Jujuboside a improved energy metabolism in senescent H9c2 cells injured by ischemia, hypoxia, and reperfusion through the CD38/Silent mating type information regulation 2 homolog 3 signaling pathway


  Table of Contents ORIGINAL ARTICLE Year : 2023  |  Volume : 9  |  Issue : 3  |  Page : 322-329

Jujuboside a improved energy metabolism in senescent H9c2 cells injured by ischemia, hypoxia, and reperfusion through the CD38/Silent mating type information regulation 2 homolog 3 signaling pathway

Yi-Ran Hu1, Hui-Yan Qu2, Jia-Ying Guo1, Tao Yang3, Hua Zhou3
1 Shuguang Hospital, Shanghai University of Traditional Chinese Medicine; Institute of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
2 Institute of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
3 Institute of Cardiovascular Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine; Institute of Cardiovascular Disease, Shanghai Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shanghai, China

Date of Submission07-Nov-2021Date of Acceptance28-Mar-2022Date of Web Publication29-Mar-2023

Correspondence Address:
Dr. Hua Zhou
No. 528, Zhangheng Road, Pudong New Area, Shanghai
China
Login to access the Email id

Source of Support: None, Conflict of Interest: None

DOI: 10.4103/2311-8571.372731

Rights and Permissions


Objective: This study explored the myocardial protection role of Jujuboside A through an ischemia–hypoxia–reperfusion (IHR) model. Materials and Methods: H9c2 cells were induced by D-galactose (D-gal) and IHR to establish an aging and IHR model. There are four groups of experiments: Control, IHR, D-gal + IHR, and D-gal + IHR + Jujuboside A. Cells viability, Adenosine triphosphate (ATP), reactive oxygen species (ROS), nicotinamide adenine dinucleotide (NAD+), nicotinamide adenine dinucleotide hydride (NADH) content, and NAD+/NADH ratio were detected using biochemical methods. Inflammatory cytokines level was detected by enzyme-linked immunosorbent assay. The expression of CD38, Recombinant NLR Family, pyrin domain-containing protein 3 (NLRP3), and silent mating type information regulation 2 homolog 3 (SIRT3) protein was detected by Western blotting. Results: Compared to the IHR group, cell viability, ATP content, NAD + content, NAD+/NADH ratio, and SIRT3 protein expression decreased, ROS level and inflammatory cytokines increased, and CD38 and NLRP3 proteins raised in the D-gal + IHR group. Compared to the D-gal + IHR group, cell viability, ATP content, NAD + content, NAD+/NADH ratio, and expression of SIRT3 protein increased, ROS level and inflammatory cytokines level decreased, and expression of the CD38 and NLRP3 proteins decreased in the D-gal + IHR + Jujuboside A group. Conclusions: Jujuboside A inhibited the expression of CD38, improved energy metabolism disorder, and mitochondrial function, and decreased inflammation in D-gal-induced H9c2 cells.

Keywords: CD38, D-galactose, ischemia hypoxia and reperfusion, Jujuboside A, nicotinamide adenine dinucleotide


How to cite this article:
Hu YR, Qu HY, Guo JY, Yang T, Zhou H. Jujuboside a improved energy metabolism in senescent H9c2 cells injured by ischemia, hypoxia, and reperfusion through the CD38/Silent mating type information regulation 2 homolog 3 signaling pathway. World J Tradit Chin Med 2023;9:322-9
How to cite this URL:
Hu YR, Qu HY, Guo JY, Yang T, Zhou H. Jujuboside a improved energy metabolism in senescent H9c2 cells injured by ischemia, hypoxia, and reperfusion through the CD38/Silent mating type information regulation 2 homolog 3 signaling pathway. World J Tradit Chin Med [serial online] 2023 [cited 2023 Sep 11];9:322-9. Available from: https://www.wjtcm.net/text.asp?2023/9/3/322/372731   Introduction Top

Cardiovascular disease is the significant cause of pathological factors responsible for the largest number of deaths worldwide, and myocardial infarction is the most common cardiovascular disease.[1] Myocardial ischemia, hypoxia, and reperfusion (IHR) injury are common clinical pathophysiological phenomena of myocardial infarction. These mainly manifest as arrhythmia, lack of blood perfusion, myocardial stunning, and an increase in necrotic cardiomyocytes.[2] Currently, there is no effective treatment to improve myocardial IHR. Therefore, the development of new therapeutic drugs and strategies is critical to improving myocardial ischemia-reperfusion. Aging is the primary risk factor for cardiovascular disease.[3] Cardiovascular aging refers to the degenerative changes in the tissue structure and physiological functions of the cardiovascular system with age. In the aging heart, typical features are decreased systolic and diastolic function, left ventricular wall thickening, myocardial hypertrophy and death, increased interstitial fibrosis, metabolic remodeling, proteostasis imbalance, and mitochondrial dysfunction. In the aging heart, blood vessels also age, and vascular aging is mainly manifested by increased vascular stiffness, vascular calcification and remodeling, endothelial cell dysfunction, intravascular plaque formation, and increased oxidative stress signals. Similar to the common features of body aging, the major molecular and cellular phenotypes of cardiovascular aging also include dysregulated nutrient sensing, protein homeostasis, mitochondrial dysfunction, telomere shortening, epigenetic changes, genomic instability, intercellular communication changes, and cellular senescence.[4] An in-depth understanding of the occurrence and development mechanism of cardiovascular aging and related diseases, and the exploration of new intervention methods for cardiovascular aging and related diseases are important foundations for promoting the prevention and treatment of cardiovascular diseases and achieving healthy aging. D-galactose (D-gal) is a reducing sugar that accumulates in the body. However, high levels of D-gal can be converted to aldose and hydrogen peroxide under the catalysis of galactose oxidase.[5] The use of D-gal to construct animal aging models has been widely used in the study of aging mechanisms and drug anti-aging. Numerous studies have found that D-gal may shorten lifespan in rodent models of aging and induce cell cycle arrest, morphological and metabolic changes, chromatin remodeling, altered gene expression, age-related secretory phenotypes in various organs senescence-associated secretory phenotype, mitochondrial dysfunction, and oxidative stress.[6],[7],[8]

Mitochondria are the main source of cardiac energy. Adenosine triphosphate (ATP) is a form of energy that is directly used in cardiac myocytes. An adequate energy supply is key to the survival of cardiomyocytes.[9] In the process of heart aging, mitochondrial function decreases significantly and reactive oxygen species (ROS) and other toxic substances are produced, which results in oxidative stress injury and cell aging.[10] In the process of cardiac energy metabolism, the nicotinamide adenine dinucleotide (NAD+)/nicotinamide adenine dinucleotide hydride (NADH) ratio is a key factor in mitochondrial electron transmission, oxidative stress, and intracellular signal transduction. It has been shown that the NAD+/NADH ratio is decreased in the hearts of patients with experimental pathological hypertrophy and heart failure.[11] NAD+ also plays an important role in regulating mitochondrial survival, partly through the NAD+-dependent deacetylase silent mating type information regulation 2 homolog 3 (SIRT3).[12] NAD+ homeostasis is maintained by the balance of NAD+ synthase and NAD+ degrading enzymes. CD38 is the main NAD+ degrading enzyme, and overactivation of CD38 is closely related to the sharp decline of NAD+ in aging.[13] CD38 is also an important regulator of inflammation and the innate immune response.[14]

With the development of traditional Chinese medicine (TCM), TCM has been used in the clinic. Through continuous practice and summary, a large number of studies have proved that TCM has a good effect on myocardial infarction. At present, the commonly used TCM methods mainly include TCM compounds, proprietary Chinese medicines and Chinese herbal monomers. Ziziphi Spinosi Semen is the natural seed of Zizyphus jujuba of Rhamnaceae Zizyphus, which has the effects of relieving mental strain, invigorating the heart and nourishing the liver, promoting the secretion of saliva or body fluid, and arresting sweating. It has been used for thousands of years in China, Japan, South Korea, and other Eastern countries. There are a variety of pharmacologically active ingredients in Ziziphi Spinosi Semen, including flavonoids, alkaloids, and saponins. Most studies have shown that saponins are the effective chemical constituents of Ziziphi Spinosi Semen. Jujuboside A has been proved to be the main active component of Ziziphi Spinosi Semen. Jujuboside A is an important saponin compound with various physiological and pharmacological activities that play a potential value in the prevention of diseases in the cardiovascular system.[15] Numerous previous studies have fully confirmed that Jujuboside A has complicated pharmacological activities including antioxidant, antianxiety, anti-inflammatory, and anti-hypoxia functions.[16],[17],[18] However, the pharmacological activity and the effect of Jujuboside A on the energy metabolism of aging cardiomyocytes injured by IHR are unclear.

This study explored whether Jujuboside A affects the energy metabolism of aging H9c2 cells (rat embryonic cardiomyocytes) with IHR injury and whether it might be a novel therapeutic agent. We also explored the role and mechanism of Jujuboside A and the CD38/SIRT3 pathway in aging cardiomyocytes with IHR injury.

  Materials and Methods Top

Establishment of the cell model

We first generated an IHR injury model of aging H9c2 cells (Procell Life Technology Co., Ltd., Wuhan, China) to establish an in vitro model of myocardial infarction.[19] H9c2 cells were treated with 20 mM D-gal (Beyotime Biotechnology Co., Ltd., Shanghai, China) for 12 h, and a serum-free medium was used as an ischemic buffer. The ischemic buffer was cultured in an anaerobic tank containing a 95% N2 and 5% CO2 gas mixture (Mitsubishi Gas Chemical Company, Inc., Japan) for 2 h. H9c2 cells were cultured on medium containing 10% fetal bovine serum at 37°C, and the culture dish was placed in a 95% O2 and 5% CO2 gas mixture to establish the IHR cell model.

Cell grouping

The cell suspension was fully mixed and divided into four groups: the control group (H9c2 cells without intervention) (control group), the IHR group (H9c2 cells after IHR, called the IHR group), the D-gal + IHR group (H9c2 cells injured by D-gal for 12 h, and then subjected to IHR injury) (D-gal + IHR group), and the D-gal + IHR + Jujuboside A group (Solarbio Technology Co., Ltd., Beijing, China) (H9c2 cells injured by D-gal for 12 h, subjected to IHR injury, and finally treated with Jujuboside A for 12 h) (D-gal + IHR + J A).

Cell viability

The cells viability was determined by CCK-8 assay (Solarbio).[19] Briefly, cells were inoculated into 96-well plates. Following the treatment of each group, the cells were washed with phosphate-buffered saline. After washing, 100 μL culture medium and 10 μL CCK-8 solution were added and incubated for 4 h. After incubation, the plates were measured by microplate reader at 450 nm.

Adenosine triphosphate detection

The ATP level was detected using an ATP detection kit (Solarbio).[20] The cells were collected after the intervention, and an appropriate amount of ATP extract was added. After ultrasonic crushing for 1 min, the cell suspension was centrifuged (10,000 g × 10 min) at 4°C to obtain the supernatant. Next, an appropriate amount of chloroform was added, and the supernatant was centrifuged (10,000 g × 3 min) at 4°C and collected. Then, 640 μL reagent I and 260 μL working solution were added to 100 μL sample or standard solution. The absorbance value A1 at 340 nm was measured immediately for 10 s. After the measurement, the samples were incubated at 37°C for 3 min to allow the reaction to proceed. Following incubation, the absorbance value A2 at 340 nm was measured immediately for 10 s. ATP content was calculated according to the formula: ΔAsample = A2sample − A1sample, ΔAstandard = A2standard − A1standard.

Reactive oxygen species detection

ROS levels in H9c2 cells were detected using a ROS detection kit (Solarbio).[21] Briefly, H9c2 cells were inoculated into 96-well plates. Following the treatment of each group, the cells of each group were washed. Next, 100 μL 20,7′00 tedentdxygen speciesrmati (DCFH-DA) medium (final concentration of 10 μM) was added to each well. After 20 min, the cells were washed with serum-free culture medium three times to remove free DCFH-DA. After washing, the 96-well plate was measured by microplate reader (excitation wavelength: 488 nm; emission wavelength: 525 nm).

Interleukin-1β, Interleukin-6, and tumor necrosis factor-alpha levels

Interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α) levels were detected.[22] Following the treatment of each group, the cells were centrifuged (1000 g × 10 min) at 4°C. Next, 100 μL standard and test samples were added per well and incubated at 37°C for 90 min. Following incubation, the plates were washed four times. After washing, 100 μL biotinylated antibody solution was added and incubated for 60 min. After washing four times, 100 μL enzyme conjugate solution was added and incubated at 37°C for 30 min. After washing, 100 μL color-developing substrate was added and incubated in the dark for 14 min. Finally, a 50 μL termination solution was added. The 96-well plate was measured in the microplate reader at 450 nm.

Nicotinamide adenine dinucleotide + and nicotinamide adenine dinucleotide hydride levels

NAD+ and NADH levels in H9c2 cells were detected using an NAD+/NADH detection kit (Beyotime) as previously described.[23] The cells were inoculated into 6-well plates. After the intervention, the cells were washed. After washing, 1 ml NAD+/NADH extract was added to fully decompose the cells, and the supernatant was collected. Next, a 100 μL sample was heated (60°C × 30 min) to decompose NAD+. Next, 20 μL supernatant was transferred to a 96-well plate to measure the content of NADH in the sample. The plate was incubated (37°C × 10 min). Following incubation, 10 μL chromogenic solution was added to each well. After mixing, plates were incubated (37°C × 30 min) in the dark. Finally, the absorbance of the cells was measured at 450 nm with an enzyme-labeling instrument, and the total concentration of NAD+ and NADH was measured according to a standard curve.

CD38, silent mating type information regulation 2 homolog 3, recombinant NLR family, and pyrin domain-containing protein 3 protein expression

CD38, SIRT3, and Recombinant NLR Family, pyrin domain-containing protein 3 (NLRP3) protein expression were detected through Western blotting.[23] Cells were inoculated into 6-well plates and treated, and total protein was extracted and determined. Next, equal amounts of protein were subjected to electrophoresis and membrane transfer using 12% separation gels and 5% concentrated gels. An appropriate amount of BSA blocking solution was added to block the membranes for 60 min and the primary antibodies, including anti-CD38 (1:1000) (Beyotime), anti-β-actin (1:1000) (Beyotime), anti-NLRP3 (1:1000) (Beyotime), and anti-SIRT3 (1:1000) (Beyotime) were added and incubated overnight at 4°C. After incubation, the membranes were washed with a washing solution three times for 10 min/wash. After washing, anti-rabbit (1:10000) (Beyotime) or anti-mouse (1:10000) (Beyotime) secondary antibodies were added, and membranes were incubated at room temperature for 60 min. Following incubation, the membranes were washed three times for 10 min/wash and the configured developer drops were added to the polyvinylidene fluoride film for exposure imaging.

Statistical analysis

Data were analyzed using GraphPad Prism 8.0 software (GraphPad Software, Inc., San Diego, CA, USA),[23] and were expressed as the mean ± standard deviation. All of the data were tested for homogeneity of variance and normality. An independent t-test was used for comparisons between two groups of data. One-way ANOVA was used for comparisons between multiple groups. P values were two-sided (α = 0.05), and P < 0.05 indicated statistical significance.

  Results Top

Jujuboside A improved the viability of D-galactose-induced H9c2 cells injured by ischemia–hypoxia–reperfusion

We first determine the effect of Jujuboside A on the viability of D-gal-induced H9c2 cells injured by IHR. The chemical composition of jujuboside A is shown in [Figure 1]a.The results showed that the cell viability in the IHR group was significantly lower than the control group (P < 0.01). Moreover, compared to the IHR group, cell viability in the D-gal + IHR group significantly descended (P < 0.01). D-gal + IHR injured H9c2 cells were treated with Jujuboside A (10, 20, 30, 40, and 50 μM). The results showed that cell viability increased with increasing Jujuboside A concentration. Jujuboside A at 40 μM significantly improved the viability of D-gal-induced H9c2 cells injured by IHR (P < 0.01). After treatment with 40 μM Jujuboside A, the increase in cell viability was more significant than in the 30 μM and 50 μM of Jujuboside A groups [[Figure 1]b; P < 0.05]. Therefore, we chose 40 μM Jujuboside A for subsequent experiments.

Figure 1: Jujuboside A improved the viability of D-gal-induced H9c2 cells injured by ischemia–hypoxia–reperfusion (a) Jujuboside A. (b) Effects of Jujuboside A (10, 20, 30, 40, and 50 μM) and exposed to D-gal and ischemia–hypoxia–reperfusion. **P < 0.01 versus control group; ##P < 0.01 versus IHR group; △△P < 0.01 and △P < 0.05 versus D-gal + IHR group; ▴P < 0.05. D-gal: D-galactose

Click here to view

Jujuboside A improved mitochondrial function of D-galactose-induced H9c2 cells injured by ischemia–hypoxia–reperfusion

Mitochondrial function was detected in each group. The results showed ATP content decreased and ROS levels raised in the IHR group compared to the control group (P < 0.05). Compared to the IHR group, ATP production decreased and ROS level increased in the D-gal + IHR group (P < 0.01). Moreover, Jujuboside A increased the ATP content and reduced ROS levels in the D-gal + IHR + Jujuboside A group [Figure 2]a and [Figure 2]b; P < 0.05].

Figure 2: Jujuboside A improved mitochondrial function of D-gal-induced H9c2 cells injured by ischemia–hypoxia–reperfusion. (a) Effect of Jujuboside A treatment on the ATP level. (b) Effect of Jujuboside A treatment on the ROS level. D-gal: D-galactose, ROS: Reactive oxygen species, ATP: Adenosine triphosphate

Click here to view

Jujuboside A improved energy metabolism in D-galactose-induced H9c2 cells injured by ischemia–hypoxia–reperfusion

We next detected the levels of NAD+ and NADH in the experimental groups. The results showed that NAD+ levels decreased (P < 0.01), NADH levels did not change significantly, and the NAD+/NADH ratio decreased in the IHR group compared to the control group (P < 0.01). Compared to the IHR group, NAD+ levels decreased (P < 0.01), the NADH level did not change significantly, and the NAD+/NADH ratio decreased in the D-gal + IHR group (P < 0.01). Moreover, Jujuboside A increased NAD+ content and the NAD+/NADH ratio in D-gal-induced H9c2 cells injured by IHR [[Figure 3]a, [Figure 3]ab, [Figure 3]ac; P < 0.05].

Figure 3: Jujuboside A improved energy metabolism in D-gal-induced H9c2 cells injured by ischemia–hypoxia–reperfusion. (a) Effect of Jujuboside A treatment on the NAD+ content. (b) Effect of Jujuboside A treatment on the NADH content. (c) Effect of Jujuboside A treatment on the NAD+/NADH ratio. NAD+: Nicotinamide adenine dinucleotide, NADH: Nicotinamide adenine dinucleotide hydride, D-gal: D-galactose

Click here to view

Jujuboside A improved the inflammatory response of D-galactose-induced H9c2 cells injured by ischemia–hypoxia–reperfusion

We also evaluated the effect of Jujuboside A on the levels of IL-1 β, IL-6, and TNF-α in D-gal-induced H9c2 cells injured by IHR. The results showed that compared to the control group, the levels of IL-1 β, IL-6, and TNF-α increased in the IHR group (P < 0.01). Compared to the IHR group, the levels of IL-1 β, IL-6, and TNF-α increased in the D-gal + IHR group (P < 0.01). Moreover, Jujuboside A significantly reduced the levels of IL-1 β, IL-6, and TNF-α in D-gal-induced H9c2 cells injured by IHR [Figure 4]a, [Figure 4]b, [Figure 4]c; P < 0.05].

Figure 4: Jujuboside A improved the inflammatory response of D-gal-induced H9c2 cells injured by ischemia–hypoxia–reperfusion. (a) Effect of Jujuboside A treatment on the IL-1β content. (b) Effect of Jujuboside A treatment on the IL-6 content. (c) Effect of Jujuboside A treatment on the TNF-α content. D-gal: D-galactose, IL-1β: Interleukin 1β

Click here to view

Jujuboside A mediated the protein expression of CD38 and silent mating type information regulation 2 homolog 3 in D-galactose-induced H9c2 cells injured by ischemia–hypoxia–reperfusion

The CD38, SIRT3, and NLRP3 expression levels were detected by Western blotting. The results showed that compared to the control group, the CD38 and NLRP3 protein expression increased, and SIRT3 protein expression decreased in the IHR group (P < 0.01). Compared to the IHR group, the protein expression of CD38 and NLRP3 increased and SIRT3 decreased in the D gal + IHR group (P < 0.05). Moreover, Jujuboside A decreased the protein expression of CD38 and NLRP3 and increased SIRT3 protein expression in D gal induced H9c2 cells injured by IHR [[Figure 5]; P < 0.05].

Figure 5: Jujuboside A mediated the protein expression of CD38 and SIRT3 in D-gal-induced H9c2 cells injured by ischemia–hypoxia–reperfusion. Western blot analyses of CD38, SIRT3, and NLRP3 protein after treatment with Jujuboside A. D-gal: D-galactose

Click here to view

  Discussion Top

Myocardial ischemia and hypoxia cause cardiomyocyte apoptosis and necrosis, which ultimately leads to heart injury.[24] Therefore, it is critical to restore blood flow and reperfusion as soon as possible to prevent myocardial infarction. However, reperfusion causes secondary injury including arrhythmia, myocardial stunning, and myocardial energy metabolism disorder.[25] Consequently, the prevention and treatment of myocardial cell injury caused by IHR is essential, as is the development for effective therapeutic drugs.[26] Aging is a hazardous risk factor of cardiovascular disease. During aging, the structure and function of the heart change and cardiac reserve declines gradually.[27] Therefore, studying the biological changes in cardiac aging may help to identify new targets for the treatment of aging-related cardiovascular diseases. Indeed, an increasing number of studies have proved that targeting aging-related pathways can improve the structural and functional changes driving age-related heart failure, thereby reducing the adverse effects caused by aging and prolonging heart health.[28]

Due to its high energy demand, the heart is vulnerable to energy metabolism disorders. Therefore, a sufficient ATP supply is important to maintaining normal heart function. During the aging process, cardiomyocyte function decreases, including the decline of myocardial contractility and dysfunction of mitochondrial function, both of which negatively impact cardiac function.[29] In an aging heart, the accumulation of aging cardiomyocytes occurs, interfering with normal cell-to-cell communication and further impairing cardiac function.[30] Our results showed that cell viability and ATP production decreased, and ROS levels increased in D-gal-induced H9c2 cells injured by IHR. Without D-gal induction, the viability of H9c2 cells and the level of ATP increased after IHR, while the level of ROS decreased. These results suggest that H9c2 cells induced by D-gal after IHR exhibit more obvious mitochondrial dysfunction than H9c2 cells without D-gal. Therefore, senescent H9c2 cells are more vulnerable to mitochondrial dysfunction.

NAD+ plays a central role in cell metabolism, energy production, and survival. NAD+ is mainly involved in the redox reaction of energy production.[31] NAD+ plays an important role in cell survival, aging, and the maintenance of normal function. The enzymes include those with single ADP ribotransferase and poly ADP ribose polymerase (PARP) activities (including CD38 and CD157), which catalyze the ADP ribotransferase reaction.[32] NAD+ is a rate-limiting cosubstrate for sirtuins, which is a key regulator of promoting cell survival pathways and maintaining the mitochondrial function and using ADP ribose in NAD as a receptor to catalyze the production of acylated proteins removal of acyl groups.[31] However, evidence has shown that the availability of intracellular NAD+ decreases with aging. The mechanisms of aging-related NAD+ decline are multifaceted, and may include decreased expression of the rate-limiting enzyme of NAD+ biosynthesis, increased usage of NAD+, caused by PARP activation, and the overactivation of NAD+ degrading enzyme CD38.[33] CD38 is a type II transmembrane glycoprotein, which is widely distributed on the surface of the brain, heart, and immune cells.[34] CD38 is the main degrading enzyme of NAD+ and mediates the plasma membrane receptor signal in immune cells.[35] Therefore, CD38 cannot only regulate the proliferation, activation, and migration of a variety of immune cells but also an important inflammatory regulator that plays an important role in maintaining homeostasis.[36] Recent studies have shown that protein expression and enzymatic activity of CD38 gradually increase with aging, which may be responsible for the rapid decline of NAD+ with aging. Our results showed that in D-gal-induced H9c2 cells injured by IHR, CD38 protein expression increased, SIRT3 protein expression decreased, NAD+ levels decreased, and the NAD+/NADH ratio decreased, resulting in energy metabolism disorder and an increase in IL-1 β, IL-6, and TNF-α level and the inflammasome NLRP3.

In China, there are many kinds of Chinese herbal medicines, the clinical foundation of TCM is solid, and Chinese herbal medicines have the characteristics of multicomponent, multilink, and multitarget. Due to the complex composition and wide range of effects of Chinese herbal medicine, it has become one of the key points and hot spots in the research and development of drugs for the treatment of acute myocardial infarction. Ziziphi Spinosi Semen belongs to the first batch of medicinal and edible dual-purpose products issued by the Ministry of Health. As of 2016, there were >260 health products to improve sleep, using Zizyphus jujuba kernel and its extract as raw materials. Jujuboside A is the main active substance of Ziziphi Spinosi Semen, which plays a pharmacological role. It has sedative and hypnotic, antidepressant, improving memory. In our previous studies, we found that Jujuboside A participates in myocardial cell injury induced by IHR. Therefore, the specific mechanism of the cardioprotective effect of Jujuboside A was further studied. In this study, we found that Jujuboside A exhibited anti-aging effects. It improved the viability of D-gal-induced H9c2 cells injured by IHR, reduced expression of the CD38 protein, increased expression of the SIRT3 protein, increased NAD+ level and NAD+/NADH ratio to improve mitochondrial dysfunction, increase ATP production, reduce ROS levels, and decrease the IL-1 β, IL-6 and TNF-α level and inflammasome NLRP3 expression.

  Conclusions Top

We studied the role of the CD38/SIRT3 signaling pathway in mitochondrial energy metabolism and the inflammatory response. Our experimental results show that reducing the expression of CD38 and increasing the expression of SIRT3 improved mitochondrial energy metabolism and decreased the inflammatory response. We observed the effects of Jujuboside A on H9c2 cells and H9c2 cells induced by D-gal subjected to IHR. Our study shows that the cardioprotection effect of Jujuboside A on H9c2 cells was achieved through regulation of the CD38/SIRT3 signaling pathway. Our results provide new information for the study of the pathogenesis of IHR and the mechanisms of Jujuboside A in senescent H9c2 cells.

Acknowledgments

We thank LetPub (www.letpub.com) for its linguistic assistance during the preparation of this manuscript.

Author's contributions

ZH and HYR designed the study. HYR, QHY, GJY, and YT performed the experiments. HYR and YT collected the research data and drafted the manuscript, which was finally checked by ZH. All the authors approved the final manuscript.

Availability of data and materials

The dataset used and analyzed during the current study is available from the corresponding author on reasonable request.

Financial support and sponsorship

Construction of atrial fibrillation-specific disease database (shdc2020cr6012-003); 3 years action plan of Shanghai Shenkang Medical Development Center (shdc2020cr1053b); Science and technology support project of Shanghai Municipal Commission of Science and Technology (18401932800); Shanghai Shenkang medical development center emerging frontier technology joint research project (shdc12018125).

Conflicts of interest

There are no conflicts of interest.

 

  References Top
1.Wang Y, Li J, Zheng X, Jiang ZH, Hu S, Wadhera RK, et al. Risk factors associated with major cardiovascular events 1 year after acute myocardial infarction. JAMA Netw Open 2018;1:e181079.  Back to cited text no. 1
    2.Deng X, Ye F, Zeng LX, Luo WZ, Tu S, Wang XY, et al. Dexmedetomidine mitigates myocardial ischemia/reperfusion-induced mitochondrial apoptosis through targeting lncRNA HCP5. Am J Chin Med 2022;50:1529-51.  Back to cited text no. 2
    3.Alfaras I, Di Germanio C, Bernier M, Csiszar A, Ungvari Z, Lakatta EG, et al. Pharmacological strategies to retard cardiovascular aging. Circ Res 2016;118:1626-42.  Back to cited text no. 3
    4.Li HB, Hastings MH, Rhee J, Trager LE, Roh JD, Rosenzweig A. Targeting age-related pathways in heart failure. Circ Res 2020;126:533-51.  Back to cited text no. 4
    5.Bo-Htay C, Palee S, Apaijai N, Chattipakorn SC, Chattipakorn N. Effects of d-galactose-induced ageing on the heart and its potential interventions. J Cell Mol Med 2018;22:1392-410.  Back to cited text no. 5
    6.Wang LF, Cao Q, Wen K, Xiao YF, Chen TT, Guan XH, et al. CD38 deficiency alleviates d-galactose-induced myocardial cell senescence through nad+/sirt1 signaling pathway. Front Physiol 2019;10:1125.  Back to cited text no. 6
    7.Du HM, Wang YJ, Liu X, Wang SL, Wu SM, Yuan Z, et al. miRNA-146a-5p mitigates stress-induced premature senescence of D-galactose-induced primary thymic stromal cells. Cytokine 2021;137:155314.  Back to cited text no. 7
    8.Sun KY, Yang PY, Zhao R, Bai YT, Guo ZJ. Matrine attenuates d-galactose-induced aging-related behavior in mice via inhibition of cellular senescence and oxidative stress. Oxid Med Cell Longev 2018;2018:7108604.  Back to cited text no. 8
    9.Fernandez-Caggiano M, Kamynina A, Francois AA, Prysyazhna O, Eykyn TR, Krasemann S, et al. Mitochondrial pyruvate carrier abundance mediates pathological cardiac hypertrophy. Nat Metab 2020;2:1223-31.  Back to cited text no. 9
    10.Giorgi C, Marchi S, Simoes IC, Ren Z, Morciano G, Perrone M, et al. Mitochondria and reactive oxygen species in aging and age-related diseases. Int Rev Cell Mol Biol 2018;340:209-344.  Back to cited text no. 10
    11.Berthiaume JM, Kurdys JG, Muntean DM, Rosca MG. Mitochondrial NAD+/NADH redox state and diabetic cardiomyopathy. Antioxid Redox Signal 2019;30:375-98.  Back to cited text no. 11
    12.Lu Y, Wang YD, Wang XY, Chen H, Cai ZJ, Xiang MX. SIRT3 in cardiovascular diseases: Emerging roles and therapeutic implications. Int J Cardiol 2016;220:700-5.  Back to cited text no. 12
    13.Boslett J, Helal M, Chini E, Zweier JL. Genetic deletion of CD38 confers post-ischemic myocardial protection through preserved pyridine nucleotides. J Mol Cell Cardiol 2018;118:81-94.  Back to cited text no. 13
    14.Lischke T, Heesch K, Schumacher V, Schneider M, Haag F, Koch-Nolte F, et al. CD38 controls the innate immune response against Listeria monocytogenes. Infect Immun 2013;81:4091-9.  Back to cited text no. 14
    15.Wan CR, Han DD, Xu JQ, Yin P, Xu XL, Mei C, et al. Jujuboside A attenuates norepinephrine-induced apoptosis of H9c2 cardiomyocytes by modulating MAPK and AKT signaling pathways. Mol Med Rep 2018;17:1132-40.  Back to cited text no. 15
    16.Bian ZH, Zhang WM, Tang JY, Fei QQ, Hu MM, Chen XW, et al. Mechanisms underlying the action of ziziphi spinosae semen in the treatment of insomnia: A study involving network pharmacology and experimental validation. Front Pharmacol 2021;12:752211.  Back to cited text no. 16
    17.Wang XX, Ma GI, Xie JB, Pang GC. Influence of JuA in evoking communication changes between the small intestines and brain tissues of rats and the GABAA and GABAB receptor transcription levels of hippocampal neurons. J Ethnopharmacol 2015;159:215-23.  Back to cited text no. 17
    18.Wang WS, Huang QJ, Chen Y, Huang ZY, Huang YF, Wang Y, et al. The novel FAT4 activator jujuboside A suppresses NSCLC tumorigenesis by activating HIPPO signaling and inhibiting YAP nuclear translocation. Pharmacol Res 2021;170:105723.  Back to cited text no. 18
    19.Zhao YB, Guo YX, Chen YQ, Liu S, Wu N, Jia DL. Curculigoside attenuates myocardial ischemia-reperfusion injury by inhibiting the opening of the mitochondrial permeability transition pore. Int J Mol Med 2020;45:1514-24.  Back to cited text no. 19
    20.Yang GZ, Xue FS, Liu YY, Li HX, Liu Q, Liao X. Feasibility analysis of oxygen-glucose deprivation-nutrition resumption on h9c2 cells in vitro models of myocardial ischemia-reperfusion injury. Chin Med J (Engl) 2018;131:2277-86.  Back to cited text no. 20
    21.Jiang WF, Song JG, Zhang ST, Ye YY, Wang J, Zhang YL. CTRP13 protects H9c2 cells against hypoxia/Reoxygenation (h/r)-induced injury via regulating the Ampk/Nrf2/Are signaling pathway. Cell Transplant 2021;30:9636897211033275.  Back to cited text no. 21
    22.Qiu Z, He Y, Ming H, Lei S, Leng Y, Xia ZY. Lipopolysaccharide (LPS) aggravates high glucose- and hypoxia/reoxygenation-induced injury through activating ros-dependent nlrp3 inflammasome-mediated pyroptosis in h9c2 cardiomyocytes. J Diabetes Res 2019;2019:8151836.  Back to cited text no. 22
    23.Hu YR, Xing SL, Chen C, Shen DZ, Chen JL. Codonopsis pilosula polysaccharides alleviate Aβ 1-40-induced PC12 cells energy dysmetabolism via CD38/NAD+signaling pathway. Curr Alzheimer Res 2021;18:208-21.  Back to cited text no. 23
    24.Hausenloy DJ, Yellon DM. Ischaemic conditioning and reperfusion injury. Nat Rev Cardiol 2016;13:193-209.  Back to cited text no. 24
    25.Yang XY, He K, Pan CS, Li Q, Liu YY, Yan L, et al. 3, 4-dihydroxyl-phenyl lactic acid restores NADH dehydrogenase 1 α subunit 10 to ameliorate cardiac reperfusion injury. Sci Rep 2015;5:10739.  Back to cited text no. 25
    26.Mishra PK, Adameova A, Hill JA, Baines CP, Kang PM, Downey JM, et al. Guidelines for evaluating myocardial cell death. Am J Physiol Heart Circ Physiol 2019;317:H891-922.  Back to cited text no. 26
    27.Steenman M, Lande G. Cardiac aging and heart disease in humans. Biophys Rev 2017;9:131-7.  Back to cited text no. 27
    28.Ren J, Sowers JR, Zhang YM. Metabolic stress, autophagy, and cardiovascular aging: From pathophysiology to therapeutics. Trends Endocrinol Metab 2018;29:699-711.  Back to cited text no. 28
    29.Ramaccini D, Montoya-Uribe V, Aan FJ, Modesti L, Potes Y, Wieckowski MR, et al. Mitochondrial function and dysfunction in dilated cardiomyopathy. Front Cell Dev Biol 2020;8:624216.  Back to cited text no. 29
    30.Elorza AA, Soffia JP. mtDNA heteroplasmy at the core of aging-associated heart failure. An integrative view of oxphos and mitochondrial life cycle in cardiac mitochondrial physiology. Front Cell Dev Biol 2021;9:625020.  Back to cited text no. 30
    31.Hershberger KA, Martin AS, Hirschey MD. Role of NAD+ and mitochondrial sirtuins in cardiac and renal diseases. Nat Rev Nephrol 2017;13:213-25.  Back to cited text no. 31
    32.Cantó C, Menzies KJ, Auwerx J. NAD(+) metabolism and the control of energy homeostasis: A balancing act between mitochondria and the nucleus. Cell Metab 2015;22:31-53.  Back to cited text no. 32
    33.Aman Y, Qiu YM, Tao J, Fang EF. Therapeutic potential of boosting NAD+ in aging and age-related diseases. Transl Med Aging 2018;2:30-7.  Back to cited text no. 33
    34.Chen LM, Diao LX, Yang YB, Yi XH, Rodriguez BL, Li YL, et al. CD38-mediated immunosuppression as a mechanism of tumor cell escape from pd-1/pd-l1 blockade. Cancer Discov 2018;8:1156-75.  Back to cited text no. 34
    35.Konen JM, Fradette JJ, Gibbons DL. The good, the bad and the unknown of cd38 in the metabolic microenvironment and immune cell functionality of solid tumors. Cells 2019;9:52.  Back to cited text no. 35
    36.Malavasi F, Deaglio S, Funaro A, Ferrero E, Horenstein AL, Ortolan E, et al. Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology. Physiol Rev 2008;88:841-86.  Back to cited text no. 36
    
  [Figure 1], [Figure 2], [Figure 3], [Figure 4], [Figure 5]

 

Top

留言 (0)

沒有登入
gif