1
|
Jin SE, Lee MY, Ha H, Shin HK, Seo CS. Safety evaluation of Gamisoyo-san: genotoxicity, acute toxicity, and influence on drug-metabolizing enzymes. Drug Chem Toxicol 2024:1-10. [PMID: 38291610 DOI: 10.1080/01480545.2024.2308830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
Gamisoyo-san is an herbal formula widely used to treat psychological issues, menopausal symptoms, and dysmenorrhea. However, there is insufficient information on its safety profile. This study aimed to confirm the genotoxic and acute toxic potential of Gamisoyo-san. We performed a battery of tests, which included a bacterial reverse mutation test (Ames test) using five bacterial strains, an in vitro chromosomal aberration test using Chinese hamster lung (CHL) cells, an in vivo micronucleus test in mice, and human Cytochrome P450 (CYP450) and UDP-glucuronosyltransferase (UGT) assays. In the acute toxicity study, male and female rats were orally administered Gamisoyo-san 1000, 2000, or 5000 mg/kg and observed for 14 days. The activities of human CYP450s and UGTs were evaluated using recombinant baculosomes. Gamisoyo-san showed no signs of genotoxicity in the five bacterial strains, CHL cells, or mouse bone marrow cells. The acute toxicity test showed that the median lethal dose (LD50) of Gamisoyo-san was greater than 5000 mg/kg in rats. Gamisoyo-san inhibited the activities of CYP1A2, CYP2C19, and UGT1A1. In conclusion, Gamisoyo-san may not exert severe toxicological events or genotoxic effects at doses up to 5000 mg/kg in rats.
Collapse
Affiliation(s)
- Seong Eun Jin
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Mee-Young Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Hyekyung Ha
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Hyeun-Kyoo Shin
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Chang-Seob Seo
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| |
Collapse
|
2
|
Sartini I, Vercelli C, Lebkowska-Wieruszewska B, Lisowski A, Fadel C, Poapolathep A, Dessì F, Giorgi M. Pharmacokinetics and antibacterial activity of tiamulin after single and multiple oral administrations in geese. Vet Anim Sci 2023; 22:100317. [PMID: 37920697 PMCID: PMC10618762 DOI: 10.1016/j.vas.2023.100317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023] Open
Abstract
Tiamulin is an antibiotic approved exclusively in veterinary medicine, active against G-positive bacteria as well as Mycoplasma spp. and Leptospirae spp. The study was aimed to establish its pharmacokinetics and to evaluate drug effects on resistance in cloacal flora in vivo in geese. Eight healthy geese underwent to a two-phase longitudinal study (60 mg/kg single oral administration vs 60 mg/kg/day for 4 days) with a two-week wash-out period. Blood samples and cloacal swabs were collected at pre-assigned times. Minimal inhibitory concentration (MIC) has been evaluated for each isolated bacterial species. The pharmacokinetic parameters that significantly differed between the groups were Cmax (p = 0.024), AUC0-t (p = 0.031), AUC0-inf (p = 0.038), t1/2kel (p = 0.021), Cl/F (p = 0.036), and Vd/F (p = 0.012). Tiamulin exhibited a slow to moderate terminal half-life (3.13 h single; 2.62 h multiple) and a rapid absorption (1 h single; 0.5 h multiple) in geese, with an accumulation ratio of 1.8 after multiple doses. An in-silico simulation of multiple dosing did not reflect the results of the in vivo multiple dosage study. In both treatments, the MIC values were very high demonstrating a resistance (> 64 μg/ml) against tiamulin that can be present prior the drug administration for some strains, or emerge shortly after the commencing of treatment for some others.
Collapse
Affiliation(s)
- Irene Sartini
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Cristina Vercelli
- Department of Veterinary Sciences, University of Turin, Torino, Italy
| | | | - Andrzej Lisowski
- Department of Biology and Animal Breeding, University of Life Sciences, Lublin, Poland
| | - Charbel Fadel
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Amnart Poapolathep
- Department of Pharmacology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Filomena Dessì
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Mario Giorgi
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
- Department of Veterinary Sciences, University of Pisa, Via Livornese (lato monte), 56122, San Piero a Grado, Pisa, Italy
| |
Collapse
|
3
|
Spanakis M, Alon-Ellenbogen D, Ioannou P, Spernovasilis N. Antibiotics and Lipid-Modifying Agents: Potential Drug-Drug Interactions and Their Clinical Implications. PHARMACY 2023; 11:130. [PMID: 37624085 PMCID: PMC10457919 DOI: 10.3390/pharmacy11040130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/30/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Evidence-based prescribing requires taking into consideration the many aspects of optimal drug administration (e.g., dosage, comorbidities, co-administered drugs, etc.). A key issue is the administration of drugs for acute disorders that may potentially interfere with previously prescribed long-term medications. Initiating an antibiotic for an acute bacterial infection constitutes a common example. Hence, appropriate knowledge and awareness of the potential DDIs of antibiotics would lead to proper adjustments, thus preventing over- or under-treatment. For example, some statins, which are the most prescribed lipid-modifying agent (LMA), can lead to clinically important drug-drug interactions (DDIs) with the concurrent administration of antibiotics, e.g., macrolides. This review discusses the clinically significant DDIs of antibiotics associated with co-administrated lipid-lowering therapy and highlights common cases where regimen modifications may or may not be necessary.
Collapse
Affiliation(s)
- Marios Spanakis
- Department Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece;
- Computational Biomedicine Laboratory, Institute of Computer Science, Foundation for Research & Technology-Hellas (FORTH), 70013 Heraklion, Greece
| | - Danny Alon-Ellenbogen
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, 2417 Nicosia, Cyprus;
| | - Petros Ioannou
- Department of Internal Medicine & Infectious Diseases, University Hospital of Heraklion, 71110 Heraklion, Greece;
| | | |
Collapse
|
4
|
Acute toxicity and genotoxicity studies on new melatonergic antidepressant GW117. Heliyon 2023; 9:e14026. [PMID: 36915542 PMCID: PMC10006472 DOI: 10.1016/j.heliyon.2023.e14026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/08/2023] [Accepted: 02/20/2023] [Indexed: 02/27/2023] Open
Abstract
GW117, a novel derivate compound of agomelatine that acts as both a 5-HT2C receptor antagonist and a MT1/MT2 receptor agonist, likely underlines the potent antidepressant action with less hepatotoxicity than agomelatine. We evaluated the acute toxicity of GW117, and the genotoxicity of GW117 using bacterial reverse mutation test, mammalian chromosomal aberration test in Chinese hamster lung cells (CHL) and mouse bone marrow micronucleus test. The acute toxicity test results showed that maximum tolerated dose (MTD) of GW117 was 2000 mg/kg, under which mean Cmax and AUC0→t was 10,782 ng/mL and 81,046 ng/mL × h, respectively. The result of bacterial reverse mutation test showed that the number of bacterial colonies in each dose group of GW117 did not increase significantly compared with that in the solvent control group with or without S9 metabolic activation system. In vitro chromosome aberration test of CHL cells, the chromosome aberration rate of each dose group of GW117 did not increase with or without S9 metabolic activation system. In mouse micronucleus test, the highest dose was 2000 mg/kg, the micronucleus rate did not increase significantly. Under the conditions of this study, the MTD of a single GW117 administration was 2000 mg/kg, there was no genotoxicity effect of GW117.
Collapse
|
5
|
Fu Y, Yang Z, Zhang H, Liu Y, Hao B, Shang R. 14-O-[(4,6-Diamino-pyrimidine-2-yl) thioacetyl] mutilin inhibits α-hemolysin and protects Raw264.7 cells from injury induced by methicillin-resistant S. aureus. Microb Pathog 2021; 161:105229. [PMID: 34624494 DOI: 10.1016/j.micpath.2021.105229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/12/2021] [Accepted: 09/17/2021] [Indexed: 10/20/2022]
Abstract
A new pleuromutilin derivative, 14-O-[(4,6-Diaminopyrimidine-2-yl) thioacetyl] mutilin (DPTM), has been synthesized and proven to be a potent agent against Gram-positive pathogens, especially for Staphylococcus aureus (S. aureus). However, its pharmacological activities against α-hemolysin (Hla), a major virulence factor produced by S. aureus, and inflammations related to S. aureus are still unknown. In the present study, we investigated the DPTM inhibition activities against methicillin-resistant S. aureus (MRSA) Hla and protective efficacy of Raw264.7 cells from injury induced by MRSA. The results showed that DPTM with sub-inhibitory concentrations significantly inhibited Hla on the hemolysis of rabbit erythrocytes and down-regulated the gene expressions of Hla and agrA with a dose-dependent fashion. In Raw264.7 cells infected with MRSA, DPTM efficiently attenuated the productions of lactate dehydrogenase (LDH), nitric oxide (NO) and pro-inflammatory cytokines, as well as the express levels of nuclear factor-kappaB (NF-κB), nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Furthermore, DPTM inhibited the translocation of p-65 to nucleus in RAW264.7 cells infected by MRSA.
Collapse
Affiliation(s)
- Yunxing Fu
- Key Laboratory of New Animal Drug Project, Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, 730050, Lanzhou, PR China; College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, 450046, Zhengzhou, PR China.
| | - Zhen Yang
- Key Laboratory of New Animal Drug Project, Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, 730050, Lanzhou, PR China
| | - Hongjuan Zhang
- Key Laboratory of New Animal Drug Project, Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, 730050, Lanzhou, PR China
| | - Yu Liu
- Key Laboratory of New Animal Drug Project, Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, 730050, Lanzhou, PR China
| | - Baocheng Hao
- Key Laboratory of New Animal Drug Project, Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, 730050, Lanzhou, PR China
| | - Ruofeng Shang
- Key Laboratory of New Animal Drug Project, Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, 730050, Lanzhou, PR China.
| |
Collapse
|
6
|
Huang HJ, Lee YH, Hsu YH, Liao CT, Lin YF, Chiu HW. Current Strategies in Assessment of Nanotoxicity: Alternatives to In Vivo Animal Testing. Int J Mol Sci 2021; 22:4216. [PMID: 33921715 PMCID: PMC8073679 DOI: 10.3390/ijms22084216] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/01/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Millions of experimental animals are widely used in the assessment of toxicological or biological effects of manufactured nanomaterials in medical technology. However, the animal consciousness has increased and become an issue for debate in recent years. Currently, the principle of the 3Rs (i.e., reduction, refinement, and replacement) is applied to ensure the more ethical application of humane animal research. In order to avoid unethical procedures, the strategy of alternatives to animal testing has been employed to overcome the drawbacks of animal experiments. This article provides current alternative strategies to replace or reduce the use of experimental animals in the assessment of nanotoxicity. The currently available alternative methods include in vitro and in silico approaches, which can be used as cost-effective approaches to meet the principle of the 3Rs. These methods are regarded as non-animal approaches and have been implemented in many countries for scientific purposes. The in vitro experiments related to nanotoxicity assays involve cell culture testing and tissue engineering, while the in silico methods refer to prediction using molecular docking, molecular dynamics simulations, and quantitative structure-activity relationship (QSAR) modeling. The commonly used novel cell-based methods and computational approaches have the potential to help minimize the use of experimental animals for nanomaterial toxicity assessments.
Collapse
Affiliation(s)
- Hung-Jin Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Yu-Hsuan Lee
- Department of Cosmeceutics, China Medical University, Taichung 406040, Taiwan;
| | - Yung-Ho Hsu
- Division of Nephrology, Department of Internal Medicine, Hsin Kuo Min Hospital, Taipei Medical University, Taoyuan City 320001, Taiwan;
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
| | - Chia-Te Liao
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Yuh-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| | - Hui-Wen Chiu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei 11031, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Department of Medical Research, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| |
Collapse
|