Skip to main content

Glucose fluctuations aggravate cardiomyocyte apoptosis by enhancing the interaction between Txnip and Akt

Abstract

Background

Glucose fluctuations may be involved in the pathophysiological process of cardiomyocyte apoptosis, but the exact mechanism remains elusive. This study focused on exploring the mechanisms related to glucose fluctuation-induced cardiomyocyte apoptosis.

Methods

Diabetic rats established via an injection of streptozotocin were randomized to five groups: the controlled diabetic (CD) group, the uncontrolled diabetic (UD) group, the glucose fluctuated diabetic (GFD) group, the GFD group rats with the injection of 0.9% sodium chloride (NaCl) (GFD + NaCl) and the GFD group rats with the injection of N-acetyl-L-cysteine (NAC) (GFD + NAC). Twelve weeks later, cardiac function and apoptosis related protein expressions were tested. Proteomic analysis was performed to further analyze the differential protein expression pattern of CD and GFD.

Results

The left ventricular ejection fraction levels and fractional shortening levels were decreased in the GFD group, compared with those in the CD and UD groups. Positive cells tested by DAB-TUNEL were increased in the GFD group, compared with those in the CD group. The expression of Bcl-2 was decreased, but the expressions of Bax, cleaved caspase-3 and cleaved caspase-9 were increased in response to glucose fluctuations. Compared with CD, there were 527 upregulated and 152 downregulated proteins in GFD group. Txnip was one of the differentially expressed proteins related to oxidative stress response. The Txnip expression was increased in the GFD group, while the Akt phosphorylation level was decreased. The interaction between Txnip and Akt was enhanced when blood glucose fluctuated. Moreover, the application of NAC partially reversed glucose fluctuations-induced cardiomyocyte apoptosis.

Conclusions

Glucose fluctuations lead to cardiomyocyte apoptosis by up-regulating Txnip expression and enhancing Txnip-Akt interaction.

Peer Review reports

Introduction

Diabetes mellitus (DM) has been recognized as one of the major health issues, posing a serious threat to human health worldwide [1, 2]. An accumulating body of studies has shown that blood glucose fluctuations may be a greater risk of the development of cardiovascular diseases than persistent hyperglycemia [3, 4]. Glucose fluctuation has been confirmed in several recent studies to promote cardiomyocyte apoptosis more significantly compared with persistent hyperglycemia [5, 6]. However, the underlying mechanisms remain unknown.

Recently, several studies have reported that glucose fluctuations could lead to excessive oxidative stress [7]. Under normal physiological condition, there is a balance between oxidants and antioxidants. While in various pathological processes, overproduction of reactive oxygen species (ROS) contribute to excessive oxidative stress, resulting in cellular dysfunction, such as cell apoptosis [8]. Thioredoxin is an antioxidant protein and involved in maintaining the balance of oxidative stress [9, 10]. Thioredoxin-interacting protein (Txnip) negatively regulates thioredoxin, which functions as pro-oxidant and sensitizes myocytes to ROS-associated apoptosis [11]. The increase level of Txnip and excessive oxidative stress contribute to myocardium apoptosis in diabetes [12]. Moreover, several studies have reported that the level of Txnip is upregulated significantly in state of glucose fluctuations [3, 13].

Protein kinase B (PKB)/Akt is a serine–threonine kinase, and the Akt signaling plays important roles in cell growth, proliferation and apoptosis [14, 15]. The Akt signaling participates in regulating apoptosis through multiple mechanisms, including directly phosphorylating several anti-apoptotic factors and activating the transcriptional genes supporting cell survival [16]. The activation of Akt alleviates myocardium apoptosis in streptozotocin (STZ)-induced diabetes [17]. A previous study reported that Txnip might regulate the activity of Akt via direct interaction, leading to glucose-mediated metabolic stress [18]. However, whether Txnip regulates Akt activity after exposure to glucose fluctuations and the mechanism remains unknown.

In the present study, we explored the influences of glucose fluctuations on the process of cardiomyocyte apoptosis and the underlying mechanisms. We found that glucose fluctuations upregulated Txnip and enhanced the interaction between Txnip and Akt, which play a vital role in the promotion of cardiomyocyte apoptosis.

Materials and methods

Experimental animal models

Male Sprague–Dawley (SD) rats (6–8 weeks) were purchased from Changzhou Cavens Laboratory Animal Company in China. The rats were raised at a standard of care specific pathogen-free (SPF) conditions (temperature 22–24℃ and humidity 55–65%) in a light/dark cycle adapted to 12 h. After acclimatization, intraperitoneal injection of streptozotocin (STZ, 60 mg/kg; S0130, Sigma-Aldrich) was performed to establish type 1 diabetic rat model, as previously reported [19]. Blood glucose was measured in the tail vein of rats after one week. If the blood glucose was > 300.6 mg/dL, the rat was enrolled and then randomized to five groups. Rats in the controlled diabetic (CD) group (n = 8) were performed with a subcutaneous injection of long-acting insulin (20 IU/kg; Sanofi-Aventis) at 8 a.m. and 8 p.m. every day. Rats in the uncontrolled diabetic (UD) group (n = 8) were raised without any treatments. Rats in the glucose fluctuated diabetic (GFD) group (n = 8) were treated with a 24 h of starvation, and would receive regular insulin (0.5 IU/kg; Novo Nordisk) if their blood glucose levels were > 99 mg/dL. Then, rats were treated with a 24 h of consumption with adequate food. Rats in the GFD group rats with the injection of 0.9% sodium chloride (NaCl, GFD + NaCl, n = 3) were injected with 0.9% NaCl daily, while rats in the GFD group rats with the injection of N-acetyl-L-cysteine (NAC, GFD + NAC, n = 3) were intraperitoneally injected with NAC (150 mg/kg; A7250, Sigma-Aldrich). Twelve weeks later, after being exposed to adequate food for the last two days, rats were sacrificed with an intraperitoneally injection of sodium pentobarbital (60 mg/kg). Hearts were harvested and stored at -80℃. The ratios of heart weight/body weight (HW/BW) were calculated. Animal care and experimental protocol were approved by the Institutional Animal Care of the Wuxi People’s Hospital Affiliated to Nanjing Medical University (Approval number: IACUC-1712028). The study was performed in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals, and this study was carried out in compliance with the ARRIVE guidelines.

Tandem Mass Tags (TMT) labeled quantitative proteomics analysis

The quantitative proteomics analysis of rat cardiac ventricle tissues was performed in Luming Biotechnology Co., Ltd (Shanghai, China). Briefly, proteins were extracted from rat cardiac ventricle tissues and quantified with bicinchonininc acid kit (BCA). Then, 100 µg proteins were hydrolyzed using trypsin and labeled with TMT reagent. Then, separation of the labeled peptides was done on a reverse phase chromatography (Agilent Zorbax Extend-C18) with an Agilent 1100 HPLC and the identification of proteins was carried out with a Q-Exactive HF MS (ThermoFisher, U.S.A). Proteome Discoverer software (v2.4, Thermo Fisher Scientific) was used to search all of the Q Exactive raw data thoroughly against the sample protein database. The global FDR was set to 0.01, and for protein groups to be considered for quantification at least 2 peptides were required. The identity of proteins in a set was verified on the basis of the following two values: a Sequest HT score > 0 and unique peptide > 1. Differentially expressed or secreted proteins were identified on the basis of a FC > 1.2 or < 1/1.2 and p-value < 0.05. A GO enrichment analysis was carried out online (https://david.ncifcrf.gov/). Figures of cluster heatmap and GO enrichment analysis were made using bioinformatics (https://www.bioinformatics.com.cn/).

Echocardiography

Rats were anesthetized with isoflurane (2%) before the echocardiography (ie33, Philip) was performed. Images were obtained using M-mode echocardiography to calculate the left ventricular ejection fraction (LVEF), left ventricular fractional shortening (LVFS), left ventricular internal diameter at end-diastole (LVIDd), and left ventricular internal diameter at end-systole (LVIDs). All measurements were made by a sonologist who was blinded to the identity of the tracings.

ELISA of malondialdehyde (MDA)

Rat blood samples were collected from the inferior vena cava and the levels of MDA in rat plasma were measured using a MDA ELISA kit (ER1878, Wuhan Fine Biotech Co.).

Cell culture and treatment

H9C2 cells were purchased from Chinese Academy of Science, and were cultured in DMEM (11885-084, Gibco) which contains 10% fetal bovine serum (FBS) (12664025, Gibco). Primary cultures of neonatal rat ventricular cardiaomyocytes (NRVCMs) were obtained from 1 to 3 days old SD rats according to a procedure published previously [20, 21]. In brief, the hearts of neonatal rats were cut off atrial tissue and cut into small pieces, and then were digested with cardiaomyocyte isolation enzyme 1 and enzyme 2 (88281, Thermo). Cardiaomyocytes were collected and resuspended with DMEM (88287, Thermo) which contains 10% FBS (12664025, Gibco). H9C2 cells and NRVCMs were cultured in 37℃ with 5% CO2 incubator. Then, the cells were divided into three groups: the normal glucose (NG) group which was cultured with 5.5 mmol/L glucose, the high glucose (HG) group which was cultured with 25 mmol/L glucose, and the glucose fluctuation (GF) group which was cultured with glucose alternating between 5.5 and 25 mmol/L every 12 h for 72 h.

Glucose monitoring

The blood glucose levels of all diabetic rats were measured with a blood glucose meter (Roche) daily.

DAB-TUNLE

Hearts were fixed in 4% paraformaldehyde, and then dehydrated in graded ethanol. After soaking xylene and paraffin imbedding, heart issues were cut into 5 μm slices, followed by dewaxing and rehydration. Firstly, the paraffin slides were deparaffinized and rehydrated, and then endogenous peroxidase was blocked. After equilibrium at room temperature, the slides were treated with the TUNEL kit (G1507, Servicebio). DAB chromogenic reagent to marked tissue followed by counterstaining with hematoxylin. The images were captured under a light microscope (Leica Microsystems). The positive apoptosis cells developed by DAB reagent have brown nucleus.

Confocal microscopy

H9C2 cells and NRVCMs were respectively incubated over night at 4℃ with the mixture of anti-Txnip antibody (ab210826, Abcam, raised in mouse) and anti-AKT antibody (4691, Cell Signaling Technology, raised in rabbit). Then, cells were incubated with the mixture of Alexa Fluor 594 AffiniPure Donkey Anti-Mouse IgG (H + L) (34112ES60, Yeasen Biotechnology), Alexa Fluor 488 AffiniPure Donkey Anti-Rabbit IgG (H + L) (34206ES60, Yeasen Biotechnology) and together with DAPI (P0131, Beyotime).

Western blot

Heart tissues were homogenized and lysed in lysis buffer (89900, Pierce), which contains protease and phosphatase inhibitors (04693159001, Roche). After being added with protein SDS-PAGE loading buffer (9173, Takara), sample proteins were transferred onto polyvinylidene difluoride membranes (IPVH00010, Immobilon). Then, polyvinylidene difluoride membranes were incubated with the specific primary antibodies to α-tubulin (2144, Cell Signaling Technology), β-actin (4970, Cell Signaling Technology), Bax (2772, Cell Signaling Technology), Bcl-2 (sc-7382, Santa Cruz Biotechnology), cleaved caspase-3 (9661, Cell Signaling Technology), caspase-9 (10380-1-AP, Proteintech), NADPH oxidase 4 (Nox4, ab154244, Abcam), Txnip (14715, Cell Signaling Technology), Akt (4691, Cell Signaling Technology), p-AKT (4060, Cell Signaling Technology). The optical density of immunoblot bands were analyzed using ImageJ software. Densities were normalized to control treatment and relative folds were normalized to α-tubulin or β-actin.

Co-immunoprecipitation

H9C2 cells were lysed by NP-40 Lysis Buffer (P0013F, Beyotime). Primary antibodies and protein A/G magnetic beads (HY-K0202, MCE) were mixed together with lysates for 12 h in 4℃. Magnetic beads were washed by lysis buffer for 3 times, followed by boiled with loading buffer (9173, Takara). The next procedure was western blot as described above. Then, polyvinylidene difluoride membranes were incubated with the specific primary antibodies to Txnip (ab210826, Abcam), Akt (4691, Cell Signaling Technology) and β-actin (4970, Cell Signaling Technology).

Statistical analysis

Data were presented as the mean ± SEM. Student’s t-test was used to compare data between two groups. One-way analysis of variance (ANOVA) followed by Tukey’s multiple-comparison tests was used to compare data from multiple groups. SPSS 22.0 was used for analyzing statistical data. Statistical significance was defined as a p value of < 0.05.

Results

Glucose fluctuations aggravate the LVEF in diabetic rats

Figure 1A shows the blood glucose levels in the CD group (around 90 mg/dL), UD group (higher than 450 mg/dL) and GFD group (between 90 mg/dL and 450 mg/dL). Figure 1B shows the body weights of rats in the CD, UD and GFD groups. The fluctuated weight in the GFD group was resulted from repeated starvation. Figure 1C shows the representative echocardiographic images. After twelve weeks, heart-to-body weight ratios among the three groups show no significant differences (Fig. 1D). As shown in Fig. 1E-H, the LVEF level in the UD group (86.74 ± 1.25%) was lower than that in the CD group (91.74 ± 0.94%), and was to a greater extent in the GFD group (81.93 ± 2.00%). While the FS level in the UD group (51.20 ± 1.55%) decreased compared with the CD group (58.81 ± 1.81%), which was more pronounced than that in the GFD group (45.90 ± 2.12%). The LVIDd data showed no significant differences among the three groups. However, the LVIDs data was increased in the UD group compared with the CD group, which was more pronounced in the GFD group. These results suggested that left ventricular systolic function was impaired in the GFD group rats.

Fig. 1
figure 1

Effects of glucose fluctuations on cardiac function (A) Blood glucose levels in the CD, UD and GFD groups (n = 8 per group). (B) Body weights in the CD, UD and GFD groups (n = 8 per group). (C-H) Representative echocardiographic images, heart weight/body weight (HW/BW), left ventricular ejection fraction (% EF), left ventricular fractional shortening (% FS), left ventricular internal diameter at end-diastole (LVIDd), and left ventricular internal diameter at end-systole (LVIDs) examined by echocardiography in the CD, UD and GFD groups (n = 8 per group). N.S. No significant difference

Glucose fluctuations promote cardiomyocyte apoptosis

As shown in Fig. 2A, compared with the CD group, the positive apoptosis cells tested by DAB-TUNEL were increased in the GFD group. There was a decreasing trend of Bcl-2 protein expression, while there was an increasing trend of Bax, cleaved caspase-3 and cleaved caspase-9 protein expressions in the UD group, and these trends showed more pronounced in the GFD group (Fig. 2B). Figure 2C-E show that the protein level of Bcl-2 was downregulated, while the protein levels of Bax and cleaved caspase-3 were upregulated in the GF group, compared with these changes in the NG and HG groups. These results indicated that glucose fluctuations could lead to cardiomyocyte apoptosis.

Fig. 2
figure 2

Glucose fluctuations promote cardiomyocyte apoptosis in diabetic rats (A) Representative DAB-TUNEL staining images of heart tissues in the CD and GFD groups. (B) The protein expressions of Bcl-2, Bax, cleaved caspase-3 and cleaved caspase-9 in rat hearts of the three groups (n = 3 per group). (C-E) The protein expressions of Bcl-2 (n = 3), Bax (n = 4) and cleaved caspase-3 (n = 4) in H9C2 cells of the NG, HG and GF groups

Glucose fluctuations upregulate the expression of Txnip

Proteomic analysis was done to further analyze the differential protein expression pattern of the CD and GFD groups. Compared with CD, there were 527 upregulated and 152 downregulated proteins in GFD group. Gene Ontology analysis showed that differentially expressed proteins were involved in several biological processes such as RNA splicing, metabolism, response to oxidative stress and so on (Fig. 3A). Oxidative stress was associated with cell death. The differentially expressed proteins related to oxidative stress response were presented in Fig. 3B. Moreover, the MDA level was significantly increased in the GFD group than that in the CD group (Fig. 3C). The protein expressions of Nox4 and Txnip were higher in the GFD group than that in the CD and UD groups, and the Akt phosphorylation level was decreased due to glucose fluctuations (Fig. 3D). Moreover, the protein expression of Txnip was increased and the Akt phosphorylation level was decreased in H9C2 cells which exposed to the fluctuated glucose concentrations (Fig. 3E-F).

Fig. 3
figure 3

Glucose fluctuations lead to the upregulation of Txnip (A) Gene Ontology analysis-biological process of differentially expressed proteins (n = 3 per group). (B) The heatmap of differentially expressed proteins involved in response to oxidative stress between CD and GFD (n = 3 per group). (C) The MDA levels in rat plasma of the CD and GFD groups (n = 5 per group). (D)The protein expressions of Nox4, Txnip and the Akt phosphorylation level in rat hearts of the three groups (n = 3 per group). (E-F) The protein expression of Txnip (n = 3) and the Akt phosphorylation level (n = 4) in H9C2 cells of the NG, HG and GF groups

Glucose fluctuations could enhance the interaction between Txnip and Akt

To identify the interaction between Txnip and Akt, we performed co-immunoprecipitation (IP) experiments in H9C2 cells. As shown in Fig. 4A, glucose fluctuations enhanced the Txnip-Akt interaction. Immunofluorescence results show that Txnip and Akt were distributed in the cytoplasm and overlapped, the Txnip-Akt interaction was observed both in the H9C2 cells and NRVCMs of the GF group (Fig. 4B-C).

Fig. 4
figure 4

Glucose fluctuations enhance the Txnip-Akt interaction (A) Co-immunoprecipitation (Co-IP) of Txnip and Akt in H9C2 cells of the NG, HG and GF groups (n = 3 per group). IgG represents a control antibody used for IPs. (B) Immunofluorescence staining of intracellular Txnip–Akt interaction examined by confocal microscopy in H9C2 cells of the GF groups. The cells were labeled with anti-Txnip (red), anti-Akt (green), and DAPI (blue) (n = 3 per group). (C) Immunofluorescence staining of intracellular Txnip–Akt interaction examined by confocal microscopy in NRVCMs of the GF groups. The cells were labeled with anti-Txnip (red), anti-Akt (green), and DAPI (blue) (n = 3 per group)

NAC treatment reversed myocardium apoptosis in glucose fluctuation

The LVEF and FS levels were increased in the GFD + NAC group, compared with those in the GFD + NaCl group (Fig. 5A-C). As shown in Fig. 5D, the protein expression of Bcl-2 was increased, while the protein expressions of Bax and cleaved caspase 3 were decreased in the GFD + NAC group. Additionally, Txnip expression was significantly decreased, and the AKT phosphorylation level was increased in the GFD + NAC group, compared with those in the GFD + NaCl group (Fig. 5E-F). These results suggested that NAC treatment partially reversed glucose fluctuations-induced left ventricular systolic dysfunction and cardiomyocyte apoptosis.

Fig. 5
figure 5

NAC treatment declined glucose fluctuation-induced cardiomyocyte apoptosis (A-C) Representative echocardiographic images, the EF and FS of rats in the GFD + NaCl and GFD + NAC groups (n = 3 per group). (D) The protein expressions of Bcl-2 (n = 6), Bax (n = 3) and cleaved caspase-3 (n = 6) in rat hearts of the two groups. (E-F) The protein expression of Txnip and the Akt phosphorylation level in rat hearts of the two groups (n = 3 per group)

Discussion

Recently, an accumulating number of studies have reported that glucose fluctuation is an important contributor to cardiomyocyte damage in DM [6, 22,23,24]. Our study was focused on the influence and underlying mechanisms of glucose fluctuations in development of myocardium apoptosis. The most important findings in this study are: (1) Glucose fluctuations could result in the upregulation of Txnip and the enhanced the interaction between Txnip and Akt, leading to cardiomyocyte apoptosis; (2) The application of NAC partially reversed cardiomyocyte apoptosis in diabetic rats with fluctuated blood glucose.

DM is regarded as a vital contributor to the development of cardiac dysfunction, including left ventricle diastolic and systolic dysfunction [25]. Moreover, glucose fluctuation has been reported to have more deleterious effects on cardiac dysfunction than persistent hyperglycemia. We found that the LVEF and LVFS levels were reduced to a greater extent in the GFD rats than those in the CD and UD rats, indicating that glucose fluctuations promoted the development of left ventricle systolic dysfunction. The diabetes-associated cardiac remolding is responsible for cardiac dysfunction, including myocardium hypertrophy, myocardium apoptosis and interstitial fibrosis [26, 27]. Hyperglycemia has been considered as a pathophysiological trigger of myocardium apoptosis in the diabetic heart. Several studies have reported that glucose fluctuations aggravate myocardium apoptosis than persistent hyperglycemia [5, 6]. Caspase-3 is a key enzyme in the execution of apoptosis, which belongs to endoprotease and mediates apoptosis by cleaved caspase-3. Bcl-2 and Bax regulate apoptosis together to keep cell homeostasis [28]. In our study, we found that the protein expressions of Bax, cleaved caspase-3 and cleaved caspase-9 were increased, while the protein expression of Bcl-2 was decreased in the GFD group, compared with the CD and UD groups.

Proteomic analysis results showed that Compared with CD, there were 527 upregulated and 152 downregulated proteins in GFD group. Gene Ontology analysis showed that differentially expressed proteins were involved in several biological processes such as RNA splicing, metabolism, response to oxidative stress and so on. Oxidative stress in diabetes is involved in the process of myocardium apoptosis [29, 30]. MDA has been confirmed as a biomarker of lipid peroxidation caused by ROS overproduction, thus we tested the MDA level in rat plasma and MDA level was increased in GFD group. Moreover, NADPH oxidases are the major sources of ROS production, Nox4 protein expression was higher in the diabetic rats with glucose fluctuations, indicating that glucose fluctuations could contribute to excessive oxidative stress. Oxidative stress is the imbalance between oxidation and anti-oxidation and tends to oxidize. Txnip and thioredoxin interact with each other directly and maintain the oxidative balance in cells to a certain extent. Txnip mediates redox homeostasis by increasing ROS production, besides ROS promotes the activation of Txnip pathway [31]. Our proteomic analysis results showed that Txnip was one of the differentially expressed proteins related to oxidative stress response. In this study, we found that the Txnip level was increased significantly in the GFD group.

Akt is involved in various kinds of cellular processes including cell survival, growth and metabolism. Akt signaling regulates cell apoptosis in multiple ways, such as directly phosphorylating several anti-apoptotic factors (caspase and Bcl2 associated agonist of cell death) [32]. In diabetes, Akt activation is reduced due to hyperglycemia, leading to the increase of cardiomyocyte apoptosis [17]. Our data showed that the Akt phosphorylation level declined significantly in the GFD group, indicating that Akt is involved in glucose fluctuation-induced cardiomyocyte apoptosis. The structural of Akt comprises of an N-terminal pleckstrin homology (PH) domain, a kinase domain (KD), and a C-terminal hydrophobic regulatory region. In an inactive state, PH domain binds with KD through an intramolecular interaction. Phosphoinositide 3-kinase activates Akt through interacting with the PH domain, thereby dislodging the PH domain from the KD. Moreover, PH domain can interact with other proteins, modulating the structure of Akt to allow for its activation [14]. It has been reported that Txnip negatively regulates the activity of Akt under glucose stress and oxidative stress through directly interacting with the PH domain [18]. In our study, we found that the Txnip-Akt interaction was enhanced in response to glucose fluctuations. We suppose that the increase level of Txnip induced by glucose fluctuations negatively regulated Akt activity through their direct interaction.

NAC, acting as an inhibitor of ROS, can eliminate ROS in oxidative stress and inhibit the activation of Txnip pathway [33, 34]. In this study, our results showed that NAC treatment could partially reversed glucose fluctuations-induced left ventricle systolic dysfunction and cardiomyocyte apoptosis.

Conclusions

In sum, our study demonstrated that glucose fluctuations up-regulate the Txnip expression and enhance the Txnip-Akt interaction, aggravating cardiomyocyte apoptosis. While the NAC treatment can reverse these changes in response to glucose fluctuation and function as a potential therapeutic option, suggesting a new therapy target to slow or reverse the cardiovascular complications of diabetes in future.

Data availability

The datasets generated and analyzed during the current study are not publicly available due the principle of funding confidentiality but are available from the corresponding author upon reasonable request.

Abbreviations

BCA:

bicinchonininc acid

BW:

body weight

CD:

controlled diabetic

DAB:

diaminobenzidine

DM:

diabetes mellitus

FBS:

fetal bovine serum

GF:

glucose fluctuation

GFD:

glucose fluctuated diabetic

HG:

high glucose

HW:

heart weight

IACUC:

Institutional Animal Care and Use Committee

IOD:

integrated optical density

IP:

immunoprecipitation

KD:

kinase domain

LVEF:

left ventricular ejection fraction

LVFS:

left ventricular fractional shortening

LVIDd:

left ventricular internal diameter at end-diastole

LVIDs:

left ventricular internal diameter at end-systole

NAC:

N-acetyl-L-cysteine

NaCl:

sodium chloride

NG:

normal glucose

NRVCMs:

neonatal rat ventricular cardiaomyocytes

PH:

pleckstrin homology

PKB:

protein kinase B

ROS:

reactive oxygen species

SD:

Sprague–Dawley

SPF:

specific pathogen-free

STZ:

streptozotocin

TMT:

Tandem Mass Tags

Txnip:

thioredoxin-interacting protein

UD:

uncontrolled diabetic

References

  1. Ruze R, Liu T, Zou X, Song J, Chen Y, Xu R, et al. Obesity and type 2 diabetes mellitus: connections in epidemiology, pathogenesis, and treatments. Front Endocrinol (Lausanne). 2023;14:1161521. https://doi.org/10.3389/fendo.2023.1161521. eCollection 2023.

    Article  PubMed  Google Scholar 

  2. Cheng F, Carroll L, Joglekar MV, Januszewski AS, Wong KK, Hardikar AA, et al. Diabetes, metabolic disease, and telomere length. Lancet Diabetes Endocrinol. 2021;9:117–26. https://doi.org/10.1016/S2213-8587(20)30365-X.

    Article  CAS  PubMed  Google Scholar 

  3. Saito S, Teshima Y, Fukui A, Kondo H, Nishio S, Nakagawa M, et al. Glucose fluctuations increase the incidence of atrial fibrillation in diabetic rats. Cardiovasc Res. 2014;104:5–14. https://doi.org/10.1093/cvr/cvu176.

    Article  CAS  PubMed  Google Scholar 

  4. Zhang ZY, Miao LF, Qian LL, Wang N, Qi MM, Zhang YM, et al. Molecular mechanisms of glucose fluctuations on Diabetic complications. Front Endocrinol (Lausanne). 2019;10:640. https://doi.org/10.3389/fendo.2019.00640.

    Article  PubMed  Google Scholar 

  5. Zhang W, Zhao S, Li Y, Peng G, Han P. Acute blood glucose fluctuation induces myocardial apoptosis through oxidative stress and nuclear factor-kB activation. Cardiology. 2013;124:11–7. https://doi.org/10.1159/000345436.

    Article  CAS  PubMed  Google Scholar 

  6. Wu LD, Liu Y, Li F, Chen JY, Zhang J, Qian LL, et al. Glucose fluctuation promotes cardiomyocyte apoptosis by triggering endoplasmic reticulum (ER) stress signaling pathway in vivo and in vitro. Bioengineered. 2022;13:13739–51. https://doi.org/10.1080/21655979.2022.2080413.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Klimontov VV, Saik OV, Korbut AI. Glucose variability: how does it work? Int J Mol Sci. 2021;22(15):7783. https://doi.org/10.3390/ijms22157783.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhang P, Li T, Wu X, Nice EC, Huang C, Zhang Y. Oxidative stress and diabetes: antioxidative strategies. Front Med. 2020;14:583–600. https://doi.org/10.1007/s11684-019-0729-1. Epub 2020 Apr 4.

    Article  PubMed  Google Scholar 

  9. Ji L, Wang Q, Huang F, An T, Guo F, Zhao Y et al. FOXO1 Overexpression Attenuates Tubulointerstitial Fibrosis and Apoptosis in Diabetic Kidneys by Ameliorating Oxidative Injury via TXNIP-TRX. Oxid Med Cell Longev. 2019; 2019: 3286928. https://doi.org/10.1155/2019/3286928

  10. Alhawiti NM, Al Mahri S, Aziz MA, Malik SS, Mohammad S. TXNIP in metabolic regulation: physiological role and therapeutic Outlook. Curr Drug Targets. 2017;18:1095–103. https://doi.org/10.2174/1389450118666170130145514.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Pan M, Zhang F, Qu K, Liu C, Zhang J. TXNIP: A Double-Edged Sword in Disease and Therapeutic Outlook. Oxid Med Cell Longev. 2022; 2022:7805115. https://doi.org/10.1155/2022/7805115. eCollection 2022.

  12. Wei H, Bu R, Yang Q, Jia J, Li T, Wang Q et al. Exendin-4 Protects against Hyperglycemia-Induced Cardiomyocyte Pyroptosis via the AMPK-TXNIP Pathway. J Diabetes Res. 2019; 2019: 8905917. https://doi.org/10.1155/2019/8905917

  13. Wang H, Deng J, Chen L, Ding K, Wang Y. Acute glucose fluctuation induces inflammation and neurons apoptosis in hippocampal tissues of diabetic rats. J Cell Biochem. 2019. https://doi.org/10.1002/jcb.29523.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Ghafouri-Fard S, Khanbabapour Sasi A, Hussen BM, Shoorei H, Siddiq A, Taheri M, et al. Interplay between PI3K/AKT pathway and heart disorders. Mol Biol Rep. 2022;49(10):9767–81. https://doi.org/10.1007/s11033-022-07468-0. Epub 2022 May 2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Manning BD, Toker A. AKT/PKB signaling: navigating the network. Cell. 2017;169:381–405. https://doi.org/10.1016/j.cell.2017.04.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kma L, Baruah TJ. The interplay of ROS and the PI3K/Akt pathway in autophagy regulation. Biotechnol Appl Biochem. 2022;69(1):248–64. https://doi.org/10.1002/bab.2104. Epub 2021 Jan 28.

    Article  CAS  PubMed  Google Scholar 

  17. Sedaghat M, Choobineh S, Ravasi AA. Taurine with combined aerobic and resistance exercise training alleviates myocardium apoptosis in STZ-induced diabetes rats via akt signaling pathway. Life Sci. 2020;258:118225. https://doi.org/10.1016/j.lfs.2020.118225.

    Article  CAS  PubMed  Google Scholar 

  18. Huy H, Song HY, Kim MJ, Kim WS, Kim DO, Byun JE, et al. TXNIP regulates AKT-mediated cellular senescence by direct interaction under glucose-mediated metabolic stress. Aging Cell. 2018;17:e12836. https://doi.org/10.1111/acel.12836.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang ZY, Dang SP, Li SS, Liu Y, Qi MM, Wang N, et al. Glucose fluctuations aggravate myocardial fibrosis via the Nuclear Factor-κB-Mediated nucleotide-binding oligomerization domain-like receptor protein 3 Inflammasome activation. Front Cardiovasc Med. 2022;9:748183. https://doi.org/10.3389/fcvm.2022.748183. eCollection 2022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fu C, Lizhao J, Luo Z, Wang T, Grapperhaus CA, Ding X, et al. Active uptake of hydrophilic copper complex cu(ii)-TETA in primary cultures of neonatal rat cardiomyocytes. Metallomics. 2019;11:565–75. https://doi.org/10.1039/c8mt00277k.

    Article  CAS  PubMed  Google Scholar 

  21. Watkins SJ, Borthwick GM, Arthur HM. The H9C2 cell line and primary neonatal cardiomyocyte cells show similar hypertrophic responses in vitro. Vitro Cell Dev Biol Anim. 2011;47:125–31. https://doi.org/10.1007/s11626-010-9368-1.

    Article  CAS  Google Scholar 

  22. Chai Q, Miao J, Liu M, Zhang Z, Meng Z, Wu W. SGLT1 knockdown prevents glucose fluctuation-induced apoptosis of cardiomyocytes through attenuating oxidative stress and mitochondrial dysfunction. Biochem Cell Biol. 2020. https://doi.org/10.1139/bcb-2020-0491.

    Article  PubMed  Google Scholar 

  23. Saito S, Thuc LC, Teshima Y, Nakada C, Nishio S, Kondo H, et al. Glucose fluctuations aggravate Cardiac susceptibility to Ischemia/Reperfusion Injury by modulating MicroRNAs expression. Circ J. 2016;80:186–95. https://doi.org/10.1253/circj.CJ-14-1218.

    Article  CAS  PubMed  Google Scholar 

  24. Mita T, Katakami N, Okada Y, Yoshii H, Osonoi T, Nishida K, et al. Protocol of a prospective observational study on the relationship between glucose fluctuation and Cardiovascular events in patients with type 2 diabetes. Diabetes Ther. 2019;10:1565–75. https://doi.org/10.1007/s13300-019-0665-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. De Blasio MJ, Huynh N, Deo M, Dubrana LE, Walsh J, Willis A, et al. Defining the Progression of Diabetic Cardiomyopathy in a mouse model of type 1 diabetes. Front Physiol. 2020;11:124. https://doi.org/10.3389/fphys.2020.00124.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Ritchie RH, Abel ED. Basic mechanisms of Diabetic Heart Disease. Circ Res. 2020;126(11):1501–25. https://doi.org/10.1161/CIRCRESAHA.120.315913. Epub 2020 May 21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Rajesh M, Mukhopadhyay P, Batkai S, Patel V, Saito K, Matsumoto S, et al. Cannabidiol attenuates cardiac dysfunction, oxidative stress, fibrosis, and inflammatory and cell death signaling pathways in diabetic cardiomyopathy. J Am Coll Cardiol. 2010;56:2115–25. https://doi.org/10.1016/j.jacc.2010.07.033.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. D’Arcy MS. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 2019;43:582–92. https://doi.org/10.1002/cbin.11137.

    Article  PubMed  Google Scholar 

  29. Luo J, Yan D, Li S, Liu S, Zeng F, Cheung CW, et al. Allopurinol reduces oxidative stress and activates Nrf2/p62 to attenuate diabetic cardiomyopathy in rats. J Cell Mol Med. 2020;24:1760–73. https://doi.org/10.1111/jcmm.14870.

    Article  CAS  PubMed  Google Scholar 

  30. Yu M, Shan X, Liu Y, Zhu J, Cao Q, Yang F, et al. RNA-Seq analysis and functional characterization revealed lncRNA NONRATT007560.2 regulated cardiomyocytes oxidative stress and apoptosis induced by high glucose. J Cell Biochem. 2019;120:18278–87. https://doi.org/10.1002/jcb.29134.

    Article  CAS  PubMed  Google Scholar 

  31. Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010;11(2):136–40. https://doi.org/10.1038/ni.1831.

    Article  CAS  PubMed  Google Scholar 

  32. Hers I, Vincent EE, Tavare JM. Akt signalling in health and disease. Cell Signal. 2011;23:1515–27. https://doi.org/10.1016/j.cellsig.2011.05.004.

    Article  CAS  PubMed  Google Scholar 

  33. Devi TS, Hosoya K, Terasaki T, Singh LP. Critical role of TXNIP in oxidative stress, DNA damage and retinal pericyte apoptosis under high glucose: implications for diabetic retinopathy. Exp Cell Res. 2013;319:1001–12. https://doi.org/10.1016/j.yexcr.2013.01.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhang W, Tang R, Ba G, Li M, Lin H. Anti-allergic and anti-inflammatory effects of resveratrol via inhibiting TXNIP-oxidative stress pathway in a mouse model of allergic rhinitis. World Allergy Organ J. 2020;13:100473. https://doi.org/10.1016/j.waojou.2020.100473.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the National Natural Science Foundation of China (82370342, 82200357), the Natural Science Foundation of Jiangsu Province (BK20231145), Research Foundation from Wuxi Health Commission for the Youth (Q202105), General Program of Wuxi Medical Center, Nanjing Medical University (WMCG202314).

Author information

Authors and Affiliations

Authors

Contributions

Z-YZ, LP and R-XW were involved in the experiment design. Z-YZ, LP, S-PD, NW, S-YZ, F-L, L-DW, LZ, H-HL and NZ performed the experiments. Y-JY and L-LQ analyzed the data. Z-YZ wrote the manuscript. L-LQ, TL and R-XW edited the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Ling-Ling Qian, Tong Liu or Ru-Xing Wang.

Ethics declarations

Ethics approval and consent to participate

Animal care and experimental protocol were approved by the Institutional Animal Care of the Wuxi People’s Hospital Affiliated to Nanjing Medical University (Approval number: IACUC-1712028). The study was performed in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals, and this study was carried out in compliance with the ARRIVE guidelines.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, ZY., Pan, L., Dang, S. et al. Glucose fluctuations aggravate cardiomyocyte apoptosis by enhancing the interaction between Txnip and Akt. BMC Cardiovasc Disord 24, 470 (2024). https://doi.org/10.1186/s12872-024-04134-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12872-024-04134-0

Keywords