Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Journal of Functional Foods 76 (2021) 104319

Contents lists available at ScienceDirect

Journal of Functional Foods


journal homepage: www.elsevier.com/locate/jff

Total flavonoids extracted from the leaves of Murraya paniculata (L.) Jack
alleviate oxidative stress, inflammation and apoptosis in a rat model of
diabetic cardiomyopathy
Jingtao Zou, Dayun Sui, Wenwen Fu, Yuangeng Li, Ping Yu, Xiaofeng Yu, Huali Xu *
Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun 130021, PR China

A R T I C L E I N F O A B S T R A C T

Keywords: The leaves of Murraya paniculata (L.) Jack were used as spices in China for many years. Flavonoids are the main
Murraya paniculata (L.) Jack components of it. However, no study has determined whether the total flavonoids of Murraya paniculata (L.) Jack
Diabetic cardiomyopathy (TFMP) has preventive effects on diabetic cardiomyopathy (DCM).The present study investigated the effect of
Oxidative stress
TFMP on DCM. STZ was administered along with a high-fat diet to induce diabetes in rats. Parallel experiments
Inflammation
were performed using H9c2 cells exposed to high glucose. Diabetic rats showed serious histological changes and
increased levels of cardiac function markers. The markers of ROS, inflammation and apoptosis were also
increased significantly, but the antioxidant defenses were decreased. Treatment with TFMP alleviated histo­
logical alteration in the myocardium. TFMP suppressed oxidative stress, inflammation and apoptosis. Our results
demonstrate that the protective of TFMP on DCM is associated with increasing Nrf2 and HO-1 gene expression
and inhibiting oxidative stress, inflammation and apoptosis.

1. Introduction development is unclear. Moreover, several studies have shown that


unbalanced energy metabolism, oxidative stress, inflammation, and
Diabetes mellitus (DM) is a group of chronic metabolic disorders apoptosis contribute to DCM development (Althunibat et al., 2019; H. Li
characterized by a high blood glucose level over a prolonged period of et al., 2019; K. Li et al., 2019). Table 1.
time. With the number of people developing DM increasing exponen­ The leaves of Murraya paniculata (L.) Jack were used as spices in
tially, DM has become one of the most common public health concerns China for many years. Murraya paniculata (L.) Jack, named Qianlixiang,
globally. An estimated 382 million people worldwide have diabetes, and belongs to the family of Rutaceae. It is widely distributed in South China
this number is expected to reach 592 million by 2035 (Guariguata et al., (Fujian, Guangdong, Guangxi and Yunan provinces), India, and
2014). High blood glucose levels can lead to the development of serious Thailand. Qianlixiang exhibits several anti-anxiety, anti-depression
life-threatening complications such as stroke, retinopathy, neuropathy, (Sharma, Batra, Kumar, & Sharma, 2017), anti-inflammation(X. Wang
nephropathy, and cardiomyopathy (Low Wang, Hess, Hiatt, & Goldfine, et al., 2019; Wu, Liu, & Shi, 2016), and anti-microbial activities(I. R.
2016). Cardiovascular complications such as atherosclerosis, myocar­ Menezes et al., 2015). Menezes showed that the hydroalcoholic extract
dial infarction, and cardiomyopathy are the major cause of death in of M. paniculata had a glucose-lowering effect on alloxan-induced dia­
nearly 50% of DM patients. The incidence rate of cardiomyopathy in DM betic rats, and it reduced tissue lesions developed in diabetes(C. D. A.
patients is 2 to 5 times higher than that in non-diabetic patients (Kannel, Menezes et al., 2017). Many active components, such as flavonoids(J. Y.
Hjortland, & Castelli, 1974). Diabetic cardiomyopathy (DCM) is char­ Zhang et al., 2011), coumarins(Saied, Nizami, & Anis, 2008), and al­
acterized by myocardial systolic and/or diastolic dysfunction, oxidative kaloids(Wang et al., 2018), have been isolated from Murraya paniculata.
stress, inflammation, cardiomyocyte apoptosis, interstitial fibrosis, and Our previous study showed that total flavonoids extracted from the
was first defined by Rubler in 1972(Rubler et al., 1972). The cardiac leaves of M. paniculata (TFMP) can effectively prevent kidney damage in
function of DCM patients declines over time. The process is irreversible diabetic rats by regulating oxidative stress and levels of inflammation
and ultimately leads to heart failure. However, the mechanism of DCM cytokines(Zou et al., 2014).

* Corresponding author at: Departments of Pharmacology, School of Pharmaceutical Sciences, Jilin University. 1266 Fujin road, Changchun 130021, Jilin Province,
PR China.
E-mail address: xhl@jlu.edu.cn (H. Xu).

https://doi.org/10.1016/j.jff.2020.104319
Received 28 August 2020; Received in revised form 23 November 2020; Accepted 29 November 2020
Available online 11 December 2020
1756-4646/© 2020 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
J. Zou et al. Journal of Functional Foods 76 (2021) 104319

Table 1 (Shanghai, China). All chemicals used in this study were of analytical
Primers for qPCR. grade. Rat H9c2 cardiomyocyte cells were obtained from Shanghai
Gene Forward primer (5′ -3′ ) Reverse primer (5′ -3′ ) Institute of Cell Biology, Chinese Academic of Sciences (Shanghai,
China). Dulbecco’s modified Eagle’s medium (DMEM) and fetal bovine
Nrf2 TTGTAGATGACCATGAGTGC TGTCCTGCTGTATGCTGCTT
HO-1 CACGCATATACCCGCTACCT AAGGCGGTCTTAGCCTCTTC serum were purchased from Life Technologies (Grand Island, NY, USA).
TNF-α AGCATGATCCGAGTGTGGAA TAGACAGAAGAGCGTGGTGGC
IL-6 GTTGCCTTCTTGGGACTGATG ATACTGGTCTGTTGTGGGTGGT 2.2. Extraction of TFMP
β-actin CAGGGTGTGATGGTGGGTATG ATGCCTCTCTTGCTCTGGG

The leaves of Murraya paniculata (L.) Jack were procured from


Flavonoids are a vast group of polyphenols found ubiquitously in Kunming (Yunnan Province, China), and identified by Professor Minglu
plant and have received considerable attention because of their car­ Deng of Changchun University of Traditional Chinese medicine. The
dioprotective function(Liu et al., 2017; Lv, Cheng, Tang, & Jiang, 2017). dried leaves were crushed, and 85% ethanol was added and the mixture
However, no study has so far determined whether TFMP has potential was refluxed extraction for three times to afford the ethanol concentrate.
preventive effects on DCM induced by a high-fat diet combined with Ethanol concentrate was adsorbed by D4020 macroporous resin column
streptozotocin (STZ) and the potential mechanisms of action. Therefore, and desorbed with 85% ethanol to obtain ethanol desorption solution.
the present study was designed to confirm the cardioprotective effect of After decolorization by D941 macroporous resin column, ethanol was
TFMP on high glucose (HG)-induced H9c2 cardiac myoblasts and dia­ recovered under reduced pressure and dried at 80 ℃ for 2 h to afford the
betic rats. crude extract of total flavonoids .The crude of total flavonoids (crude
flavonoids: silica gel mass = 1:10), eluted with eluent, recovered under
2. Materials and methods reduced pressure, dried at 105 ℃ for 2 h to afford the total flavonoids of
Murraya paniculata (L.) Jack (TFMP). The total amount of flavonoids
2.1. Reagents and antibodies should not be<50% by HPLC (Fig. 1).

TFMP was provided by Dr. Yongri Jin (College of Chemistry, Jilin 2.3. Culturing of H9c2 cardiomyocyte cells
University). Streptozotocin (STZ) was purchased from Sigma Chemicals
(St. Louis, MO, USA). Malondialdehyde (MDA), superoxide dismutase H9c2 cells were cultured in DMEM medium containing 5.5 mM
(SOD), glutathione peroxidase (GSH-Px), troponin I (cTnI), creatine glucose, supplemented with 10% fetal calf serum. Cells in the HG-
kinase-MB (CK-MB), and lactate dehydrogenase (LDH) were purchased treated group were incubated in DMEM medium containing 35 mM
from Nanjing Jiancheng Bioengineering Institute (Nanjing, China). An­ glucose. The culture was maintained at 37 ◦ C with a gas mixture of 5%
tibodies against Bax and Bcl-2 were obtained from Cell Signaling CO2/95% air. All media were supplemented with 100 U/mL penicillin
Technology (Beverly, MA, USA). Antibody against β-actin was obtained and 100 µg/mL streptomycin.
from ZSGB-BIO (Beijing, China). Goat anti-rabbit and goat anti-mouse
secondary antibodies were purchased from Beijing Dingduo Chang­ 2.4. Cell viability assay
sheng Biotechnology (Beijing, China). A quantitative polymerase chain
reaction (qPCR) kit was obtained from Beijing TransGen Biotech (Bei­ The viability of H9c2 rat cardiomyocytes was measured by per­
jing, China). A TUNEL kit was obtained Institute of Biotechnology forming the CCK-8 assay. H9c2 cells at a concentration of 5 × 104 cells/
(Jiangsu, China). An annexin V–fluorescein isothiocyanate (FITC) mL were collected and resuspended in DMEM medium, and 100 µL ali­
apoptosis detection kit was obtained from Tianjin Sungene Biotech Co., quots were added to each well of 96-well flat-bottomed microtiter
Ltd. (Tianjin, China). The CCK-8 was purchased from Yeasen Biotech plates. TFMP was dissolved in DMSO. H9c2 cells were treated with
TFMP (25, 50, 75, 100, 200 µg/mL) for 24 h. Three replicate wells were

Fig. 1. HPLC of TFMP.

2
J. Zou et al. Journal of Functional Foods 76 (2021) 104319

used for each data point in the experiments. Then 10 µL of CCK-8 was 0.5% CMC-Na; TFMP 35 mg/kg group: the diabetic rats were orally
added to each well. Absorption was measured at 450 nm with a micro­ treated with TFMP at a dose of 35 mg/kg; and TFMP 70 mg/kg group:
plate reader (SpectraMax Plus384, Molecular Devices, USA). diabetic rats were orally administered with TFMP at a dose of 70 mg/kg.
TFMP was dissolved in 0.5% CMC-Na. Meanwhile, the control rats were
2.5. Lactate dehydrogenase release assay orally administered with 0.5% CMC-Na. The rats were treated with
TFMP or 0.5% CMC-Na once a day for 12 weeks.
Cytotoxicity was measured by using the lactate dehydrogenase
(LDH) assay kit. The supernatant (20 µL/well) was incubated with 2.10. Physical and biochemical analyses
250 µL LDH reaction agent at room temperature for 5 min. Absorbance
value at 450 nm was measured using the microplate reader. At 13 weeks, rats were anesthetized with ketamine (80 mg/kg, i.p),
heart weight (HW), body weight (BW), and levels of blood glucose, cTn
2.6. DAPI staining I, CK-MB, and LDH were measured. MDA level, and SOD and GSH-Px
activities in the heart tissue and H9c2 cells were assayed using
H9c2 cells were seeded on 6-well plates and treated with TFMP for commercially available kits and in accordance with the manufacturer’s
24 h. The cells were collected and washed twice with PBS, permeabilized instructions.
with 0.1% Triton X-100, and stained with 2 µg/mL DAPI for 10 min at
room temperature. The cells were subsequently observed using a fluo­ 2.11. Histopathological examination
rescence microscope (magnification, x100; Nikon TE-2000U; Nikon
Corporation, Tokyo, Japan). Heart tissues were fixed in 10% phosphate-buffered formalin,
dehydrated, and embedded in paraffin. Sections about 4-µm thickness
2.7. Apoptosis assay were sliced and stained with hematoxylin and eosin (HE). The slides
were examined under a light microscope with a magnification of
Apoptosis was determined by staining cells with annexin V-FITC and 200 × by a pathologist who was blinded to the experiments.
propidium iodide (PI) labeling. H9c2 cells were treated with TFMP for The heart tissues were fixed in 4% glutaraldehyde in sodium phos­
24 h. After treatment, the cells were collected and washed twice in ice- phate buffer (0.2 M, PH 7.4) for 3 h at 4 ◦ C and dehydrated using graded
cold PBS and resuspended in 300 µL of binding buffer at 2 × 105 cells/ ethanol. The samples were embedded in resin and polymerized at 72 ◦ C
mL. The samples were incubated with 5 µL of annexin V-FITC and 5 µL for 48 h. Ultra-thin sections (50–70 nm) were stained with uranyl ace­
propidium iodide in the dark for 15 min at room temperature. Finally, tate and lead acetate. The images were captured using a JEM 1200 EX
the samples were analyzed by flow cytometry and evaluated on the basis electron microscope (JEM 1200 EX; Jeol, Tokyo, Japan).
of the percentage of cells that were annexin V positive.
2.12. TUNEL staining
2.8. Quantitative real time-polymerase chain reaction
The apoptotic analysis was performed by TUNEL staining, as
Total RNA from H9c2 cells was extracted using Trizol reagent described previously. Heart tissues were pretreated in accordance with
(Invitrogen Inc, Carlsbad, CA, USA) according to the manufacturer’s the pretreatment procedure followed for HE staining. Fixed tissue sec­
instructions. RNA was quantitated by optical density measurements at tions (4 µm) were minced. Based on the instructions given with the
260 and 280 nm. TransScript Green Two-Step aPT-PCR SuperMix was TUNEL staining kit, the apoptotic level was determined.
used for cDNA synthesis and amplification. qPCR reaction was per­
formed using the Mx 3000P Real-Time PCR system (Agilent, Santa Clara, 2.13. Western blotting
USA). The samples were heated to 95 ◦ C for 10 min. The cycles were
performed at 95 ◦ C for 15 s, 60 ◦ C for 60 s, and 72 ◦ C for 40 s. Relative Rat heart tissue and H9c2 cells were lysed in RIPA buffer (150 mM
expression of genes was performed by Mx 3000P software to calculate NaCl, 1% Triton X-100, 0.5% sodium deoxycholate, 0.1% SDS, 50 mM
the Ct values for each sample by comparing the Ct values of the target Tris-HCl pH 7.4) for 30 min on ice. After centrifugation (13, 000 × g
gene with those of the β-actin constitutive gene product. The primers 4 ◦ C, 15 min), the supernatants were collected. Samples were loaded
(Nrf2, HO-1, TNF-α, IL-6) were synthesized by the Sangon company onto 12% polyacrylamide SDS gels. After electrophoresis, the gels were
(Shanghai, China). blotted onto a PVDF membrane, blocked with 5% (w/v) milk for 1 h on a
shaker at room temperature, washed twice with TBST, and probed
2.9. Rat model of DCM overnight at 4 ◦ C. The blots were incubated with primary antibodies
against Bax and Bcl-2 overnight. Next, the blots were incubated with a
Male Wistar rats weighing 160–180 g were purchased from the secondary antibody for 1 h at room temperature, and bands were
Experimental Animal Center of Jilin University. They were housed in a detected by enhanced chemiluminescence (ECL).
room with an ambient temperature of 22 to 25 ◦ C and a 12-h light/dark
cycle, and fed a standard pelleted diet (SPD) with water ad libitum. The 2.14. Statistical analyses
animal studies are done according to ethical procedures and experi­
mental protocols were approved in accordance with nationally approved Results are expressed as mean ± SD. The statistical significances of
guidelines for “The Institutional Animal Care and Use Committee” (No. all data were determined using one-way analysis of variance followed by
20180041). After one week of acclimatization, the rats were randomly Turkey’s post-hoc test for multiple comparisons. A P value < 0.05 was
divided into two groups and were fed with either SPD (control) or a considered statistically significant.
high-fat diet (HFD) consisting of 40% fat, 41% carbohydrates, and 18%
protein for eight weeks. After overnight fasting, the HFD-fed rats were 3. Results
intravenously (i.v.) injected with STZ (30 mg/kg). Two weeks later, the
fasting blood glucose (FBG) level was measured in these rats, and those 3.1. TFMP enhanced the viability of HG-induced H9c2 cells
with an FBG level of ≥ 11.1 mmol/L were considered diabetic. Then, the
diabetic rats were randomly divided into three groups according to the Cell viability and cytotoxicity were measured by CCK-8 and LDH
FBG levels, with 12 rats in each group. The experimental groups were as assays. TFMP treatment (200 µg/mL) significantly decreased the
follows: Diabetic group: the diabetic rats were orally administered with viability of H9c2 cells (Fig. 2A). However, TFMP treatment of < 200 µg/

3
J. Zou et al. Journal of Functional Foods 76 (2021) 104319

Fig. 2. Effect of TFMP on HG-induced cell injury in H9c2 cells. (A) Cells incubated with different concentrations (25, 50, 75, 100 and 200 µg/mL) of TFMP for 24 h,
Cell viability was detected by CCK-8 assay. (B) Effect of TFMP on LDH leakage in the H9c2 cells was measured by LDH assay. #P < 0.05 versus the control group;
*P < 0.05 versus the HG group.

mL did not affect cell viability. As shown in Fig. 2B, HG significantly (P < 0.01), while TFMP reduced the expression of Bax and increased the
increased LDH release, which was alleviated by 50 and 100 µg/mL TFMP expression of Bcl-2. The ratio of Bax/Bcl-2 expression was markedly
treatment. The use of 25 µg/mL TFMP was less effective. Based on these decreased after treatment with 50 and 100 µg/mL TFMP (Fig. 3C-F).
results, TFMP was used at concentrations of 50 and 100 µg/mL in the To further investigate the effect of TFMP on oxidative stress in H9c2
subsequent experiments. cells induced by HG, the MDA content and SOD and GSH-Px activities
were determined. As shown in Fig. 4, the activities of SOD (Fig. 4A) and
GSH-Px (Fig. 4B) were decreased significantly in the HG group. TFMP
3.2. TFMP suppressed apoptosis and oxidative stress in H9c2 cells effectively increased the activities of SOD and GSH-Px, while the MDA
content was notably increased in the HG group, which was restored after
Apoptosis plays a critical role in the development of diabetic car­ 50 and 100 µg/mL TFMP treatment. To identify the mechanism of action
diomyopathy, and hence, the effects of TFMP on HG-induced apoptosis of TFMP in relation to oxidative stress, Nrf2 and HO-1 mRNA levels were
was investigated. As shown in Fig. 3A, morphological changes typical of determined by real-time PCR, and these levels were increased after 50 or
apoptosis such as cell shrinkage, nuclear condensation, and apoptotic 100 µg/mL TFMP treatment (Fig. 4D, E).
body formation were observed, and the apoptotic rate was also
increased (mean = 38.10%, P < 0.05) in HG cells. The apoptotic rate was
alleviated after treatment with 50 and 100 µg/mL TFMP 3.3. TFMP inhibited TNF-α and IL-6 mRNA expression in H9c2 cells
(mean = 21.28% and 16.36%, P < 0.05) (Fig. 3B). The expression of Bax
was upregulated and that of Bcl-2 was downregulated in HG cells To determine the effect of TFMP on inflammation in H9c2 cells

Fig. 3. Effect of TFMP on apoptosis in HG-induced H9c2 cells. (A) Annexin V-FITC/PI staining. (B) Apoptosis rate in the control, HG, and TFMP groups. (C) DAPI
staining showing the apoptotic cells. Scale bar = 100 µm. (D–G) Western blot showing Bax, Bcl-2, and the ratio of Bax to Bcl-2. #P < 0.05 vs. the control group;
*P < 0.05 vs. the HG group.

4
J. Zou et al. Journal of Functional Foods 76 (2021) 104319

Fig. 4. Effect of TFMP on oxidative stress in HG-induced H9c2 cells. (A) content of MDA, (B) activity of SOD, (C) activity of GSH-Px, (D) Nrf2 mRNA level and (E)
HO-1 mRNA level, (F) TNF-α mRNA level, (G) IL-6 mRNA level.#P < 0.05 versus control group; * P < 0.05 versus the HG group.

Fig. 5. Effect of TFMP on blood glucose levels, body weight, and HW/BW ratio in DCM rats. (A) Blood glucose levels, (B) Body weight, (C) HW/BW ratio, (D) CK-MB
activity, (E) LDH level, and (F) cTnI activity. #P < 0.05 versus the control group; * P < 0.05 versus the DCM group.

5
J. Zou et al. Journal of Functional Foods 76 (2021) 104319

induced by HG, the mRNA levels of TNF-α and IL-6 were determined by TFMP. Treatment with TFMP inhibited the production and mRNA level
real-time PCR. As shown in Fig. 4F, G, the mRNA levels of TNF-α and IL- of TNF-α and IL-6 (Fig. 7F-I).
6 were significantly increased in H9c2 cells after HG treatment. How­
ever, TFMP efficiently inhibited HG-induced expression of TNF-α and IL-
6 in H9c2 cells. 3.7. TFMP decreases apoptotic rate in DCM rats

To evaluate the cardioprotective effect of TFMP on apoptosis in DCM


3.4. TFMP attenuated blood glucose levels and HW/BW ratio
rats, the TUNEL assay was performed. As shown in Fig. 8A-B, the
number of TUNEL-positive cells was significantly increased in the DCM
At 13 weeks, BW in the DCM group showed a significant reduction
group, which was decreased after TFMP treatment. The expression of
compared with that in the control group. TFMP prevented BW loss
Bax was higher, while that of Bcl-2 was lower in the DCM group, and this
(Fig. 5A). In addition, blood glucose levels and HW/BW ratio were
expression was reversed after TFMP treatment (Fig. 8C-E).
higher in the DCM group. TFMP treatment thus significantly inhibited
the increase in glucose levels and HW/BW (Fig. 5B, 5C).
4. Discussion

3.5. TFMP alleviated myocardial damage in DCM rats In the present study, we induced cardiomyopathy in vitro by using a
high glucose concentration and in vivo using a rat model of type 2 dia­
To evaluate the effect of TFMP on myocardial damage in DCM rats, betes. Consistent with previous studies, high glucose levels resulted in
CK-MB, LDH, and cTnI levels were measured. As shown in Fig. 5D-F, CK- oxidative stress, inflammation, apoptosis, and myocardial pathological
MB, LDH, and cTnI levels in the DCM group remarkably increased changes in DCM(Badran et al., 2018; Tang et al., 2019). For the first
compared with those in the control group. TFMP treatment significantly time, our study showed that the treatment with TFMP significantly
reduced the levels of CK-MB, LDH, and cTnI. attenuated HG-induced oxidative stress, fibrosis, inflammation, and
The cardioprotective effect of TFMP was further confirmed by apoptosis in H9c2 cells in vitro and prevented structural alterations in
morphological examination. As showed in Fig. 6A, the rats in normal the cardiac tissues of DM rats. This result suggests that TFMP protects
group exhibited a normal heart histology. On the contrary, the animals against DCM.
in DCM group showed a disorder of the arrangement of cardiomyocytes, DM is a complex metabolic disease characterized by hyperglycemia
the uneven color, the degeneration of muscle fiber, the swelling of or other metabolic alternations such as hyperlipidemia, hypercholes­
interstitial muscle and neutrophil infiltration. In the ultrastructural terolemia, and ketoacidemia. Several studies have indicated that
morphologies examination, the rats of normal group exhibited well- oxidative stress plays a crucial role in the occurrence and development
arranged myofibrils (Fig. 6 B). In the rats of DCM group, myofibrils of cardiomyopathy in DM (Faria & Persaud, 2017; Liao et al., 2017).
arrangement was disordered, broken and disappeared. Sarcomere Hyperglycemia and/or hyperlipidemia could stimulate reactive oxygen
arrangement was disorderly, Z line was unclear. Mitochondrion was species (ROS) generation in DM (Zheng et al., 2020). Under the condi­
broken, swollen and vacuolated. However, TFMP treatment markedly tions of DCM, a constantly elevated status of oxidative stress depletes
alleviated the pathological changes mentioned above. endogenous antioxidants such as SOD and glutathione peroxidase (GSH-
Px) in the heart (Jimenez et al., 2018). The decrease in antioxidant ca­
3.6. TFMP suppressed oxidative stress and inflammation in DCM rats pacity in the myocardium can, therefore, lead to myocardial cell dam­
age. To combat the damage caused by oxidative stress, a series of
As shown in Fig. 7, the activities of SOD (Fig. 7A) and GSH-Px endogenous antioxidative mechanisms in these cells is activated, one of
(Fig. 7B) were decreased significantly in the DCM group. TFMP treat­ which involves nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 is
ment effectively increased the activities of SOD and GSH-Px, while the a member of the CNC transcription factor family that controls the
MDA content was notably increased in the DCM group, which was expression of several antioxidant genes including heme oxygenase (HO-
restored after TFMP treatment (Fig. 7C). Nrf2 and HO-1 mRNA levels 1), nicotinamide adenine dinucleotide phosphate-H (NADPH),
were increased in the cardiac tissue after TFMP treatment compared quinineoxidoreductase-1 (NQO-1), and glutamate-cysteine ligase cata­
with that in the control group (Fig. 7D,E). lytic (GCLC) to protect against oxidative damage and ROS-induced
A previous study showed that DCM may also be associated with oxidative damage (HAS, Alotaibi, Bin-Jumah, Elgebaly, & Mahmoud,
inflammation, and hence, we determined the anti-inflammatory effect of 2019; Jimenez et al., 2018; Tonelli, Chio, & Tuveson, 2018). In our

Fig. 6. Effect of TFMP on pathogenic changes in DCM rats. (A) Representative HE staining (magnification = 200 × ). Scale bar = 100 µm. (B) Representative
Transmission Electron Microscopy images of the left ventricular tissues from the different groups (magnification = 15000 × ). Scale bar = 500 nm.

6
J. Zou et al. Journal of Functional Foods 76 (2021) 104319

Fig. 7. Effect of TFMP on oxidative stress and inflammation in DCM rats. (A) SOD activity, (B) GSH-Px activity, (C) MDA level, (D) Nrf2 mRNA level, (E) HO-1 mRNA
level, (F) IL-6 content, (G) TNF-α content, (H) IL-6 mRNA level, (I) TNF-α mRNA level. #P < 0.05 versus control group; *P < 0.05 versus the DCM group.

results, the damage to cardiac function was related to the increase in the show the effect of TFMP on the cardiac fibrosis in diabetic rats. We
levels of blood glucose, myocardial enzymes, and ROS. TFMP reduced openly accept this as a limitation of our study.
the release of myocardial enzymes and restored cardiac function. As Apoptosis plays a vital role in the pathogenesis of DCM (Ouyang,
expected, TFMP increased the activities of SOD and GSH-Px, and You, & Xie, 2014). In DM rats, hyperglycemia, hyperlipidemia, excessive
decreased the MDA content. Moreover, TFMP activated Nrf2 and HO-1 ROS production, and inflammation could induce apoptosis in the heart
gene expression. These findings demonstrated that TFMP had a protec­ (Althunibat et al., 2019). To further elucidate the protective effect of
tive effect on myocardial damage induced by oxidative stress in DM rats. TFMP, the present study examined its effect on apoptosis. Our results
Hyperglycemia could stimulate the ROS generation, which in turn showed that apoptosis in the heart decreased significantly compared
promotes the secretion of pro-inflammatory cytokines in the diabetic with diabetic rats, indicating that TFMP can alleviate the process of
myocardium(Luo et al., 2019). Additionally, ROS inhibitors dramati­ apoptosis in the heart of DM.
cally reduce the secretion of IL-1β and IL-18(H. Zhang et al., 2018). In summary, our results show that oxidative stress, inflammation,
Moreover, a few studies showed that anti-inflammatory agents prevent and apoptosis play critical roles in the pathophysiology of DCM. This
the development of cardiac injury in DM rats. TNF-α, a pro- study shows for the first time that long-term treatment with TFMP for a
inflammatory cytokine, resulted in cardiac inflammation and dysfunc­ period of 12 weeks clearly exhibits therapeutic potential for the treat­
tion. It also aggravates the inflammatory response by promoting the ment of DCM by decreasing oxidative stress and inhibiting inflammatory
production of other inflammatory cytokines. In our results, the levels of response and apoptosis. Also, inhibition of oxidative stress was related to
IL-6 and TNF-α were increased in the cardiac tissue of DM rats. Treat­ the increased Nrf2 and HO-1 gene expressions. We have already sepa­
ment of diabetic rats with TFMP markedly decreased the level of those rated two monomer compounds (Ja and Jb) in TFMP. Further studies are
inflammatory cytokines in the cardiac tissues. needed to determine the exact molecular mechanism of TFMP.
Cardiac fibrosis is a characteristic of pathological changes in the
diabetic heart (Kai, Kuwahara, Tokuda, & Imaizumi, 2005; Qi, Zheng, 5. Conclusion
Jiang, Yuan, & Dong, 2020). Several studies have shown that the
inflammation and ROS contributes to the pathogenesis of cardiac The present study demonstrated that TFMP alleviated the develop­
fibrosis (Y. Wang et al., 2019; Zheng et al., 2020). TGF β1, an inflam­ ment of DCM in type 2 diabetes rats. The protective effect is related to
matory cytokine, is a key player in the extracellular matrix (ECM) increasing Nrf2 and HO-1 gene expression and inhibiting oxidative
accumulation. Overexperssion of TGF β1 results in the production of stress, inflammation and apoptosis. These findings suggest that TFMP
granulation consisting connective tissue cells and excessive of ECM might have significant therapeutic potential for treating DCM.
(Voelker et al., 2017; Qi et al., 2020). However, the present study did not

7
J. Zou et al. Journal of Functional Foods 76 (2021) 104319

Fig. 8. Effect of TFMP on apoptosis in DCM rats. (A) TUNEL staining showing the apoptotic cells. Scale bar = 100 µm. (B-E) Western blot showing Bax, Bcl-2, and the
ratio of Bax to Bcl-2. *P < 0.05 vs the control group; #P < 0.05 vs the DCM group.

Declaration of Competing Interest Kannel, W. B., Hjortland, M., & Castelli, W. P. (1974). Role of diabetes in congestive
heart failure: The Framingham study. Am J Cardiol, 34(1), 29–34. https://doi.org/
10.1016/0002-9149(74)90089-7.
The authors declare that they have no known competing financial Li, H., Shi, Y., Wang, X., Li, P., Zhang, S., Wu, T., … Wu, R. (2019). Piceatannol alleviates
interests or personal relationships that could have appeared to influence inflammation and oxidative stress via modulation of the Nrf2/HO-1 and NF-kappaB
the work reported in this paper. pathways in diabetic cardiomyopathy. Chem Biol Interact, 310, Article 108754.
https://doi.org/10.1016/j.cbi.2019.108754.
Li, K., Zhai, M., Jiang, L., Song, F., Zhang, B., Li, J., … Wang, S. (2019).
References Tetrahydrocurcumin Ameliorates Diabetic Cardiomyopathy by Attenuating High
Glucose-Induced Oxidative Stress and Fibrosis via Activating the SIRT1 Pathway.
Althunibat, O. Y., Al Hroob, A. M., Abukhalil, M. H., Germoush, M. O., Bin-Jumah, M., & Oxid Med Cell Longev, 2019, 6746907. https://doi.org/10.1155/2019/6746907.
Mahmoud, A. M. (2019). Fisetin ameliorates oxidative stress, inflammation and Liao, H. H., Zhu, J. X., Feng, H., Ni, J., Zhang, N., Chen, S., … Tang, Q. Z. (2017).
apoptosis in diabetic cardiomyopathy. Life Sci, 221, 83–92. https://doi.org/ Myricetin Possesses Potential Protective Effects on Diabetic Cardiomyopathy
10.1016/j.lfs.2019.02.017. through Inhibiting IkappaBalpha/NFkappaB and Enhancing Nrf2/HO-1. Oxid Med
Badran, M. M., Alomrani, A. H., Harisa, G. I., Ashour, A. E., Kumar, A., & Yassin, A. E. Cell Longev, 2017, 8370593. https://doi.org/10.1155/2017/8370593.
(2018). Novel docetaxel chitosan-coated PLGA/PCL nanoparticles with magnified Liu, X. Y., Xu, L., Wang, Y., Li, J. X., Zhang, Y., Zhang, C., … Zhang, X. M. (2017).
cytotoxicity and bioavailability. Biomed Pharmacother, 106, 1461–1468. https://doi. Protective effects of total flavonoids of Astragalus against adjuvant-induced arthritis
org/10.1016/j.biopha.2018.07.102. in rats by regulating OPG/RANKL/NF-kappaB pathway. Int Immunopharmacol, 44,
Faria, A., & Persaud, S. J. (2017). Cardiac oxidative stress in diabetes: Mechanisms and 105–114. https://doi.org/10.1016/j.intimp.2017.01.010.
therapeutic potential. Pharmacol Ther, 172, 50–62. https://doi.org/10.1016/j. Low Wang, C. C., Hess, C. N., Hiatt, W. R., & Goldfine, A. B. (2016). Clinical Update:
pharmthera.2016.11.013. Cardiovascular Disease in Diabetes Mellitus: Atherosclerotic Cardiovascular Disease
Guariguata, L., Whiting, D. R., Hambleton, I., Beagley, J., Linnenkamp, U., & Shaw, J. E. and Heart Failure in Type 2 Diabetes Mellitus - Mechanisms, Management, and
(2014). Global estimates of diabetes prevalence for 2013 and projections for 2035. Clinical Considerations. Circulation, 133(24), 2459–2502. https://doi.org/10.1161/
Diabetes Res Clin Pract, 103(2), 137–149. https://doi.org/10.1016/j. CIRCULATIONAHA.116.022194.
diabres.2013.11.002. Luo, W., Jin, Y., Wu, G., Zhu, W., Qian, Y., Zhang, Y., … Liang, G. (2019). Blockage of
Has, A. L., Alotaibi, M. F., Bin-Jumah, M., Elgebaly, H., & Mahmoud, A. M. (2019). Olea ROS and MAPKs-mediated inflammation via restoring SIRT1 by a new compound
europaea leaf extract up-regulates Nrf2/ARE/HO-1 signaling and attenuates LF10 prevents type 1 diabetic cardiomyopathy. Toxicol Appl Pharmacol, 370, 24–35.
cyclophosphamide-induced oxidative stress, inflammation and apoptosis in rat https://doi.org/10.1016/j.taap.2019.03.005.
kidney. Biomed Pharmacother, 111, 676-685. doi: 10.1016/j.biopha.2018.12.112. Lv, D., Cheng, X., Tang, L., & Jiang, M. (2017). The cardioprotective effect of total
Jimenez, R., Toral, M., Gomez-Guzman, M., Romero, M., Sanchez, M., Mahmoud, A. M., flavonoids on myocardial ischemia/reperfusion in rats. Biomed Pharmacother, 88,
& Duarte, J. (2018). The Role of Nrf2 Signaling in PPARbeta/delta-Mediated 277–284. https://doi.org/10.1016/j.biopha.2017.01.060.
Vascular Protection against Hyperglycemia-Induced Oxidative Stress. Oxid Med Cell Menezes, C. D. A., de Oliveira Garcia, F. A., de Barros Viana, G. S., Pinheiro, P. G.,
Longev, 2018, 5852706. https://doi.org/10.1155/2018/5852706. Felipe, C. F. B., de Albuquerque, T. R., … de Menezes, I. R. A. (2017). Murraya
Kai, H., Kuwahara, F., Tokuda, K., & Imaizumi, T. (2005). Diastolic dysfunction in paniculata (L.) (Orange Jasmine): Potential Nutraceuticals with Ameliorative Effect
hypertensive hearts: Roles of perivascular inflammation and reactive myocardial in Alloxan-Induced Diabetic Rats. Phytother Res, 31(11), 1747–1756. https://doi.
fibrosis. Hypertens Res, 28(6), 483–490. https://doi.org/10.1291/hypres.28.483. org/10.1002/ptr.5903.
Menezes, I. R., Santana, T. I., Varela, V. J., Saraiva, R. A., Matias, E. F., Boligon, A. A., …
Rocha, J. B. (2015). Chemical composition and evaluation of acute toxicological,

8
J. Zou et al. Journal of Functional Foods 76 (2021) 104319

antimicrobial and modulatory resistance of the extract of Murraya paniculata. Pharm Wang, X., Liang, H., Zeng, K., Zhao, M., Tu, P., Li, J., & Jiang, Y. (2019). Panitins A-G:
Biol, 53(2), 185–191. https://doi.org/10.3109/13880209.2014.913068. Coumarin derivatives from Murraya paniculata from Guangxi Province, China show
Ouyang, C., You, J., & Xie, Z. (2014). The interplay between autophagy and apoptosis in variable NO inhibitory activity. Phytochemistry, 162, 224–231. https://doi.org/
the diabetic heart. J Mol Cell Cardiol, 71, 71–80. https://doi.org/10.1016/j. 10.1016/j.phytochem.2019.03.012.
yjmcc.2013.10.014. Wang, X. T., Zeng, K. W., Zhao, M. B., Tu, P. F., Li, J., & Jiang, Y. (2018). Three new
Qi, S. S., Zheng, H. X., Jiang, H., Yuan, L. P., & Dong, L. C. (2020). Protective Effects of indole alkaloid derivatives from the roots of Murraya paniculata. J Asian Nat Prod
Chromium Picolinate Against Diabetic-Induced Renal Dysfunction and Renal Res, 20(3), 201–208. https://doi.org/10.1080/10286020.2017.1327950.
Fibrosis in Streptozotocin-Induced Diabetic Rats. Biomolecules, 10(3). https://doi. Wang, Y., Li, H., Li, Y., Zhao, Y., Xiong, F., Liu, Y., … Wang, L. (2019). Coriolus
org/10.3390/biom10030398. versicolor alleviates diabetic cardiomyopathy by inhibiting cardiac fibrosis and
Qi, S. S., He, J., Dong, L. C., Yuan, L. P., Wu, J. L., Zu, Y. X., & Zheng, H. X. (2020). NLRP3 inflammasome activation. Phytother Res, 33(10), 2737–2748. https://doi.
Cyanidin-3-glucoside from black rice prevents renal dysfunction and renal fibrosis in org/10.1002/ptr.6448.
streptozotocin-diabetic rats. J Funct Foods, 72. https://doi.org/10.1016/j. Wu, J., Liu, K., & Shi, X. (2016). The anti-inflammatory activity of several flavonoids
jff.2020.104062. isolated from Murraya paniculata on murine macrophage cell line and gastric
Rubler, S., Dlugash, J., Yuceoglu, Y. Z., Kumral, T., Branwood, A. W., & Grishman, A. epithelial cell (GES-1). Pharm Biol, 54(5), 868–881. https://doi.org/10.3109/
(1972). New type of cardiomyopathy associated with diabetic glomerulosclerosis. 13880209.2015.1089294.
Am J Cardiol, 30(6), 595–602. https://doi.org/10.1016/0002-9149(72)90595-4. Zhang, H., Chen, X., Zong, B., Yuan, H., Wang, Z., Wei, Y., … Ma, Y. (2018). Gypenosides
Saied, S., Nizami, S. S., & Anis, I. (2008). Two new coumarins from Murraya paniculata. improve diabetic cardiomyopathy by inhibiting ROS-mediated NLRP3
J Asian Nat Prod Res, 10(5–6), 515–519. https://doi.org/10.1080/ inflammasome activation. J Cell Mol Med, 22(9), 4437–4448. https://doi.org/
10286020801967292. 10.1111/jcmm.13743.
Sharma, P., Batra, S., Kumar, A., & Sharma, A. (2017). In vivo antianxiety and Zhang, J. Y., Li, N., Che, Y. Y., Zhang, Y., Liang, S. X., Zhao, M. B., … Tu, P. F. (2011).
antidepressant activity of Murraya paniculata leaf extracts. J Integr Med, 15(4), Characterization of seventy polymethoxylated flavonoids (PMFs) in the leaves of
320–325. https://doi.org/10.1016/S2095-4964(17)60352-2. Murraya paniculata by on-line high-performance liquid chromatography coupled to
Tang, S. G., Liu, X. Y., Wang, S. P., Wang, H. H., Jovanovic, A., & Tan, W. (2019). photodiode array detection and electrospray tandem mass spectrometry. J Pharm
Trimetazidine prevents diabetic cardiomyopathy by inhibiting Nox2/TRPC3- Biomed Anal, 56(5), 950–961. https://doi.org/10.1016/j.jpba.2011.08.019.
induced oxidative stress. J Pharmacol Sci, 139(4), 311–318. https://doi.org/ Zheng, H. X., Qi, S. S., He, J., Hu, C. Y., Han, H., Jiang, H., & Li, X. S. (2020). Cyanidin-3-
10.1016/j.jphs.2019.01.016. glucoside from Black Rice Ameliorates Diabetic Nephropathy via Reducing Blood
Tonelli, C., Chio, I. I. C., & Tuveson, D. A. (2018). Transcriptional Regulation by Nrf2. Glucose, Suppressing Oxidative Stress and Inflammation, and Regulating
Antioxid Redox Signal, 29(17), 1727–1745. https://doi.org/10.1089/ars.2017.7342. Transforming Growth Factor beta1/Smad Expression. J Agric Food Chem, 68(15),
Voelker, J., Berg, P. H., Sheetz, M., Duffin, K., Shen, T., Moser, B., … Lewis, J. B. (2017). 4399–4410. https://doi.org/10.1021/acs.jafc.0c00680.
Anti-TGF-beta1 Antibody Therapy in Patients with Diabetic Nephropathy. J Am Soc Zou, J., Yu, X., Qu, S., Li, X., Jin, Y., & Sui, D. (2014). Protective effect of total flavonoids
Nephrol, 28(3), 953–962. https://doi.org/10.1681/ASN.2015111230. extracted from the leaves of Murraya paniculata (L.) Jack on diabetic nephropathy in
rats. Food Chem Toxicol, 64, 231–237. https://doi.org/10.1016/j.fct.2013.11.043.

You might also like