Journal List > Korean J Physiol Pharmacol > v.28(2) > 1516086323

Yang, Yang, Liu, Liu, Liu, Nie, Chu, and Yang: Effects of gas signaling molecule SO2 in cardiac functions of hyperthyroid rats

Abstract

Sulfur dioxide (SO2), a novel endogenous gas signaling molecule, is involved in the regulation of cardiac function. Exerting a key role in progression of hyperthyroidism-induced cardiomyopathy (HTC), myocardial fibrosis is mainly caused by myocardial apoptosis, leading to poor treatment outcomes and prognoses. This study aimed to investigate the effect of SO2 on the hyperthyroidism-induced myocardial fibrosis and the underlying regulatory mechanisms. Elisa, Masson staining, Western-Blot, transmission electron microscope, and immunofluorescence were employed to evaluate the myocardial interstitial collagen deposition, endoplasmic reticulum stress (ERS), apoptosis, changes in endogenous SO2, and Hippo pathways from in vitro and in vivo experiments. The study results indicated that the hyperthyroidism-induced myocardial fibrosis was accompanied by decreased cardiac function, and down-regulated ERS, apoptosis, and endogenous SO2-producing enzyme aspartate aminotransferase (AAT)1/2 in cardiac myocytes. In contrast, exogenous SO2 donors improved cardiac function, reduced myocardial interstitial collagen deposition, up-regulated AAT1/2, antagonized ERS and apoptosis, and inhibited excessive activation of Hippo pathway in hyperthyroid rats. In conclusion, the results herein suggested that SO2 inhibited the overactivation of the Hippo pathway, antagonized ERS and apoptosis, and alleviated myocardial fibrosis in hyperthyroid rats. Therefore, this study was expected to identify intervention targets and new strategies for prevention and treatment of HTC.

INTRODUCTION

Hyperthyroidism is mainly characterized by excessive synthesis and release of thyroid hormones (THs) by the thyroid gland, causing hyper-metabolism in the body [1,2]. Hyperthyroidism-induced cardiomyopathy (HTC), one of the most common complications of hyperthyroidism, is harmful to the cardiac conduction system and cardiomyocytes caused by the direct or indirect toxic effects of chronic excess THs on the heart [3,4]. It is the main cause of death in hyperthyroidism other than hyperthyroid crisis [5]. In contrast, myocardial fibrosis is a key factor affecting the progression of HTC. THs induce endoplasmic reticulum stress (ERS) [6] and apoptosis in cardiomyocytes [7] while increasing the force and speed of myocardial contraction [8] and the burden on the heart. Thus, it can lead to cardiac hypertrophy and left ventricular dysfunction [9], resulting in heart failure [10,11] and dilated cardiomyopathy [12]. Currently, the mechanism of hyperthyroidism-induced myocardial fibrosis remains unclear, and there are few studies on clear and effective clinical interventions for hyperthyroidism-induced myocardial fibrosis.
Apoptosis is a unique and important mode of “programmed” cell death that involves the genetically determined cell elimination [13]. Hyperthyroidism can induce apoptosis in liver cells [14] and myocardium of hyperthyroid rats [7], but the specific intrinsic regulatory mechanisms are not clear. Homeostatic imbalance of the endoplasmic reticulum (ER), an important organelle in eukaryotic cells, will lead to ERS [15], which is closely associated with several cardiovascular diseases, such as myocardial infarction, myocardial ischemia, dilated cardiomyopathy, and heart failure [16]. As an ERS protein, C/EBP homologous protein (CHOP) can inhibit the expression of anti-apoptotic proteins, activate the apoptotic proteins, and initiate the Caspase signaling pathway to induce apoptosis [17]. In addition, excessive ERS can induce apoptosis [18]. Whether ERS can mediate the hyperthyroidism-induced myocardial fibrosis is still unclear and needs to be further investigated.
The Hippo signaling pathway, which plays a crucial role in regulating cellular homeostasis and is a significant mediator of apoptosis, represents a promising target for intervention in various many cardiovascular diseases [19]. Blocking the Hippo pathway has been demonstrated to reduce apoptosis and thus counteract fibrosis [20]. Specifically, the activation of Yes-associated protein (YAP) is a critical component of Hippo signal transduction, which is essential for apoptosis during ERS [21]. Furthermore, recent research suggests that downregulation of the Hippo pathway could ameliorate myocardial injury by suppressing ERS [22,23]. However, the precise contribution of the Hippo pathway-mediated apoptosis to the development of HTC remains unclear.
Sulfur dioxide (SO2), once considered a toxic gas with an unpleasant odor, is now recognized as a novel endogenous gas signaling molecule skin to nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S). In mammals, SO2 is produced endogenously through the transamination of aspartate aminotransferase (AAT) during the metabolism of sulfur-containing amino acids [24]. Studies have demonstrated that SO₂ can ameliorate myocardial fibrosis caused by myocardial infarction or diabetes by inhibiting the proliferation and migration of cardiac fibroblasts [25]. Nonetheless, it remains unclear whether SO2 can attenuate hyperthyroidism-induced myocardial fibrosis and whether endogenous SO2 is involved in the regulatory mechanisms of ERS and apoptosis in hyperthyroid rat cardiomyocytes.
Thus, this study aims to establish an animal model of hyperthyroidism to observe the changes in myocardial interstitial fibrosis and investigate the effects of exogenous SO2 donor (Na2SO3/NaHSO3 solution) and SO2-producing enzyme inhibitor (hydrogen deuterium exchange [HDX]) on myocardial fibrosis in hyperthyroid rats. Meanwhile, it intends to explore whether the underlying mechanisms of SO2 involve downregulating Hippo pathway to antagonize ERS and inhibit apoptosis. The findings of this study are expected to yield novel intervention targets and therapeutic approaches for the prevention and treatment of HTC.

METHODS

Animals

Animal experiments were conducted in accordance with the Guidelines for the Welfare and Use of Experimental Animals and the animal management regulations and specifications of the University of South China Laboratory Animal Center. Meanwhile, they were approved by the Animal Ethics Committee of the University of South China and conformed (Granted ID: USC201912SD23).

Study design

Forty male Sprague Dawley (SD) rats weighing (200 ± 20) g at 8 weeks of age were firstly acclimatized and fed for 1 week. After that, they were divided into 4 groups with equal number: a control group, a levothyroxine (L-Thy) group, a L-Thy + SO2 group, and a L-Thy + SO2 + HDX group. On a normal diet, L-Thy (0.5 mg/kg, Sigma-Aldrich) was intraperitoneally injected for rats in all groups except the control group [26]. Rats in the control group were intraperitoneally injected with the same dose of solvent as the model group. Both the L-Thy + SO2 and L-Thy + SO2 + HDX groups were administered intraperitoneally with SO2 donor (Na2SO3/NaHSO3, 85 mg/kg/d, Sigma-Aldrich) [27]. Furthermore, those in the L-Thy + SO2 + HDX group were additionally injected with SO2 production enzyme inhibitor (HDX, 25 mg/kg, Sigma-Aldrich) intraperitoneal with once a week for continuous 5 weeks [28].

Materials

L-Thy, Na2SO3, NaHSO3, and HDX (serine racemase inhibitor) purchased from Sigma-Aldrich. AAT1 (14886-1-AP), AAT2 (14800-1-AP), CHOP (15204-1-AP), Caspase3 (19677-1-AP), Caspase9 (10380-1-AP), Bax (50599-2-Ig), Bcl2 (26593-1-AP), MMP2 (10373-2-AP), MMP3 (66338-1-Ig), Collagen I (14695-1-AP), Collagen III (22734-1-AP), YAP1 (13584-1-AP) and GAPDH (10494-1-AP) primary antibodies and mouse/rabbit secondary antibodies were purchased from Proteintech. MST1 (3682T), GRP78/BIP (3177P), ERP72 (5033P), and Phosphor-YAP (P-YAP) (13008S) primary antibodies were purchased from Cell Signaling Technology; TIMP2 (A1558) and LATS1 (A17992) primary antibodies were purchased from ABclonal. Meanwhile, gel configuration kits and BCA protein quantification kits were purchased from Beyotime Institute of Biotechnology. Electrochemiluminescence (ECL) kit was purchased from New Cell and Molecular Biotech Co, Ltd. Total-triiodothyronine (TT3) and total thyroxine (TT4) ELISA kits were purchased from Wuhan Purity Biotech Co, Ltd. SO2 ELISA kit was purchased from Shanghai Enzyme Link Biotech Co, Ltd. Scrambled control siRNA oligonucleotide (si-NC) or siRNA for YAP1 were purchased from Hanbio Tech.

Cell culture and transfection

H9c2 cardiomyocyte cell lines, from ATCC were incubated in Dulbecco’s modified Eagle’s medium containing 5.5 mM D-glucose (10% fetal bovine serum and 1% penicillin/streptomycin) and 5% CO2 at 37°C. When the density was approximate 1 × 106, the cells were inoculated in 6-well plates. The cells were divided into a control group, a L-Thy group, a L-Thy + SO2 group, a L-Thy + SO2 + HDX group, and a SO2 group according to different treatments. The cells in the control group and the SO2 group were cultured with a normal medium, and those in the L-Thy group and L-Thy + SO2 group were treated with 2 μM L-Thy at 37°C for 24 h [29]. The cells in the L-Thy + SO2 and L-Thy + SO2 + HDX groups were pretreated for 0.5 h by adding exogenous SO2 donor Na2SO3/NaHSO3 (100 μM, 3:1 mol rate), as described previously [30]. Next, they were co-incubated with a normal medium. In contrast, the cells in the L-Thy + SO2 + HDX group were pretreated with the endogenous SO2 produced enzyme inhibitor HDX (200 μM) for 24 h [31].
In order to knock down YAP1 expression, YAP1-specific siRNA was mixed with the transfection reagent and incubated at room temperature for 15 min, as per manufacturer’s instructions. The culture medium was removed, and the cells were washed once before adding fresh antibiotic-free culture medium. The pre-mixed siRNA and transfection reagent were then introduced to the respective cell groups. The cells were then incubated at 37°C in a 5% CO2 atmosphere for 24 h. After 24 h, the culture medium was replaced to remove the siRNA and transfection reagent, and the cells continued to be cultured. Another 24 h later, cellular protein samples were extracted, and Western blot technique was employed to detect protein expression levels, thereby verifying the transfection efficiency. After successful transfection, subsequent experiments were continued.

Echocardiography

At the end of the drug intervention period, the rats were anaesthetized with 2.5% isoflurane (RWD Life Science, R510-22-10), followed by an anaesthetic respiratory system (1.5% isoflurane to maintain anaesthesia), a M-mode ultrasound machine (GE Versana Premier) and L8-18i-RS ultrahigh-frequency line-array probe were performed to measure heart rate (HR), left ventricular fractional shortening (LVFS) and left ventricular ejection fraction (LVEF) from the left parasternal axis using two-dimensional echocardiography for five consecutive cardiac cycles.

Serum TT3 and TT4 levels

Before euthanizing the rats, 2 ml of blood was collected from the renal vein and allowed to coagulate at room temperature. The blood was then centrifuged at 4°C and 3,000 r/min for 10 min to separate the serum. The serum was stored in a –80°C refrigerator until measurement. TT3 and TT4 levels in serum were quantified using the TT3 Elisa kit (CD28783; Chundubio) and TT4 Elisa kit (CD28784; Chundubio), respectively, following the instructions provided in the kit.

Calculation of heart weight (HW) index

First, the rats were weighed individually prior to the experiment. After anesthesia, the rats were placed on the operating table in a supine position and secured in place. Next, the hearts were dissected quickly and immediately rinsed with low-temperature saline to remove all blood and impurities. The hearts were then blotted with filter paper to remove excess moisture and weighed using an electronic balance to calculate the HW index. The specimens were then collected according to the required procedures.

SO2 content assay

The myocardial tissue was mashed with the appropriate amount of saline and then centrifugated. The supernatant was collected and assayed with the rat SO2 Elisa kit (YJ832161; Mlbio) according to the instruction manual.

Masson staining

The myocardial tissues were fixed using 4% paraformaldehyde, dehydrated by gradient alcohol, and embedded in paraffin. The tissues were then sectioned into 4-um-thin sections by a microtome, stained using a Masson staining kit (Abiowell Biotech Co, Ltd) according to the manufacturer’s instructions. Next, the tissues were covered with a coverslip using mounting medium. Finally, the stained tissues were examined under a light microscope.

Transmission electron microscopy (TEM)

Myocardial tissues were fixed with 2.5% glutaraldehyde, segmented into 50–100 nm thin slices, and rinsed with phosphoric acid (Beyotime Institute of Biotechnology). After fixed immersion in 1% osmium tetroxide (Absin Biosciences, Inc), the tissues were rinsed with phosphoric acid again, followed by gradient acetone immersion, dehydration, and drying. Then, they were stained with 3% uranyl acetate and lead nitrate for 10–20 min and rinsed with distilled water. Finally, they were observed under TEM to study their ultrastructure.

Western blotting

Rat myocardial tissues and cell precipitates were lysed using cold radioimmunoprecipitation assay lysis buffer containing protease inhibitors. After centrifugation, the supernatant was collected, and the protein concentration was quantified and normalized using the BCA kit, following the manufacturer’s instructions. The samples were denatured by heating at 95°C for 10 min and separated on 10%–12% polyacrylamide electrophoresis gels. The proteins were then transferred electrophoretically onto a polyvinylidene fluoride membrane (Millipore). The membranes were blocked with 5% skim milk at room temperature for 1 h and then incubated overnight at 4°C with the primary antibody. Next, they were washed three times (5 min × 3) with tris-buffered saline containing Tween 20 (TBST) and incubated with the corresponding horseradish peroxidase-conjugated secondary antibody at room temperature for 1 h. After washing with TBST (5 min × 3), the membrane was exposed to ECL chemiluminescent solution, and the image signals were acquired using the BIO-RAD XRS+ imaging system. Finally, the relative data were analyzed using ImageJ software.

Immunofluorescence staining

Cells were fixed with 4% perhalooxy-alkanes for 40 min, permeabilized with 0.1% Triton X-100 for 5 min × 2, and blocked with 5% BSA for 1 h. Next, they were incubated with rabbit anti-Bax (1:100) at 4°C overnight and then with secondary antibody coupled with FITC (1:200) at room temperature for 1 h. After that, the cells were incubated with rabbit anti-Caspase3 (1:100) at 4°C overnight and with a secondary antibody coupled with Cy3 (1:200) at room temperature for 1 h. Finally, they were re-stained using DAPI. The fluorescence was analyzed using a fluorescence microscope (Olympus IX71; Olympus).

Statistical analysis

Values are presented as the mean ± standard deviation for data that were normally distributed or median and interquartile range for data that were not normally distributed for continuous variables and number (%) for categorical variables. The representative image for each group was selected based on the mean value after quantitative analysis. Statistical analyses were performed with GraphPad Prism software (version 9.5.1). For comparisons of two groups, Student’s t-test was used. For comparisons of more than two groups, one-way analysis of variance was used, with Bonferroni’s or Dunnett’s multiple-comparisons test for normally distributed data or the Mann–Whitney test for non-normally distributed data. Unless otherwise stated, p < 0.05 was considered statistically significant.

RESULTS

TT3 and TT4 levels in the serum of rats in each group

The TT3 and TT4 levels in the serum of rats in each group were detected by the Elisa method (Table 1). Compared with the control group, the TT3 and TT4 levels in the L-Thy, L-Thy + SO2, and L-Thy + SO2 + HDX groups were significantly increased (p < 0.05). However, there was no significant differences in TT3 and TT4 levels among the three groups (p > 0.05), indicating the successful modeling of hyperthyroid rats.

Changes in SO2 content and SO2-producing enzyme (AAT1/2) expression in myocardial tissues of rats in each group

In this experiment, the SO2 contents in the myocardial tissues of rats in each group were detected by the Elisa method (Fig. 1A). The results showed that the SO2 content in the L-Thy group was significantly lower than that in the control group (p < 0.05). Additionally, compared with the L-Thy group, the SO2 content in the L-Thy + SO2 group was significantly increased (p < 0.05). Furthermore, the SO2 content in the L-Thy + SO2 + HDX group was significantly decreased compared with that in the L-Thy + SO2 group (p < 0.05). In addition, the changes in expressions of AAT1/2 in the myocardial tissues of rats in each group were measured using Western-Blot (Fig. 1B–D). Compared with the control group, the expressions of AAT1/2 in the myocardial tissues of rats in the L-Thy group were significantly lower (p < 0.05), whereas those in the L-Thy + SO2 group were upregulated (p < 0.05) after the intervention of exogenous SO2 donor (Na2SO3/NaHSO3). Besides, expressions of AAT1/2 in the myocardial tissues of rats in the L-Thy + SO2 + HDX group were significantly lower than those in the L-Thy + SO2 group after intervention with HDX (p < 0.05).
The results from in vitro cellular experiments (Fig. 1E–G) were consistent with those from the in vivo findings. The expressions of AAT1/2 were significantly decreased in H9c2 cardiomyocytes in the L-Thy group compared to those in the control group (p < 0.05). In contrast, the expressions of AAT1/2 were significantly upregulated in the L-Thy + SO2 group compared to those in the L-Thy group (p < 0.05). Moreover, H9c2 cardiomyocytes in the L-Thy + SO2 + HDX group exhibited significantly lower AAT1/2 expressions after intervention with HDX compared with those in the L-Thy + SO2 group (p < 0.05). The above results suggest that high thyroxine stimulation reduces endogenous SO2 production in myocardial tissues of hyperthyroid rats, whereas the administration of exogenous SO2 donor can increase the SO2 content and the AAT1/2 expressions in myocardial tissues of hyperthyroid rats.

Effect of SO2 on general condition and heart weight index (HW/BW) of hyperthyroid rats

The values of body weight (BW), HW, and HW/BW of rats in each group are presented in Table 2. The results indicated that the rats in the L-Thy group had significantly lower BW values and increased HW/BW values compared to those in the control group (p < 0.05). However, the rats in the L-Thy + SO2 group showed a significant increase in BW values compared with the L-Thy group (p < 0.05), while the HW/BW values demonstrated a decreasing trend without statistically significant differences (p > 0.05). Furthermore, compared with the L-Thy + SO2 group, the BW values of rats in the L-Thy + SO2 + HDX group decreased (p < 0.05), while the HW/BW values showed an increasing trend without statistically significant differences (p > 0.05).

Effect of SO2 on cardiac function in hyperthyroid rats

The hyperthyroid rats presented significant left ventricular dysfunction, which was revealed by echocardiographic analysis after 5 weeks of continuous intraperitoneal injection of L-Thy (Fig. 2, Table 3). Rats in the L-Thy group showed faster HR and decreased LVFS and LVEF values compared to those in the control group. In contrast to the rats in the L-Thy group, those in the L-Thy + SO2 group exhibited significantly slowed HR and increased LVFS and LVEF values. Besides, the rats in the L-Thy + SO2 + HDX group possessed a significantly faster HR and decreased LVFS and LVEF values in contrast to those in the L-Thy + SO2 group. All these differences above were statistically significant (p < 0.05). In summary, these results demonstrate that high thyroxine leads to changes in the left ventricular structure and impairs the cardiac function in SD rats. However, intervention with SO2 improved the cardiac structure and function of hyperthyroid rats, while this improvement was reversed after HDX intervention.

Effect of SO2 on myocardial fibrosis in hyperthyroid rats

The deposition of myocardial collagen fibers was observed and as displayed in Fig. 3A, B. The results showed that the blue-stained collagen fibers were about 5 times in the L-Thy group compared with those in the control group (p < 0.05). The number of cardiomyocytes was reduced and disordered, and myocardial interstitial collagen deposition was significantly increased. However, in the L-Thy + SO2 group, the area of blue-stained collagen fibers was reduced by about 1/2 compared to that in the L-Thy group (p < 0.05). Besides, the number of cardiomyocytes increased, and they were relatively arranged in order. Compared with the L-Thy + SO2 group, the L-Thy + SO2 + HDX group exhibited a significant increase in the area of blue-stained collagen fibers, approximately 1-fold increase (p < 0.05) and a more disordered arrangement of cardiomyocytes. Meanwhile, changes in expressions of fibrosis-related proteins of Collagen I, Collagen III, MMP2, MMP3, and TIMP2 were evaluated through Western-Blot analysis (Fig. 3C–H). The results showed that the expressions of all these proteins were significantly higher in myocardial tissues in the L-Thy group than those in the control group, while the expression of TIMP2 protein was significantly reduced (p < 0.05). Compared with the L-Thy group, the expressions of Collagen I, Collagen III, MMP2, and MMP3 in the myocardium of rats in the L-Thy + SO2 group were decreased, while the expression of TIMP2 protein was significantly increased (p < 0.05). However, the effect of SO2 on the above protein expressions was significantly reversed in the L-Thy + SO2 + HDX group compared with the L-Thy + SO2 group after HDX intervention (p < 0.05). All these results indicate that SO2 can alleviate the dysregulation of MMPs/TIMPs expression and reduce collagen fiber deposition in myocardial tissues of hyperthyroid rats. Reduced the expression of fibrosis-related proteins in cardiomyocytes. Thus, improving myocardial fibrosis in hyperthyroid rats.

SO2 alleviated the mitochondrial damage and inhibited hyperthyroidism-induced apoptosis in cardiac myocytes of hyperthyroid rats

Fig. 4A–D illustrated the TEM results for the ultrastructure of myocardial tissues of rats in each group. The figures revealed that the myocardial fiber arrangement was disordered in the L-Thy group compared with that in the control group. Meanwhile, the mitochondrial swelling, mitochondrial cristae disappearance, mitochondrial outer membrane rupture, and ER swelling were observed. Compared with the L-Thy group, the myocardial fiber disorder mitochondrial swelling and ER swelling were alleviated in the L-Thy + SO2 group. On the other hand, the results in the L-Thy + SO2 + HDX group were similar to or even more severe than those in the L-Thy group. Apoptosis-related proteins, including BAX, Bcl2, Caspase3, and Caspase9, were detected by Western-Blot, as demonstrated in Fig. 4E–I. The results showed that the expressions of BAX, Caspase3, and Caspase9 were all significantly higher in the L-Thy group compared to those in the control group (p < 0.05), while the Bcl2 was significantly down-regulated (p < 0.05). However, the expressions of these pro-apoptotic proteins were down-regulated and the expression of Bcl2 was up-regulated after SO2 intervention (p < 0.05). In addition, anti-apoptotic effect of SO2 was significantly reversed after intervention with HDX (p < 0.05).
With the in vitro experiments, expressions of BAX and Caspase3 in each group of H9c2 cardiomyocytes were detected using immunofluorescence (Fig. 5A), and the expressions of apoptosis-related proteins were measured by Western-Blot (Fig. 5B–D). The results indicated that BAX was significantly up-regulated in H9c2 cardiomyocytes in the L-Thy group while Bcl2 was significantly down-regulated compared with that in the control group (p < 0.05). Moreover, the immunofluorescence detection revealed a significant increase in the fluorescence intensities of BAX and Caspase3 in this group of cardiomyocytes. However, the BAX was downregulated in the L-Thy + SO2 group after SO2 intervention (p < 0.05), while Bcl2 showed a significant upregulation (p < 0.05). The addition of HDX significantly reversed the above effects of SO2. Similarly, immunofluorescence detection revealed similar changes in expressions of BAX and Caspase3 after SO2 and HDX interventions (p < 0.05). It suggests that thyroxine can induce mitochondrial damage in cardiomyocytic rats and lead to cardiomyocyte apoptosis, while SO2 intervention can alleviate thyroxine-induced mitochondrial damage and antagonize cardiomyocyte apoptosis.

Association of SO2 with ERS-related proteins and its potential role in cardiomyocytes

The changes in expressions of ERS-related proteins in the myocardial tissues of rats in each group were analyzed, as observed by Western-Blot (Fig. 6A–D). The figures disclosed that the expressions of CHOP, GRP78/BIP, and ERP72 were significantly up-regulated in the myocardial tissues of rats in the L-Thy group compared with those in the control group (p < 0.05). Nevertheless, SO2 significantly antagonized the up-regulation of the above proteins (p < 0.05). Meanwhile, the changes in expressions of ERS-related proteins in H9c2 cardiomyocytes were observed by Western-Blot in vitro experiments (Fig. 6E–G). The results revealed that CHOP and ERP72 were significantly upregulated in the L-Thy group compared with those in the control group under high TH stimulation (p < 0.05), but were significantly down-regulated after SO2 intervention (p < 0.05). Furthermore, HDX intervention can reverse the inhibitory effect of SO2 on the above ERS-related proteins in vitro and in vivo experiments (p < 0.05). The results indicate that TH can induce myocardial fibrosis and excessive ERS. Additionally, we observed an association between SO2 and the expression of proteins related to ERS, suggesting a potential protective role of SO2 in cardiomyocytes. However, the exact mediating mechanisms warrant further investigation.

SO2 improved ERS by downregulating MST1, LATS1, and P-YAP to antagonize cardiomyocyte apoptosis

This study observed the changes in expressions of MST1, LATS1, and P-YAP in myocardial tissues of rats in each group, as observed by Western-Blot (Fig. 7A–D). The MST1, LATS1, and P-YAP were significantly up-regulated in the L-Thy group compared with those in the control group (p < 0.05), but were down-regulated in the myocardium of rats in the L-Thy + SO2 group (p < 0.05). Meanwhile, the in vitro experiments by Western-Blot (Fig. 7E–H) indicated that MST1, LATS1, and P-YAP were up-regulated in cardiomyocytes of the L-Thy group compared with those in the control group (p < 0.05), but were down-regulated in L-Thy + SO2 group (p < 0.05). Therefore, the in vitro and in vivo experiments demonstrated that HDX reversed the down-regulation of MST1, LATS1, and P-YAP by SO2 intervention (p < 0.05).
In our in vitro experiments, we further investigated the expression of ERS-related proteins by inhibiting the activation of the Hippo pathway via the transfection of YAP1-specific si-RNA. Our results revealed that (Fig. 7K–L), compared to the control group, the expression of ERP72 protein was significantly upregulated in the L-Thy group (p < 0.05). After transfection with YAP1-specific si-RNA, the expression of ERP72 protein in the L-Thy + si-YAP1-1 group of H9c2 cardiomyocytes was downregulated, similar to the Thy + SO2 group (p < 0.05).

DISCUSSION

Biochemically, hyperthyroidism is a condition characterized by low levels of TSH and high levels of THs (T4 and T3) [32]. THs mainly affect the heart and peripheral vascular system, including increased oxygen consumption of myocardial tissues, vascular resistance, blood volume, cardiac contractility, and HR [8]. Hyperthyroidism can lead to several complications such as HTC and compensated heart failure [33]. HTC is a group of cardiovascular diseases dominated by cardiomyocyte hypertrophy and myocardial fibrosis due to the positive inotropic and chronotropic effects of THs on the heart, as well as elevated adrenergic sensitivity and activation of the renin angiotensin aldosterone system [3]. In this study, a hyperthyroid rat model was established by continuous intraperitoneal injection of L-Thy for 5 weeks. The Masson staining and Western-Blot results revealed that myocardial collagen synthesis was significantly increased in hyperthyroid rats, accompanied by impaired synthesis and clearance of extracellular matrix and obviously increased myocardial fibrosis. Similarly, Bao et al. [29] reported a significant increase in interstitial fibrosis in cardiac myocytes of hyperthyroid rats, which is accompanied by weakened cardiac function and cardiac hypertrophy. In addition, Khamis et al. [34] found significant upregulations of cardiac fibrosis-related genes of SMAD-2, SMAD-3, SMAD-4, TGF-β, and NF-κβ in myocardial tissues of hyperthyroid rats, and severely disturbed fibrotic changes in myocardial histopathology. Myocardial fibrosis is a key part in progression of HTC, and is mainly characterized by increased accumulation of fibrillar collagen in the cardiac interstitium and myocardial structural disorders [35]. Meanwhile, it greatly affects the systolic and diastolic function of the heart and the prognosis of patient [36]. Furthermore, this study showed that myocardial fibrosis is an important pathophysiological change and a potentially important target for intervention in hyperthyroid rats.
Apoptosis, a highly conserved physiological process of programmed cell death, plays an important role in normal organism development and maintenance of overall organismal homeostasis. It is mainly divided into endogenous pathways (mitochondrial pathways), exogenous pathways (death receptor pathways), and ERS-induced apoptotic pathways [15,37]. Cardiomyocytes, which are permanent cells with a limited capacity for regeneration in adults, are particularly susceptible to apoptosis. Apoptosis of cardiomyocytes can lead to the loss of these cells, thus activating myofibroblasts can increase the collagen fiber synthesis, which in turn induces myocardial fibrosis [38,39]. Therefore, effectively inhibiting cardiomyocyte apoptosis is important to prevent and treat myocardial fibrosis. Numerous studies have shown that Caspase3 is one of the most frequently activated cysteases during apoptosis [40,41]. BAX can act as a pro-apoptotic signal to increase cell permeability and promote apoptosis, while Bcl2 is an anti-apoptotic signal to promote cell survival and inhibit apoptosis [42]. In this study, the expressions of Caspase3 and BAX were significantly increased in the myocardial tissues of hyperthyroid rats and H9c2 cardiomyocytes after thyroxine intervention, while the expression of Bcl2 was downregulated. Besides, Wang et al. [7] reported a significant increase in apoptosis in the myocardium of hyperthyroid rats after 4 weeks of intervention with thyroxine T4, indicating that high thyroxine can increase the susceptibility of cardiomyocytes to apoptosis [7]. Teixeira et al. [43] found the Bcl2 was downregulated and the BAX/Bcl2 ratio was increased in the hyperthyroid rat model. Additionally, the hyperthyroidism-induced myocardial fibrosis was associated with increased apoptotic protein signaling [43]. Taken together, these results suggest that high thyroxine can induce apoptosis in cardiomyocytes, leading to myocardial fibrosis in hyperthyroid rats.
ERS refers to the accumulation of large amounts of unfolded or misfolded proteins caused by environmental stress in ER, which may inhibit the synthesis of new proteins [44]. ERS can activate the adaptive unfolded protein response, but prolonged ERS can induce apoptosis [45]. ERS-mediated apoptosis may be triggered by three signaling pathways: the CHOP, Caspase-12, and JNK [46]. This study investigated whether the thyroxine-induced apoptosis is associated with excessive ERS and found that hyper thyroxine intervention can cause ER swelling in myocardial tissues, as observed by TEM. Moreover, CHOP, GRP78/BIP, and ERP72 were significantly upregulated in the myocardial tissues of hyperthyroid rats. CHOP is an important mediator of ERS-induced apoptosis and can be upregulated under severe or sustained ERS. Meanwhile, it can regulate the expressions of many pro- and anti-apoptotic genes [47]. Luo et al. [48] demonstrated experimentally that sustained ERS is associated with cardiomyocyte apoptosis and post-infarction fibrosis. Some other studies have reported that excessive ERS can cause cardiac dilatation and even congestive heart failure and myocardial fibrosis [47,49]. These findings are similar to the observation in the hyperthyroid rat model in this study. The in vitro experiments revealed that hyper thyroxine intervention can result in elevated ERS-related proteins (including CHOP and ERP72) in H9c2 cardiomyocytes. This suggests that high thyroxine can affect the ER homeostasis to cause ERS, which in turn induces apoptosis and myocardial fibrosis in cardiomyocytes. There are few studies on hyperthyroidism-induced ERS in cardiomyocytes. Meng et al. [50] studied the thyroid specimens from five hyperthyroid patients and five healthy people and revealed that the ERS-related proteins are involved in hyperthyroidism. This suggests that thyroxine can cause excessive ERS and may be an important mechanism leading to apoptosis and myocardial fibrosis.
The Hippo pathway plays an essential role in regulating the cell proliferation and apoptosis [51]. It consists of the kinase cascade, MST1/2, LATS1/2, the downstream effectors, and transcriptional co-activators YAP and TAZ [52]. The upregulation of the Hippo pathway can induce apoptosis and reduce cell viability in cardiomyocytes [53], which is closely associated with myocardial fibrosis [54]. This study found that high thyroxine induced apoptosis in cardiomyocytes, significantly increased the levels of MST1, LATS1, and P-YAP in myocardial tissues of hyperthyroid rats as well as MST1 protein in H9c2 cardiomyocytes. These findings suggest that the Hippo pathway may play a crucial role in mediating the thyroxine-induced cardiomyocyte apoptosis. Zhang et al. [55] also demonstrated that MST1 overexpression can enhance the cardiomyocyte apoptosis and aggravate the cardiac dysfunction in diabetic rats. YAP is a key downstream transcription factor of the Hippo signaling pathway and is very important in regulating the cell proliferation and apoptotic signaling. P-YAP can be degraded, while unphosphorylated YAP can enter the nucleus to mediate the regulatory mechanisms that antagonize apoptosis. The findings in this study showed that P-YAP and LATS1 expressions were upregulated in myocardial tissues of hyperthyroid rats and H9c2 cardiomyocytes under high thyroid induction. What’s more, MST1 may promote high thyroxine-induced apoptosis in cardiomyocytes by activating LATS1 and mediating YAP1 phosphorylation. Su et al. [56] reported that in a high glucose environment, MST1 can indirectly mediate the phosphorylation of YAP1 at Ser127 and Ser397 sites through LATS1/2, thus regulating the high glucose-induced apoptosis in cardiomyocytes. These results suggest that excessive ERS can activate the Hippo pathway to induce cardiomyocyte apoptosis and mediate the regulatory mechanisms of myocardial fibrosis in hyperthyroid rats.
Previously, SO2 was regarded as an air pollutant that could endanger human health. However, it has now been recognized as a fourth class of endogenous gas signaling molecules, similar to NO, CO, and H2S [24]. An increasing number of studies have revealed that certain levels of endogenous SO2 can exert myocardial protective effects on various diseases, including hypertension, atherosclerosis, myocardial hypertrophy, diabetic myocardial fibrosis, sepsis-induced cardiac dysfunction, and pulmonary hypertension [57]. AAT is the key enzyme responsible for production of the endogenous SO2 in mammals, which comprises two isozymes: AAT1 and AAT2. AAT1 is predominantly located in the cytoplasm, while AAT2 is mainly found in the mitochondria. Meanwhile, AAT presents the highest activity in cardiac tissues [58]. Numerous studies have demonstrated that endogenous SO2 is involved in regulating cardiac function. For instance, Jin et al. [59] indicated that SO2 donors could inhibit isoprenaline-induced myocardial injury and cardiac dysfunction. According to a previous study, SO2 can inhibit myocardial fibrosis in diabetic rats and is associated with the inhibition of cardiomyocyte apoptosis [28]. Nevertheless, the underlying mechanism by which SO2 antagonizes the myocardial fibrosis remains unclear. This study revealed that expressions of AAT1/2 in myocardial tissue were significantly reduced in hyperthyroid rats, indicating that high thyroxine-induced myocardial fibrosis might be associated with the down-regulation of endogenous SO2. However, administration of exogenous SO2 donors up-regulated the AAT1/2. Zhao et al. [60] also reported a decrease in endogenous SO2 production in cardiomyocytes following high glucose stimulation. The results observed in this study suggested that administration of exogenous SO2 donors significantly improved the myocardial fibrosis in hyperthyroid rats, restricted the ERS-activated, and inhibited apoptosis in cardiomyocytes. These outcomes may be attributed to the antagonistic effect of SO2 on excessive activation of the Hippo pathway. In our in vitro experiments, we confirmed that inhibiting the activation of the Hippo pathway can ameliorate ERS in cardiomyocytes induced by elevated THs. Suggesting that SO2 may improve myocardial fibrosis in hyperthyroid rats by suppressing excessive ERS and Hippo pathway antagonizing cardiomyocyte apoptosis.
In conclusion, this study has demonstrated that SO2 can act as a novel endogenous gas signaling molecule to inhibit excessive ERS and cardiomyocyte apoptosis, thus ameliorating the myocardial fibrosis in hyperthyroid rats. The intrinsic mechanism of this effect may be related to antagonizing excessive activation of the Hippo pathway. Furthermore, this study has suggested that downregulation of endogenous SO2 may be involved in the hyperthyroidism-induced myocardial fibrosis, which can be mediated by the Hippo pathway-mediated apoptosis. In contrast, exogenous SO2 may improve myocardial fibrosis in hyperthyroid rats by inhibiting excessive ERS and antagonizing apoptosis of the overactivated Hippo pathway. These results offer new insights into the mechanism of hyperthyroidism-induced myocardial fibrosis and may aid in the identification of novel therapeutic targets and strategies for HTC. However, the underlying mechanism of endogenous SO2 destabilization that contributes to hyperthyroidism-induced myocardial fibrosis and the strategies for regulating endogenous SO2 homeostasis to inhibit myocardial fibrosis require further investigation.

ACKNOWLEDGMENTS

None.

Notes

FUNDING

This project was supported by the National Natural Science Foundation of China (No. 81870230 and No. 82074236), Clinical Major Projects of Hunan Provincial Health Commission (No. 20201913), the Natural Science Foundation of Hunan Province (No. 2021JJ70035, No. 2021JJ40499 and No. 2021JJ70116), the Hunan Provincial Graduate Student Research and Innovation Program (No. CX20231002).

CONFLICTS OF INTEREST

The authors declare no conflicts of interest.

REFERENCES

1. De Leo S, Lee SY, Braverman LE. 2016; Hyperthyroidism. Lancet. 388:906–918. DOI: 10.1016/S0140-6736(16)00278-6. PMID: 27038492.
crossref
2. Garmendia Madariaga A, Santos Palacios S, Guillén-Grima F, Galofré JC. 2014; The incidence and prevalence of thyroid dysfunction in Europe: a meta-analysis. J Clin Endocrinol Metab. 99:923–931. DOI: 10.1210/jc.2013-2409. PMID: 24423323.
crossref
3. Osuna PM, Udovcic M, Sharma MD. 2017; Hyperthyroidism and the heart. Methodist Debakey Cardiovasc J. 13:60–63. DOI: 10.14797/mdcj-13-2-60. PMID: 28740583. PMCID: PMC5512680.
4. Danzi S, Klein I. 2014; Thyroid disease and the cardiovascular system. Endocrinol Metab Clin North Am. 43:517–528. DOI: 10.1016/j.ecl.2014.02.005. PMID: 24891175.
crossref
5. Razvi S, Jabbar A, Pingitore A, Danzi S, Biondi B, Klein I, Peeters R, Zaman A, Iervasi G. 2018; Thyroid hormones and cardiovascular function and diseases. J Am Coll Cardiol. 71:1781–1796. DOI: 10.1016/j.jacc.2018.02.045. PMID: 29673469.
crossref
6. Bektur Aykanat NE, Şahin E, Kaçar S, Bağcı R, Karakaya Ş, Burukoğlu Dönmez D, Şahintürk V. 2021; Cardiac hypertrophy caused by hyperthyroidism in rats: the role of ATF-6 and TRPC1 channels. Can J Physiol Pharmacol. 99:1226–1233. DOI: 10.1139/cjpp-2021-0260. PMID: 34283935.
crossref
7. Wang YY, Jiao B, Guo WG, Che HL, Yu ZB. 2010; Excessive thyroxine enhances susceptibility to apoptosis and decreases contractility of cardiomyocytes. Mol Cell Endocrinol. 320:67–75. DOI: 10.1016/j.mce.2010.01.031. PMID: 20122986.
8. Klein I, Ojamaa K. 2001; Thyroid hormone and the cardiovascular system. N Engl J Med. 344:501–509. DOI: 10.1056/NEJM200102153440707. PMID: 11172193.
9. Freitas F, Estato V, Carvalho VF, Torres RC, Lessa MA, Tibiriçá E. 2013; Cardiac microvascular rarefaction in hyperthyroidism-induced left ventricle dysfunction. Microcirculation. 20:590–598. DOI: 10.1111/micc.12057. PMID: 23510303.
crossref
10. Woeber KA. 1992; Thyrotoxicosis and the heart. N Engl J Med. 327:94–98. DOI: 10.1056/NEJM199207093270206. PMID: 1603141.
11. Yue WS, Chong BH, Zhang XH, Liao SY, Jim MH, Kung AW, Tse HF, Siu CW. 2011; Hyperthyroidism-induced left ventricular diastolic dysfunction: implication in hyperthyroidism-related heart failure. Clin Endocrinol (Oxf). 74:636–643. DOI: 10.1111/j.1365-2265.2011.03981.x. PMID: 21470287.
12. Wang YY, Morimoto S, Du CK, Lu QW, Zhan DY, Tsutsumi T, Ide T, Miwa Y, Takahashi-Yanaga F, Sasaguri T. 2010; Up-regulation of type 2 iodothyronine deiodinase in dilated cardiomyopathy. Cardiovasc Res. 87:636–646. DOI: 10.1093/cvr/cvq133. PMID: 20453157.
13. Elmore S. 2007; Apoptosis: a review of programmed cell death. Toxicol Pathol. 35:495–516. DOI: 10.1080/01926230701320337. PMID: 17562483. PMCID: PMC2117903.
14. Kumar A, Sinha RA, Tiwari M, Singh R, Koji T, Manhas N, Rastogi L, Pal L, Shrivastava A, Sahu RP, Godbole MM. 2007; Hyperthyroidism induces apoptosis in rat liver through activation of death receptor-mediated pathways. J Hepatol. 46:888–898. DOI: 10.1016/j.jhep.2006.12.015. PMID: 17321637.
15. Hu H, Tian M, Ding C, Yu S. 2019; The C/EBP homologous protein (CHOP) transcription factor functions in endoplasmic reticulum stress-induced apoptosis and microbial infection. Front Immunol. 9:3083. DOI: 10.3389/fimmu.2018.03083. PMID: 30662442. PMCID: PMC6328441.
16. Groenendyk J, Agellon LB, Michalak M. 2013; Coping with endoplasmic reticulum stress in the cardiovascular system. Annu Rev Physiol. 75:49–67. DOI: 10.1146/annurev-physiol-030212-183707. PMID: 23020580.
17. Li Y, Guo Y, Tang J, Jiang J, Chen Z. 2015; New insights into the roles of CHOP-induced apoptosis in ER stress. Acta Biochim Biophys Sin (Shanghai). 47:146–147. Erratum for: Acta Biochim Biophys Sin (Shanghai). 2014;46:629-640. DOI: 10.1093/abbs/gmu048. PMID: 25016584.
18. Zhang J, Guo J, Yang N, Huang Y, Hu T, Rao C. 2022; Endoplasmic reticulum stress-mediated cell death in liver injury. Cell Death Dis. 13:1051. DOI: 10.1038/s41419-022-05444-x. PMID: 36535923. PMCID: PMC9763476.
19. He L, Yuan L, Yu W, Sun Y, Jiang D, Wang X, Feng X, Wang Z, Xu J, Yang R, Zhang W, Feng H, Chen HZ, Zeng YA, Hui L, Wu Q, Zhang Y, Zhang L. 2020; A regulation loop between YAP and NR4A1 balances cell proliferation and apoptosis. Cell Rep. 33:108284. DOI: 10.1016/j.celrep.2020.108284. PMID: 33086070.
20. Zhang J, Liu J, Gao S, Lin W, Gao P, Gao K, Zhang Y, Du K, Yang X, Wang W, Zhu R, Wang Y. 2019; Antiapoptosis and antifibrosis effects of Qishen granules on heart failure rats via Hippo pathway. Biomed Res Int. 2019:1642575. Erratum in: Biomed Res Int. 2020;2020: 8569251. DOI: 10.1155/2019/1642575. PMID: 31915683. PMCID: PMC6930732.
21. Wu H, Wei L, Fan F, Ji S, Zhang S, Geng J, Hong L, Fan X, Chen Q, Tian J, Jiang M, Sun X, Jin C, Yin ZY, Liu Q, Zhang J, Qin F, Lin KH, Yu JS, Deng X, et al. 2015; Integration of Hippo signalling and the unfolded protein response to restrain liver overgrowth and tumorigenesis. Nat Commun. 6:6239. DOI: 10.1038/ncomms7239. PMID: 25695629.
22. Ashokkumar R, Jamuna S, Sakeena Sadullah MS, Niranjali Devaraj S. 2018; Vitexin protects isoproterenol induced post myocardial injury by modulating hipposignaling and ER stress responses. Biochem Biophys Res Commun. 496:731–737. DOI: 10.1016/j.bbrc.2018.01.104. PMID: 29406244.
23. Ouyang H, Zhong J, Lu J, Zhong Y, Hu Y, Tan Y. 2019; Inhibitory effect of melatonin on Mst1 ameliorates myocarditis through attenuating ER stress and mitochondrial dysfunction. J Mol Histol. 50:405–415. DOI: 10.1007/s10735-019-09836-w. PMID: 31256303.
24. Huang Y, Tang C, Du J, Jin H. 2016; Endogenous sulfur dioxide: a new member of gasotransmitter family in the cardiovascular system. Oxid Med Cell Longev. 2016:8961951. DOI: 10.1155/2016/8961951. PMID: 26839635. PMCID: PMC4709694.
25. Wang XB, Cui H, Du JB. 2019; Potential therapeutic effect of SO₂ on fibrosis. Histol Histopathol. 34:1289–1297.
26. Hwang JH, Kang SY, Kang AN, Jung HW, Jung C, Jeong JH, Park YK. 2017; MOK, a pharmacopuncture medicine, regulates thyroid dysfunction in L-thyroxin-induced hyperthyroidism in rats through the regulation of oxidation and the TRPV1 ion channel. BMC Complement Altern Med. 17:535. DOI: 10.1186/s12906-017-2036-1. PMID: 29246135. PMCID: PMC5732465.
27. Yu W, Liu D, Liang C, Ochs T, Chen S, Chen S, Du S, Tang C, Huang Y, Du J, Jin H. 2016; Sulfur dioxide protects against collagen accumulation in pulmonary artery in association with downregulation of the transforming growth factor β1/Smad pathway in pulmonary hypertensive rats. J Am Heart Assoc. 5:e003910. DOI: 10.1161/JAHA.116.003910. PMID: 27792648. PMCID: PMC5121494.
28. Liu M, Liu S, Tan W, Tang F, Long J, Li Z, Liang B, Chu C, Yang J. 2017; Gaseous signalling molecule SO2 via HippoMST pathway to improve myocardial fibrosis of diabetic rats. Mol Med Rep. 16:8953–8963. DOI: 10.3892/mmr.2017.7714. PMID: 28990064. PMCID: PMC5779980.
crossref
29. Bao M, Hua X, Mo H, Sun Z, Xu B, Chen X, Xu M, Xu X, Song J. 2022; N-acetylcysteine, an ROS inhibitor, alleviates the pathophysiology of hyperthyroidism-induced cardiomyopathy via the ROS/Ca2+ pathway. Biomolecules. 12:1195. DOI: 10.3390/biom12091195. PMID: 36139036. PMCID: PMC9496499.
30. Lv B, Peng H, Qiu B, Zhang L, Ge M, Bu D, Li K, Yu X, Du J, Yang L, Tang C, Huang Y, Du J, Jin H. 2022; Sulphenylation of CypD at cysteine 104: a novel mechanism by which SO2 inhibits cardiomyocyte apoptosis. Front Cell Dev Biol. 9:784799. DOI: 10.3389/fcell.2021.784799. PMID: 35118072. PMCID: PMC8805922.
31. Zhang D, Wang X, Tian X, Zhang L, Yang G, Tao Y, Liang C, Li K, Yu X, Tang X, Tang C, Zhou J, Kong W, Du J, Huang Y, Jin H. 2018; The increased endogenous sulfur dioxide acts as a compensatory mechanism for the downregulated endogenous hydrogen sulfide pathway in the endothelial cell inflammation. Front Immunol. 9:882. DOI: 10.3389/fimmu.2018.00882. PMID: 29760703. PMCID: PMC5936987.
32. Grais IM, Sowers JR. 2014; Thyroid and the heart. Am J Med. 127:691–698. DOI: 10.1016/j.amjmed.2014.03.009. PMID: 24662620. PMCID: PMC4318631.
33. Fadel BM, Ellahham S, Ringel MD, Lindsay J Jr, Wartofsky L, Burman KD. 2000; Hyperthyroid heart disease. Clin Cardiol. 23:402–408. DOI: 10.1002/clc.4960230605. PMID: 10875028. PMCID: PMC6654928.
crossref
34. Khamis T, Alsemeh AE, Abdullah DM. 2022; Sacubitril/valsartan (LCZ696) ameliorates hyperthyroid-induced cardiac hypertrophy in male rats through modulation of miR-377, let-7 b, autophagy, and fibrotic signaling pathways. Sci Rep. 12:14654. DOI: 10.1038/s41598-022-18860-y. PMID: 36030321. PMCID: PMC9420135.
crossref
35. Kong P, Christia P, Frangogiannis NG. 2014; The pathogenesis of cardiac fibrosis. Cell Mol Life Sci. 71:549–574. DOI: 10.1007/s00018-013-1349-6. PMID: 23649149. PMCID: PMC3769482.
36. Frangogiannis NG. 2021; Cardiac fibrosis. Cardiovasc Res. 117:1450–1488. DOI: 10.1093/cvr/cvaa324. PMID: 33135058. PMCID: PMC8152700.
37. Goldblatt ZE, Cirka HA, Billiar KL. 2021; Mechanical regulation of apoptosis in the cardiovascular system. Ann Biomed Eng. 49:75–97. DOI: 10.1007/s10439-020-02659-x. PMID: 33169343. PMCID: PMC7775273.
crossref
38. Li YY, McTiernan CF, Feldman AM. 2000; Interplay of matrix metalloproteinases, tissue inhibitors of metalloproteinases and their regulators in cardiac matrix remodeling. Cardiovasc Res. 46:214–224. DOI: 10.1016/S0008-6363(00)00003-1. PMID: 10773225.
39. Foster CR, Daniel LL, Daniels CR, Dalal S, Singh M, Singh K. 2013; Deficiency of ataxia telangiectasia mutated kinase modulates cardiac remodeling following myocardial infarction: involvement in fibrosis and apoptosis. PLoS One. 8:e83513. DOI: 10.1371/journal.pone.0083513. PMID: 24358288. PMCID: PMC3865210.
crossref
40. Jiang M, Qi L, Li L, Li Y. 2020; The caspase-3/GSDME signal pathway as a switch between apoptosis and pyroptosis in cancer. Cell Death Discov. 6:112. DOI: 10.1038/s41420-020-00349-0. PMID: 33133646. PMCID: PMC7595122.
crossref
41. Boudreau MW, Peh J, Hergenrother PJ. 2019; Procaspase-3 overexpression in cancer: a paradoxical observation with therapeutic potential. ACS Chem Biol. 14:2335–2348. DOI: 10.1021/acschembio.9b00338. PMID: 31260254. PMCID: PMC6858495.
42. Czabotar PE, Lessene G, Strasser A, Adams JM. 2014; Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy. Nat Rev Mol Cell Biol. 15:49–63. DOI: 10.1038/nrm3722. PMID: 24355989.
crossref
43. Teixeira RB, Barboza TE, DE Araujo CC, Siqueira R, DE Castro AL, Bonetto JHP, DE Lima-Seolin BG, Carraro CC, Bello-Klein A, Singal PK, Araujo ASDR. 2018; Decreased PGC1-α levels and increased apoptotic protein signaling are associated with the maladaptive cardiac hypertrophy in hyperthyroidism. J Biosci. 43:887–895. DOI: 10.1007/s12038-018-9816-8. PMID: 30541949.
44. Liu MQ, Chen Z, Chen LX. 2016; Endoplasmic reticulum stress: a novel mechanism and therapeutic target for cardiovascular diseases. Acta Pharmacol Sin. 37:425–443. DOI: 10.1038/aps.2015.145. PMID: 26838072. PMCID: PMC4820795.
crossref
45. Bravo R, Gutierrez T, Paredes F, Gatica D, Rodriguez AE, Pedrozo Z, Chiong M, Parra V, Quest AF, Rothermel BA, Lavandero S. 2012; Endoplasmic reticulum: ER stress regulates mitochondrial bioenergetics. Int J Biochem Cell Biol. 44:16–20. DOI: 10.1016/j.biocel.2011.10.012. PMID: 22064245. PMCID: PMC4118286.
crossref
46. Shi Y, Wang S, Peng H, Lv Y, Li W, Cheng S, Liu J. 2019; Fibroblast growth factor 21 attenuates vascular calcification by alleviating endoplasmic reticulum stress mediated apoptosis in rats. Int J Biol Sci. 15:138–147. DOI: 10.7150/ijbs.28873. PMID: 30662354. PMCID: PMC6329919.
crossref
47. Lenna S, Trojanowska M. 2012; The role of endoplasmic reticulum stress and the unfolded protein response in fibrosis. Curr Opin Rheumatol. 24:663–668. DOI: 10.1097/BOR.0b013e3283588dbb. PMID: 22918530. PMCID: PMC3828639.
crossref
48. Luo T, Kim JK, Chen B, Abdel-Latif A, Kitakaze M, Yan L. 2015; Attenuation of ER stress prevents post-infarction-induced cardiac rupture and remodeling by modulating both cardiac apoptosis and fibrosis. Chem Biol Interact. 225:90–98. DOI: 10.1016/j.cbi.2014.10.032. PMID: 25450231. PMCID: PMC4684183.
49. Hamada H, Suzuki M, Yuasa S, Mimura N, Shinozuka N, Takada Y, Suzuki M, Nishino T, Nakaya H, Koseki H, Aoe T. 2004; Dilated cardiomyopathy caused by aberrant endoplasmic reticulum quality control in mutant KDEL receptor transgenic mice. Mol Cell Biol. 24:8007–8017. DOI: 10.1128/MCB.24.18.8007-8017.2004. PMID: 15340063. PMCID: PMC515036.
50. Meng S, Zhang W, Guan LJ, Muhali FS, Zhou JZ, Song RH, Xu J, Zhang JA. 2017; Proteomic analysis reveals aberrant expression of CALR and HSPA5 in thyroid tissues of Graves' disease. Clin Biochem. 50:40–45. DOI: 10.1016/j.clinbiochem.2016.08.014. PMID: 27566407.
crossref
51. Zhou Q, Li L, Zhao B, Guan KL. 2015; The hippo pathway in heart development, regeneration, and diseases. Circ Res. 116:1431–1447. DOI: 10.1161/CIRCRESAHA.116.303311. PMID: 25858067. PMCID: PMC4394208.
crossref
52. Ma S, Meng Z, Chen R, Guan KL. 2019; The hippo pathway: biology and pathophysiology. Annu Rev Biochem. 88:577–604. DOI: 10.1146/annurev-biochem-013118-111829. PMID: 30566373.
53. Deshpande A, Borlepawar A, Rosskopf A, Frank D, Frey N, Rangrez AY. 2021; SH3-binding glutamic acid rich-deficiency augments apoptosis in neonatal rat cardiomyocytes. Int J Mol Sci. 22:11042. DOI: 10.3390/ijms222011042. PMID: 34681711. PMCID: PMC8541172.
54. Francisco J, Zhang Y, Jeong JI, Mizushima W, Ikeda S, Ivessa A, Oka S, Zhai P, Tallquist MD, Del Re DP. 2020; Blockade of fibroblast YAP attenuates cardiac fibrosis and dysfunction through MRTF-A inhibition. JACC Basic Transl Sci. 5:931–945. Erratum in: JACC Basic Transl Sci. 2021;6:629. DOI: 10.1016/j.jacbts.2020.07.009. PMID: 33015415. PMCID: PMC7524792.
55. Zhang M, Zhang L, Hu J, Lin J, Wang T, Duan Y, Man W, Feng J, Sun L, Jia H, Li C, Zhang R, Wang H, Sun D. 2016; MST1 coordinately regulates autophagy and apoptosis in diabetic cardiomyopathy in mice. Diabetologia. 59:2435–2447. DOI: 10.1007/s00125-016-4070-9. PMID: 27510910.
56. Su D, Li Y, Guan L, Li Q, Shi C, Ma X, Song Y. 2021; Elevated MST1 leads to apoptosis via depletion of YAP1 in cardiomyocytes exposed to high glucose. Mol Med. 27:13. DOI: 10.1186/s10020-021-00267-6. PMID: 33568044. PMCID: PMC7874454.
crossref
57. Huang Y, Zhang H, Lv B, Tang C, Du J, Jin H. 2022; Sulfur dioxide: endogenous generation, biological effects, detection, and therapeutic potential. Antioxid Redox Signal. 36:256–274. DOI: 10.1089/ars.2021.0213. PMID: 34538110.
58. Liu J, Huang Y, Chen S, Tang C, Jin H, Du J. 2016; Role of endogenous sulfur dioxide in regulating vascular structural remodeling in hypertension. Oxid Med Cell Longev. 2016:4529060. DOI: 10.1155/2016/4529060. PMID: 27721913. PMCID: PMC5046050.
59. Jin H, Liu AD, Holmberg L, Zhao M, Chen S, Yang J, Sun Y, Chen S, Tang C, Du J. 2013; The role of sulfur dioxide in the regulation of mitochondrion-related cardiomyocyte apoptosis in rats with isopropylarterenol-induced myocardial injury. Int J Mol Sci. 14:10465–10482. DOI: 10.3390/ijms140510465. PMID: 23698774. PMCID: PMC3676849.
60. Zhao J, Wu Q, Yang T, Nie L, Liu S, Zhou J, Chen J, Jiang Z, Xiao T, Yang J, Chu C. 2022; Gaseous signal molecule SO2 regulates autophagy through PI3K/AKT pathway inhibits cardiomyocyte apoptosis and improves myocardial fibrosis in rats with type II diabetes. Korean J Physiol Pharmacol. 26:541–556. DOI: 10.4196/kjpp.2022.26.6.541. PMID: 36302628. PMCID: PMC9614393.

Fig. 1

SO2 contents and AAT1/2 expressions in myocardial tissues of rats in each group.

(A) ELISA assay for SO2 contents. (B–D) Western-Blot results of changes in AAT1/2 expressions in myocardial tissues of hyperthyroid rats (n = 3); *, #, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. (E–G) Western-Blot results of changes in AAT1/2 expressions in myocardial tissues of each group of H9c2 cardiomyocytes (n = 3); *, #, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. SO2, sulfur dioxide; AAT, aspartate aminotransferase; L-Thy, levothyroxine; HDX, hydrogen deuterium exchange.
kjpp-28-2-129-f1.tif
Fig. 2

Echocardiograms of rats in each group.

L-Thy, levothyroxine; SO2, sulfur dioxide; HDX, hydrogen deuterium exchange.
kjpp-28-2-129-f2.tif
Fig. 3

Effect of SO2 on myocardial fibrosis in hyperthyroid rats.

(A) Masson staining results of myocardial tissues in the control, L-Thy, L-Thy + SO2, and L-Thy + SO2 + HDX groups under 10 × 40x field of view (blue-stained part referred to the collagen fibers, and black arrows marked the collagen fibers), n = 3. Scale bar = 50 um. (B) Masson staining results of collagen volume fraction, *,#, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. (C–H) Western-Blot results of changes in expressions of Collagen I, Collagen III, MMP2, MMP3, and TIMP2 in myocardial tissues of rats in each group, n = 3. *, #, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. L-Thy, levothyroxine; SO2, sulfur dioxide; HDX, hydrogen deuterium exchange.
kjpp-28-2-129-f3.tif
Fig. 4

SO2 reduces hyperthyroidism apoptosis in rat cardiomyocytes.

(A–D) TEM results of myocardial tissues of rats in control group, L-Thy group, L-Thy + SO2 group, and L-Thy + SO2 + HDX group, respectively (red arrow: mitochondria; ×7,000). Scale bar = 0.5 μM. (E–I) Western-Blot results for changes in expressions of BAX, Bcl2, Caspase3, and Caspase9 in myocardial tissues (n = 3). *, #, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. TEM, transmission electron microscopy; L-Thy, levothyroxine; SO2, sulfur dioxide; HDX, hydrogen deuterium exchange.
kjpp-28-2-129-f4.tif
Fig. 5

SO2 inhibits hyperthyroidism-induced apoptosis in cardiomyocytes.

(A) Immunofluorescence observation for the expressions of BAX and Caspase3 in each group of H9c2 cardiomyocytes. Scale bar = 50 μM. (B–D) Western-Blot results of changes in expressions of BAX and Bcl2 in each group of H9c2 cardiomyocytes (n = 3). *, #, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. L-Thy, levothyroxine; SO2, sulfur dioxide; HDX, hydrogen deuterium exchange.
kjpp-28-2-129-f5.tif
Fig. 6

SO2 reduces endoplasmic reticulum stress in cardiomyocytes of hyperthyroid rats.

(A–D) Western-Blot results for changes in expressions CHOP, GRP78/BIP, and ERP72 in cardiomyocytes of each group (n = 3). *, #, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. (E–G) Western-Blot results for changes in expressions of CHOP and ERP72 in each group of H9c2 cardiomyocytes (n = 3). *, #, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. L-Thy, levothyroxine; SO2, sulfur dioxide; HDX, hydrogen deuterium exchange.
kjpp-28-2-129-f6.tif
Fig. 7

Reversing effect of SO2 in up-regulation of Hippo pathway expression induced by hyperthyroid.

(A–D) Western-Blot results for changes in expressions of MST1, LATS1, and P-YAP in myocardial tissues of rats (with n = 3); *, #, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. (E–H) Western-Blot results for changes in expressions of MST1, LATS1, and P-YAP in H9c2 cardiomyocytes in each group; n = 3. *,#, and & meant p < 0.05 vs. the values in the control, L-Thy, and L-Thy + SO2 groups, respectively. (I, J) Cells were transfected with siRNA negative control (si-NC) or si-YAP1 for 24 h, and the expression of YAP1 was measured by Western blot analysis (I) and quantitatively analyzed (J). n = 3. * meant p < 0.05 vs. the values in the si-NC groups, respectively. (K, L) Western-Blot results for changes in expressions of ERP72 in H9c2 cardiomyocytes in each group; n = 3. * and # meant p < 0.05 vs. the values in the control, and L-Thy groups, respectively. L-Thy, levothyroxine; SO2, sulfur dioxide; HDX, hydrogen deuterium exchange.
kjpp-28-2-129-f7.tif
Table 1
Comparison of TT3 and TT4 levels in the serum of rats in each group
Groups Control L-Thy L-Thy + SO2 L-Thy + SO2 + HDX
Number 10 8 9 8
TT3 (ng/l) 62.82 ± 3.64 89.15 ± 7.71* 82.53 ± 3.58* 88.12 ± 6.12*
TT4 (ng/l) 120.06 ± 9.99 172.78 ± 6.05* 163.77 ± 8.08* 176.98 ± 7.25*

Values are presented as mean ± standard deviation. TT3, total-triiodothyronine; TT4, total thyroxine; L-Thy, levothyroxine; SO2, sulfur dioxide; HDX, hydrogen deuterium exchange. *p < 0.05 vs. control.

Table 2
Comparison of BW, HW, and HW/BW values of rats in each group
Groups Number BW (g) HW (mg) HW/BW (mg/g)
Control 10 259.50 ± 1.74 955.80 ± 20.92 3.68 ± 0.62
L-Thy 8 202.50 ± 4.82* 875.25 ± 18.87* 4.34 ± 0.12*
L-Thy + SO2 9 231.11 ± 2.47# 937.78 ± 14.41# 4.06 ± 0.07
L-Thy + SO2 + HDX 8 209.38 ± 2.20& 882.13 ± 23.20 4.22 ± 0.15

Values are presented as mean ± standard deviation. BW, body weight; HW, heart weight; L-Thy, levothyroxine; SO2, sulfur dioxide; HDX, hydrogen deuterium exchange. *p < 0.05 vs. control; #p < 0.05 vs. L-Thy; &p < 0.05 vs. L-Thy + SO2.

Table 3
Levels of echocardiographic parameters in each group of rats
Groups Number HR (times/min) LVFS (%) LVEF (%)
Control 10 380.20 ± 15.33 42.30 ± 0.95 73.50 ± 0.43
L-Thy 8 520 ± 23.25* 31.82 ± 1.02* 59.53 ± 0.19*
L-Thy + SO2 9 420 ± 13.85# 39.52 ± 2.33# 69.40 ± 1.16#
L-Thy + SO2 + HDX 8 489 ± 19.77& 28.91 ± 2.14& 55.31 ± 3.18&

Values are presented as mean ± standard deviation. HR, heart rate; LVFS, left ventricular fractional shortening; LVEF, left ventricular ejection fraction; L-Thy, levothyroxine; SO2, sulfur dioxide; HDX, hydrogen deuterium exchange. *p < 0.05 vs. control group; #p < 0.05 vs. L-Thy group; &p < 0.05 vs. L-Thy + SO2 group.

TOOLS
Similar articles