Open Access

miR‑127 aggravates myocardial failure by promoting the TGF‑β1/Smad3 signaling

Retraction in: /10.3892/mmr.2019.10442

  • Authors:
    • Hainian Xu
    • Fengmei Li
  • View Affiliations

  • Published online on: September 27, 2018     https://doi.org/10.3892/mmr.2018.9514
  • Pages: 4839-4846
  • Copyright: © Xu 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

Myocardial failure has a negative impact on the quality of human life. MicroRNA (miR) expression abnormalities lead to the development of many pathological conditions, including myocardial failure, and therefore the present study primarily focused on the investigation of the functions of miR‑127 in the development of myocardial failure. The miR‑127 expression levels in serum samples from patients with myocardial failure were examined. Oil red O staining was used to analyze the characteristics of the myocardium of the patients. Immunohistochemistry was used to detect fatty acid synthase (FASN), stearoyl‑CoA desaturase‑1 (SCD1) and mitochondrial brown fat uncoupling protein 1 (UCP1) protein expression in the myocardium of the patients. Furthermore, C57BL/6J (B6) mice were induced with 15 mg/kg of doxorubicin. Echocardiography was used to detect the histopathological alterations of the myocardial cells by comparison of the myocardial tissues from the myocardial failure animal model and normal C57BL/6 mice. Reverse transcription‑quantitative polymerase chain reaction was used to detect the expression levels of miR‑127 following different induction periods and immunohistochemistry was used to detect the expression of transforming growth factor‑β1 (TGF‑β1) and mothers against decapentaplegic homolog 3 (Smad3). Immunofluorescence was used to detect the expression alterations TGF‑β1/Smad3 when miR‑127 overexpression or inhibition was established. The results of the present study indicated that myocardial failure resulted in an upregulated expression of miR‑127 and severe fat accumulation. FASN, SCD1 and UCP1 were highly expressed in the myocardial failure group compared with the control. Abdominal artery contraction and the ejection fraction were significantly reduced in the DOX‑induced B6 mice. The cardiomyocytes became hypertrophic, and left ventricular systolic pressure and left ventricular maximum ejection pressure were altered following DOX induction in B6 mice. The results confirmed that miR‑127 regulates the expression of TGF‑β1/Smad3. The potential pathological mechanism of the effect of miR‑127 may be based on the upregulation of the TGF‑β1/Smad3 signaling pathway.

Introduction

Myocardial failure is a heart disease with high mortality. The morbidity and mortality of myocardial failure in developing countries is significantly increased compared with developed countries (1). The incidence of heart failure is a multifactorial, multi-step and complex process closely associated with abnormal expression of genes (2,3). Although many studies have focused on the development of myocardial failure, the exact underlying mechanisms remain to be elucidated. The heart is unable to offer the same vein reflow and blood supply when myocardial failure occurs, and a number of patients with myocardial failure rapidly succumb to mortality. Heart failure causes a variety of diseases and results in a series of symptoms and signs, such as dyspnea, pulmonary edema and cardiogenic shock. Heart valve disease, coronary atherosclerosis, hypertension, endocrine disorders, bacterial toxins, acute pulmonary infarction, emphysema and other chronic lung diseases can lead to heart failure (4). Pregnancy, fatigue and rapid intravenous infusion can also increase the burden of heart disease and induce myocardial failure (5,6).

Stearoyl-CoA desaturase-1 (SCD1) is a key enzyme in the synthesis of monounsaturated fatty acids (7). SCD1 primarily catalyzes the formation of saturated fatty acids and a previous study demonstrated that it may serve a role in the development of heart failure (8). Fatty acid synthase (FASN) uses substrates and intermediates channeled into the functional domain to catalyze the synthesis of fatty acids. Cells use mitochondrial brown fat uncoupling protein 1 (UCP1) to generate heat, which allows infants and animals that hibernate to maintain body temperature without muscle tremor. Previous researchers have found that a particular compound, an aldehyde which activates UCP1, can cause fat burning under certain conditions (9,10). For instance, Liu et al (9) discovered a series of potent, selective, orally bioavailable SCD1 inhibitors based on a known pyridazine carboxamide template. Another study (7) regarding lung cancer revealed that the inhibition of SCD1 activity in human lung cancer cells with the small molecule SCD inhibitor CVT-11127 reduces lipid synthesis and impairs proliferation by blocking the progression of the cell cycle through the G(1)/S boundary and by triggering programmed cell death.

MicroRNAs (miRs) are small non-coding RNAs which regulate the transcription of mRNA and thereby promote or inhibit the expression of oncogenes and tumor suppressor genes. Currently, more than a 1,000 miRs have been identified and reported in the miRBase (www.mirbase.org), and a number of them are used as molecular biomarkers for diagnosis, prognosis and treatment (11,12). miR-127, located at chromosome 20q13.32, has been reported to be associated with angiogenesis, and, as a tumor-promoting factor, it is involved in the regulation of cell polarity (13).

The transforming growth factor-β (TGF-β) family consists of a variety of isomers, including three subtypes: TGF-β1, TGF-β2 and TGF-β3 (14). Mothers against decapentaplegic homolog 3 (Smad) is a downstream factor of the TGF-β superfamily signaling, which mediates the TGF-β signaling from the cytoplasm into the nucleus and specifically regulates the expression of the target genes (15). Smad has a highly conserved N-terminal domain (MH1) and a C-terminal domain (MH2) (16). According to the different roles served in the TGF-β signal transduction, the Smad family is sub-divided into receptor-activated Smads (R-Smads), common type Smads (Co-Smads) and inhibitory Smads (I-Smads). R-Smads, including Smad1, 2, 3, 5 and 8, are substrates for the TGF-β1 receptor kinases, where Smad2 and 3 mediate the TGF-β1 signaling (17). Co-Smads, including Smad4 and Smad10, are involved in signaling through binding to R-Smads. TGF-β1/Smad3 is a signaling pathway associated with inflammation, and it has been reported that the occurrence and development of myocardial failure was associated with myocarditis and inflammation. The development of myocardial failure may be associated with the activation of certain key factors in the TGF-β1/Smad3 signaling pathway (18,19). The aim of the present study was to determine the role of miR-127 in myocardial failure and its potential regulatory mechanism.

Materials and methods

Clinical data and animal induction

A total of 51 tissue samples from patients with myocardial failure (mean age, 67.8±17.4 years; 31 males and 20 females) were collected, as well as 50 normal myocardial tissue specimens (mean age, 64.6±16.9 years; 27 males and 23 females), from July to December 2014 at Weifang People's Hospital (Weifang, China). No differences in the age (P=0.352) and sex (P=0.849) distributions between the myocardial failure and control group were detected. All participants provided written informed consent following institutional review board approval at the Weifang People's Hospital. All aspects of this research were approved by the Ethics Committee of The Weifang People's Hospital. The tissue samples were collected and frozen in liquid nitrogen. Complete patient records and the pathological data were obtained. The following inclusion criteria were used: i) >20 years of age; ii) suspected symptoms or signs of heart failure without obvious alterations within 1 week; and iii) New York Heart Association (NYHA) class II or III symptoms. The diagnosis of heart failure was based on both the symptoms and the echocardiography results. The exclusion criteria included severe valvular heart disease, acute pulmonary embolism, severe infection or sepsis, acute decompensated heart failure or diagnosis of heart failure by echocardiography and radionuclide imaging or magnetic resonance imaging within the past 3 months. The normal controls were included from the organ donation.

A total of 24 male C57BL/6J (B6) mice (4 weeks old) were obtained from Vital River Laboratories Co., Ltd. (Beijing, China) and were routinely housed under specific pathogen free (SPF) conditions at 22±2°C with 40–60% humidity in a 12 h light/dark cycle. Animals had free access to food and water. Doxorubicin (DOX), an anthracycline antibiotic commonly used as chemotherapy medication, is frequently used to establish animal models of cardiomyopathy (20). The animal model was established as previously described using 6 intraperitoneal injections with 2.5 mg/kg DOX (Adriamycin®; Pfizer, Inc., New York, NY, USA) every 48 h to achieve an accumulative dose of 15 mg/kg (21). The control group was fed a normal diet and injected with physiological saline. This total dose was selected according to a study by Zhang et al (13). Following the observation period (1 and 6 months following treatment) and echocardiography, the mice were euthanized by placing them into a chamber filled with isoflurane vapor until respiration ceased, and the heart tissue was collected for examination. Histological alterations in normal B6 mice and the ranolazine treated group were detected and the arteries were measured by echocardiography contrast at 1 and 6 months post-AAC. All animal experiments were undertaken in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals, with the approval of the Weifang People's Hospital Animal Care and Use Committee.

Abdominal artery contraction (AAC)

Under ketamine (70 mg/kg)/xylazine (5 mg/kg) anesthesia, 28 rats were subjected to restrictive banding of the suprarenal abdominal aorta at 16 to 18 weeks of age using a blunt 23G needle as a template. The needle was tied tightly against the aorta above the renal arteries (causing visible kidney blanching), and then removed, leaving the suture in place to partially restore blood flow (visually confirmed).

Cell transfection

Recombinant adenovirus amplification and cell transfection were subsequently performed. The 293T cells were plated in fresh Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at a density of 1×105 cells/ml. When the adherent cells reached 60% confluence, they were infected with an empty recombinant adenovirus AdGFP (Vector Laboratories, Inc., Burlingame, CA, USA), recombinant adenovirus AdCx3 expressing miR-127, or recombinant adenovirus AdSmad1 expressing small interfering (si)-miR-127 (both Invitrogen; Thermo Fisher Scientific, Inc.). When the floating cells accounted for 50% of all cells, the cells were harvested, collected following centrifugation at 250 × g for 5 min in 37°C, and resuspended in 500 µl of DMEM. Following incubation in liquid nitrogen for 1–2 min, the tubes with cells were placed in a 37°C water bath with continuous oscillation. Following cell lysis, the samples were vortexed for another 1–2 min. The above steps were repeated 4–5 times, followed by centrifugation at 1,000 × g for 10 min in 4°C, and the supernatant containing the recombinant adenoviral extract was used to transfect the H9C2 cells (American Type Culture Collection, Manassas, VA, USA).

A total of 2 ml of the H9C2 cells at a density of 0.5×105 cells/ml were seeded into 6-well plates and cultured in DMEM with 10% FBS for 24 h, followed by the addition of the corresponding recombinant adenovirus for transfection. 0, 2.5, 5 and 10 µM siRNA were used in the transfection. siRNAs were transfected with Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) for 6 h according to the manufacturer's manual. The sequences of miR-127 and si-miR-127 were CUGAAGCUCAGAGGGCUCUGAU and CACTTCGAGTCTCCCGAGACUUA, respectively. After transfection for 12 h, cells were washed with culture media and used for subsequent experiments. Total RNA was extracted after culturing the cells for 30 h to detect the expression of target genes, using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

The expression of miR-127 was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) in tissue samples from patients with myocardial failure and normal control tissue, as well as H9C2 cells. RNA was extracted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA), and then centrifuged for 10 min at 4,000 × g and 4°C. The supernatant was transferred into a new Eppendorf Tube (EP; Eppendorf, Hamburg, Germany) and allowed to stand for 5 min at room temperature. Subsequently, the following steps were conducted: 200 µl of chloroform was added, the samples were vortexed for 15 sec and then left to stand for 3 min at room temperature; the samples were centrifuged at 4,000 × g at 4°C for 15 min; the top layer was transferred to a new EP and an equal volume of isopropanol was added into each EP at room temperature for 10 min; the samples were centrifuged at 4,000 × g and 4°C for 10 min, and the supernatant is discarded; the pellets were washed with 75% ethanol, centrifuged at 2,500 × g and 4°C for 5 min and supernatant was discarded; RNA was left to dry and 20 µl DEPC water was subsequently added to dissolve the extracted RNA. The reverse transcription kit was purchased from Takara Biotechnology Co., Ltd. (Dalian, China). The temperature protocol for reverse transcription was as follows: 37°C for 15 min; 85°C for 5 sec and 4°C for 5 min. qPCR was subsequently used to detect the expression of miR-127. The following primer sequences were used: miR-127 forward, 5′-GCGAGCTACATTGTCTGCTGGGTT-3′ and reverse, 5′-GTCGAGGGTCCGAGGTATTCCG-3′; U6 forward, 5′-CGGCGGTAGCTTATCAGACTGATG-3′ and reverse, 5′-CCAGTCGAGGGTCCGAGGTATT-3′. The following thermocycling conditions were used for the PCR: Initial denaturation at 95°C for 10 min; 40 cycles of denaturation at 95°C for 30 sec, annealing at 55°C for 30 sec and extension at 72°C for 30 sec; followed by melting curve analysis. All experiments were performed in triplicate. Finally, the 2−ΔΔCq method was performed to calculate the relative expression (22).

Western blotting (WB)

The TGF-β1 and Smad3 protein expression levels were detected by WB in mice with myocardial failure. The total proteins were extracted using RIPA lysis buffer (OP-0003, Epigentek Group, Inc., Farmingdale, NY, USA). A bicinchoninic acid protein assay was performed to determine protein concentration. Equal amounts of protein (50 µg/lane) were separated by 8–10% SDS-PAGE and transferred to polyvinylidene difluoride membranes. Prior to incubation with primary antibodies, the PVDF membrane was treated with 0.1% Tween-20 in Tris-buffered saline (TBST) containing 50 g/l skimmed milk at room temperature for 4 h. The primary antibodies, including rabbit monoclonal anti-TGF-β1 (1:1,000; cat. no. ab25121; Abcam, Cambridge, MA, USA), rabbit polyclonal anti-Smad3 (1:500; cat. no. 9523), rabbit polyclonal anti-p-Smad3 (1:500; cat. no. 8769), rabbit polyclonal anti-Smad2 (1:500; cat. no. 5678; all Cell Signaling Technology, Inc., Danvers, MA, USA), mouse monoclonal α-tubulin (1:1,000, cat. no. sc-398103) and mouse monoclonal anti-GAPDH (1:1,000; sc-51907; both Santa Cruz Biotechnology, Inc., Dallas, TX, USA) were used in the current study. After treatment with primary antibodies for 1 h at room temperature, the membranes were rinsed with TBST and incubated with anti-rabbit horseradish peroxidase-conjugated secondary antibody (1:2,000; cat. no. SC-2004, Santa Cruz Biotechnology, Inc.) at room temperature for 1 h. Chemiluminescent detection was performed with the Enhanced Chemiluminescence kit (Pierce; Thermo Fisher Scientific, Inc.). The amount of the protein of interest, which was expressed using arbitrary densitometric units, was normalized to the densitometric units of GAPDH.

Hematoxylin and eosin stain (H&E)

The cardiac tissue (4 µm thick) was obtained from the mice, fixed with 10% formaldehyde at room temperature for 12 h, dehydrated, embedded in paraffin at room temperature for 1 h and stored at 4°C. The tissue section was incubated at 65°C for 1 h according to a procedure for dewaxing (23,24). Samples were stained with hematoxylin for 1 min, followed by eosin staining for 10 sec at room temperature, and, then the samples were dried at room temperature and sealed. The middle part of the visual field was examined by light microscopy (magnification, ×20) and the field was assessed by three different pathologists (25).

Immunofluorescence analysis

The tissue from myocardial failure patients and controls were fixed with neutralized formaldehyde overnight at 4°C and dehydrated the next day. Subsequently, the tissue was 4 µm and placed at 4°C. The film was cooled and placed in a slicing machine for 1 h washed with PBS three times for 5 min. Following washing, sodium citrate buffer (pH 6.0) was boiled at high-pressure for antigen repair for 5 min and washed with PBS 3 times for 5 min. The tissue sample was then soaked in hydrogen peroxide for 30–60 min at room temperature, washed with PBS three times for 5 min, and blocked with 10% fetal bovine serum. The samples were incubated with anti-FASN (1:100; LS-C104946; BD Biosciences, Franklin Lakes, NJ, USA), anti-SCD1 (1:50; sc-515844) and anti-UCP1 (1:100; sc-293418; both Santa Cruz Biotechnology, Inc., Dallas, TX, USA) antibodies overnight at 4°C. The samples were washed with PBS 3 times for 5 min. Following washing, goat-anti-mouse secondary antibody was added (1:100, A32723; Invitrogen; Thermo Fisher Scientific, Inc.) with 1% bovine serum albumin (BSA; Baoman Biological Technology Co., Ltd, Shanghai, China) at 37°C for 1 h. DAB was used to subsequently stain the samples at 37°C for 5 min, which were then washed with PBS 3 times for 5 min and the reaction was terminated. The samples were stained with hematoxylin for 3 min at room temperature, sealed and images were captured. PBS instead of the primary antibody was used as a negative control.

When the H9c2 cells reached 70% confluency, they were cultured in DMEM with or without DOX (10 µM) for 24 h. The H9C2 cells in the treatment and control groups were washed with PBS 3 times and fixed with 4% paraformaldehyde for 10 min at room temperature. Following 3 washes with PBS, the cultured cells were blocked with 10% BSA (MP Biomedicals, LLC, Santa Ana, CA, USA) for 1 h and incubated overnight at 4°C with the Smad3 and TGF-β1 primary antibodies described above. The anti-TGF-β1 antibody was diluted 1:200 (cat. no. ab25121, Abcam) and anti-Smad3 antibody was diluted 1:500 (cat. no. 51-1500; Thermo Fisher Scientific, Inc. USA). The cells were incubated with Alexa Fluor 488-conjugated goat anti-rabbit IgG (H+L) highly cross-adsorbed secondary antibody (1:100; cat. no. A-11034; Invitrogen) at 37°C for 1 h and stained with 4,6-diamino-2-phenyl indole at 37°C for 5 min to visualize the nuclei. The images were captured using a fluorescence microscope (model DMI-4000B; Leica Microsystems GmbH, Wetzlar, Germany).

Oil red O staining

Lipid distribution in heart tissues was examined by oil red O staining. Mouse samples were fixed with 10% formalin for 30 min in room temperature and sliced into 10-µm-thick sections with a cryostat. Next, the tissues were washed in PBS for 30 sec, washed in 60% isopropyl alcohol for 1 min, and stained by 0.5% oil red O for 10 min at 37°C. Thereafter, slices were fractionated with 60% iso-propyl alcohol for 2 min, washed with PBS solution for 2 min, and stained with 0.25% hematoxylin for 5 min in room temperature. Following a further 2 min wash in PBS, slices were colored with 0.1% lithium carbonate for 30 sec at room temperature, washed with PBS solution for 5 min and coverslipped (26). The slices were visualized using a Nikon 80i light microscope (Nikon Corporation, Tokyo, Japan) at magnification, ×20.

Mouse electrocardiographic detection and echocardiography

To anaesthetize each mouse, avertin (2,2,2 trimethylethanol; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany; 240 mg/kg) was administered into the intraperitoneal space prior to electrocardiographic detection and echocardiography. Subsequently, the mice were fixed on a test bed according to the connection for the right upper limb, left upper limb, left lower limb, and right lower limb. Subsequently, the electrode needle was injected subcutaneously into the animal limbs and the electrocardiography was recorded. Transthoracic echocardiography was performed at the end of the observation period to determine heart function using an ultrasonic apparatus (Voluson E8; GE Healthcare, Chicago, IL, USA; 15-MHz probe) with mice being lightly sedated by 1.5–2% isoflurane. Cardiac relaxation was assessed by first derivative of left ventricular pressure (dP/dt). The left ventricular systolic pressure (LVSP) was detected through the BL-420 multi-channel physiological system. A 2-dimensional echocardiogram from the apical view was used to determine the systolic ejection fraction by planimetry of the left ventricle.

Statistical analysis

All experimental data are presented as the mean ± standard deviation and were analyzed using Image-Pro-Plus 6.0 (Media Cybernetics, Inc., Rockville, MD, USA) and Graph Pad Prism 5 (GraphPad Software, Inc., La Jolla, CA, USA) statistical software. One-way analysis of variance followed by Bonferroni's multiple comparisons test was used to compare multiple groups and paired t-test was used to compare two groups. P<0.05 was considered to indicate a statistically significant difference.

Results

miR-127 is highly expressed in patients with myocardial failure

To detect the expression of miR-127 in patients with myocardial failure, an RT-qPCR method was used to measure the expression levels of miR-127 in tissue samples of 51 patients with myocardial failure and 50 normal controls. The results indicated that the expression of miR-127 in the myocardium significantly increased compared with the control group (P<0.001; Fig. 1A). The overexpression of miR-127 in myocardial failure suggests that miR-127 may serve an important role in the development of myocardial failure. Oil red O staining indicated accumulation of large amounts of lipids in patients with myocardial failure, compared with healthy controls, which suggested that heart lipid accumulation may affect the normal function of myocardial cells (Fig. 1B). Detection of FASN, SCD1 and UCP1 by immunohistochemistry suggested that the accumulation of fat was more severe in patients with myocardial failure compared with the control group (Fig. 1C).

Chronic pressure overload leads to myocardial failure

In the present study, a drug-induced mouse model of myocardial failure was successfully established. An advanced study of heart function was conducted by echocardiography. AAC and the ejection fraction were significantly reduced in the heart failure model compared with the control mice (Fig. 2A). To detect the pathological alterations, H&E staining was performed using the tissues from both the heart failure model and the control mice. The results indicated that cardiomyocytes were significantly hypertrophic following induction, especially after 6 months of induction (Fig. 2B). The left ventricular systolic pressure (LVSP) in B6 mice after induction was significantly increased after 1 month compared with the control mice, as indicated by the results of the echocardiographic analysis (Fig. 2C). The maximum left ventricular ejection pressure was significantly decreased in mice after 6 months of induction (P<0.001; Fig. 2D).

Overexpression of miR-127 promotes myocardial failure

A model of myocardial failure was successfully established in mice and it was observed that miR-127 expression significantly increased following DOX induction compared with the untreated mice (Fig. 3A). Expression of TGF-β1 and Smad3 in myocardial tissue induced by DOX markedly increased compared with the control group (Fig. 3B and C).

miR-127 regulates the expression of TGF-β1/Smad3

Following transfection with si-miR-127, RT-qPCR demonstrated that the expression of miR-127 was decreased in the 6 months post-AAC siRNA group, compared with 6 months post-AAC group alone (Fig. 4A) and the expression of TGF-β1 and Smad3 was positively correlated with miR-127 (Fig. 4B). TGF-β, as a transcription factor, is located in the nucleus and involved in the regulation of cell function. When the expression levels of miR-127 were decreased by siRNA, the TGF-β1 expression in the nucleus was significantly reduced, which inhibited the Smad3 signal transduction (Fig. 4C).

Discussion

The present study demonstrated that miR-127 was upregulated in cardiac samples from patients with heart failure. The in vivo and in vitro experiments indicated that miR-127 aggravated the development of heart failure. The potential pathological mechanisms of the effect of miR-127 may be based on the upregulation of the TGF-β1/Smad3 pathway.

The hypothesis that circulating miRNAs and incident myocardial failure may be associated has been suggested in a previous study (14). In one study, among the 19 miRNAs investigated in the plasma of patients, individuals with high levels of miR-126 had a 2.7-fold higher risk of incident myocardial failure compared with healthy patients, and patients with low levels of miR-223 and miR-197 had a high risk of incident myocardial failure (15). The results of the present study also indicated that miR-127 is highly expressed in patients with myocardial failure. Therefore, whether miR-127 could be used as a potential biomarker of myocardial failure requires further study.

Previous studies have indicated that the expression of miR-127 is increased in malignant tumor tissues compared with normal and benign tissue samples, and that the overexpressed miR-127 may contribute to tumor formation and progression (27,28). In the present study, miR-127 was significantly overexpressed in patients with myocardial failure compared with the control group, and the myocardial failure may have been due to the excessive fat accumulation. It has been reported that elevated expression of miR-127 was associated with activation of the inflammatory signaling pathway, such as nuclear factor-κB (29) and JNK pathway (30). The results of the present study demonstrated that miR-127 was highly expressed in the drug-induced mouse model of myocardial failure and the expression increased in a time-dependent manner.

TGF-β1 is the most potent pro-fibrotic cytokine identified to date and a key mediator of fibroblast activation and fibrosis in the diseased heart (18). TGF-β1 was highly expressed following drug induction and caused the upregulation of Smad3 in the mouse model of myocardial failure. In the canonical TGF-β signaling, activation of the TGF-β receptor leads to p-Smad2/3 (31). Activated Smads can bind to transcription factors and regulate a variety of biological effects resulting in cell-specific transcriptional regulation (32,33). In the present study, the expression levels of TGF-β1/Smad3 were upregulated in myocardial failure tissues and inhibited when miR-127 was knocked down with siRNAs. Therefore, miR-127 may regulate myocardial failure by activating the TGF-β1/Smad3 signaling pathways.

A limitation of the present study was that p-Smad3 was not detected by immunohistochemistry. The present study also did not use the antagomir-127 to verify the results. Further studies, such as antagomir-127 application in both in-vivo and in-vitro experiments, are required to validate the function of miR-127 in myocardial failure.

In conclusion, miR-127 was highly expressed and fat was severely accumulated in myocardial failure, in both human and mouse tissue. The present study also indicated that the expression levels of TGF-β1 and Smad3 significantly increased in the myocardial tissue following treatment with DOX. The potential pathological mechanism of the effect of miR-127 may be based on the upregulation of the TGF-β1/Smad3 signaling pathway.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

FL conceived and designed the experiments. HX and FL performed the experiments, and HX and FL analyzed the data. HX and FL wrote the manuscript.

Ethics approval and consent to participate

All aspects of this research were approved by the Ethics Committee of Weifang People's Hospital.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wan G, Ji L, Xia W, Cheng L and Zhang Y: Screening genes associated with elevated neutrophiltolymphocyte ratio in chronic heart failure. Mol Med Rep. 18:1415–1422. 2018.PubMed/NCBI

2 

Kannel WB: Incidence and epidemiology of heart failure. Heart Fail Rev. 5:167–173. 2000. View Article : Google Scholar : PubMed/NCBI

3 

Neubauer S: The failing heart-an engine out of fuel. N Engl J Med. 356:1140–1151. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Gulsin GS, Shetye A, Khoo J, Swarbrick DJ, Levelt E, Lai FY, Squire IB, Arnold JR and McCann GP: Does stress perfusion imaging improve the diagnostic accuracy of late gadolinium enhanced cardiac magnetic resonance for establishing the etiology of heart failure? BMC Cardiovasc Disord. 17:982017. View Article : Google Scholar : PubMed/NCBI

5 

Ingwall JS and Weiss RG: Is the failing heart energy starved? On using chemical energy to support cardiac function. Circ Res. 95:135–145. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Ashrafian H, Frenneaux MP and Opie LH: Metabolic mechanisms in heart failure. Circulation. 116:434–448. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Murakami A, Nagao K, Juni N, Hara Y and Umeda M: An N-terminal di-proline motif is essential for fatty acid-dependent degradation of Δ9-desaturase in Drosophila. J Biol Chem. 292:19976–19986. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Dobrzyn A and Ntambi JM: The role of stearoyl-CoA desaturase in body weight regulation. Trends Cardiovasc Med. 14:77–81. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Liu G, Lynch JK, Freeman J, Liu B, Xin Z, Zhao H, Serby MD, Kym PR, Suhar TS, Smith HT, et al: Discovery of potent, selective, orally bioavailable stearoyl-CoA desaturase 1 inhibitors. J Med Chem. 50:3086–3100. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Hess D, Chisholm JW and Igal RA: Inhibition of stearoylCoA desaturase activity blocks cell cycle progression and induces programmed cell death in lung cancer cells. PLoS One. 5:e113942010. View Article : Google Scholar : PubMed/NCBI

11 

Theocharisa S, Margeli A and Kouraklis G: Peroxisome proliferator activated receptor-gamma ligands as potent antineoplastic agents. Curr Med Chem Anticancer Agents. 3:239–251. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Wei JJ, Wu X, Peng Y, Shi G, Basturk O, Yang X, Daniels G, Osman I, Ouyang J, Hernando E, et al: Regulation of HMGA1 expression by microRNA-296 affects prostate cancer growth and invasion. Clin Cancer Res. 17:1297–1305. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Zhang Z, Wan F, Zhuang Q, Zhang Y and Xu Z: Suppression of miR-127 protects PC-12 cells from LPS-induced inflammatory injury by downregulation of PDCD4. Biomed Pharmacother. 96:1154–1162. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Ciebiera M, Wlodarczyk M, Wrzosek M, Męczekalski B, Nowicka G, Łukaszuk K, Ciebiera M, Słabuszewska-Jóźwiak A and Jakiel G: Role of transforming growth factor β in uterine fibroid biology. Int J Mol Sci. 18:pii: E2435. 2017. View Article : Google Scholar

15 

Yoshida K, Murata M, Yamaguchi T and Matsuzaki K: TGF-β/Smad signaling during hepatic fibro-carcinogenesis (review). Int J Oncol. 45:1363–1371. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Kamato D, Burch ML, Piva TJ, Rezaei HB, Rostam MA, Xu S, Zheng W, Little PJ and Osman N: Transforming growth factor-β signalling: Role and consequences of Smad linker region phosphorylation. Cell Signal. 25:2017–2024. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Wei Q, Liu Q, Ren C, Liu J, Cai W, Zhu M, Jin H, He M and Yu J: Effects of bradykinin on TGFbeta1induced epithelialmesenchymal transition in ARPE19 cells. Mol Med Rep. 17:5878–5886. 2018.PubMed/NCBI

18 

Liu ZY, Qiu HO, Yuan XJ, Ni YY, Sun JJ, Jing W and Fan YZ: Suppression of lymphangiogenesis in human lymphatic endothelial cells by simultaneously blocking VEGF-C and VEGF-D/VEGFR-3 with norcantharidin. Int J Oncol. 41:1762–1772. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Lequerica-Fernandez P, Astudillo A and de Vicente JC: Expression of vascular endothelial growth factor in salivary gland carcinomas correlates with lymph node metastasis. Anticancer Res. 27:3661–3666. 2007.PubMed/NCBI

20 

Panagiotaki KN, Sideratou Z, Vlahopoulos SA, Paravatou-Petsotas M, Zachariadis M, Khoury N, Zoumpourlis V and Tsiourvas D: A Triphenylphosphonium-Functionalized Mitochondriotropic nanocarrier for efficient Co-Delivery of doxorubicin and chloroquine and enhanced antineoplastic activity. Pharmaceuticals (Basel). 10:pii: E91. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Chen X, Guo Z, Wang P and Xu M: Erythropoietin modulates imbalance of matrix metalloproteinase-2 and tissue inhibitor of metalloproteinase-2 in doxorubicin-induced cardiotoxicity. Heart Lung Circ. 23:772–777. 2014. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Adult Acute Myeloid Leukemia Treatment (PDQ(R)): Patient VersionPDQ Cancer Information Summaries. Bethesda (MD): 2002

24 

Magadum A, Ding Y, He L, Kim T, Vasudevarao MD, Long Q, Yang K, Wickramasinghe N, Renikunta HV, Dubois N, et al: Live cell screening platform identifies PPARδ as a regulator of cardiomyocyte proliferation and cardiac repair. Cell Res. 27:1002–1019. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Hu J, Cheng P, Huang GY, Cai GW, Lian FZ, Wang XY and Gao S: Effects of Xin-Ji-Er-Kang on heart failure induced by myocardial infarction: Role of inflammation, oxidative stress and endothelial dysfunction. Phytomedicine. 42:245–257. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Andrés-Manzano MJ, Andrés V and Dorado B: Oil Red O and hematoxylin and eosin staining for quantification of atherosclerosis burden in mouse aorta and aortic root. Methods Mol Biol. 1339:85–99. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Tedesco FS, Dellavalle A, Diaz-Manera J, Messina G and Cossu G: Repairing skeletal muscle: Regenerative potential of skeletal muscle stem cells. J Clin Invest. 120:11–19. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Charge SB and Rudnicki MA: Cellular and molecular regulation of muscle regeneration. Physiol Rev. 84:209–238. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Huan L, Bao C, Chen D, Li Y, Lian J, Ding J, Huang S, Liang L and He X: MicroRNA-127-5p targets the biliverdin reductase B/nuclear factor-kB pathway to suppress cell growth in hepatocellular carcinoma cells. Cancer Sci. 107:258–266. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Ying H, Kang Y, Zhang H, Zhao D, Xia J, Lu Z, Wang H, Xu F and Shi L: MiR-127 modulates macrophage polarization and promotes lung inflammation and injury by activating the JNK pathway. J Immunol. 194:1239–1251. 2015. View Article : Google Scholar : PubMed/NCBI

31 

D'Arpino MC, Fuchs AG, Sánchez SS and Honoré SM: Extracellular matrix remodeling and TGF-β1/Smad signaling in diabetic colon mucosa. Cell Biol Int. 42:443–456. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Gressner AM and Weiskirchen R: Modern pathogenetic concepts of liver fibrosis suggest stellate cells and TGF-beta as major players and therapeutic targets. J Cell Mol Med. 10:76–99. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Liu X, Hu H and Yin JQ: Therapeutic strategies against TGF-beta signaling pathway in hepatic fibrosis. Liver Int. 26:8–22. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 18 Issue 6

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
Xu H and Xu H: miR‑127 aggravates myocardial failure by promoting the TGF‑β1/Smad3 signaling Retraction in /10.3892/mmr.2019.10442 . Mol Med Rep 18: 4839-4846, 2018
APA
Xu, H., & Xu, H. (2018). miR‑127 aggravates myocardial failure by promoting the TGF‑β1/Smad3 signaling Retraction in /10.3892/mmr.2019.10442 . Molecular Medicine Reports, 18, 4839-4846. https://doi.org/10.3892/mmr.2018.9514
MLA
Xu, H., Li, F."miR‑127 aggravates myocardial failure by promoting the TGF‑β1/Smad3 signaling Retraction in /10.3892/mmr.2019.10442 ". Molecular Medicine Reports 18.6 (2018): 4839-4846.
Chicago
Xu, H., Li, F."miR‑127 aggravates myocardial failure by promoting the TGF‑β1/Smad3 signaling Retraction in /10.3892/mmr.2019.10442 ". Molecular Medicine Reports 18, no. 6 (2018): 4839-4846. https://doi.org/10.3892/mmr.2018.9514