Open Access

Shengxian decoction decreases doxorubicin‑induced cardiac apoptosis by regulating the TREM1/NF‑κB signaling pathway

Retraction in: /10.3892/mmr.2024.13159

  • Authors:
    • Lei Yao
    • Mingtai Gui
    • Jianhua Li
    • Bo Lu
    • Jing Wang
    • Xunjie Zhou
    • Deyu Fu
  • View Affiliations

  • Published online on: January 20, 2021     https://doi.org/10.3892/mmr.2021.11858
  • Article Number: 219
  • Copyright: © Yao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Shengxian decoction (SXT) is a traditional Chinese medicine that is clinically used for treating cardiovascular diseases. It is known for its beneficial effect on cardiomyocyte injuries, some of which can be induced by anticancer agents including doxorubicin (DOX). To determine the molecular mechanisms involved in the cardioprotective effects of SXT, DOX‑induced H9c2 cells were analyzed for apoptosis and expression levels of apoptosis biomarkers. Cell viability and apoptosis were measured by CCK‑8 and flow cytometry. Triggering receptors expressed on myeloid cells 1 (TREM1), cleaved caspase‑3, survivin and NF‑κBp65 expression levels were measured by reverse transcription‑quantitative PCR and/or western blotting. A total of 30 adult male Sprague‑Dawley rats were randomly allocated into five groups (n=6 each); control group receiving 0.9% saline, 1 DOX group receiving 2.5 mg/kg of DOX and 3 DOX + SXT groups, receiving a DOX dose equivalent to the DOX‑only group and either 0.4, 0.8 or 1.6 g/kg of SXT. It was found that DOX increased apoptosis and NF‑κB activation of H9c2 cells by increasing TREM1 expression and that SXT inhibited apoptosis and NF‑κB activation of H9c2 cells induced by DOX or Trem1 overexpression. SXT also significantly reversed DOX‑induced cardiotoxicity in rats. The results suggested that the protective effects of SXT against DOX‑induced apoptosis may be attributed to its downregulation of TREM1.

Introduction

Doxorubicin (DOX) is one of the most effective and widely-accepted anticancer drugs available in clinic (1,2). Unfortunately, its cardiotoxic effects limit its use (3,4), affect the quality of life of patients and may even lead to mortality. Once the cumulative dose of DOX reaches a certain level, the risk of congestive heart failure, dilated cardiomyopathy and even mortality significantly increases (5,6). Previous pharmacological and molecular studies reveal that apoptosis regulated by NF-κB is one of the causes for cardiotoxicity in DOX use (7,8).

One approach used to protect cardiomyocytes includes the use of traditional Chinese medicine. SXT, in particular, has been clinically used in China to cure a variety of heart and lung diseases (9,10). SXT also serves an important role in the treatment of cerebral watershed infarcts (11), autoimmune myasthenia gravis (12), hypothyroidism (13) and cauda equina injuries (14). In addition, it has been reported that SXT possesses cardioprotective effects from chronic heart failure during metabolic profiling of rats (15). However, its cardioprotective effects in DOX-induced cardiovascular disease (CVD) remain to be elucidated.

A recent molecular and cellular study emphasized the important role of triggering receptors expressed on myeloid cells 1 (TREM1), as it is involved in several mechanisms that are responsible for both acute and chronic CVD (16). TREM1 prevents macrophage apoptosis by maintaining mitochondrial function (17), while downregulation of TREM1 inhibits apoptosis by suppressing the NF-κB pathway during chondrocyte injury (18). Thus, the present study intended to determine whether the protective effects of SXT can prevent, reverse, or at least ameliorate, the deleterious cardiac effects of DOX-induced apoptosis by regulating TREM1, which can improve tolerance of this widely-used chemotherapeutic agent. Accordingly, the present study was designed to investigate the cardioprotective effects of SXT against DOX-induced cardiotoxicity in H9c2 myoblast cells in rats and identify possible underlying pathways that might be involved.

Materials and methods

SXT preparation

SXT (1,300 g): Powders of Astragali Radix (600 g), Bupleuri Radix (150 g), Cimicifugae Rhizoma (100 g), Anemarrhenae Rhizoma (300 g) and Platycodonis Radix (150 g) were suspended in water in a beaker (5 l) for 24 h and then were boiled with water (5 l × 3) as previously described (19). The combined water decoction (~15 l) was concentrated (13,000, 6,500, 2,600, 1,300, 650 and 325 ml) by a rotary evaporator under vacuum and the concentrated decoction (SXT, 0.1, 0.2, 0.5, 1.0, 2.0 and 4.0 mg/ml) was stored in a refrigerator at 4°C prior to analysis.

Cell culture, treatment and collection

H9c2 myoblast cells (Rattus norvegicus, rat; American Type Culture Collection; CRL-1446) were incubated in Dulbecco's Modified Eagle's Medium (HyClone; Cytiva) with addition of 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 1% Penicillin-Streptomycin (Beijing Solarbio Science & Technology Co., Ltd.) at 37°C in a sterile thermostatic incubator with 5% CO2. In contrast to the control group, which had no pretreatment and no chemical exposure, a chemical-induced heart failure (HF) model was achieved by incubating H9c2 cells in the above-mentioned conditions and exposing them to 2 µmol/l doxorubicin (DOX; Cell Signaling Technology, Inc.) for 12 h. In order to test the potential protective effects of SXT, cells were pretreated with SXT at the indicated concentration for 3 h before being exposed to DOX. Following incubation, cell cultures were collected and adherent cells were washed with PBS. H9c2 cells were digested by 0.25% Trypsin/EDTA and collected by centrifuging the culture and washing solution at 1,000 × g for 5 min at 4°C.

Cell transfection

small interfering (si)Trem1 (siTrem1-1; 5′-CCUACCGAGGCCAUGUUAUUU-3′; siTrem1-2; 5′-CCUUCAAGUGACAGACUCUUU-3′; siTrem1-3; 5′-GACUCUGGAUUAUAUCGUUUU-3′;) and control siNC (5′-CAGUACUUUUGUGUAGUACAA-3′) were purchased from Shanghai GenePharma Co., Ltd. and transfected into the H9c2 cells with Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) following manufacturer's protocol for 6 h at 37°C. The cDNA encoding the full-length coding regions of human Trem1 was subcloned into the pLVX-Puro lentiviral vector (Clontech Laboratories, Inc.) for constructing the Trem1 overexpressing vector. To produce transducer plasmids, the recombinant plasmids (1,000 ng) were transfected together with the packaging plasmids psPAX2 (100 ng) and pMD2G (900 ng; Addgene, Inc.) and amplified in 293 cells (ATCC; ACS-4500) with Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) following manufacturer's protocol for 6 h at 37°C. At 48 h following transfection, the recombinant lentivirus in the cell supernatant was collected by centrifugation at 5,000 × g for 5 min at 25°C and the purification and titration of recombinant lentivirus was performed as previously described (20). H9c2 cells were infected with the recombinant lentivirus-transducing units at an MOI of 20 in the presence of 8 µg/ml polybrene (Sigma-Aldrich; Merck KGaA) for 24 h at 37°C. Stable cells were selected by puromycin (3 µg/ml; Thermo Fisher Scientific, Inc.) for four more days and then used for subsequent experiments.

Cytotoxicity assay

The Cell Counter Kit-8 (CCK-8) assay (Signalway Antibody LLC) was used to assess the viability of H9c2 cells as a parameter for DOX-induced apoptosis. Following treatment of cells as described above, 10 µl of CCK-8 solution was added to each well and cells were incubated for 1 h at 37°C with 5% CO2. Optical density (OD) was measured at 450 nm with a DNM-9602 enzyme-labeled analyzer (Beijing Perlong New Technology Co., Ltd.). Then three wells were measured as replicates for the calculation of average cell viability for the control and DOX-exposed groups. The suppression rate was calculated as (1-ODsample/ODcontrol) ×100%.

Flow cytometry measurement of apoptosis level

Precipitated cells were re-suspended in PBS and double-stained by Annexin V-FITC and propidium iodide (PI) using an Annexin V-FITC apoptosis detection kit (Beyotime Institute of Biotechnology) according to the manufacturer's protocol. Data were obtained by detecting different signals emitted from normal cells (AnnexinV-FITC/PI), early (AnnexinV-FITC+/PI) and late (AnnexinV-FITC+/PI+) apoptotic cells and necrotic cells (AnnexinV-FITC/PI+) using an Accuri™ C6 flow cytometer (BD Biosciences) and Accuri™ C6 Software (version 1.0.264; BD Biosciences). The apoptosis level was quantified by the sum of apoptotic and necrotic cells in percentages, shown in a column plot.

RNA extraction and reverse transcription-quantitative (RT-q) PCR analyses

Total RNA was extracted from H9c2 cell lines (1×107) using TRIzol® (Thermo Fisher Scientific, Inc.) and transcribed into cDNA using a RevertAid First Strand cDNA Synthesis kit (Fermentas; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. cDNA (2 µl) was amplified using Maxima SYBR Green/ROX qPCR Master Mix (2X; Thermo Fisher Scientific, Inc.) on an ABI Prism 7300 Real-Time PCR system (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol and each sample was tested in triplicate. The following thermocycling conditions were used for qPCR: 95°C for 10 min; followed by 40 cycles at 95°C for 15 sec and 60°C for 45 sec; final extension at 95°C for 15 sec, 60°C for 1 min, 95°C for 15 sec and 60°C for 15 sec. GADPH was used as control gene for normalization of expression level. The Trem1 gene-specific primers were Trem1 forward (F) 5′-GAAGTATGCCAGAAGCAGGAAG-3′ and Trem1 reverse (R) 5′-GGAAGAGCAGAACAGGGTCG-3′. Rat GAPDH gene was used as the internal reference gene and the GAPDH gene-specific primers are GAPDH F 5′-GGAGTCTACTGGCGTCTTCAC-3′ and GAPDH R 5′-ATGAGCCCTTCCACGATGC-3′. mRNA expression levels were quantified using the 2−ΔΔCq method (21).

Protein preparation and western blot assay

H9c2 cells were collected as described and homogenized using RIPA lysis buffer (Jrdun Biotech). Nuclear protein was extracted using the Nuclear and Cytoplasmic Protein Extraction kit (Beyotime Institute of Biotechnology) and the protein concentration determined by a bicinchoninic acid assay kit (Thermo Fisher Scientific, Inc.). Loading samples for sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) were generated by boiling the loading buffer-containing proteins in a waterbath for 10 min. Proteins (30 µg) were separately fractionated by 10 and 15% gel, then transferred onto polyvinylidene difluoride membranes. Membranes were blocked for 1 h at room temperature in fresh blocking buffer [0.1% Tween-20 in Tris-buffered saline (TBS-T) containing 5% fat-free milk] and then immunostained with primary antibodies, including anti-TREM1 (1:1,000; cat. no. ab104413; Abcam), anti-cleaved caspase-3 (1:5,000; cat. no. ab2302; Abcam), anti-Survivin (1:1,000; cat. no. 2808; Cell Signaling Technology, Inc.), anti-NF-κB p65 (1:1,000; cat. no. 8242; Cell Signaling Technology, Inc.), anti-GAPDH (1:2,000; cat. no. 5174; Cell Signaling Technology, Inc.) and anti-histone H3 (1:1,000; cat. no. 4499; Cell Signaling Technology, Inc.) at 4°C overnight. After thrice washing with TBS-T buffer, membranes were stained with HRP-labeled Goat Anti-Rabbit IgG secondary antibody (1:1,000; cat. no. A0208; Beyotime Institute of Biotechnology) at 37°C for 1 h. The proteins were visualized using Immobilon Western Chemiluminescent HRP Substrate (EMD Millipore). The bands were quantified by the densitometry with ImageJ software (version 1.51; National Institutes of Health).

Animal experiment and drug administration

A total of 30 adult male Sprague-Dawley rats, aged 6–8 weeks and weighing 200–220 g, were obtained from Vital River Laboratory Animal Technology, housed in groups of three and given five days to acclimate to the housing facility. The rats were kept in the animal facility at 25°C (humidity, 60–70%) with a 12-h light/dark cycle, in 595×380×200 mm cages (1354G Eurostandard Type IV; Techniplast) and received food and water ad libitum. During housing, animals were monitored twice daily for health status and no adverse events were observed. Rats were randomly assigned to five groups (six per group): A control group which received 0.5 ml of 0.9% saline intraperitoneally (i.p.) daily over two weeks and four HF model groups which received doxorubicin HCL (Abmole Bioscience Inc.) at a dose of 2.5 mg/kg i.p. thrice weekly for two weeks (cumulative dose 15 mg/kg). Rats of three HF groups were also treated with SXT particle solution at a dose of 0.4, 0.8 and 1.6 g/kg/daily, respectively, via intragastric administration (i.g.) over two weeks. At 48 h from the last drug administration, echocardiographic examination was performed using AVevo 770 high-resolution ultrasound imaging system (FUJIFILM VisualSonics) to monitor cardiac functions (22). Left ventricular internal diameter (LVID) in diastole (d) and systole (s) were measured in vivo. Left ventricular end-diastolic and end-systolic volumes (LVEDV and LVESV)=1.04×LVIDd3 and 1.04×LVIDs3, respectively (23). Left ventricular ejection fraction (EF) and fractional shortening (FS) as EF (%)=(LVEDV-LVESV)/LVEDVx100% and FS (%)=(LVIDd-LVIDs)/LVIDdx100%, respectively (23). Blood pressure (mmHg) and heartbeat (bpm) were also measured. Animals were then anesthetized with a mixture of 100 mg/kg of ketamine (Sigma-Aldrich; Merck KGaA) and 10 mg/kg of xylazine (Sigma-Aldrich; Merck KGaA) i.p. Blood samples were then taken from the abdominal aorta to detect brain natriuretic peptide (BNP) using a Rat BNP ELISA kit (cat. no. orb-EHJ137053; Xiamen Huijia Biotechnology Co., Ltd.). At the end of the treatment, animals were sacrificed with an intravenous bolus of 2 ml of pentobarbital (182.2 mg/ml; Dolethal; Vétoquinol SA) followed by cervical dislocation and the heart was removed, washed with cold saline, then weighed. Myocardial tissue was embedded in paraffin and stained using haemotoxylin and eosin stain (Baso Diagnostic Inc.) and TUNEL kit (Roche Diagnostics) following manufacturer's protocol as previously described (24). Frozen tissue specimens were stored in liquid nitrogen. TUNEL was calculated from six replicates from each experiment group. Scores of histopathological changes were obtained according to the Hafez (25) method. All experiments were performed in accordance with the international guidelines of the Principles of Laboratory Animals Care and were approved by the Animal Care Committee of Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine (approval no. 201911).

Statistical analysis

All data are expressed as mean ± standard deviation of triplicate dependent experiments. Statistical analyses were performed using one-way analysis of variance followed by Dunnett's and Tukey's multiple comparisons test. P<0.05 was considered to indicate a statistically significant difference.

Results

Quantification of DOX-induced cytotoxicity and TREM1 expression

DOX cardiotoxicity was quantified by CCK-8 assay on H9c2 cells, which revealed that rate of suppression of cell proliferation followed a dosage-dependent pattern when DOX concentration was between 0.5 and 8 µmol/l (Fig. 1A). TREM1 was found to be upregulated in the DOX-exposed cells, which was expected as TREM1 is known to be one of the most crucial receptor proteins that can induce apoptosis upon activation (17,18) (Fig. 1B and C). The expression level of TREM1 increased when the DOX concentration was also increased.

Protective effect of SXT on DOX-induced apoptosis in H9c2 cells

In contrast to the suppressed cell proliferation rate induced by DOX, the suppression was observed to decrease when the concentration of pretreatment SXT increased (Fig. 2A). Significant deviations from the control group were observed in H9c2 cells pretreated with 1 mg/ml or higher concentration of SXT. Further evidence for the reduced apoptosis level exhibited by the SXT pretreated cells were obtained from cytometric analysis, which revealed that all SXT pretreated groups had a significantly lower proportion of apoptotic and necrotic cells compared with groups without SXT pretreatment (Fig. 2B and C). However, SXT had no effect on the cell viability, apoptosis and TREM1 expression in Hc9c cells under DOX-free condition (Fig. S1).

As the eventual regulator of the cellular pathway in response to chemical stress, the proteomic profile revealed by western blot assay suggested that DOX-exposed cells possessed higher levels of TREM1, cleaved caspase-3 and nuclear NF-κB p65 but lower levels of survivin and cytoplasmic NF-κB p65 in comparison with the control group (Fig. 2D). Notably, the SXT pretreated groups showed that the difference in the expression levels of these proteins can be reduced by 0.5 to 2 mg/ml with SXT pretreatment (Fig. 2D).

DOX induces H9c2 cells apoptosis by increasing expression of TREM1

The TREM1-induced apoptosis pathway was blocked by siRNA silencing in H9c2 cells, resulting in stable transfected cell lines, referred to as siTrem1, in which significant downregulation of TREM1 expression could be detected by western blotting and qPCR analysis (Fig. 3A and B). When exposed to DOX, two replicates of siTrem1 transfected groups exhibited a significant decrease in apoptotic and necrotic H9c2 cells in comparison with DOX-exposed groups that were transfected with non-sense RNA (siNC), indicating decreased apoptosis level (Fig. 3C and D). Western blot analysis on siTrem1 transfected groups suggested that TREM1, cleaved caspase-3 and nuclear NF-κB p65, which were upregulated in DOX-exposed siNC cells, can be downregulated (Fig. 3E and F). Meanwhile, the survivin and cytoplasmic NF-κB p65 that were downregulated in DOX-exposed siNC group was upregulated.

SXT protects H9c2 cells from TREM1 overexpression-induced apoptosis

Complementation test for the role of SXT in regulating TREM1-induced apoptosis was achieved by transfecting H9c2 cells with overexpression vector (oeTrem1; Fig. 3A and B). Consistent with the hypothesis of the present study, oeTrem1 exhibited a higher apoptotic level compared with vector control and the increased apoptosis could be inhibited by SXT treatment (Fig. 4A and B). Western blot analysis suggested that expression levels of TREM1, cleaved caspase-3 and nuclear NF-κB p65 were reduced while expression levels of the downregulated survivin and cytoplasmic NF-κB p65 were increased following SXT treatment (Fig. 4C).

Protective effect of SXT in DOX-induced HF animal models

To further investigate the effect of SXT on DOX-induced cardiac injury, an in vivo HF model was established in rats. Histological assessment showed that necrosis, edema and disorganization of muscle fibers observed in HF rats were alleviated with SXT treatment (Fig. 5A-C). It was also observed that HF also increased end-diastolic pressure and decreased EF, FS, end-systolic pressure, heart weight and heart rate (Tables SI and I). However, treatment with SXT could improve DOX-induced cardiac function. This was demonstrated when the BNP level in blood was markedly increased in rats following HF, but decreased when treated with SXT (Fig. 5D). SXT treatment also significantly inhibited the upregulation of TREM1, caspase 3 and nuclear NF-κB and downregulation of survivin and cytoplasmic NF-κB, which were induced by DOX (Fig. 5E). These data further supported the findings in DOX-induced H9c2 cells.

Discussion

The commonest clinical manifestations of DOX-induced cardiotoxicity are myocardial dilatation and severe weakening of left ventricular systolic function, leading to congestive heart failure (26). Mechanisms of DOX-induced cardiotoxicity include oxidative stress, calcium dysregulation, extracellular matrix remodeling and cell apoptosis (2729). The present study determined the benefits of SXT treatment and its protective effect on DOX-induced cardiotoxicity in vitro and in vivo. First, TREM1 was found to be upregulated in the DOX-induced H9c2 cells and rat models. Second, TREM1 had a role in DOX-induced apoptosis of H9c2 cells through regulation of the expression of cleaved caspase-3, survivin and NF-κB p65. Third, SXT was found to have cardioprotective functions by inhibiting the upregulation of TREM1 in DOX-induced H9c2 cells. The present study also provided a strategy of targeting TREM1 for the prevention and/or treatment of DOX-induced cardiotoxicity.

A previous study demonstrated that DOX preferentially accumulates in mitochondria after entering cardiomyocytes, which is probably due to the high affinity of DOX to cardiolipin (30). Therefore, mitochondria are considered as one of the main targets of DOX-induced cardiotoxicity. Compared with other tissues, myocardia have higher and sustained metabolic activity and relatively lower levels of antioxidant enzymes such as peroxidase, catalase and superoxide dismutase (31). It is for this reason that the heart is more likely to produce DOX-dependent reactive oxygen species (ROS), which can induce cardiomyocyte death mainly by apoptosis and necrosis. DOX can directly or indirectly promote mitochondrial release of cytochrome c to initiate the endogenous apoptosis pathway by upregulating caspase-3 and/or downregulating survivin (32,33). DOX can also increase the expression of TNF receptor, which is associated with the NF-κB pathway and contributes to the activation of caspase cascade receptors to induce apoptosis (33,34). Consistent with these findings, the data from the present study suggested that DOX may induce cardiotoxicity and apoptosis through the regulation of the caspase 3/survivin and NF-κB pathway and mitochondrial dysregulation.

The potential biomarkers involves in fatty acid metabolism and sphingolipid metabolism were also identified in HF rats following SXT treatment (15). Increasing the levels of sphinganine 1-phosphate, an anti-apoptotic protein which protects against ischemia-reperfusion injury and inflammation, also highlights the regulatory and cardioprotective role of SXT (35). Since defects in the structure and function of the cardiomyocytes as a result of a disruption in mitochondrial fatty acid metabolism may lead to HF (36), the reduction of long-chain fatty acid levels induced by SXT is associated with a decreased risk for HF in rats. Therefore, it was hypothesized that the cardioprotective role of SXT in response to DOX-induced H9c2 cells in rats may be a result of its anti-apoptotic effect.

TREM1 overexpression leads to increased expression levels of mitofusin, which suggests that TREM1 serves a role in mitochondrial structure and function (37). TREM1 ligation also increases the production of ROS and pro-inflammatory cytokines by activating NF-κB via the caspase recruitment domain-containing protein 9/Bcl10 complex (18). TREM1 can promote the activation of several transcription complexes which synergistically act with NF-κB to increase transcription of target genes (38). One of these target genes is survivin, which has been found to bind to caspase-3, inhibiting its catalytic activities and thereby also inhibiting its apoptotic effects (39,40). The data from the present study also revealed that SXT treatment was able to ameliorate DOX-induced cardiomyocyte injury and reduce tissue inflammation and apoptosis, suggesting that SXT possessed cardioprotective properties. In the present study, TREM1 expression was increased by DOX but inhibited by SXT in DOX-induced cells in rats and SXT inhibited apoptosis through the NF-κB pathway, which is induced by TREM1 overexpression. This suggested that SXT may inhibit DOX-induced apoptosis by inhibiting TREM1 expression and subsequent inhibition of the TREM1-mediated NF-κB pathway. STX also inhibited the DOX-induced decrease in heart rate. The decreased heart rate induced by DOX is also observed in the study by Jafarinezhad et al (41), which demonstrates increased serum troponin I, QT interval and QRS complex in rats treated with DOX. The increase of cardiac troponin I level following DOX treatment is a strong predictor of ventricular dysfunction and poor cardiac outcome both in rats and in patients (42,43). Ji et al (44) indicate that NF-κB might be responsible for transcriptional regulation of the TNNI1 gene, coding troponin I. These data suggest that STX may inhibit DOX-induced decrease in the heart rate through the TREM1-mediated NF-κB pathway. This issue will be further examined in a forthcoming study. Although the use of SXT may improve DOX-induced cardiotoxicity in a clinical setting, more animal experiments and clinical studies need to be performed to establish this.

In conclusion, TREM-1 expression is increased by DOX, but inhibited by SXT. TREM1 downregulation exerts anti-apoptotic effects on DOX-exposed chondrocytes, while its overexpression exerts pro-apoptotic effects via the regulation of the NF-κB pathway. SXT may inhibit DOX-induced apoptosis by inhibiting TREM1 expression and subsequent inhibition of the TREM1-mediated NF-κB pathway. The mechanism of its action remains ambiguous and requires further study. These data may also give consideration to TREM1 as a potential target of therapy in these particular disease settings.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by grant from the National nature science foundation of China (grant no. 81774111), Shanghai Three Year Project of Traditional Chinese Medicine (grant no. 2018-2020) and the Cardiology of Traditional Chinese Medicine and Shanghai Three Year Project of Traditional Chinese Medicine (grant no. ZY3-CCCX-3-3026).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

LY and MG were involved in experimental design and drafting the manuscript. JL, BL, JW, XZ and DF were involved in designing the experiments, writing, reviewing and editing the manuscript, and supervising the study. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

All experiments were performed in accordance with the international guidelines Principles of Laboratory Animals Care and were approved by the Animal Care Committee of Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine (approval no. 201911).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Rivankar S: An overview of doxorubicin formulations in cancer therapy. J Cancer Res Ther. 10:853–858. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Zhang Y, Yang C, Wang W, Liu J, Liu Q, Huang F, Chu L, Gao H, Li C, Kong D, et al: Co-delivery of doxorubicin and curcumin by pH-sensitive prodrug nanoparticle for combination therapy of cancer. Sci Reps. 6:212252016. View Article : Google Scholar

3 

Kalyanaraman B: Teaching the basics of the mechanism of doxorubicin-induced cardiotoxicity: Have we been barking up the wrong tree? Redox Biol. 29:1013942019. View Article : Google Scholar : PubMed/NCBI

4 

Hu X, Liu H, Wang Z, Hu Z and Li L: miR-200a attenuated doxorubicin-induced cardiotoxicity through upregulation of Nrf2 in Mice. Oxid Med Cell Longev. 2019:15123262019. View Article : Google Scholar : PubMed/NCBI

5 

Yerebakan C, Boltze J, Elmontaser H, Yoruker U, Latus H, Khalil M, Ostermayer S, Steinbrenner B, Apitz C, Schneider M, et al: Effects of pulmonary artery banding in doxorubicin-induced left ventricular cardiomyopathy. J Thorac Cardiovasc Surg. 157:2416–2428.e4. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Azamthulla M, Mukherjee D, Roy J and Ravishankar C: Evaluation of cardiomyocyte targeted novel nano-formulation of pirfenidone on doxorubicin induced congestive heart failure in rats. Conference on Drug Design and Discovery Technologies. 212–218. 2019. View Article : Google Scholar

7 

Zhang DX, Ma DY, Yao ZQ, Fu CY, Shi YX, Wang QL and Tang QQ: ERK1/2/p53 and NF-κB dependent-PUMA activation involves in doxorubicin-induced cardiomyocyte apoptosis. Eur Rev Med Pharmacol Sci. 20:2435–2442. 2016.PubMed/NCBI

8 

Imam F, Al-Harbi NO, Al-Harbi MM, Ansari MA, Al-Asmari AF, Ansari MN, Al-Anazi WA, Bahashwan S, Almutairi MM, Alshammari M, et al: Apremilast prevent doxorubicin-induced apoptosis and inflammation in heart through inhibition of oxidative stress mediated activation of NF-κB signaling pathways. Pharmacol Rep. 70:993–1000. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Kang HY, Zhang FH, Liu XM and Yu W: Preliminary research of the effects of Shengxian decoction in rats acute anoxemia of cardiac muscle. Chin J Hos Phar. 27:6172007.

10 

Gu HR, Zhou MX and Liu QC: Experience of GU Wei-chao in treating heart and lung diseases through application of modified shengxian decoction. Chin J Inform Traditional Chin Med. 25:108–110. 2018.

11 

Gao YH and Zheng Y: Effect of modified shengxian decoction on treating cerebral watershed infarctionion. Chin Arch Traditional Chin Med. 812012.

12 

Junyao X, Zhu J, Cheng Y, Wu ZY, Chen YD, Xia BM and Wu HX: Research on immune mechanism of Shengxian decoction in experimental autoimmune myasthenia gravis rats. Chin J Immunol. 32:1462–1466. 2016.(In Chinese).

13 

Guangshan L, Ren Z, Zheng Y, Huang D and Mingdi L: Clinical observation on 32 cases of hypothyroidism of thyroid cancer after operation treated with Shengxian decoction combined with levothyroxine sodium tablets. Int J Traditional Chin Med. 35:692–694. 2013.

14 

Zhang L, Xiong Y and Wang Z: Study on interference of miao medicine sheng xian decoction on protein expression of brain-derived neuro-trophic factor in the sacral spinal cord of rats with cauda equina injury. J Med Postgraduates. 1042–1046. 2014.

15 

Zhang F, Zhan Q, Dong X, Jiang B, Sun L, Gao S, He Z, Tao X and Chen W: Shengxian decoction in chronic heart failure treatment and synergistic property of Platycodonis Radix: A metabolomic approach and its application. Mol Biosyst. 10:2055–2063. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Kouassi KT, Gunasekar P, Agrawal DK and Jadhav GP: TREM-1; Is it a pivotal target for cardiovascular diseases? J Cardiovasc Dev Dis. 5:452018. View Article : Google Scholar

17 

Campbell GR, To RK and Spector SA: TREM-1 protects HIV-1-infected macrophages from apoptosis through maintenance of mitochondrial function. mBio. 10:e02638–19. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Tang J and Dong Q: Knockdown of TREM-1 suppresses IL-1β-induced chondrocyte injury via inhibiting the NF-κB pathway. Biochem Biophys Res Commun. 482:1240–1245. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Zhang F, Zhan Q, Gao S, Dong X, Jiang B, Sun L, Tao X and Chen WS: Chemical profile- and pharmacokinetics-based investigation of the synergistic property of platycodonis radix in traditional Chinese medicine formula Shengxian decoction. J Ethnopharmacol. 152:497–507. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Xiong S, Zheng Y, Jiang P, Liu R, Liu X and Chu Y: MicroRNA-7 inhibits the growth of human non-small cell lung cancer A549 cells through targeting BCL-2. Int J Biol Sci. 7:805–814. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Jansen CHP, Brangsch J, Reimann C, Adams L, Hamm B, Botnar RM and Makowski MR: In vivo high-frequency ultrasound for the characterization of thrombi associated with aortic aneurysms in an experimental mouse model. Ultrasound Med Biol. 43:2882–2890. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Devereux RB and Reichek N: Echocardiographic determination of left ventricular mass in man. Anatomic validation of the method. Circulation. 55:613–618. 1977. View Article : Google Scholar : PubMed/NCBI

24 

Wang X, Liu F, Xu M and Wu L: Penehyclidine hydrochloride alleviates lipopolysaccharide-induced acute respiratory distress syndrome in cells via regulating autophagy-related pathway. Mol Med Rep. 23:1002021. View Article : Google Scholar : PubMed/NCBI

25 

Hafez HM and Hassanein H: Montelukast ameliorates doxorubicin-induced cardiotoxicity via modulation of p-glycoprotein and inhibition of ROS-mediated TNF-α/NF-κB pathways. Drug Chem Toxicol. 1–12. 2020.doi: 10.1080/01480545.2020.1730885 (Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

26 

Volkova M and Russell R III: Anthracycline cardiotoxicity: Prevalence, pathogenesis and treatment. Curr Cardiol Rev. 7:214–220. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Wang Y, Lei T, Yuan J, Wu Y, Shen X, Gao J, Feng W and Lu Z: GCN2 deficiency ameliorates doxorubicin-induced cardiotoxicity by decreasing cardiomyocyte apoptosis and myocardial oxidative stress. Redox Biol. 17:25–34. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Polegato BF, Minicucci MF, Azevedo PS, Carvalho RF, Chiuso-Minicucci F, Pereira EJ, Paiva SA, Zornoff LA, Okoshi MP, Matsubara BB and Matsubara LS: Acute doxorubicin-induced cardiotoxicity is associated with matrix metalloproteinase-2 alterations in rats. Cell Physiol Biochem. 35:1924–1933. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Renu K, V G A, P B TP and Arunachalam S: Molecular mechanism of doxorubicin-induced cardiomyopathy-An update. Eur J Pharmacol. 818:241–253. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Ichikawa Y, Ghanefar M, Bayeva M, Wu R, Khechaduri A, Naga Prasad SV, Mutharasan RK, Naik TJ and Ardehali H: Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation. J Clin Invest. 124:617–630. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Vejpongsa P and Yeh ET: Prevention of anthracycline-induced cardiotoxicity: Challenges and opportunities. J Am Coll Cardiol. 64:938–945. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Li J, Wu Y, Wang D, Zou L, Fu C, Zhang J and Leung GP: Oridonin synergistically enhances the anti-tumor efficacy of doxorubicin against aggressive breast cancer via pro-apoptotic and anti-angiogenic effects. Pharmacol Res. 146:1043132019. View Article : Google Scholar : PubMed/NCBI

33 

Ibrahim KM, Mantawy EM, Elanany MM, Abdelgawad HS, Khalifa NM, Hussien RH, El-Agroudy NN and El-Demerdash E: Protection from doxorubicin-induced nephrotoxicity by clindamycin: Novel antioxidant, anti-inflammatory and anti-apoptotic roles. Naunyn Schmiedebergs Arch Pharmacol. 393:739–748. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Zhao L and Zhang B: Doxorubicin induces cardiotoxicity through upregulation of death receptors mediated apoptosis in cardiomyocytes. Sci Repo. 7:447352017. View Article : Google Scholar

35 

Yang J, Wang H, Xu W, Hao D, Du L, Zhao X and Sun C: Metabolomic analysis of rat plasma following chronic low-dose exposure to dichlorvos. Hum Exp Toxicol. 32:196–205. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Marín-García J and Goldenthal MJ: Fatty acid metabolism in cardiac failure: Biochemical, genetic and cellular analysis. Cardiovasc Res. 54:516–527. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Yuan Z, Syed MA, Panchal D, Joo M, Colonna M, Brantly M and Sadikot RT: Triggering receptor expressed on myeloid cells 1 (TREM-1)-mediated Bcl-2 induction prolongs macrophage survival. J Biol Chem. 289:15118–15129. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Liu F, Zhang X, Zhang B, Mao W, Liu T, Sun M and Wu Y: TREM1: A positive regulator for inflammatory response via NF-κB pathway in A549 cells infected with Mycoplasma pneumoniae. Biomed Pharmacother. 107:1466–1472. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Raninga PV, Di Trapani G, Vuckovic S and Tonissen KF: TrxR1 inhibition overcomes both hypoxia-induced and acquired bortezomib resistance in multiple myeloma through NF-κβ inhibition. Cell Cycle. 15:559–572. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Shin S, Sung BJ, Cho YS, Kim HJ, Ha NC, Hwang JI, Chung CW, Jung YK and Oh BH: An anti-apoptotic protein human survivin is a direct inhibitor of caspase-3 and −7. Biochemistry. 40:1117–1123. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Jafarinezhad Z, Rafati A, Ketabchi F, Noorafshan A and Karbalay-Doust S: Cardioprotective effects of curcumin and carvacrol in doxorubicin-treated rats: Stereological study. Food Sci Nutr. 7:3581–3588. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Reagan WJ, York M, Berridge B, Schultze E, Walker D and Pettit S: Comparison of cardiac troponin I and T, including the evaluation of an ultrasensitive assay, as indicators of doxorubicin-induced cardiotoxicity. Toxicol Pathol. 41:1146–1158. 2013. View Article : Google Scholar : PubMed/NCBI

43 

El-Sayed el SM, Mansour AM and Abdul-Hameed MS: Thymol and carvacrol prevent doxorubicin-induced cardiotoxicity by abrogation of oxidative stress, inflammation, and apoptosis in rats. J Biochem Mol Toxicol. 30:37–44. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Ji GG, Shu JT, Zhang M, Ju XJ, Shan YJ, Liu YF and Tu YJ: Transcriptional regulatory region and DNA methylation analysis of TNNI1 gene promoters in Gaoyou duck skeletal muscle (Anas platyrhynchos domestica). Br Poult Sci. 60:202–208. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2021
Volume 23 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yao L, Gui M, Li J, Lu B, Wang J, Zhou X and Fu D: Shengxian decoction decreases doxorubicin‑induced cardiac apoptosis by regulating the TREM1/NF‑κB signaling pathway Retraction in /10.3892/mmr.2024.13159. Mol Med Rep 23: 219, 2021
APA
Yao, L., Gui, M., Li, J., Lu, B., Wang, J., Zhou, X., & Fu, D. (2021). Shengxian decoction decreases doxorubicin‑induced cardiac apoptosis by regulating the TREM1/NF‑κB signaling pathway Retraction in /10.3892/mmr.2024.13159. Molecular Medicine Reports, 23, 219. https://doi.org/10.3892/mmr.2021.11858
MLA
Yao, L., Gui, M., Li, J., Lu, B., Wang, J., Zhou, X., Fu, D."Shengxian decoction decreases doxorubicin‑induced cardiac apoptosis by regulating the TREM1/NF‑κB signaling pathway Retraction in /10.3892/mmr.2024.13159". Molecular Medicine Reports 23.3 (2021): 219.
Chicago
Yao, L., Gui, M., Li, J., Lu, B., Wang, J., Zhou, X., Fu, D."Shengxian decoction decreases doxorubicin‑induced cardiac apoptosis by regulating the TREM1/NF‑κB signaling pathway Retraction in /10.3892/mmr.2024.13159". Molecular Medicine Reports 23, no. 3 (2021): 219. https://doi.org/10.3892/mmr.2021.11858