1.Integrated multiomics reveal mechanism of Aidi Injection in attenuating doxorubicin-induced cardiotoxicity.
Yan-Li WANG ; Yu-Jie TU ; Jian-Hua ZHU ; Lin ZHENG ; Yong HUANG ; Jia SUN ; Yong-Jun LI ; Jie PAN ; Chun-Hua LIU ; Yuan LU
China Journal of Chinese Materia Medica 2025;50(8):2245-2259
The combination of Aidi Injection(ADI) and doxorubicin(DOX) is a common strategy in the treatment of cancer, which can achieve synergistic anti-tumor effects while attenuating the cardiotoxicity caused by DOX. This study aims to investigate the mechanism of ADI in attenuating DOX-induced cardiotoxicity by multi-omics. DOX was used to induce cardiotoxicity in mice, and the cardioprotective effects of ADI were evaluated based on biochemical indicators and pathological changes. Based on the results, transcriptomics, proteomics, and metabolomics were employed to analyze the changes of endogenous substances in different physiological states. Furthermore, data from multiple omics were integrated to screen key regulatory pathways by which ADI attenuated DOX-induced cardiotoxicity, and important target proteins were selected for measurement by ELISA kits and immunohistochemical analysis. The results showed that ADI significantly reduced the levels of cardiac troponin T(cTnT) and N-terminal pro-B-type natriuretic peptide(NT-proBNP) and effectively ameliorated myocardial fibrosis and intracellular vacuolization, indicating that ADI showed therapeutic effect on DOX-induced cardiotoxicity. The transcriptomics analysis screened out a total of 400 differentially expressed genes(DEGs), which were mainly enriched in inflammatory response, oxidative stress, and myocardial fibrosis. After proteomics analysis, 70 differentially expressed proteins were selected, which were mainly enriched in the inflammatory response, cardiac function, and energy metabolism. A total of 51 differentially expressed metabolites were screened by the metabolomics analysis, and they were mainly enriched in multiple signaling pathways, including the inflammatory response, lipid metabolism, and energy metabolism. The integrated data of multiple omics showed that linoleic acid metabolism, arachidonic acid metabolism, and glycerophosphate metabolism pathways played an important role in DOX-induced cardiotoxicity, and ADI may exert therapeutic effects by modulating these pathways. Target validation experiments suggested that ADI significantly regulated abnormal protein levels of cyclooxygenase-1(COX-1), cyclooxygenase-2(COX-2), prostaglandin H2(PGH2), and prostaglandin D2(PGD2) in the model group. In conclusion, ADI may attenuate DOX-induced cardiotoxicity by regulating linoleic acid metabolism, arachidonic acid metabolism, and glycerophosphate metabolism, thus alleviating inflammation of the body.
Doxorubicin/toxicity*
;
Animals
;
Mice
;
Cardiotoxicity/genetics*
;
Drugs, Chinese Herbal/administration & dosage*
;
Male
;
Proteomics
;
Metabolomics
;
Injections
;
Humans
;
Multiomics
2.Protective effect of achyranthes bidentata against doxorubicin-induced spermatogenic disorder in mice: An investigation based on the glycolytic metabolic pathway.
Man-Yu WANG ; Yang FU ; Pei-Pei YUAN ; Li-Rui ZHAO ; Yan ZHANG ; Qing-Yun MA ; Yan-Jun SUN ; Wei-Sheng FENG ; Xiao-Ke ZHENG
National Journal of Andrology 2025;31(2):99-107
OBJECTIVE:
To investigate the protective effect of achyranthes bidentata (AB) on sperm quality in mice with spermatogenic disorder through the glycolytic metabolic pathway and its action mechanism.
METHODS:
We equally randomized 40 Kunming mice into a normal control, a model control, a low-dose AB (3.5 g/kg) and a high-dose AB group (7.0 g/kg), and established the model of spermatogenic disorder in the latter three groups of mice by intraperitoneal injection of doxorubicin (30 mg/kg). Two days after modeling, we collected the testis and kidney tissues and blood samples from the mice for observation of the pathological changes in the testis tissue by HE staining, detection of perm motility with the sperm quality analyzer, examination of the apoptosis of testis cells by flow cytometry, measurement of the levels of testosterone (T), malondialdehyde (MDA), superoxide dismutase (SOD) and catalase (CAT) in the serum and testis tissue by ELISA, and determination of expressions of the key enzymes of glycolysis hexokinase Ⅱ (HK2), pyruvate kinase M2 (PKM2), platelet phosphofructokinase (PFKP), lactate dehydrogenase A (LDHA) and the meiosis proteins REC8 and SCP3 by Western blot, and the mRNA expressions of glycolytic phosphofructokinase 1 (PFK1), phosphoglycerate kinase 1 (PGK1), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) by fluorescence quantitative PCR (FQ-PCR).
RESULTS:
Compared with the model controls, the mice in the AB groups showed significant increases in the testis coefficient, kidney index, sperm concentration, sperm motility, spermatogonia, primary spermatocytes, spermatids, sperm count and the serum T level (P<0.05 or P<0.01), but dramatic decreases in the apoptosis of testis cells and percentage of morphologically abnormal sperm (P<0.01). Achyranthes bidentata also significantly elevated the levels of SOD and CAT, and down-regulated the mRNA expressions of MDA, TNF-α and IL-1β (P<0.05 or P<0.01), and up-regulated the protein expressions of HK2, PKM2, PFKP, LDHA, REC8 and SCP3, and expressions of the glycolysis key genes Pfk1 and Pgk1 (P<0.05 or P<0.01).
CONCLUSION
Achyranthes bidentata ameliorates doxorubicin-induced spermatogenic disorder in mice by regulating the glycolytic pathway and reducing oxidative stress and the expressions of inflammatory factors.
Glycolysis/drug effects*
;
Doxorubicin/toxicity*
;
Spermatogenesis/drug effects*
;
Random Allocation
;
Male
;
Animals
;
Mice
;
Disease Models, Animal
;
Achyranthes/chemistry*
;
Spermatozoa/pathology*
;
Oxidative Stress/drug effects*
;
Primary Cell Culture
;
Apoptosis/drug effects*
;
Sperm Motility/drug effects*
;
Testis/pathology*
;
Infertility, Male/prevention & control*
;
Medicine, Chinese Traditional/methods*
;
Animals, Outbred Strains
3.Hesperetin alleviates doxorubicin-induced cardiotoxicity by regulating the AMPK/NLRP3 pathway.
Aili YAN ; Mengyao LUO ; Jinrui CHANG ; Xinhua LI ; Juanxia ZHU
Journal of Southern Medical University 2025;45(9):1850-1858
OBJECTIVES:
To verify whether hesperetin (Hes) alleviates doxorubicin (DOX)-induced cardiotoxicity by reducing inflammation via regulating the AMPK/NLRP3 pathway.
METHODS:
C57/bl6 mice and H9c2 cells treated with DOX to mimic cardiotoxicity were randomly divided into Sham (or control) group, DOX group, DOX+Hes group, DOX+Hes+compound C (CC, an AMPK inhibitor) group. Cardiac function and myocardial pathologies of the mice were evaluated, and the changes in H9c2 cell morphology and viability were assessed. Lactate dehydrogenase (LDH) activity in mouse myocardial tissues and H9c2 cells was measured using ELISA, and H9c2 cell apoptosis was detected with TUNEL staining. In both H9c2 cells and the myocardial tissues of the mice, cellular expression levels of TNF-α, IL-6 and IL-1β mRNAs and cleaved caspase-3, Bcl2, Bax, IL-1β, IL-18, p-AMPK, AMPK, p-mTOR, mTOR, NLRP3, ASC and caspase-1 proteins were detected using RT-PCR and Western blotting.
RESULTS:
DOX treatment caused cell swelling, decreased cell viability and increased LDH activity in H9c2 cells, resulting also in significantly increased cell apoptosis and cleaved caspase-3 expression and decreased Bcl2/Bax ratio. The DOX-treated mice showed obvious myocardial fiber swelling and inflammatory infiltration, decreased cardiac function and significantly increased myocardial LDH activity. In H9c2 cells, DOX treatment significantly increased the mRNA expressions of TNF-α, IL-6 and IL-1β and protein expressions of IL-1β and IL-18, lowered the expressions of p-AMPK and p-mTOR, and increased the expressions of NLRP3, ASC and caspase-1. Hes treatment obviously reduced these toxic effects of DOX in H9c2 cells, but its protective effects were blocked by application of compound C.
CONCLUSIONS
Hes reduces DOX-induced cardiotoxicity by inhibiting inflammation via regulating the AMPK/NLRP3 pathway.
Animals
;
Doxorubicin/toxicity*
;
NLR Family, Pyrin Domain-Containing 3 Protein/metabolism*
;
Mice, Inbred C57BL
;
Mice
;
Signal Transduction/drug effects*
;
Cardiotoxicity
;
AMP-Activated Protein Kinases/metabolism*
;
Apoptosis/drug effects*
;
Cell Line
;
Myocytes, Cardiac/drug effects*
;
Rats
4.Study on the effects of telomerase reverse transcriptase in alleviating doxorubicin induced cardiotoxicity.
Qingqing GU ; Qianwe CHEN ; Yu WANG ; Dabei CAI ; Tingting XIAO ; Qingjie WANG ; Ling SUN
Chinese Critical Care Medicine 2025;37(6):583-589
OBJECTIVE:
To investigate the role of telomerase reverse transcriptase (TERT) in alleviating doxorubicin (DOX)-induced cardiotoxicity.
METHODS:
(1) Cell experiments: rat H9c2 cardiomyocytes were divided into control group (CON group), null adenovirus transfection group (NC group), TERT overexpression adenovirus transfection group (TERT group), DOX group (treated with 1 μmol/L DOX for 12 hours), DOX+NC group, and DOX+TERT group (null adenovirus or TERT overexpression adenovirus were transfected for 24 hours and then treated with 1 μmol/L DOX for 12 hours). The mRNA expression of TERT in cardiomyocytes was detected by real-time fluorescence quantitative polymerase chain reaction (RT-qPCR). The level of mitochondrial membrane potential was detected by immunofluorescence. The expression levels of intracellular Bax, Bcl-2, microtubule-associated protein 1 light chain 3 (LC3) and p62 were detected by Western blotting. (2) Animal experiments: male C57BL/6 mice were randomly divided into a sham operation group (Sham group), DOX group (acute cardiotoxicity model was constructed by intraperitoneal injection of DOX 15 mg/kg), DOX+NC group and DOX+TERT group (modeled after transfection with airborne adenovirus or TERT overexpression adenovirus for 7 days). After 7 days of modeling, the area of myocardial fibrosis was detected by Sirius scarlet staining, and cardiac function was detected by echocardiography.
RESULTS:
(1) Cellular experiments: the mRNA expression level of TERT was significantly higher in the TERT group compared with the CON and NC groups. Compared with the CON group, the TERT mRNA expression level of cardiomyocytes in the DOX group and the DOX+NC group were significantly lower, the level of mitochondrial membrane potential was significantly lower, the protein expressions of Bax and LC3 were significantly increased, and the protein expressions of Bcl-2 and p62 were significantly decreased. No significant differences were found between the DOX group and DOX+NC group. Compared with the DOX group and DOX+NC group, the TERT mRNA expression level was increased in the DOX+TERT group (relative expression: 1.02±0.10 vs. 0.61±0.05, 0.54±0.03, both P < 0.05), the level of mitochondrial membrane potential was significantly increased (1.14±0.05 vs. 0.96±0.01, 0.96±0.01, both P < 0.05), the protein expressions of Bax and LC3 were significantly decreased, and the protein expressions of Bcl-2 and p62 were significantly increased (Bax/β-actin: 0.88±0.01 vs. 1.31±0.02, 1.26±0.01; LC3-II/I: 2.16±0.05 vs. 2.64±0.06, 2.58±0.02; Bcl-2/β-actin: 0.65±0.01 vs. 0.40±0.01, 0.41±0.01; p62/β-actin: 0.45±0.01 vs. 0.23±0.02, 0.29±0.01; all P < 0.05). (2) Animal experiments: compared with the Sham group, the percentage of myocardial fibrosis area was significantly increased and left ventricular ejection fraction (LVEF) and fractional shortening (FS) were significantly decreased in the DOX group and DOX+NC group. Compared with the DOX group and DOX+NC group, the percentage of myocardial fibrotic area was significantly decreased in the DOX+TERT group (%: 2.33±0.06 vs. 3.76±0.07, 3.87±0.06, both P < 0.05), and the LVEF and FS were significantly increased [LVEF (%): 67.00±1.14 vs. 54.60±1.57, 53.40±2.18; FS (%): 38.60±0.51 vs. 30.60±1.10, 30.00±0.71; all P < 0.05].
CONCLUSION
Up-regulation of TERT expression can inhibit DOX-induced cardiomyocyte autophagy and apoptosis, attenuate DOX-induced myocardial fibrosis in mice, improve cardiac function, and thus alleviate DOX-induced cardiotoxicity.
Animals
;
Doxorubicin/toxicity*
;
Telomerase/metabolism*
;
Myocytes, Cardiac/metabolism*
;
Rats
;
Male
;
Cardiotoxicity
;
Mice, Inbred C57BL
;
Mice
;
Membrane Potential, Mitochondrial
;
Adenoviridae
;
bcl-2-Associated X Protein/metabolism*
;
Proto-Oncogene Proteins c-bcl-2/metabolism*
;
Transfection
;
Apoptosis
5.Kaixin San ameliorating doxorubicin-induced neurotoxicity by activating AMPK signaling pathway.
Ying-Chao WU ; Jia-Qi CUI ; Hui WANG ; Da-Jin PI ; Li-Guo CHEN ; Ming-Zi OUYANG ; Qian-Jun CHEN
China Journal of Chinese Materia Medica 2024;49(24):6763-6772
The study explored the pathological mechanism of doxorubicin chemotherapy-induced neurotoxicity and the intervention methods of traditional Chinese medicine. BALB/c mice were selected to establish tumor-bearing mouse models by orthotopic injection of 4T1 triple-negative breast cancer cells. After randomization, the mice were treated with doxorubicin chemotherapy or doxorubicin chemotherapy + Kaixin San(KXS). The lesions in the prefrontal cortex of mice were observed by pathological examination, and the lesion information was obtained by long non-coding RNA sequencing. The occurrence of lesions was determined by Western blot and biochemical indicators. In addition, neuroblastoma cells and microglia cells were used to construct in vitro models, and drug-containing serum and p-AMPK dephosphorylation inhibitors were used to further verify the accuracy of animal experiments. Pathological results showed that KXS could alleviate doxorubicin-induced neuronal degeneration in the prefrontal cortex. The long non-coding RNA sequencing suggested that neuronal degeneration and the intervention process of KXS were related to ferroptosis, immune diseases, AMPK signaling pathway, etc. Western blot and biochemical indicators confirmed that this process was directly related to the activation of the AMPK/HIF-1α/ACSL4 signaling pathway to alleviate ferroptosis of neurons and immune response of glial cells. In conclusion, KXS could alleviate doxorubicin-induced neurotoxicity by activating the AMPK signaling pathway and reducing the ferroptosis of neurons and immune response of glial cells.
Animals
;
Doxorubicin/toxicity*
;
Mice
;
AMP-Activated Protein Kinases/genetics*
;
Signal Transduction/drug effects*
;
Mice, Inbred BALB C
;
Drugs, Chinese Herbal/administration & dosage*
;
Female
;
Humans
;
Cell Line, Tumor
;
Neurotoxicity Syndromes/genetics*
6.Astragalus polysaccharides affects multidrug resistance gene 1 and P-glycoprotein 170 in adriamycin nephropathy rats via regulating microRNA-16/NF-κB axis.
Xiaoli ZUO ; Lingyun BI ; Hongmin CAO
Journal of Central South University(Medical Sciences) 2022;47(1):26-34
OBJECTIVES:
Nephrotic syndrome is a common disease of the urinary system. The aim of this study is to explore the effect of astragalus polysaccharides (APS) on multidrug resistance gene 1 (MDR1) and P-glycoprotein 170 (P-gp170) in adriamycin nephropathy rats and the underlying mechanisms.
METHODS:
A total of 72 male Wistar rats were divided into a control group, a model group, an APS low-dose group, an APS high-dose group, an APS+micro RNA (miR)-16 antagomir group and an APS+miR-16 antagomir control group, with 12 rats in each group. Urine protein (UP) was detected by urine analyzer, and serum cholesterol (CHOL), albumin (ALB), blood urea nitrogen (BUN), and creatinine (SCr) were detected by automatic biochemical analyzer; serum interleukin-6 (IL-6), IL-1β, tumor necrosis factor α (TNF-α) levels were detected by ELISA kit; the morphological changes of kidney tissues were observed by HE staining; the levels of miR-16 and MDR1 mRNA in kidney tissues were detected by real-time RT-PCR; the expression levels of NF-κB p65, p-NF-κB p65, and P-gp170 protein in kidney tissues were detected by Western blotting; and dual luciferase was used to verify the relationship between miR-16 and NF-κB.
RESULTS:
The renal tissue structure of rats in the control group was normal without inflammatory cell infiltration. The renal glomeruli of rats in the model group were mildly congested, capillary stenosis or occlusion, and inflammatory cell infiltration was obvious. The rats in the low-dose and high-dose APS groups had no obvious glomerular congestion, the proliferation of mesangial cells was significantly reduced, and the inflammatory cells were reduced. Compared with the high-dose APS group and the APS+miR-16 antagomir control group, there were more severe renal tissue structure damages in the APS + miR-16 antagomir group. Compared with the control group, the levels of UP, CHOL, BUN, SCr, IL-6, IL-1β, TNF-α, and MDR1 mRNA, and the protein levels of p-NF-κB p65 and P-gp170 in the model group were significantly increased (all P<0.05); the levels of ALB and miR-16 were significantly decreased (both P<0.05). Compared with the model group, the levels of UP, CHOL, BUN, SCr, IL-6, IL-1β, TNF-α, and MDR1 mRNA, and the protein levels of pNF-κB p65 and P-gp170 in the low-dose and high-dose APS groups were significant decreased (all P<0.05); and the levels of ALB and miR-16 were significantly increased (both P<0.05). Compared with APS+miR-16 antagomir control group, the UP, CHOL, BUN, SCr, IL-6, IL-1β, and TNF-α levels, MDR1 mRNA, and the protein levels of p-NF-κB p65 and P-gp170 were significantly increased (all P<0.05). The levels of ALB and miR-16 were significantly decreased in the APS+miR-16 antagomir group compared with the APS+miR-16 antagomir control group (both P<0.05).
CONCLUSIONS
APS can regulate the miR-16/NF-κB signaling pathway, thereby affecting the levels of MDR1 and P-gp170, and reducing the inflammation in the kidney tissues in the adriamycin nephropathy rats.
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics*
;
Animals
;
Antagomirs
;
Doxorubicin/toxicity*
;
Genes, MDR
;
Interleukin-6/metabolism*
;
Kidney Diseases/genetics*
;
Male
;
MicroRNAs/metabolism*
;
NF-kappa B/metabolism*
;
Polysaccharides/pharmacology*
;
RNA, Messenger
;
Rats
;
Rats, Wistar
;
Tumor Necrosis Factor-alpha/metabolism*
7.Serum Metabolic Profiling in a Mouse Model of Adriamycin-Induced Focal Segmental Glomerulosclerosis.
Li LYU ; Cai-Li WANG ; Zeng-Yan LI ; Ying-Jin SHI ; Yan-Hui ZHANG ; Yan MI ; Zhao HU
Chinese Medical Journal 2018;131(22):2743-2746
Animals
;
Body Weight
;
physiology
;
Computational Biology
;
methods
;
Disease Models, Animal
;
Doxorubicin
;
toxicity
;
Fatty Acids, Monounsaturated
;
blood
;
metabolism
;
Glomerulosclerosis, Focal Segmental
;
blood
;
chemically induced
;
metabolism
;
Male
;
Methoxyhydroxyphenylglycol
;
analogs & derivatives
;
blood
;
metabolism
;
Mice
;
Mice, Inbred BALB C
;
Pyridoxine
;
blood
;
metabolism
;
Valine
;
analogs & derivatives
;
blood
;
metabolism
;
Vanillic Acid
;
blood
;
metabolism
8.Reversal of multidrug resistance by icaritin in doxorubicin-resistant human osteosarcoma cells.
Zhen-Dong WANG ; Rui-Zhi WANG ; Yuan-Zheng XIA ; Ling-Yi KONG ; Lei YANG
Chinese Journal of Natural Medicines (English Ed.) 2018;16(1):20-28
Multidrug resistance (MDR) is one of the major obstacles in cancer chemotherapy. Our previous study has shown that icariin could reverse MDR in MG-63 doxorubicin-resistant (MG-63/DOX) cells. It is reported that icariin is usually metabolized to icariside II and icaritin. Herein, we investigated the effects of icariin, icariside II, and icaritin (ICT) on reversing MDR in MG-63/DOX cells. Among these compounds, ICT exhibited strongest effect and showed no obvious cytotoxicity effect on both MG-63 and MG-63/DOX cells ranging from 1 to 10 μmol·L. Furthermore, ICT increased accumulation of rhodamine 123 and 6-carboxyfluorescein diacetate and enhanced DOX-induced apoptosis in MG-63/DOX cells in a dose-dependent manner. Further studies demonstrated that ICT decreased the mRNA and protein levels of multidrug resistance protein 1 (MDR1) and multidrug resistance-associated protein 1 (MRP1). We also verified that blockade of STAT3 phosphorylation was involved in the reversal effect of multidrug resistance in MG-63/DOX cells. Taken together, these results indicated that ICT may be a potential candidate in chemotherapy for osteosarcoma.
ATP Binding Cassette Transporter, Subfamily B
;
drug effects
;
genetics
;
metabolism
;
Antineoplastic Agents
;
pharmacology
;
Apoptosis
;
drug effects
;
Cell Line, Tumor
;
Cell Survival
;
drug effects
;
Dose-Response Relationship, Drug
;
Doxorubicin
;
metabolism
;
pharmacology
;
toxicity
;
Drug Resistance, Multiple
;
drug effects
;
Drug Resistance, Neoplasm
;
drug effects
;
Flavonoids
;
pharmacology
;
Gene Expression Regulation, Neoplastic
;
drug effects
;
Humans
;
Multidrug Resistance-Associated Proteins
;
drug effects
;
genetics
;
metabolism
;
Osteosarcoma
;
drug therapy
;
metabolism
;
pathology
;
Phosphorylation
;
drug effects
;
Rhodamine 123
;
metabolism
;
STAT3 Transcription Factor
;
antagonists & inhibitors
;
metabolism
;
Triterpenes
;
pharmacology
9.Metabolomics study of doxorubicin induced hepatotoxicity.
Qian-yun NIU ; Yue-tao LIU ; Zhen-yu LI ; Xue-mei QIN
Acta Pharmaceutica Sinica 2015;50(6):708-713
To reveal the underlying mechanism of doxorubicin induced hepatotoxicity, an NMR-based metabolomic approach combined with multivariate statistical analysis was used to observe its metabolic alternations of rat liver. Sixteen differential metabolites between model rats and normal rats were characterized as potential pathological biomarkers related to doxorubicin induced hepatotoxicity. Six pathways, including phenylalanine, tyrosine and tryptophan biosynthesis, valine, leucine and isoleucine biosynthesis, phenylalanine metabolism, glycine, serine and threonine metabolism, alanine, aspartate and glutamate metabolism, and tyrosine metabolism were regarded as the targeted metabolic pathways according to Metabolic Pathway Analysis (MetPA). The results suggested that the metabolic perturbations in rats with doxorubicin induced hepatotoxicity were mainly involved in amino acid metabolism, lipid pathways, purine metabolism, energy metabolism, dysfunction of biotransformation and oxidative stress. The investigation revealed the effects of doxorubicin on liver in a holistic metabolic way, which laid a foundation for further studies on its toxicity mechanism.
Animals
;
Biomarkers
;
metabolism
;
Doxorubicin
;
toxicity
;
Energy Metabolism
;
Liver
;
drug effects
;
metabolism
;
Magnetic Resonance Imaging
;
Magnetic Resonance Spectroscopy
;
Metabolic Networks and Pathways
;
Metabolomics
;
Multivariate Analysis
;
Oxidative Stress
;
Rats
10.A Novel Angiotensin Type I Receptor Antagonist, Fimasartan, Prevents Doxorubicin-induced Cardiotoxicity in Rats.
Sung A CHANG ; Byung Kwan LIM ; You Jung LEE ; Mi Kyung HONG ; Jin Oh CHOI ; Eun Seok JEON
Journal of Korean Medical Science 2015;30(5):559-568
Angiotensin receptor blockers (ARBs) have organ-protective effects in heart failure and may be also effective in doxorubicin-induced cardiomyopathy (DOX-CMP); however, the efficacy of ARBs on the prevention of DOX-CMP have not been investigated. We performed a preclinical experiment to evaluate the preventive effect of a novel ARB, fimasartan, in DOX-CMP. All animals underwent echocardiography and were randomly assigned into three groups: treated daily with vehicle (DOX-only group, n=22), 5 mg/kg of fimasartan (Low-fima group, n=22), and 10 mg/kg of fimasartan (High-fima group, n=19). DOX was injected once a week for six weeks. Echocardiography and hemodynamic assessment was performed at the 8th week using a miniaturized conductance catheter. Survival rate of the High-fima group was greater (100%) than that of the Low-fima (75%) and DOX-only groups (50%). Echocardiography showed preserved left ventricular (LV) ejection fraction in the High-fima group, but not in the DOX-only group (P=0.002). LV dimensions increased in the DOX-only group; however, remodeling was attenuated in the Low-fima and High-fima groups. Hemodynamic assessment showed higher dP/dt in the High-fima group compared with the DOX-only group. A novel ARB, fimasartan, may prevent DOX-CMP and improve survival rate in a dose-dependent manner in a rat model of DOX-CMP and could be a treatment option for the prevention of DOX-CMP.
Angiotensin Receptor Antagonists/*therapeutic use
;
Animals
;
Biphenyl Compounds/*therapeutic use
;
Cardiomyopathies/chemically induced/mortality/*prevention & control
;
Doxorubicin/*toxicity
;
Echocardiography
;
Hemodynamics
;
Pyrimidines/*therapeutic use
;
Rats
;
Rats, Sprague-Dawley
;
Receptor, Angiotensin, Type 1/chemistry/*metabolism
;
Survival Rate
;
Tetrazoles/*therapeutic use
;
Ventricular Function, Left/physiology

Result Analysis
Print
Save
E-mail