University Hospital, Turku, Finland
| | - Fabricio Espinosa-Ortega
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Laura Pirilä
- Department of Rheumatology, Turku University Hospital, Turku, Finland
| | - Ingrid E Lundberg
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tanja Savukoski
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Kim Pettersson
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| |
Collapse
|
13
|
Oda S, Yokoi T. Recent progress in the use of microRNAs as biomarkers for drug-induced toxicities in contrast to traditional biomarkers: A comparative review. Drug Metab Pharmacokinet 2021; 37:100372. [PMID: 33461055 DOI: 10.1016/j.dmpk.2020.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 02/09/2023]
Abstract
microRNAs (miRNAs) are small non-coding RNAs with 18-25 nucleotides. They play key regulatory roles in versatile biological process including development and apoptosis, and in disease pathogenesis, for example carcinogenesis, by negatively regulating gene expression. miRNAs often exhibit characteristics suitable for biomarkers such as tissue-specific expression patterns, high stability in serum/plasma, and change in abundance in circulation immediately after toxic injury. Since the discovery of circulating miRNAs in extracellular biological fluids in 2008, there have been many reports on the use of miRNAs as biomarkers for various diseases including cancer and organ injury in humans and experimental animals. In this review article, we have summarized the utility and limitation of circulating miRNAs as safety/toxicology biomarkers for specific tissue injuries including liver, skeletal muscle, heart, retina, and pancreas, by comparing them with conventional protein biomarkers. We have also covered the discovery of miRNAs in serum/plasma and their stability, the knowledge of which is essential for understanding the kinetics of miRNA biomarkers. Since numerous studies have reported the use of these circulating miRNAs as safety biomarkers with high sensitivity and specificity, we believe that circulating miRNAs can promote pre-clinical drug development and improve the monitoring of tissue injuries in clinical pharmacotherapy.
Collapse
Affiliation(s)
- Shingo Oda
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| | - Tsuyoshi Yokoi
- Department of Drug Safety Sciences, Division of Clinical Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| |
Collapse
|
14
|
Khodabukus A, Kaza A, Wang J, Prabhu N, Goldstein R, Vaidya VS, Bursac N. Tissue-Engineered Human Myobundle System as a Platform for Evaluation of Skeletal Muscle Injury Biomarkers. Toxicol Sci 2020; 176:124-136. [PMID: 32294208 PMCID: PMC7643536 DOI: 10.1093/toxsci/kfaa049] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traditional serum biomarkers used to assess skeletal muscle damage, such as activity of creatine kinase (CK), lack tissue specificity and sensitivity, hindering early detection of drug-induced myopathies. Recently, a novel four-factor skeletal muscle injury panel (MIP) of biomarkers consisting of skeletal troponin I (sTnI), CK mass (CKm), fatty-acid-binding protein 3 (Fabp3), and myosin light chain 3, has been shown to have increased tissue specificity and sensitivity in rodent models of skeletal muscle injury. Here, we evaluated if a previously established model of tissue-engineered functional human skeletal muscle (myobundle) can allow detection of the MIP biomarkers after injury or drug-induced myotoxicity in vitro. We found that concentrations of three MIP biomarkers (sTnI, CKm, and Fabp3) in myobundle culture media significantly increased in response to injury by a known snake venom (notexin). Cerivastatin, a known myotoxic statin, but not pravastatin, induced significant loss of myobundle contractile function, myotube atrophy, and increased release of both traditional and novel biomarkers. In contrast, dexamethasone induced significant loss of myobundle contractile function and myotube atrophy, but decreased the release of both traditional and novel biomarkers. Dexamethasone also increased levels of matrix metalloproteinase-2 and -3 in the culture media which correlated with increased remodeling of myobundle extracellular matrix. In conclusion, this proof-of-concept study demonstrates that tissue-engineered human myobundles can provide an in vitro platform to probe patient-specific drug-induced myotoxicity and performance assessment of novel injury biomarkers to guide preclinical and clinical drug development studies.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708-90281
| | - Amulya Kaza
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708-90281
| | - Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708-90281
| | - Neel Prabhu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708-90281
| | | | - Vishal S Vaidya
- Drug Research and Development, Pfizer, Groton, Connecticut 06340
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708-90281
| |
Collapse
|
15
|
Dowling P, Gargan S, Zweyer M, Swandulla D, Ohlendieck K. Proteomic profiling of fatty acid binding proteins in muscular dystrophy. Expert Rev Proteomics 2020; 17:137-148. [PMID: 32067530 DOI: 10.1080/14789450.2020.1732214] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Duchenne muscular dystrophy is a neuromuscular disorder, which is caused by abnormalities in the DMD gene that encodes the membrane cytoskeletal protein dystrophin. Besides progressive skeletal muscle wasting, dystrophinopathy also affects non-skeletal muscle tissues, including cells in the cardio-respiratory system, the central nervous system, the liver and the kidney.Areas covered: This review summarizes the proteomic characterization of a key class of lipid chaperones, the large family of fatty acid binding proteins, and their potential role in muscular dystrophy. Recent proteomic surveys using animal models and patient specimens are reviewed. Pathobiochemical changes in specific proteoforms of fatty acid binding protein in the multi-system pathology of dystrophinopathy are discussed.Expert opinion: The mass spectrometric identification of distinct changes in fatty acid binding proteins in muscle, heart, liver, kidney and serum demonstrates that considerable alterations occur in key steps of metabolite transport and fat metabolism in muscular dystrophy. These new findings might be helpful to further develop a comprehensive biomarker signature of metabolic changes in X-linked muscular dystrophy, which should improve (i) our understanding of complex pathobiochemical changes due to dystrophin deficiency, (ii) the identification of novel therapeutic targets, and (iii) the design of differential diagnostic, prognostic and therapy-monitoring approaches.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Margit Zweyer
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital, University of Bonn, Bonn, Germany
| | | | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.,Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
16
|
Zeiss CJ, Gatti DM, Toro-Salazar O, Davis C, Lutz CM, Spinale F, Stearns T, Furtado MB, Churchill GA. Doxorubicin-Induced Cardiotoxicity in Collaborative Cross (CC) Mice Recapitulates Individual Cardiotoxicity in Humans. G3 (BETHESDA, MD.) 2019; 9:2637-2646. [PMID: 31263061 PMCID: PMC6686936 DOI: 10.1534/g3.119.400232] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/19/2019] [Indexed: 12/15/2022]
Abstract
Anthracyclines cause progressive cardiotoxicity whose ultimate severity is individual to the patient. Genetic determinants contributing to this variation are difficult to study using current mouse models. Our objective was to determine whether a spectrum of anthracycline induced cardiac disease can be elicited across 10 Collaborative Cross mouse strains given the same dose of doxorubicin. Mice from ten distinct strains were given 5 mg/kg of doxorubicin intravenously once weekly for 5 weeks (total 25 mg/kg). Mice were killed at acute or chronic timepoints. Body weight was assessed weekly, followed by terminal complete blood count, pathology and a panel of biomarkers. Linear models were fit to assess effects of treatment, sex, and sex-by-treatment interactions for each timepoint. Impaired growth and cardiac pathology occurred across all strains. Severity of these varied by strain and sex, with greater severity in males. Cardiac troponin I and myosin light chain 3 demonstrated strain- and sex-specific elevations in the acute phase with subsequent decline despite ongoing progression of cardiac disease. Acute phase cardiac troponin I levels predicted the ultimate severity of cardiac pathology poorly, whereas myosin light chain 3 levels predicted the extent of chronic cardiac injury in males. Strain- and sex-dependent renal toxicity was evident. Regenerative anemia manifested during the acute period. We confirm that variable susceptibility to doxorubicin-induced cardiotoxicity observed in humans can be modeled in a panel of CC strains. In addition, we identified a potential predictive biomarker in males. CC strains provide reproducible models to explore mechanisms contributing to individual susceptibility in humans.
Collapse
Affiliation(s)
| | | | - Olga Toro-Salazar
- Connecticut Children's Medical Center, University of Connecticut School of Medicine, Hartford, CT 06106, and
| | | | | | - Francis Spinale
- University of South Carolina School of Medicine, Columbia SC 29208
| | | | | | | |
Collapse
|
17
|
Myoglobin and troponin concentrations are increased in early stage deep tissue injury. J Mech Behav Biomed Mater 2019; 92:50-57. [DOI: 10.1016/j.jmbbm.2018.12.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/12/2018] [Accepted: 12/20/2018] [Indexed: 12/27/2022]
|
18
|
Shrivastava R, Trivedi S, Singh PK, Asif M, Chourasia MK, Khanna A, Bhadauria S. Design and development of PEGylated liposomal formulation of HER2 blocker Lapatinib for enhanced anticancer activity and diminished cardiotoxicity. Biochem Biophys Res Commun 2018; 503:677-683. [DOI: 10.1016/j.bbrc.2018.06.060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 06/12/2018] [Indexed: 10/28/2022]
|
19
|
Tochinai R, Komatsu K, Murakami J, Nagata Y, Ando M, Hata C, Suzuki T, Kado S, Kobayashi T, Kuwahara M. Histopathological and functional changes in a single-dose model of combretastatin A4 disodium phosphate-induced myocardial damage in rats. J Toxicol Pathol 2018; 31:307-313. [PMID: 30393435 PMCID: PMC6206283 DOI: 10.1293/tox.2018-0023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/13/2018] [Indexed: 11/23/2022] Open
Abstract
Cardiotoxicity is a concern in the development of microtubule-disassembling agents
(MDAs) as vascular-disrupting agents of tumors. This study investigated cardiotoxicity in
rats induced by a single-dose of combretastatin A4 disodium phosphate (CA4DP), an MDA and
discussed the use of this rat model in nonclinical studies of MDAs. First, CA4DP (120
mg/kg) was administered to rats intravenously, and cardiac histopathology and blood
biomarkers were examined after 0.5, 24, and 72 h. Next, CA4DP (120 mg/kg) was administered
to rats intravenously, and the electrocardiography and echocardiography results were
analyzed. The results showed that at 0.5 h after dosing, plasma creatine kinase (CK),
CK-muscle/brain (CK-MB), and fatty acid binding protein 3 levels increased. At 24 h,
lactate dehydrogenase (LDH)-1, CK, and CK-MB levels increased, and multifocal vacuolar
degeneration of myocardial cells was observed in the apical inner layer. At 72 h, LDH-1
levels were increased, and multifocal myocardial necrosis was observed in the
interventricular septum and inner layer of the apex of left ventricular wall. Furthermore,
at 0.5 h, heart rate (HR), ejection fraction (EF), and cardiac output (CO) decreased. At
24 h, CO decreased. Finally, at 72 h, HR, EF, and CO decreased, and depression of the
T-wave amplitude was observed. In conclusion, myocardial injury, bradycardia, and
depressed cardiac function were induced in rats by a single-dose of CA4DP. The lesion
distribution and electrocardiographic features suggested that myocardial injury was
induced by ischemia. These findings are similar to MDA-induced cardiotoxicity in humans,
and this rat model will prove useful in studies of the cardiotoxicity in humans.
Collapse
Affiliation(s)
- Ryota Tochinai
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan.,Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kayoko Komatsu
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Junta Murakami
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Yuriko Nagata
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Minoru Ando
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Chie Hata
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Tomo Suzuki
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Shoichi Kado
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Toshihide Kobayashi
- Yakult Central Institute, Yakult Honsha Co., Ltd., 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Masayoshi Kuwahara
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
20
|
Toth PP, Patti AM, Giglio RV, Nikolic D, Castellino G, Rizzo M, Banach M. Management of Statin Intolerance in 2018: Still More Questions Than Answers. Am J Cardiovasc Drugs 2018; 18:157-173. [PMID: 29318532 PMCID: PMC5960491 DOI: 10.1007/s40256-017-0259-7] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Statin therapy is generally well tolerated and very effective in the prevention and treatment of cardiovascular disease, regardless of cholesterol levels; however, it can be associated with various adverse events (myalgia, myopathy, rhabdomyolysis, and diabetes mellitus, among others). Patients frequently discontinue statin therapy without medical advice because of perceived side effects and consequently increase their risk for cardiovascular events. In patients with statin intolerance, it may be advisable to change the dose, switch to a different statin, or try an alternate-day regimen. If intolerance is associated with all statins-even at the lowest dose-non-statin drugs and certain nutraceuticals can be considered. This review focuses on the definition of statin intolerance and on the development of clinical and therapeutic strategies for its management, including emerging alternative therapies.
Collapse
Affiliation(s)
- Peter P Toth
- CGH Medical Center, Sterling, IL, USA
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angelo Maria Patti
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Rosaria Vincenza Giglio
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Dragana Nikolic
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giuseppa Castellino
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Manfredi Rizzo
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Maciej Banach
- Chair of Nephrology and Hypertension, Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, 90-549, Lodz, Poland.
- Polish Mother's Memorial Hospital Research Institute, Lodz, Poland.
- Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland.
| |
Collapse
|
21
|
miR-21-5p as a potential biomarker of inflammatory infiltration in the heart upon acute drug-induced cardiac injury in rats. Toxicol Lett 2018; 286:31-38. [PMID: 29355689 DOI: 10.1016/j.toxlet.2018.01.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/12/2018] [Accepted: 01/15/2018] [Indexed: 01/09/2023]
Abstract
Investigation of genomic changes in cardiotoxicity can provide novel biomarkers and insights into molecular mechanisms of drug-induced cardiac injury (DICI). The main objective of this study was to identify and characterize dysregulated microRNAs (miRNAs) in the heart associated with cardiotoxicity. Wistar rats were dosed once with either isoproterenol (1.5 mg/kg, i.p), allylamine (100 mg/kg, p.o.) or the respective vehicle controls. Heart tissue was collected at 24 h, 48 h and 72 h post-drug administration and used for histopathological assessment, miRNA profiling, immunohistochemical analysis and in situ hybridization. Multiplex analysis of 68 miRNAs in the heart revealed a significant upregulation of several miRNAs (miR-19a-3p, miR-142-3p, miR-155-5p, miR-208b-3p, miR-21-5p) after isoproterenol and one miRNA (miR-21-5p) after allylamine administration. Localization of miR-21-5p was specific to inflammatory cell infiltrates in the heart after both treatments. Immunohistochemical analysis of Stat3, a known miR-21-5p regulator, also confirmed its upregulation in cardiomyocytes and inflammatory cell infiltrates. The toxicity signatures based on miRNA networks, identified in vivo, can potentially be used as mechanistic biomarkers as well as to study cardiotoxicity in vitro in order to develop sensitive tools for early hazard identification and risk assessment.
Collapse
|
22
|
Dalrymple BP, Guo B. TRIENNIAL GROWTH AND DEVELOPMENT SYMPOSIUM: Intramuscular fat deposition in ruminants and pigs: A transcriptomics perspective. J Anim Sci 2017; 95:2272-2283. [PMID: 28727003 DOI: 10.2527/jas.2016.1112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The genomics era has led to an explosion in the study of gene expression in production animals. Intramuscular fat (IMF) content (both high and low) and composition are major quality attributes of meat, and more than 90 transcriptomic studies of IMF deposition have been undertaken in the ruminants and pigs since 2001, with the majority since 2008. The studies have implicated many genes involved in the control of adipogenesis, lipogenesis, and deposition of IMF, but there is relatively little consistency between the different studies. However, the genes encoding the synthesis enzymes acetyl-CoA carboxylase α, fatty acid synthase, and stearoyl-CoA desaturase; the fatty acid binding protein 4; the potential signaling protein thyroid hormone responsive; and the regulators C/EBPα, PPARγ, and sterol regulatory element binding transcription factor 1 are supported by 5 or more of the 90 studies. By combining the results of all the studies, complete pathways for long-chain fatty acid (LCFA) and triacylglyceride (TAG) synthesis are identified, as are a number of genes encoding proteins probably associated with the storage of TAG and genes encoding a number of known and potential adipokines. In contrast, support for the association of lipolytic pathways with IMF percentage is less strong. Differences in experimental design-in particular, the age of the animals, the rate of IMF deposition at sampling, the past nutritional history of the animals used, and the complexities of using a tissue with mixed cell types-have contributed to the differences in results and interpretation. Biomarkers predictive of future IMF percentage, facilitating reaching optimal IMF content at slaughter, may have industry utility, but to be useful in animal biopsy and postslaughter samples, where multiple cell types are present, genes must be carefully chosen to ensure that they are informative about the expected processes. Despite these problems, candidate biomarkers for estimation of de novo intramuscular adipocyte LCFA synthesis, LCFA uptake rate by intramuscular adipocytes, and IMF deposition rate have been identified and examples of their utility have been published. However, further work is required to demonstrate how best to apply the assays for the benefit of the relevant livestock production industries.
Collapse
|
23
|
Obayashi H, Kobayashi N, Nezu Y, Yamoto T, Shirai M, Asai F. Plasma 2-hydroxyglutarate and hexanoylcarnitine levels are potential biomarkers for skeletal muscle toxicity in male Fischer 344 rats. J Toxicol Sci 2017; 42:385-396. [PMID: 28717097 DOI: 10.2131/jts.42.385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
To identify new candidate biomarkers for skeletal muscle toxicity, an unbiased metabolomic analysis was performed in rats treated with two distinct myotoxicants, cerivastatin (CER) and tetramethyl-p-phenylenediamine (TMPD). Skeletal muscle toxicity was induced in male Fischer 344 rats by administering CER or TMPD and monitored using established endpoints, such as increased plasma creatine kinase (CK) activity and histopathology, and a metabolomic analysis of skeletal muscle and plasma samples. Plasma CK levels in CER-treated rats were markedly elevated at Day 11; however, those in TMPD-treated rats showed a statistically significant decrease at 24 hr after dosing. Light microscopy revealed that vacuolated or necrotic fibers were evident in all CER-treated rats on Day 11, and slightly vacuolated fibers were observed in TMPD-treated rats at 6 and 24 hr after dosing. Metabolomic analysis of the rectus femoris indicated increases in 2-hydroxyglutarate (2HG) in CER-treated rats and hexanoylcarnitine in CER- and TMPD-treated rats. There were also increases in plasma 2HG in CER-treated rats on Days 8 and 11 and in TMPD-treated rats at 24 hr after dosing and increases in plasma hexanoylcarnitine in CER-treated rats on Day 11 and in TMPD-treated rats at 6 and 24 hr after dosing. These experiments demonstrated the potential of plasma 2HG and hexanoylcarnitine as specific and easily detectable biomarkers for skeletal muscle toxicity in rats and demonstrated the value of metabolomics for biomarker detection and identification in toxicological studies.
Collapse
Affiliation(s)
- Hisakuni Obayashi
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University.,Medicinal Safety Research Laboratories, Daiichi-Sankyo Co., Ltd
| | - Naoko Kobayashi
- Medicinal Safety Research Laboratories, Daiichi-Sankyo Co., Ltd
| | - Yoshikazu Nezu
- Medicinal Safety Research Laboratories, Daiichi-Sankyo Co., Ltd
| | - Takashi Yamoto
- Medicinal Safety Research Laboratories, Daiichi-Sankyo Co., Ltd
| | - Mitsuyuki Shirai
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University
| | - Fumitoshi Asai
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University
| |
Collapse
|
24
|
Maliver P, Festag M, Bennecke M, Christen F, Bánfai B, Lenz B, Winter M. Assessment of Preclinical Liver and Skeletal Muscle Biomarkers Following Clofibrate Administration in Wistar Rats. Toxicol Pathol 2017; 45:506-525. [PMID: 28485676 DOI: 10.1177/0192623317707271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clofibrate is a known rodent hepatotoxicant classically associated with hepatocellular hypertrophy and increased serum activities of cellular alanine aminotransferase/aspartate aminotransferase (ALT/AST) in the absence of microscopic hepatocellular degeneration. At toxic dose, clofibrate induces liver and skeletal muscle injury. The objective of this study was to assess novel liver and skeletal muscle biomarkers following clofibrate administration in Wistar rats at different dose levels for 7 days. In addition to classical biomarkers, liver injury was assessed by cytokeratin 18 (CK18) cleaved form, high-mobility group box 1, arginase 1 (ARG1), microRNA 122 (miR-122), and glutamate dehydrogenase. Skeletal muscle injury was evaluated with fatty acid binding protein 3 (Fabp3) and myosin light chain 3 (Myl3). Clofibrate-induced hepatocellular hypertrophy and skeletal muscle degeneration (type I rich muscles) were noted microscopically. CK, Fabp3, and Myl3 elevations correlated to myofiber degeneration. Fabp3 and Myl3 outperformed CK for detection of myofiber degeneration of minimal severity. miR-122 and ARG1 results were significantly correlated and indicated the absence of liver toxicity at low doses of clofibrate, despite increased ALT/AST activities. Moreover, combining classical and novel biomarkers (Fabp3, Myl3, ARG1, and miR-122) can be considered a valuable strategy for differentiating increased transaminases due to liver toxicity from skeletal muscle toxicity.
Collapse
Affiliation(s)
- Pierre Maliver
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Matthias Festag
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Moritz Bennecke
- 2 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Penzberg, Germany
| | - Francois Christen
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Balázs Bánfai
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland.,3 Soladis GmbH, Basel, Switzerland
| | - Barbara Lenz
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Michael Winter
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
25
|
Cho YE, Im EJ, Moon PG, Mezey E, Song BJ, Baek MC. Increased liver-specific proteins in circulating extracellular vesicles as potential biomarkers for drug- and alcohol-induced liver injury. PLoS One 2017; 12:e0172463. [PMID: 28225807 PMCID: PMC5321292 DOI: 10.1371/journal.pone.0172463] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/06/2017] [Indexed: 12/21/2022] Open
Abstract
Drug- and alcohol-induced liver injury are a leading cause of liver failure and transplantation. Emerging evidence suggests that extracellular vesicles (EVs) are a source of biomarkers because they contain unique proteins reflecting the identity and tissue-specific origin of the EV proteins. This study aimed to determine whether potentially hepatotoxic agents, such as acetaminophen (APAP) and binge alcohol, can increase the amounts of circulating EVs and evaluate liver-specific EV proteins as potential biomarkers for liver injury. The circulating EVs, isolated from plasma of APAP-exposed, ethanol-fed mice, or alcoholic hepatitis patients versus normal control counterparts, were characterized by proteomics and biochemical methods. Liver specific EV proteins were analyzed by immunoblots and ELISA. The amounts of total and liver-specific proteins in circulating EVs from APAP-treated mice significantly increased in a dose- and time-dependent manner. Proteomic analysis of EVs from APAP-exposed mice revealed that the amounts of liver-specific and/or hepatotoxic proteins were increased compared to those of controls. Additionally, the increased protein amounts in EVs following APAP exposure returned to basal levels when mice were treated with N-acetylcysteine or glutathione. Similar results of increased amounts and liver-specific proteins in circulating EVs were also observed in mice exposed to hepatotoxic doses of thioacetamide or d-galactosamine but not by non-hepatotoxic penicillin or myotoxic bupivacaine. Additionally, binge ethanol exposure significantly elevated liver-specific proteins in circulating EVs from mice and alcoholics with alcoholic hepatitis, compared to control counterparts. These results indicate that circulating EVs in drug- and alcohol-mediated hepatic injury contain liver-specific proteins that could serve as specific biomarkers for hepatotoxicity.
Collapse
Affiliation(s)
- Young-Eun Cho
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland, United States of America
| | - Eun-Ju Im
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Pyong-Gon Moon
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Esteban Mezey
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland, United States of America
| | - Moon-Chang Baek
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- * E-mail:
| |
Collapse
|
26
|
Evaluation of microRNAs−208 and 133a/b as differential biomarkers of acute cardiac and skeletal muscle toxicity in rats. Toxicol Appl Pharmacol 2016; 312:53-60. [DOI: 10.1016/j.taap.2015.11.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 01/15/2023]
|
27
|
Melissa officinalis Protects against Doxorubicin-Induced Cardiotoxicity in Rats and Potentiates Its Anticancer Activity on MCF-7 Cells. PLoS One 2016; 11:e0167049. [PMID: 27880817 PMCID: PMC5120835 DOI: 10.1371/journal.pone.0167049] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 11/08/2016] [Indexed: 12/26/2022] Open
Abstract
Cardiotoxicity is a limiting factor of doxorubicin (DOX)-based anticancer therapy. Due to its beneficial effects, we investigated whether standardized extract of Melissa officinalis (MO) can attenuate doxorubicin-induced cardiotoxicity and can potentiate the efficacy of DOX against human breast cancer cells. MO was administered orally to male albino rats once daily for 10 consecutive days at doses of 250, 500 and 750 mg/kg b.wt. DOX (15 mg/kg b.wt. i.p.) was administered on the 8th day. MO protected against DOX-induced leakage of cardiac enzymes and histopathological changes. MO ameliorated DOX-induced oxidative stress as evidenced by decreasing lipid peroxidation, protein oxidation and total oxidant capacity depletion and by increasing antioxidant capacity. Additionally, MO pretreatment inhibited inflammatory responses to DOX by decreasing the expressions of nuclear factor kappa-B, tumor necrosis factor-alpha and cyclooxygenase-2 and the activity of myeloperoxidase. MO ameliorated DOX-induced apoptotic tissue damage in heart of rats. In vitro study showed that MO augmented the anticancer efficacy of DOX in human breast cancer cells (MCF-7) and potentiated oxidative damage and apoptosis. Thus, combination of DOX and MO may prove future cancer treatment protocols safer and more efficient.
Collapse
|
28
|
Kim K, Chini N, Fairchild DG, Engle SK, Reagan WJ, Summers SD, Mirsalis JC. Evaluation of Cardiac Toxicity Biomarkers in Rats from Different Laboratories. Toxicol Pathol 2016; 44:1072-1083. [PMID: 27638646 DOI: 10.1177/0192623316668276] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is a great need for improved diagnostic and prognostic accuracy of potential cardiac toxicity in drug development. This study reports the evaluation of several commercially available biomarker kits by 3 institutions (SRI, Eli Lilly, and Pfizer) for the discrimination between myocardial degeneration/necrosis and cardiac hypertrophy as well as the assessment of the interlaboratory and interplatform variation in results. Serum concentrations of natriuretic peptides (N-terminal pro-atrial natriuretic peptide [NT-proANP] and N-terminal pro-brain natriuretic peptide [NT-proBNP]), cardiac and skeletal troponins (cTnI, cTnT, and sTnI), myosin light chain 3 (Myl3), and fatty acid binding protein 3 (FABP3) were assessed in rats treated with minoxidil (MNX) and isoproterenol (ISO). MNX caused increased heart-to-body weight ratios and prominent elevations in NT-proANP and NT-proBNP concentrations detected at 24-hr postdose without elevation in troponins, Myl3, or FABP3 and with no abnormal histopathological findings. ISO caused ventricular leukocyte infiltration, myocyte fibrosis, and necrosis with increased concentrations of the natriuretic peptides, cardiac troponins, and Myl3. These results reinforce the advantages of a multimarker strategy in elucidating the underlying cause of cardiac insult and detecting myocardial tissue damage at 24-hr posttreatment. The interlaboratory and interplatform comparison analyses also showed that the data obtained from different laboratories and platforms are highly correlated and reproducible, making these biomarkers widely applicable in preclinical studies.
Collapse
Affiliation(s)
- Kyuri Kim
- 1 SRI International, Menlo Park, California, USA
| | - Naseem Chini
- 1 SRI International, Menlo Park, California, USA
| | | | - Steven K Engle
- 2 Lilly Research Laboratories, A Division of Eli Lilly and Company, Indianapolis, Indiana, USA
| | - William J Reagan
- 3 Pfizer, Drug Safety Research and Development, Groton, Connecticut, USA
| | - Sandra D Summers
- 3 Pfizer, Drug Safety Research and Development, Groton, Connecticut, USA
| | | | | |
Collapse
|
29
|
Muntean DM, Thompson PD, Catapano AL, Stasiolek M, Fabis J, Muntner P, Serban MC, Banach M. Statin-associated myopathy and the quest for biomarkers: can we effectively predict statin-associated muscle symptoms? Drug Discov Today 2016; 22:85-96. [PMID: 27634340 DOI: 10.1016/j.drudis.2016.09.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 07/28/2016] [Accepted: 09/05/2016] [Indexed: 12/21/2022]
Abstract
Over the past three decades, statins have become the cornerstone of prevention and treatment of atherosclerotic cardiovascular and metabolic diseases. Albeit generally well tolerated, these drugs can elicit a variety of muscle-associated symptoms that represent the most important reason for treatment discontinuation. Statin-associated myopathy has been systematically underestimated by randomized controlled trials as compared with the incidence observed in clinical practice and obtained from patient registries. There are several reasons for this discrepancy, among which the lack of reliable diagnostic tests and a validated questionnaire to assess muscle symptoms are recognized as unmet needs. Here, we review the cellular and molecular mechanisms underlying statin-associated myopathy and discuss the experimental and clinical data on various biomarkers to diagnose and predict muscle-related complaints.
Collapse
Affiliation(s)
- Danina M Muntean
- Department of Pathophysiology Functional Sciences, Victor Babeş University of Medicine and Pharmacy of Timisoara, Timisoara, Romania; Center for Translational Research and Systems Medicine, Victor Babeş University of Medicine and Pharmacy of Timisoara, Timisoara, Romania
| | - Paul D Thompson
- Division of Cardiology, Hartford Hospital, Hartford, CT, USA
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, IRCCS Multimedica, Milan, Italy
| | - Mariusz Stasiolek
- Department of Neurology, Polish Mother's Memorial Hospital-Research Institute in Lodz, Lodz, Poland
| | - Jaroslaw Fabis
- Department of Arthroscopy, Minimally Invasive Surgery and Sports Traumatology, Medical University of Lodz, Poland
| | - Paul Muntner
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Maria-Corina Serban
- Department of Pathophysiology Functional Sciences, Victor Babeş University of Medicine and Pharmacy of Timisoara, Timisoara, Romania; Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Poland; Healthy Aging Research Centre (HARC), Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
30
|
Sabourin PJ, Kobs CL, Gibbs ST, Hong P, Matthews CM, Patton KM, Sabourin CL, Wakayama EJ. Characterization of a Mouse Model of Oral Potassium Cyanide Intoxication. Int J Toxicol 2016; 35:584-603. [PMID: 27170682 DOI: 10.1177/1091581816646973] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Potassium cyanide (KCN) is an inhibitor of cytochrome C oxidase causing rapid death due to hypoxia. A well-characterized model of oral KCN intoxication is needed to test new therapeutics under the Food and Drug Administration Animal Rule. Clinical signs, plasma pH and lactate concentrations, biomarkers, histopathology, and cyanide and thiocyanate toxicokinetics were used to characterize the pathology of KCN intoxication in adult and juvenile mice. The acute oral LD50s were determined to be 11.8, 11.0, 10.9, and 9.9 mg/kg in water for adult male, adult female, juvenile male, and juvenile female mice, respectively. The time to death was rapid and dose dependent; juvenile mice had a shorter mean time to death. Juvenile mice displayed a more rapid onset and higher incidence of seizures. The time to observance of respiratory signs and prostration was rapid, but mice surviving beyond 2 hours generally recovered fully within 8 hours. At doses up to the LD50, there were no gross necropsy or microscopic findings clearly attributed to administration of KCN in juvenile or adult CD-1 mice from 24 hours to 28 days post-KCN challenge. Toxicokinetic analysis indicated rapid uptake, metabolism, and clearance of plasma cyanide. Potassium cyanide caused a rapid, dose-related decrease in blood pH and increase in serum lactate concentration. An increase in fatty acid-binding protein 3 was observed at 11.5 mg/kg KCN in adult but not in juvenile mice. These studies provide a characterization of KCN intoxication in adult and juvenile mice that can be used to screen or conduct preclinical efficacy studies of potential countermeasures.
Collapse
|
31
|
Zhang L, Zhou H, Peng Q, Jiang W, Qiao W, Wang G. Fatty acid binding protein 3 is associated with skeletal muscle strength in polymyositis and dermatomyositis. Int J Rheum Dis 2016; 20:252-260. [PMID: 26891180 DOI: 10.1111/1756-185x.12838] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Lu Zhang
- Department of Rheumatology; China-Japan Friendship Hospital; Beijing China
| | - Hang Zhou
- Department of Rheumatology; Beijing Friendship Hospital Capital Medical University; Beijing China
| | - Qinglin Peng
- Department of Rheumatology; China-Japan Friendship Hospital; Beijing China
| | - Wei Jiang
- Department of Rheumatology; China-Japan Friendship Hospital; Beijing China
| | - Wei Qiao
- Department of Senior Ward; China-Japan Friendship Hospital; Beijing China
| | - Guochun Wang
- Department of Rheumatology; China-Japan Friendship Hospital; Beijing China
| |
Collapse
|
32
|
Burch PM, Greg Hall D, Walker EG, Bracken W, Giovanelli R, Goldstein R, Higgs RE, King NMP, Lane P, Sauer JM, Michna L, Muniappa N, Pritt ML, Vlasakova K, Watson DE, Wescott D, Zabka TS, Glaab WE. Evaluation of the Relative Performance of Drug-Induced Skeletal Muscle Injury Biomarkers in Rats. Toxicol Sci 2015; 150:247-56. [DOI: 10.1093/toxsci/kfv328] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
33
|
Koturbash I, Tolleson WH, Guo L, Yu D, Chen S, Hong H, Mattes W, Ning B. microRNAs as pharmacogenomic biomarkers for drug efficacy and drug safety assessment. Biomark Med 2015; 9:1153-76. [PMID: 26501795 PMCID: PMC5712454 DOI: 10.2217/bmm.15.89] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Much evidence has documented that microRNAs (miRNAs) play an important role in the modulation of interindividual variability in the production of drug metabolizing enzymes and transporters (DMETs) and nuclear receptors (NRs) through multidirectional interactions involving environmental stimuli/stressors, the expression of miRNA molecules and genetic polymorphisms. MiRNA expression has been reported to be affected by drugs and miRNAs themselves may affect drug metabolism and toxicity. In cancer research, miRNA biomarkers have been identified to mediate intrinsic and acquired resistance to cancer therapies. In drug safety assessment, miRNAs have been found associated with cardiotoxicity, hepatotoxicity and nephrotoxicity. This review article summarizes published studies to show that miRNAs can serve as early biomarkers for the evaluation of drug efficacy and drug safety.
Collapse
Affiliation(s)
- Igor Koturbash
- Department of Environmental & Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - William H Tolleson
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Lei Guo
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Dianke Yu
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Si Chen
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Huixiao Hong
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - William Mattes
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Baitang Ning
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
34
|
Woolley CF, Hayes MA. Sensitive Detection of Cardiac Biomarkers Using a Magnetic Microbead Immunoassay. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2015; 7:8632-8639. [PMID: 26527562 PMCID: PMC4625556 DOI: 10.1039/c5ay01071c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
To achieve improved sensitivity in cardiac biomarker detection, a batch incubation magnetic microbead immunoassay was developed and tested on three separate human protein targets: myoglobin, heart-type fatty acid binding protein, and cardiac troponin I. A sandwich immunoassay was performed in a simple micro-centrifuge tube allowing full dispersal of the solid capture surface during incubations. Following magnetic bead capture and wash steps, samples were analyzed in the presence of a manipulated magnetic field utilizing a modified microscope slide and fluorescent inverted microscope to collect video data files. Analysis of the video data allowed for the quantitation of myoglobin, heart-type fatty acid binding protein and cardiac troponin I to levels of 360 aM, 67 fM, and 42 fM, respectively. Compared to the previous detection limit of 50 pM for myoglobin, this offers a five-fold improvement in sensitivity. This improvement in sensitivity and incorporation of additional markers, along with the small sample volumes required, suggest the potential of this platform for incorporation as a detection method in a total sample analysis device enabling multiplexed detection for the analysis of clinical samples.
Collapse
Affiliation(s)
- Christine F Woolley
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ, USA
| | - Mark A Hayes
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
35
|
Glineur SF, De Ron P, Hanon E, Valentin JP, Dremier S, da Costa AN. Paving the Route to Plasma miR-208a-3p as an Acute Cardiac Injury Biomarker: Preclinical Rat Data Supports Its Use in Drug Safety Assessment. Toxicol Sci 2015; 149:89-97. [DOI: 10.1093/toxsci/kfv222] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
36
|
Kamendi H, Zhou Y, Crosby M, Keirstead N, Snow D, Bentley P, Patel N, Barthlow H, Luo W, Dragan Y, Bialecki R. Doxorubicin: Comparison between 3-h continuous and bolus intravenous administration paradigms on cardio-renal axis, mitochondrial sphingolipids and pathology. Toxicol Appl Pharmacol 2015; 289:560-72. [PMID: 26450648 DOI: 10.1016/j.taap.2015.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 09/22/2015] [Accepted: 10/03/2015] [Indexed: 01/29/2023]
Abstract
Doxorubicin (DOX) is a potent and effective broad-spectrum anthracycline antitumor agent, but its clinical usefulness is restricted by cardiotoxicity. This study compared pharmacokinetic, functional, structural and biochemical effects of single dose DOX bolus or 3-h continuous iv infusion (3-h iv) in the Han–Wistar rat to characterize possible treatment-related differences in drug safety over a 72 h observation period. Both DOX dosing paradigms significantly altered blood pressure, core body temperature and QA interval (indirect measure of cardiac contractility); however, there was no recovery observed in the bolus iv treatment group. Following the 3-h iv treatment, blood pressures and QA interval normalized by 36 h then rose above baseline levels over 72 h. Both treatments induced biphasic changes in heart rate with initial increases followed by sustained decreases. Cardiac injury biomarkers in plasma were elevated only in the bolus iv treatment group. Tissue cardiac injury biomarkers, cardiac mitochondrial complexes I, III and V and cardiac mitochondrial sphingolipids were decreased only in the bolus iv treatment group. Results indicate that each DOX dosing paradigm deregulates sinus rhythm.However, slowing the rate of infusion allows for functional compensation of blood pressure and may decrease the likelihood of cardiac myocyte necrosis via a mechanism associated with reduced mitochondrial damage.
Collapse
Affiliation(s)
- Harriet Kamendi
- Drug Safety and Metabolism, AstraZeneca, Waltham, MA 02451, USA.
| | - Ying Zhou
- Oncology Innovative Medicines and Early Development, AstraZeneca, Waltham, MA 02451, USA.
| | - Meredith Crosby
- Drug Safety and Metabolism, AstraZeneca, Waltham, MA 02451, USA.
| | | | - Debra Snow
- Drug Safety and Metabolism, AstraZeneca, Waltham, MA 02451, USA.
| | - Patricia Bentley
- Drug Safety and Metabolism, AstraZeneca, Waltham, MA 02451, USA.
| | - Nilaben Patel
- Drug Safety and Metabolism, AstraZeneca, Waltham, MA 02451, USA.
| | - Herbert Barthlow
- Drug Safety and Metabolism, AstraZeneca, Waltham, MA 02451, USA.
| | - Wenli Luo
- Discovery Sciences, Innovative Medicines, AstraZeneca, Waltham, MA 02451, USA.
| | - Yvonne Dragan
- Drug Safety and Metabolism, AstraZeneca, Waltham, MA 02451, USA.
| | - Russell Bialecki
- Drug Safety and Metabolism, AstraZeneca, Waltham, MA 02451, USA.
| |
Collapse
|
37
|
Thoonen R, Ernande L, Cheng J, Nagasaka Y, Yao V, Miranda-Bezerra A, Chen C, Chao W, Panagia M, Sosnovik DE, Puppala D, Armoundas AA, Hindle A, Bloch KD, Buys ES, Scherrer-Crosbie M. Functional brown adipose tissue limits cardiomyocyte injury and adverse remodeling in catecholamine-induced cardiomyopathy. J Mol Cell Cardiol 2015; 84:202-11. [PMID: 25968336 DOI: 10.1016/j.yjmcc.2015.05.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/18/2015] [Accepted: 05/01/2015] [Indexed: 11/25/2022]
Abstract
Brown adipose tissue (BAT) has well recognized thermogenic properties mediated by uncoupling protein 1 (UCP1); more recently, BAT has been demonstrated to modulate cardiovascular risk factors. To investigate whether BAT also affects myocardial injury and remodeling, UCP1-deficient (UCP1(-/-)) mice, which have dysfunctional BAT, were subjected to catecholamine-induced cardiomyopathy. At baseline, there were no differences in echocardiographic parameters, plasma cardiac troponin I (cTnI) or myocardial fibrosis between wild-type (WT) and UCP1(-/-) mice. Isoproterenol infusion increased cTnI and myocardial fibrosis and induced left ventricular (LV) hypertrophy in both WT and UCP1(-/-) mice. UCP1(-/-) mice also demonstrated exaggerated myocardial injury, fibrosis, and adverse remodeling, as well as decreased survival. Transplantation of WT BAT to UCP1(-/-) mice prevented the isoproterenol-induced cTnI increase and improved survival, whereas UCP1(-/-) BAT transplanted to either UCP1(-/-) or WT mice had no effect on cTnI release. After 3 days of isoproterenol treatment, phosphorylated AKT and ERK were lower in the LV's of UCP1(-/-) mice than in those of WT mice. Activation of BAT was also noted in a model of chronic ischemic cardiomyopathy, and was correlated to LV dysfunction. Deficiency in UCP1, and accompanying BAT dysfunction, increases cardiomyocyte injury and adverse LV remodeling, and decreases survival in a mouse model of catecholamine-induced cardiomyopathy. Myocardial injury and decreased survival are rescued by transplantation of functional BAT to UCP1(-/-) mice, suggesting a systemic cardioprotective role of functional BAT. BAT is also activated in chronic ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Robrecht Thoonen
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Laura Ernande
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; DHU Ageing Thorax Vessels Blood, Inserm Unit 955 Team 08, Faculté de Medecine de Créteil, Hôpital Henri Mondor, AP-HP, Créteil, France
| | - Juan Cheng
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Department of Ultrasound, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yasuko Nagasaka
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Department of Anesthesia and Critical Pain, Massachusetts General Hospital, Boston, MA, USA
| | - Vincent Yao
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Chan Chen
- Department of Anesthesia and Critical Pain, Massachusetts General Hospital, Boston, MA, USA
| | - Wei Chao
- Department of Anesthesia and Critical Pain, Massachusetts General Hospital, Boston, MA, USA
| | - Marcello Panagia
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - David E Sosnovik
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Dheeraj Puppala
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Antonis A Armoundas
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Allyson Hindle
- Department of Anesthesia and Critical Pain, Massachusetts General Hospital, Boston, MA, USA
| | - Kenneth D Bloch
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Department of Anesthesia and Critical Pain, Massachusetts General Hospital, Boston, MA, USA
| | - Emmanuel S Buys
- Department of Anesthesia and Critical Pain, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
38
|
Miwa K, Tamai S, Kinpara Y, Komatsu S, Goto M, Iguchi T, Suzuki T, Takasaki W, Mori K. Impact of different blood sampling techniques on plasma biomarkers for skeletal myopathy in conscious rats. ACTA ACUST UNITED AC 2015. [DOI: 10.2131/fts.2.25] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Kyoko Miwa
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Satoshi Tamai
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
| | | | - Satomi Komatsu
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Mayumi Goto
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Takuma Iguchi
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Takami Suzuki
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Wataru Takasaki
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
| | - Kazuhiko Mori
- Medicinal Safety Research Laboratories, Daiichi Sankyo Co., Ltd
| |
Collapse
|
39
|
Tonomura Y, Kato Y, Hanafusa H, Morikawa Y, Matsuyama K, Uehara T, Ueno M, Torii M. Diagnostic and predictive performance and standardized threshold of traditional biomarkers for drug-induced liver injury in rats. J Appl Toxicol 2014; 35:165-72. [PMID: 25186495 DOI: 10.1002/jat.3053] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 07/01/2014] [Accepted: 07/01/2014] [Indexed: 02/05/2023]
Abstract
Traditional biomarkers such as alanine and aspartate aminotransferase (ALT, AST) and total bilirubin (TBIL) have been widely used for detecting drug-induced liver injury (DILI). Although the Food and Drug Administration (FDA) proposed standardized thresholds for human as Hy's law, those for animals have not been determined, and predictability of these biomarkers for future onset of hepatic lesions remains unclear. In this study, we investigated these diagnostic and predictive performance of 10 traditional biomarkers for liver injury by receiver-operating characteristic (ROC) curve, using a free-access database where 142 hepatotoxic or non-hepatotoxic compounds were administrated to male rats (n=5253). Standardization of each biomarker value was achieved by calculating the ratio to control mean value, and the thresholds were determined under the condition of permitting 5% false positive. Of these 10 biomarkers, AST showed the best diagnostic performance. Furthermore, ALT and TBIL also showed high performance under the situation of hepatocellular necrosis and bile duct injury, respectively. Additionally, the availability of the diagnostic thresholds in difference testing facility was confirmed by the application of these thresholds to in-house prepared dataset. Meanwhile, incorrect diagnosis by the thresholds was also observed. Regarding prediction, all 10 biomarkers showed insufficient performance for future onset of hepatic lesions. In conclusion, the standardized diagnostic thresholds enable consistent evaluation of traditional biomarkers among different facilities, whereas it was suggested that novel biomarker is required for more accurate diagnosis and prediction of DILI.
Collapse
Affiliation(s)
- Yutaka Tonomura
- Drug Safety Evaluation, Research Laboratory for Development, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Nishimura Y, Kondo C, Morikawa Y, Tonomura Y, Torii M, Yamate J, Uehara T. Plasma miR-208 as a useful biomarker for drug-induced cardiotoxicity in rats. J Appl Toxicol 2014; 35:173-80. [PMID: 25092230 DOI: 10.1002/jat.3044] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 06/09/2014] [Accepted: 06/11/2014] [Indexed: 12/13/2022]
Abstract
Cardiotoxicity is one of the major safety concerns in drug development. Therefore, detecting and monitoring cardiotoxicity throughout preclinical and clinical studies is important for pharmaceutical companies. The present study was conducted in order to explore a plasma miRNA biomarker for cardiotoxicity in rats. As organ specificity is an important factor for a biomarker, we analyzed the miRNA microarray dataset in 55 organs/tissues in normal male rats. Based on this analysis, 5 miRNAs consisting of miR-208 (heart-specific), miR-1, miR-133a, miR-133b (heart and skeletal muscle-specific) and miR-206 (skeletal muscle-specific) were selected. Next, we evaluated the usefulness of those 5 miRNAs as circulating biomarkers in rats administered with single-dose isoproterenol or doxorubicin. Plasma miR-208 was consistently increased through 24 h after dosing in rats administered with isoproterenol, whereas plasma concentrations of cardiac troponin (cTn) showed transient elevation. In contrast, the plasma levels of miR-1, miR-133a, miR-133a and miR-206 were elevated after treatment with doxorubicin, probably as a result of skeletal muscle toxicity. Additionally, the plasma miR-208 level was elevated even after repeat-dose administration (once daily for 7 days) of isoproterenol under which the pathological condition proceeded to the sub-chronic phase such as fibrosis. Thus, our data suggest that miR-208 is a promising plasma biomarker for cardiotoxicity in rats. Monitoring of plasma miR-208 levels in rats may lead to more accurate evaluation of cardiotoxicity in preclinical studies.
Collapse
Affiliation(s)
- Yoko Nishimura
- Research Laboratory for Development, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | | | | | | | | | | | | |
Collapse
|
41
|
Herman E, Knapton A, Zhang J, Estis J, Todd J, Lipshultz S. The utility of serum biomarkers to detect myocardial alterations induced by Imatinib in rats. Pharmacol Res Perspect 2014; 2:e00015. [PMID: 25505575 PMCID: PMC4186398 DOI: 10.1002/prp2.15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/10/2013] [Accepted: 10/15/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Imatinib (Imb) is a tyrosine kinase inhibitor with cardiotoxic activity (decreases in left ventricular function and congestive heart failure) in patients. Currently, clinical diagnosis of Imb cardiotoxicity relies primarily on evaluation of left ventricular function, Imb also induces cardiac lesions in rats. AIMS This study, in rats, sought to determine whether monitoring biochemical markers would be a sensitive means to detect Imb-induced changes in cardiomyocyte morphology. MATERIALS AND METHODS Groups of male Sprague-Dawley rats were dosed orally with 50, 100, 200 mg kg(-1) Imb or water daily for 28 days. Tissues and blood samples were collected 24 h after the last dosing. Cardiac biomarkers such as cardiac troponin I (cTnI), cardiac troponin T (cTnT), and fatty acid binding protein 3 (FABP3) were monitored by the Erenna, Elecsys, and Meso Scale immunoassay systems. RESULTS Imb caused microscopic myocardial lesions (myofibrillar loss, cytoplasmic vacuolization, and necrosis) at all doses as determined by unbiased histopathology analysis. The severity of the alterations was dose-related with mean lesion scores (based on a scale of 0-3) of 1.2 (50 mg kg(-1)), 2.1 (100 mg kg(-1)) and 2.9 (200 mg kg(-1)). However, the increases in cTnI, cTnT, and FABP3 levels were noted primarily in high-dose Imb treated animals. DISCUSSION AND CONCLUSION The occurrence of myocardial alterations in animals without consistent changes in cardiac troponin and FABP3 concentrations raises questions regarding the utility of these biomarkers as early indicators of Imb-induced cardiotoxicity. Due to limited numbers of animals the reasons for this discrepancy could not be determined.
Collapse
Affiliation(s)
- Eugene Herman
- Division of Drug Safety Research, Food and Drug Administration Silver Spring, Maryland
| | - Alan Knapton
- Division of Drug Safety Research, Food and Drug Administration Silver Spring, Maryland
| | - Jun Zhang
- Division of Drug Safety Research, Food and Drug Administration Silver Spring, Maryland
| | | | | | - Steven Lipshultz
- Department of Pediatrics, Mailman Center for Child Development, Leonard A. Miller School of Medicine, University of Miami Miami, Florida
| |
Collapse
|
42
|
Ahuja V, Sharma S. Drug safety testing paradigm, current progress and future challenges: an overview. J Appl Toxicol 2013; 34:576-94. [PMID: 24777877 DOI: 10.1002/jat.2935] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 08/08/2013] [Accepted: 08/22/2013] [Indexed: 12/29/2022]
Abstract
Early assessment of the toxicity potential of new molecules in pharmaceutical industry is a multi-dimensional task involving predictive systems and screening approaches to aid in the optimization of lead compounds prior to their entry into development phase. Due to the high attrition rate in the pharma industry in last few years, it has become imperative for the nonclinical toxicologist to focus on novel approaches which could be helpful for early screening of drug candidates. The need is that the toxicologists should change their classical approach to a more investigative approach. This review discusses the developments that allow toxicologists to anticipate safety problems and plan ways to address them earlier than ever before. This includes progress in the field of in vitro models, surrogate models, molecular toxicology, 'omics' technologies, translational safety biomarkers, stem-cell based assays and preclinical imaging. The traditional boundaries between teams focusing on efficacy/ safety and preclinical/ clinical aspects in the pharma industry are disappearing, and translational research-centric organizations with a focused vision of bringing drugs forward safely and rapidly are emerging. Today's toxicologist should collaborate with medicinal chemists, pharmacologists, and clinicians and these value-adding contributions will change traditional toxicologists from side-effect identifiers to drug development enablers.
Collapse
Affiliation(s)
- Varun Ahuja
- Drug Safety Assessment, Novel Drug Discovery and Development, Lupin Limited (Research Park), 46A/47A, Nande Village, MulshiTaluka, Pune, 412 115, India
| | | |
Collapse
|