Next Article in Journal
Acceleration of the Deamination of Cytosine through Photo-Crosslinking
Next Article in Special Issue
Cardiac Toxicities in Oncology: Elucidating the Dark Box in the Era of Precision Medicine
Previous Article in Journal
DDIT4 Downregulation by siRNA Approach Increases the Activity of Proteins Regulating Fatty Acid Metabolism upon Aspirin Treatment in Human Breast Cancer Cells
Previous Article in Special Issue
The Link between Magnesium Supplements and Statin Medication in Dyslipidemic Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Antioxidant Flavonoid Diosmetin Is Cardioprotective in a Rat Model of Myocardial Infarction Induced by Beta 1-Adrenergic Receptors Activation

1
Department of Pharmacy, Abbottabad Campus, COMSATS University Islamabad, University Road, Abbottabad 22060, Pakistan
2
Laboratory of Cardiovascular Research and Integrative Pharmacology, College of Pharmacy, University of Sargodha, University Road, Sargodha 40100, Pakistan
3
Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
*
Authors to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2023, 45(6), 4675-4686; https://doi.org/10.3390/cimb45060297
Submission received: 6 May 2023 / Revised: 23 May 2023 / Accepted: 24 May 2023 / Published: 29 May 2023
(This article belongs to the Special Issue Focus on Molecular Basis of Cardiac Diseases)

Abstract

:
Myocardial infarction (MI) is a common and life-threatening manifestation of ischemic heart diseases (IHD). The most important risk factor for MI is hypertension. Natural products from medicinal plants have gained considerable attention globally due to their preventive and therapeutic effects. Flavonoids have been found to be efficacious in ischemic heart diseases (IHD) by alleviating oxidative stress and beta-1 adrenergic activation, but the mechanistic link is not clear. We hypothesized that antioxidant flavonoid diosmetin is cardioprotective in a rat model of MI induced by beta 1-adrenergic receptor activation. To test this hypothesis, we evaluated the cardioprotective potential of diosmetin on isoproterenol-induced MI in rats by performing lead II electrocardiography (ECG), cardiac biomarkers including troponin I (cTnI) and creatinine phosphokinase (CPK), CK-myocardial band, (CK-MB), lactate dehydrogenase (LDH), alanine aminotransferase (ALT), and aspartate aminotranferase (AST) by using biolyzer 100, as well as histopathological analysis. We found that diosmetin (1 and 3 mg/kg) attenuated isoproterenol-induced elevation in the T-wave and deep Q-wave on the ECG, as well as heart-to-body weight ratio and infarction size. In addition, pretreatment with diosmetin attenuated the isoproterenol-induced increase in serum troponin I. These results demonstrate that flavonoid diosmetin may provide therapeutic benefit in myocardial infarction.

1. Introduction

Myocardial infarction (MI) is life-threatening and the most common manifestation of ischemic heart disease (IHD), also called coronary artery disease (CAD) [1]. According to world health organization (WHO), IHD is one of the main cause of deaths worldwide, responsible for more than nine million deaths in 2016 [2]. Almost 39% of deaths in developing countries under the age of 70 years are due to IHD and MI [3]. IHD leads to many pathological changes, including increase in oxidative stress, inflammation, necrosis, and lipid peroxidation (LPO), and decline in the nitric oxide (NO) signaling pathway [4]. The oxidative stress and uncontrolled free radicals mainly effect the myocardium of the heart by initiating the inflammatory pathway [5].The activation of this pathological pathway also initiates the lipid peroxidative products, which, in response, suppress the level of natural antioxidants, catalase (CAT), glutathione peroxidase (GPx), and superoxide dismutase (SOD), respectively. The initiation of this pathway plays major role in the development of myocardial damage in MI [6]. Atherosclerosis is also reported to actively participate in pathological changes during MI [7]. That is why atherosclerosis is considered to be the most common cause of MI. Hypertension, hyperlipidemia, diabetes, and smoking are major risk factors for atherosclerosis. In addition, inflammation, inhibition of nitric oxide synthase (NOS) activity, and decline in nitric oxide pathway lead to endothelial dysfunction, which initiates atherosclerotic plaque formation [8,9,10].
Furthermore, the expression of calcium channels [11], intracellular calcium load [12], potassium channels inhibition [10], and the increase in the level of cTnI, CPK, CK-MB, LDH, ALT, and AST are important biomarkers [13,14,15]. CTn is the gold standard to assess MI [16].
The cardioprotection mechanism(s) include all those approaches, which contribute to the prevention of pathological changes in myocardium [17]. Therapeutic approaches include drugs such as vasodilator to decrease vascular resistance, adrenergic antagonist to inhibit overactive sympathetic activity, and calcium channel antagonist. Unfortunately, some undesirable effects are also related to this therapeutic approach, such as cluster headaches with nitroglycerine, withdrawal effects and sexual dysfunction with β-adrenergic antagonist, and reflex tachycardia and hypotension with calcium channel blockers (CCBs) [18].
As a substitute approach to search for alternative drugs, many phytochemicals from medicinal plant sources have been reported as demonstrating cardioprotective properties [19,20]. Natural products including flavonoids have been reported for their protective response against IHD [21,22]. Some studies show that flavonoids reduce the risk of hypertension and CVD [23,24]. Other examples of flavonoids include hesperetin, luteolin, and naringenin, to mention but a few [24]. Flavonoids have also been reported to exert multiple biological effects, including cardioprotective [24], antioxidant [25], and anti-inflammatory [26,27].
Concerning diosmetin (5,7,3′-trihydroxy-4′-methoxyflavone), Figure 1 is a monomethoxyflavone, a flavonoid that can be found in lemon peel and citrus fruit [28,29]. Lemon (Citrus limon L.), olives (Olea europaea L.), and Rosmarinus officinalis L. are sources of diosmetin with reported anti-oxidant and cardioprotective activities [30,31,32]. Diosmetin is one of the important phytochemical constituent in these plants [30,31,32,33,34,35]. Diosmetin is reported in anti-oxidant [36,37] hypolipidemic [38], anti-inflammatory [39], antiproliferative, and pro-apoptotic [40] studies. Previous studies have proven the relationship between the mentioned activities and management of MI. Some preliminary studies by Manivannan et al., 2015 and Mo et al., 2020 [41,42] have reported the potential cardioprotective responses of diosmetin, but there is no detailed study available related to the diosmetin effect on beta 1-adrenergic receptors, cardiac biomarkers, ECG pattern, myocardial infarction size and histopathology. Hence, based on clinically reported activities of plant sources and the mentioned pharmacological effects of antioxidant flavonoid diosmetin, the present study was designed.

2. Methods

2.1. Animals Used

Preferably male adult rats (200–250 g) were used in this study and were kept at the animal house at standard conditions. The protocol of this study was approved (17-06-2013 video notification EC/PHM/07-2013/CUI/ATD) by the Ethical Committee (EC), Department of Pharmacy, COMSATS University Islamabad, Abbottabad campus, Pakistan.

2.2. Experimental Protocol

Diosmetin Response against the MI Induced by β1-Adrenergic Receptors Activation

As cited in the literature [43,44,45], the MI was induced by activating beta 1-adrenergic receptors using a synthetic catecholamine, isoproterenol (ISO), at maximal doses (80 mg/kg). The ISO was administered subcutaneously at a gap of 24 h. Before ISO administration, we administered diosmetin (1 and 3 mg/kg) for 4 days, and on the 5th and 6th day, we administered both ISO and diosmetin. The 1% DMSO was used as a solvent of diosmetin. Simultaneously, atenolol at 1 mg/kg was used as a positive standard to compare the response of diosmetin. The details of the five groups (n = 6) are shown in Table 1.
The body weights of rats were measured before and at the end of the experiment. Then anesthesia was induced by using thiopental at a dose of 80–60 mg/kg.
After that, the rats were ready to measure the ECG by using lead II ECG, consisting of needle electrodes connected to BioAmp and PowerLab (AD Instruments, Sydney, Australia). The ECG was analyzed by using Lab Chart 7. To analyze the different MI biomarkers, cTnI, CPK, CK-MB, LDH, AST, and ALT in serum, blood samples were also collected. The standard assay kits were used for analysis and were carried out through Biolyzer 100. To compare the anatomical changes in the hearts of different groups, the heart weight of all the experimental rats were noted [15,46]. The isolated hearts of different groups of rats were then preserved in the 10% formalin solution for histopathological study. For staining H and E, stains were used and heart tissue were mainly investigated under light microscope (200×) for changes in the branched appearance of cardiac muscle fibers, denoting inflammation which is characterized by edema and ultimately leads to necrosis [43,47].

2.3. Statistical Significance Analysis

The GraphPad Prism software version 9.5.1 was used to analyze the data. One-way ANOVA followed by Tukey’s test was applied, to compare the statistical significance of results. The data was declared statistically significant with p less than 0.05.

3. Results

3.1. Electrocardiograph (ECG) Analysis of Different Groups of Experimental Rats

In vehicle (1% DMSO) control group, no significant change in the ECG pattern was observed (Figure 2B). ISO-injected rats showed a significant change in the ST-segment elevation, Q wave, unequal PR interval and tachycardia (Figure 2C). Interestingly, in the atenolol- and diosmetin-treated groups, the changes in the ECG patterns were prevented significantly (Figure 2D–F). The significant different in ST-elevation among different groups is shown in Figure 2D.

3.2. Heart to Body Weight Ratio

No significant change in the body weight (BW) of all experimental rats were observed at the end of experiment. In ISO-alone-injected rats, a significant change in the ratio of heart to BW was witnessed, in comparison to the control group. The atenolol (1 mg/kg) and diosmetin (1 and 3 mg/kg) pre- and co-treated ISO group significantly prevented changes in heart to BW ratio in comparison to the ISO-alone-injected group (Figure 3A).

3.3. Anatomical and Histopathological Analysis

In the ISO-alone-administered group, a significant anatomical change in the heart shape was observed in compared to control group rats. This remodeling of rat heart shape also confirms that MI is successfully induced. The white or pale grey area in cardiac tissues is identified as an infarct area. This infarct area was significantly reduced in atenolol (15%)- and diosmetin 1 mg/kg (24%)- and 3 mg/kg (13%)-administered groups as compared to isoproterenol (55%), as shown in Figure 3B.

3.4. Protective Response of Diosmetin against Alteration in Cardiac Biomarkers

In comparison to control, in ISO-alone-administered rats, a significant escalation was witnessed in the serum levels of cardiac markers including, cTnI, CPK, CK-MB, LDH, AST, and ALT (Figure 4B–G). Atenolol at 1 mg/kg significantly prevented the pathological changes induced by ISO. Moreover, diosmetin (1 and 3 mg/kg) significantly restored the alteration in biomarkers, as compared to the ISO-alone-treated group.

3.5. Diosmetin Prevent Histopathological Changes Induced by ISO

The control group’s myocardium histology shows normal striation with a branched appearance, as shown in Figure 5A, while ISO administration alone at high doses leads to inflammation followed by edema and then ultimately necrosis (Figure 5B). The section of heart tissue sections from atenolol- (1 mg/kg) (Figure 5C) and diosmetin (1 and 3 mg/kg) (Figure 5D,E)-injected groups showed a significant decrease in inflammation, edema and necrosis and the myocardium histology was relatively well protected, with the lowest indication of necrosis compared to the ISO-alone-injected group, as shown in Figure 5F.

4. Discussion

The plant sources of diosmetin are reported for antioxidant and cardioprotective activities [31,32]. We tested the protective response of diosmetin in an ISO-induced MI rat model to confirm its cardioprotective activity. ISO is a non-selective β-adrenergic receptor agonist. Stimulation of β1-adrenergic receptor activation leads to myocardial contractility. High doses of ISO lead to cardiac toxicity. Maximal stimulation of the β1-adrenergic signaling pathway leads to the activation of L-type Ca2+ channels. This influx of calcium, in response, releases more calcium from the intracellular store(s) and increase oxidative stress [46,47]. The major finding of this study is that diosmetin significantly inhibited the pathological changes induced by submaximal administration of isoproterenol. Administration of a 80 mg/kg of ISO produced acute stress in the cardiac muscles of SD rats, resulting in necrosis, ST interval elevation, pathological Q-wave, which, looking deep in the ECG tracing and pathological remodeling of the heart shape, is very similar to those patients who are effected by MI [48,49]. Persistently increased heart rate leads to oxygen demand and speeds up myocardial necrosis [43].
The changes in the ECG pattern are considered one of the key and primary clinical markers to diagnose MI. Its accurate elucidation is commonly the basis for instant therapeutic interventions and different analyses for diagnosis. Variations in the ECG events, such as elevation of the ST-event and the deep Q-wave, may be due to the pathological changes in plasma membrane, which lead to abnormal electrical signaling in myocardium [50,51].
Pre-administration of different doses of diosmetin and atenolol (1 mg/kg) significantly restrained ISO-induced changes in ECG patterns, elevation in the ST-event, and deep Q-wave, proposing membrane protective effects. In comparison, diosmetin highly significantly restored the ECG pattern at a 3 mg/kg dose. The cardioprotective response of diosmetin was further confirmed through assessing the cardiac biomarkers.
Cardiac muscles contain copious amounts of diagnostic biomarkers; after MI, the plasma membrane leakage leads to the release of different markers into systemic circulation [52]. So, to confirm clinically the incidence of MI, validation of the cardiac injury markers is vital [53].
The potent cardiac muscle contraction by ISO initiates the release of biomarker enzymes in response to myocardial damage, hypoxia, and this subsequently leads to necrosis (Figure 6). Moreover, significant changes in the heart-to-body-weight ratio, with abnormal histopathology of cardiac tissue, confirm ISO-induced MI, which is comparable with previously reported studies [54,55,56,57]. Pretreatment with diosmetin and atenolol and co-administration of ISO significantly prevented the change in heart-to-body-wight ration and infarct size when compared to only-ISO-injected rats. The minimum infarct area was observed in animals pretreated with diosmetin (13%) at 3 mg/kg.
Furthermore, diosmetin also significantly decreased the serum levels of cTnI. A significant decline in the cTnI serum level was detected with pretreatment of diosmetin (55%) at 3 mg/kg. cTnI is a gold standard for diagnosing MI [58]. High serum levels of CPK have usually been considered to be an indirect biomarker of cardiac muscle injury. A significant decline in the CPK level was witnessed with 3 mg/kg diosmetin (41%), while diosmetin also significantly decreased (54%) the serum level of CK-MB. One more important marker which was released in serum after MI is CK-MB [59,60]). Another biomarker which indicates damage and inflammation in cardiac tissues is LDH [61]. The diosmetin (3 mg/kg) also significantly decreased (38%) the serum level of LDH. In physiological conditions, the concentration of AST and ALT is low in the serum. However, post-MI pathological changes also include high levels of markers AST and ALT in serum [62]. The diosmetin also significantly decreases (40%, 46%) the serum concentration of AST and ALT in comparison to ISO-alone-administered rats.
Moreover, a significant infarct zone in rat myocardium was observed with ISO-alone treatment, which is characterized by edema, inflammation, necrosis, and the cardiac muscle fibers’ irregular appearance. It is reported that diosmetin inhibits inflammation by resisting expression of interleukin6 (IL-6), phospho-nuclear factor-kappaB (p-NF-κB), and phospho-c-Jun N-terminal kinases (p-JNK), respectively [63]. ISO at submaximal dose is reported to deplete ATP levels, cause Ca2+ overload, increase oxygen demand, and generation of free radicals [64]. However, pre-administration of diosmetin and atenolol counter the pathological changes initiated by ISO injection. Previous studies reported that diosmetin exhibited a significant free-radical-hunting response [36,37]. The diosmetin structure (Figure 1) contains multiple hydroxyl (OH) groups, which also elucidates its capability to prevent free radical damage [65].
The most significant effects of diosmetin were noticed at 3 mg/kg. Plants containing diosmetin have shown increases in levels of SOD, GPX, and CAT [66,67,68]. In addition, diosmetin is known to restore these enzymes in myocardium and inhibit MDA formation [38]. MDA is also considered an important indicator of lipid peroxidation [69]. Lipid peroxidation plays a major role in the generation of oxidative stress [70]. This additional antioxidant effect of diosmetin might complement its cardioprotective potential. The studies by Si et al., 2020 and Mo et al., 2020 [42,71] also reported diosmetin as a cardioprotective agent, decreasing oxidative stress and inducing myocardial apoptosis. Our studies shows that diosmetin blocks the L-type calcium channel and inhibits the release of intracellular calcium in isolated porcine coronary artery and rat aorta [72,73]. So, the cardioprotective response of diosmetin might be due to calcium-channels-blocking activity. The limitation of this study is that more biological markers at molecular level can be traced, such as expression of Bcl-2, Bax, Caspase-3, and proinflammatory factors such as TNF-α in the heart of normal and ISO-administered rats.
Briefly, the data suggest that the pre-administration of diosmetin significantly prevented the pathological changes induced by ISO. The diosmetin significantly preserved the myocardial damage, which is confirmed by the ECG pattern, and the presence of cardiac biomarkers in serum within the range. Further studies would provide more insight into the molecular aspects of these mechanism(s), including the protein expression of Bcl-2, Bax, and Caspase-3 in the myocardium of control and treated rats.

Author Contributions

T.A. and A.J.S. designed the study. T.A. performed experiments and acquired the data. A.J.S., T.K. and A.K., analyzed the data. T.A., A.J.S. and A.K. wrote the manuscript. All authors reviewed and revised the manuscript. A.K. supervised the study. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Institutes of Health grants R01HL147818 (AK and TK), R03HL155041 (AK), and R01HL144941 (AK).

Institutional Review Board Statement

Experiments performed complies with the rulings of the Institute of Laboratory Animal Re-sources, Commission on Life Sciences, National Research Council (NRC, 1996) and approved by the Ethical Committee of Department of Pharmacy, COMSATS University Islamabad, Abbotta-bad campus, in its meeting held on 17-06-2013 video notification EC/PHM/07-2013/CIIT/ATD.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is available on request.

Acknowledgments

We are thankful to the Kirabo Lab for providing access to the BioRender software (https://www.biorender.com/).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Anversa, P.; Sonnenblick, E.H. Ischemic Cardiomyopathy: Pathophysiologic Mechanisms. Prog. Cardiovasc. Dis. 1990, 33, 49–70. [Google Scholar] [CrossRef] [PubMed]
  2. Nowbar, A.N.; Gitto, M.; Howard, J.P.; Francis, D.P.; Al-Lamee, R. Mortality from Ischemic Heart Disease. Circ. Cardiovasc. Qual. Outcomes 2019, 12, e005375. [Google Scholar] [CrossRef] [PubMed]
  3. Gaziano, T.A.; Bitton, A.; Anand, S.; Abrahams-Gessel, S.; Murphy, A. Growing Epidemic of Coronary Heart Disease in Low- and Middle-Income Countries. Curr. Probl. Cardiol. 2010, 35, 72–115. [Google Scholar] [CrossRef] [PubMed]
  4. Hegstad, A.C.; Ytrehus, K.; Myklebust, R.; Jørgensen, L. Ultrastructural Changes in the Myocardial Myocytic Mitochondria: Crucial Step in the Development of Oxygen Radical-Induced Damage in Isolated Rat Hearts? Basic Res. Cardiol. 1994, 89, 128–138. [Google Scholar] [CrossRef]
  5. Misra, M.K.; Sarwat, M.; Bhakuni, P.; Tuteja, R.; Tuteja, N. Oxidative Stress and Ischemic Myocardial Syndromes. Med. Sci. Monit. 2009, 15, RA209–RA219. [Google Scholar]
  6. Wu, J.; Hecker, J.G.; Chiamvimonvat, N. Antioxidant Enzyme Gene Transfer for Ischemic Diseases. Adv. Drug Deliv. Rev. 2009, 61, 351–363. [Google Scholar] [CrossRef]
  7. Rezende, P.C.; Ribas, F.F.; Serrano, C.V.; Hueb, W. Clinical Significance of Chronic Myocardial Ischemia in Coronary Artery Disease Patients. J. Thorac. Dis. 2019, 11, 1005–1015. [Google Scholar] [CrossRef]
  8. Libby, P.; Ridker, P.M.; Maseri, A. Inflammation and Atherosclerosis. Circulation 2002, 105, 1135–1143. [Google Scholar] [CrossRef]
  9. Cunningham, K.S.; Gotlieb, A.I. The Role of Shear Stress in the Pathogenesis of Atherosclerosis. Lab. Investig. J. Tech. Methods Pathol. 2005, 85, 9–23. [Google Scholar] [CrossRef]
  10. Baker, J.E.; Su, J.; Fu, X.; Hsu, A.; Gross, G.J.; Tweddell, J.S.; Hogg, N. Nitrite Confers Protection against Myocardial Infarction: Role of Xanthine Oxidoreductase, NADPH Oxidase and K(ATP) Channels. J. Mol. Cell. Cardiol. 2007, 43, 437–444. [Google Scholar] [CrossRef]
  11. Brunton, L.L.; Hilal-Dandan, R.; Bjorn, K.C. Goodman and Gillman’s The Pharmacological Basis of Therapeutics, 13th ed.; McGraw-Hill Education: New York, NY, USA, 2018. [Google Scholar]
  12. Talukder, M.A.H.; Zweier, J.L.; Periasamy, M. Targeting Calcium Transport in Ischaemic Heart Disease. Cardiovasc. Res. 2009, 84, 345–352. [Google Scholar] [CrossRef] [PubMed]
  13. Kim, J.-E.; Choi, B.-K.; Choi, J.-Y.; Ryu, T.; Roh, W.S.; Song, S.-Y. Role of Calcium Channels Responsible for Phenylephrine-Induced Contraction in Rat Aorta 3 Days after Acute Myocardial Infarction. Korean J. Anesthesiol. 2014, 66, 143–152. [Google Scholar] [CrossRef] [PubMed]
  14. Akila, P.; Vennila, L. Chlorogenic Acid a Dietary Polyphenol Attenuates Isoproterenol Induced Myocardial Oxidative Stress in Rat Myocardium: An in Vivo Study. Biomed. Pharmacother. 2016, 84, 208–214. [Google Scholar] [CrossRef] [PubMed]
  15. Ighodaro, O.M.; Akinloye, O.A. First Line Defence Antioxidants-Superoxide Dismutase (SOD), Catalase (CAT) and Glutathione Peroxidase (GPX): Their Fundamental Role in the Entire Antioxidant Defence Grid. Alex. J. Med. 2018, 54, 287–293. [Google Scholar] [CrossRef]
  16. Mythili, S.; Malathi, N. Diagnostic Markers of Acute Myocardial Infarction. Biomed. Rep. 2015, 3, 743–748. [Google Scholar] [CrossRef]
  17. Kubler, W.; Haass, M. Cardioprotection: Definition, Classification, and Fundamental Principles. Heart 1996, 75, 330–333. [Google Scholar] [CrossRef]
  18. Egred, M.; Shaw, S.; Mohammad, B.; Waitt, P.; Rodrigues, E. Under-Use of Beta-Blockers in Patients with Ischaemic Heart Disease and Concomitant Chronic Obstructive Pulmonary Disease. QJM Mon. J. Assoc. Physicians 2005, 98, 493–497. [Google Scholar] [CrossRef]
  19. Khan, M.; Gilani, A.-H. Studies on Blood Pressure Lowering, Vasodilator and Cardiac Suppressant Activities of Vitex Negundo: Involvement of K+ Channel Activation and Ca++ Channel Blockade. Available online: https://scialert.net/abstract/?doi=ijp.2015.137.142 (accessed on 1 April 2021).
  20. Afsheen, N.; Khalil-ur-Rehman; Jahan, N.; Ijaz, M.; Manzoor, A.; Khan, K.M.; Hina, S. Cardioprotective and Metabolomic Profiling of Selected Medicinal Plants against Oxidative Stress. Oxid. Med. Cell. Longev. 2018, 2018, 9819360. [Google Scholar] [CrossRef]
  21. Patwardhan, B.; Vaidya, A.D.B.; Chorghade, M. Ayurveda and Natural Products Drug Discovery. Curr. Sci. 2004, 86, 789–799. [Google Scholar]
  22. Lahlou, M. The Success of Natural Products in Drug Discovery. Pharmacol. Pharm. 2013, 4, 17–31. [Google Scholar] [CrossRef]
  23. Brüll, V.; Burak, C.; Stoffel-Wagner, B.; Wolffram, S.; Nickenig, G.; Müller, C.; Langguth, P.; Alteheld, B.; Fimmers, R.; Naaf, S.; et al. Effects of a Quercetin-Rich Onion Skin Extract on 24 h Ambulatory Blood Pressure and Endothelial Function in Overweight-to-Obese Patients with (Pre-)Hypertension: A Randomised Double-Blinded Placebo-Controlled Cross-over Trial. Br. J. Nutr. 2015, 114, 1263–1277. [Google Scholar] [CrossRef] [PubMed]
  24. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An Overview. J. Nutr. Sci. 2016, 5, E47. [Google Scholar] [CrossRef] [PubMed]
  25. Meng, Z.; Zhao, J.; Duan, H.; Guan, Y.; Zhao, L. Green and Efficient Extraction of Four Bioactive Flavonoids from Pollen Typhae by Ultrasound-Assisted Deep Eutectic Solvents Extraction. J. Pharm. Biomed. Anal. 2018, 161, 246–253. [Google Scholar] [CrossRef]
  26. Choy, K.W.; Murugan, D.; Leong, X.-F.; Abas, R.; Alias, A.; Mustafa, M.R. Flavonoids as Natural Anti-Inflammatory Agents Targeting Nuclear Factor-Kappa B (NFκB) Signaling in Cardiovascular Diseases: A Mini Review. Front. Pharmacol. 2019, 10, 1295. [Google Scholar] [CrossRef] [PubMed]
  27. Maleki, S.J.; Crespo, J.F.; Cabanillas, B. Anti-Inflammatory Effects of Flavonoids. Food Chem. 2019, 299, 125124. [Google Scholar] [CrossRef]
  28. Doostdar, H.; Burke, M.D.; Mayer, R.T. Bioflavonoids: Selective Substrates and Inhibitors for Cytochrome P450 CYP1A and CYP1B1. Toxicology 2000, 144, 31–38. [Google Scholar] [CrossRef]
  29. Spanakis, M.; Kasmas, S.; Niopas, I. Simultaneous Determination of the Flavonoid Aglycones Diosmetin and Hesperetin in Human Plasma and Urine by a Validated GC/MS Method: In Vivo Metabolic Reduction of Diosmetin to Hesperetin. Biomed. Chromatogr. BMC 2009, 23, 124–131. [Google Scholar] [CrossRef]
  30. Meirinhos, J.; Silva, B.M.; Valentão, P.; Seabra, R.M.; Pereira, J.A.; Dias, A.; Andrade, P.B.; Ferreres, F. Analysis and Quantification of Flavonoidic Compounds from Portuguese Olive (Olea Europaea L.) Leaf Cultivars. Nat. Prod. Res. 2005, 19, 189–195. [Google Scholar] [CrossRef]
  31. Boshtam, M.; Naderi, G.A.; Moshtaghian, J.; Asgary, S.; Jafari, N. Effects of Citrus Limon Burm. f. on Some Atherosclerosis Risk Factors in Rabbits with Atherogenic Diet. Atherosclerosis 2009, 5, 88–93. [Google Scholar]
  32. Riaz, A.; Khan, R.A.; Mirza, T.; Mustansir, T.; Ahmed, M. In Vitro/in Vivo Effect of Citrus Limon (L. Burm. f.) Juice on Blood Parameters, Coagulation and Anticoagulation Factors in Rabbits. Pak. J. Pharm. Sci. 2014, 27, 907–915. [Google Scholar]
  33. Tripoli, E.; La Guardia, M.; Giammanco, S.; Di Majo, D.; Giammanco, M. Citrus Flavonoids: Molecular Structure, Biological Activity and Nutritional Properties: A Review. Food Chem. 2007, 104, 466–479. [Google Scholar] [CrossRef]
  34. Kato, Y.; Domoto, T.; Hiramitsu, M.; Katagiri, T.; Sato, K.; Miyake, Y.; Aoi, S.; Ishihara, K.; Ikeda, H.; Umei, N.; et al. Effect on Blood Pressure of Daily Lemon Ingestion and Walking. J. Nutr. Metab. 2014, 2014, 912684. [Google Scholar] [CrossRef]
  35. Rašković, A.; Milanović, I.; Pavlović, N.; Ćebović, T.; Vukmirović, S.; Mikov, M. Antioxidant Activity of Rosemary (Rosmarinus officinalis L.) Essential Oil and Its Hepatoprotective Potential. BMC Complement. Altern. Med. 2014, 14, 225. [Google Scholar] [CrossRef] [PubMed]
  36. Morel, I.; Lescoat, G.; Cogrel, P.; Sergent, O.; Pasdeloup, N.; Brissot, P.; Cillard, P.; Cillard, J. Antioxidant and Iron-Chelating Activities of the Flavonoids Catechin, Quercetin and Diosmetin on Iron-Loaded Rat Hepatocyte Cultures. Biochem. Pharmacol. 1993, 45, 13–19. [Google Scholar] [CrossRef] [PubMed]
  37. Liao, W.; Ning, Z.; Chen, L.; Wei, Q.; Yuan, E.; Yang, J.; Ren, J. Intracellular Antioxidant Detoxifying Effects of Diosmetin on 2,2-Azobis(2-Amidinopropane) Dihydrochloride (AAPH)-Induced Oxidative Stress through Inhibition of Reactive Oxygen Species Generation. J. Agric. Food Chem. 2014, 62, 8648–8654. [Google Scholar] [CrossRef]
  38. Andreeva, O.A.; Ivashev, M.N.; Ozimina, I.I.; Maslikova, G.V. Diosmetin Glycosides from Caucasian Vetch: Isolation and Study of Biological Activity. Pharm. Chem. J. 1998, 32, 595–597. [Google Scholar] [CrossRef]
  39. Ge, A.; Liu, Y.; Zeng, X.; Kong, H.; Ma, Y.; Zhang, J.; Bai, F.; Huang, M. Effect of Diosmetin on Airway Remodeling in a Murine Model of Chronic Asthma. Acta Biochim. Biophys. Sin. 2015, 47, 604–611. [Google Scholar] [CrossRef]
  40. Chen, Y.; Wang, Y.; Liu, M.; Zhou, B.; Yang, G. Diosmetin Exhibits Anti-Proliferative and Anti-Inflammatory Effects on TNF-α-Stimulated Human Rheumatoid Arthritis Fibroblast-like Synoviocytes through Regulating the Akt and NF-ΚB Signaling Pathways. Phytother. Res. PTR 2020, 34, 1310–1319. [Google Scholar] [CrossRef]
  41. Manivannan, J.; Silambarasan, T.; Kadarkarairaj, R.; Raja, B. Systems Pharmacology and Molecular Docking Strategies Prioritize Natural Molecules as Cardioprotective Agents. RSC Adv. 2015, 5, 77042–77055. [Google Scholar] [CrossRef]
  42. Mo, G.; He, Y.; Zhang, X.; Lei, X.; Luo, Q. Diosmetin Exerts Cardioprotective Effect on Myocardial Ischaemia Injury in Neonatal Rats by Decreasing Oxidative Stress and Myocardial Apoptosis. Clin. Exp. Pharmacol. Physiol. 2020, 47, 1713–1722. [Google Scholar] [CrossRef]
  43. Wang, S.; Tian, S.; Yang, F.; Yang, H.; Yang, X.; Du, G. Cardioprotective Effect of Salvianolic Acid A on Isoproterenol-Induced Myocardial Infarction in Rats. Eur. J. Pharmacol. 2009, 615, 125–132. [Google Scholar] [CrossRef] [PubMed]
  44. Li, H.; Xie, Y.-H.; Yang, Q.; Wang, S.-W.; Zhang, B.-L.; Wang, J.-B.; Cao, W.; Bi, L.-L.; Sun, J.-Y.; Miao, S.; et al. Cardioprotective Effect of Paeonol and Danshensu Combination on Isoproterenol-Induced Myocardial Injury in Rats. PLoS ONE 2012, 7, e48872. [Google Scholar] [CrossRef] [PubMed]
  45. Sahu, B.D.; Anubolu, H.; Koneru, M.; Kumar, J.M.; Kuncha, M.; Rachamalla, S.S.; Sistla, R. Cardioprotective Effect of Embelin on Isoproterenol-Induced Myocardial Injury in Rats: Possible Involvement of Mitochondrial Dysfunction and Apoptosis. Life Sci. 2014, 107, 59–67. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, Y.-J.; Ahn, J.K.; Shin, K.-A.; Kim, C.-H.; Lee, Y.-H.; Park, K.-M. Correlation of Cardiac Markers and Biomarkers With Blood Pressure of Middle-Aged Marathon Runners. J. Clin. Hypertens. 2015, 17, 868–873. [Google Scholar] [CrossRef]
  47. Ahmed, S.; Gul, S.; Ze Jaafar, H.; Moga, M.; Zia-Ul-Haq, M.; Dima, L. Anti-Platelet Effects of Nimesulide in Isoproterenol-Induced Myocardial Ischaemia and Infarction in Rabbits. Acta Cardiol. 2015, 70, 401–408. [Google Scholar] [CrossRef]
  48. Wexler, B.C. Myocardial Infarction in Young vs Old Male Rats: Pathophysiologic Changes. Am. Heart J. 1978, 96, 70–80. [Google Scholar] [CrossRef]
  49. Geng, B.; Chang, L.; Pan, C.; Qi, Y.; Zhao, J.; Pang, Y.; Du, J.; Tang, C. Endogenous Hydrogen Sulfide Regulation of Myocardial Injury Induced by Isoproterenol. Biochem. Biophys. Res. Commun. 2004, 318, 756–763. [Google Scholar] [CrossRef]
  50. Sambhi, M.P.; White, F.N. The Electrocardiogram of the Normal and Hypertensive Rat. Circ. Res. 1960, 8, 129–134. [Google Scholar] [CrossRef]
  51. Rajadurai, M.; Prince, P.S.M. Preventive Effect of Naringin on Cardiac Mitochondrial Enzymes during Isoproterenol-Induced Myocardial Infarction in Rats: A Transmission Electron Microscopic Study. J. Biochem. Mol. Toxicol. 2007, 21, 354–361. [Google Scholar] [CrossRef]
  52. Suchalatha, S.; Shyamala Devi, C.S. Protective Effect of Terminalia Chebula against Experimental Myocardial Injury Induced by Isoproterenol. Indian J. Exp. Biol. 2004, 42, 174–178. [Google Scholar]
  53. Thygesen, K.; Alpert, J.S.; Jaffe, A.S.; Chaitman, B.R.; Bax, J.J.; Morrow, D.A.; White, H.D. Executive Group on behalf of the Joint European Society of Cardiology (ESC)/American College of Cardiology (ACC)/American Heart Association (AHA)/World Heart Federation (WHF) Task Force for the Universal Definition of Myocardial Infarction Fourth Universal Definition of Myocardial Infarction (2018). Circulation 2018, 138, e618–e651. [Google Scholar] [CrossRef] [PubMed]
  54. Sabeena Farvin, K.H.; Anandan, R.; Kumar, S.H.S.; Shiny, K.S.; Sankar, T.V.; Thankappan, T.K. Effect of Squalene on Tissue Defense System in Isoproterenol-Induced Myocardial Infarction in Rats. Pharmacol. Res. 2004, 50, 231–236. [Google Scholar] [CrossRef] [PubMed]
  55. Rajadurai, M.; Prince, P.S.M. Preventive Effect of Naringin on Cardiac Markers, Electrocardiographic Patterns and Lysosomal Hydrolases in Normal and Isoproterenol-Induced Myocardial Infarction in Wistar Rats. Toxicology 2007, 230, 178–188. [Google Scholar] [CrossRef] [PubMed]
  56. Panda, V.S.; Naik, S.R. Cardioprotective Activity of Ginkgo Biloba Phytosomes in Isoproterenol-Induced Myocardial Necrosis in Rats: A Biochemical and Histoarchitectural Evaluation. Exp. Toxicol. Pathol. Off. J. Ges. Toxicol. Pathol. 2008, 60, 397–404. [Google Scholar] [CrossRef] [PubMed]
  57. Roy, S.J.; Prince, P.S.M. Protective Effects of Sinapic Acid on Cardiac Hypertrophy, Dyslipidaemia and Altered Electrocardiogram in Isoproterenol-Induced Myocardial Infarcted Rats. Eur. J. Pharmacol. 2013, 699, 213–218. [Google Scholar] [CrossRef]
  58. Body, R.; McDowell, G.; Carley, S.; Wibberley, C.; Ferguson, J.; Mackway-Jones, K. A FABP-Ulous “rule out” Strategy? Heart Fatty Acid Binding Protein and Troponin for Rapid Exclusion of Acute Myocardial Infarction. Resuscitation 2011, 82, 1041–1046. [Google Scholar] [CrossRef]
  59. Karras, D.J.; Kane, D.L. Serum Markers in the Emergency Department Diagnosis of Acute Myocardial Infarction. Emerg. Med. Clin. N. Am. 2001, 19, 321–337. [Google Scholar] [CrossRef]
  60. Baird, M.F.; Graham, S.M.; Baker, J.S.; Bickerstaff, G.F. Creatine-Kinase- and Exercise-Related Muscle Damage Implications for Muscle Performance and Recovery. J. Nutr. Metab. 2012, 2012, 960363. [Google Scholar] [CrossRef]
  61. Peer, P.A.; Trivedi, P.C.; Nigade, P.B.; Ghaisas, M.M.; Deshpande, A.D. Cardioprotective Effect of Azadirachta Indica A. Juss. on Isoprenaline Induced Myocardial Infarction in Rats. Int. J. Cardiol. 2008, 126, 123–126. [Google Scholar] [CrossRef]
  62. Shen, J.; Zhang, J.; Wen, J.; Ming, Q.; Zhang, J.; Xu, Y. Correlation of Serum Alanine Aminotransferase and Aspartate Aminotransferase with Coronary Heart Disease. Int. J. Clin. Exp. Med. 2015, 8, 4399–4404. [Google Scholar]
  63. Meephat, S.; Prasatthong, P.; Potue, P.; Bunbupha, S.; Pakdeechote, P.; Maneesai, P. Diosmetin Ameliorates Vascular Dysfunction and Remodeling by Modulation of Nrf2/HO-1 and p-JNK/p-NF-ΚB Expression in Hypertensive Rats. Antioxidants 2021, 10, 1487. [Google Scholar] [CrossRef]
  64. Padmanabhan, M.; Prince, P.S.M. Preventive Effect of S-Allylcysteine on Lipid Peroxides and Antioxidants in Normal and Isoproterenol-Induced Cardiotoxicity in Rats: A Histopathological Study. Toxicology 2006, 224, 128–137. [Google Scholar] [CrossRef] [PubMed]
  65. Bendary, E.; Francis, R.R.; Ali, H.M.G.; Sarwat, M.I.; El Hady, S. Antioxidant and Structure–Activity Relationships (SARs) of Some Phenolic and Anilines Compounds. Ann. Agric. Sci. 2013, 58, 173–181. [Google Scholar] [CrossRef]
  66. Safizadeh, M.R.; Rahemi, M.; Aminlari, M. Effect of Postharvest Calcium and Hot-Water Dip Treatments on Catalase, Peroxidase and Superoxide Dismutase in Chilled Lisbon Lemon Fruit. Int. J. Agric. Res. 2007, 2, 440–449. [Google Scholar] [CrossRef]
  67. Rasoul, A.; Maryam, H.G.; Taghi, G.M.; Taghi, L.; Dehghan, R. asle Antioxidant Activity of Oral Administration of Rosmarinus Officinalis Leaves Extract on Rat’s Hippocampus Which Exposed to 6-Hydroxydopamine. Braz. Arch. Biol. Technol. 2016, 59, 354. [Google Scholar] [CrossRef]
  68. Soussi, R.; Hfaiedh, N.; Sakly, M.; Rhouma, K.B. The Aqueous Extract of Olea Europaea Leaves Protects from Haematotoxicity and Kidney Damage Induced by Diclofenac in Swiss Albino Mice. RSC Adv. 2019, 9, 23352–23361. [Google Scholar] [CrossRef] [PubMed]
  69. Godfraind, T. Antioxidant Effects and the Therapeutic Mode of Action of Calcium Channel Blockers in Hypertension and Atherosclerosis. Philos. Trans. R. Soc. B Biol. Sci. 2005, 360, 2259–2272. [Google Scholar] [CrossRef] [PubMed]
  70. Reddy, B.S.; Rao, C.V. Novel Approaches for Colon Cancer Prevention by Cyclooxygenase-2 Inhibitors. J. Environ. Pathol. Toxicol. Oncol. Off. Organ. Int. Soc. Environ. Toxicol. Cancer 2002, 21, 155–164. [Google Scholar] [CrossRef]
  71. Si, Q.; Shi, Y.; Huang, D.; Zhang, N. Diosmetin Alleviates Hypoxia-Induced Myocardial Apoptosis by Inducing Autophagy through AMPK Activation. Mol. Med. Rep. 2020, 22, 1335–1341. [Google Scholar] [CrossRef]
  72. Ahmad, T.; Shah, A.J.; Khan, T.; Roberts, R. Mechanism Underlying the Vasodilation Induced by Diosmetin in Porcine Coronary Artery. Eur. J. Pharmacol. 2020, 884, 173400. [Google Scholar] [CrossRef]
  73. Ahmad, T.; Javed, A.; Khan, T.; Althobaiti, Y.S.; Ullah, A.; Almutairi, F.M.; Shah, A.J. Investigation into the Antihypertensive Effects of Diosmetin and Its Underlying Vascular Mechanisms Using Rat Model. Pharmaceuticals 2022, 15, 951. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Research protocol of cardioprotective activity of diosmetin against. The isoproterenol induced myocardial infarction.
Figure 1. Research protocol of cardioprotective activity of diosmetin against. The isoproterenol induced myocardial infarction.
Cimb 45 00297 g001
Figure 2. Typical electrocardiogram (ECG) tracings. (A) Lead II ECG setup; (B) control group, (C) isoproterenol group; (D) atenolol (1 mg/kg); (E) diosmetin, 1 mg; (F) diosmetin, 3 mg/kg; and (G) ST-segments elevation comparison in tracings (CG). Analysis by 1-way ANOVA with a Tukey multiple comparisons post hoc test. n = 6, expressed as mean ± SEM.
Figure 2. Typical electrocardiogram (ECG) tracings. (A) Lead II ECG setup; (B) control group, (C) isoproterenol group; (D) atenolol (1 mg/kg); (E) diosmetin, 1 mg; (F) diosmetin, 3 mg/kg; and (G) ST-segments elevation comparison in tracings (CG). Analysis by 1-way ANOVA with a Tukey multiple comparisons post hoc test. n = 6, expressed as mean ± SEM.
Cimb 45 00297 g002aCimb 45 00297 g002b
Figure 3. Heart to body weight ratio and infarction size. (A) Body weight/heart weight (BW/HW) ratio of diosmetin- and ISO-treated groups in comparison to the control group. (B) The % infarction size of the heart in control and experimental rats. Results are expressed as the mean ± SEM of six determinations. Analysis by 1-way ANOVA with a Tukey multiple comparisons post hoc test.
Figure 3. Heart to body weight ratio and infarction size. (A) Body weight/heart weight (BW/HW) ratio of diosmetin- and ISO-treated groups in comparison to the control group. (B) The % infarction size of the heart in control and experimental rats. Results are expressed as the mean ± SEM of six determinations. Analysis by 1-way ANOVA with a Tukey multiple comparisons post hoc test.
Cimb 45 00297 g003
Figure 4. Levels of cardiac biomarkers in the serum of control and treated groups. (A) Experimental design of cardiac markers’ estimation in blood serum. Effect of diosmetin and atenolol on cardiac markers’ enzymes in serum of isoproterenol (ISO)-induced ischemic rats; (B) troponin I (cTnI); (C) creatine kinase-MB (CK-MB); (D) creatine phosphokinase (CPK); (E) lactate dehydrogenase (LDH); (F) alanine transaminase (ALT); and (G) aspartate transaminase (AST). Results are expressed as Mean ± SEM (n = 6). Analysis by 1-way ANOVA with a Tukey multiple comparisons post hoc test.
Figure 4. Levels of cardiac biomarkers in the serum of control and treated groups. (A) Experimental design of cardiac markers’ estimation in blood serum. Effect of diosmetin and atenolol on cardiac markers’ enzymes in serum of isoproterenol (ISO)-induced ischemic rats; (B) troponin I (cTnI); (C) creatine kinase-MB (CK-MB); (D) creatine phosphokinase (CPK); (E) lactate dehydrogenase (LDH); (F) alanine transaminase (ALT); and (G) aspartate transaminase (AST). Results are expressed as Mean ± SEM (n = 6). Analysis by 1-way ANOVA with a Tukey multiple comparisons post hoc test.
Cimb 45 00297 g004aCimb 45 00297 g004b
Figure 5. Effect of diosmetin on histopathological changes. Photomicrographs of histopathological examination of heart tissue (H and E, 200×) of the control and ISO-treated experimental rats. The heart section from (A) shows normal cardiomyocytes indicated by arrow; the heart section from (B) shows marked inflammation indicated by the arrow, edema indicated by the arrow head, and confluent necrosis indicated by the asterix; the heart section from (C) revealed moderate reduction in inflammation, edema and necrosis; and the heart section from (D,E) shows normal cardiac fibers with maximal reduction of inflammation edema and without any necrosis. (F) Qualitative analysis of different groups showing the preventive effects of diosmetin and atenolol on the degree of histological changes in comparison to ISO-alone-treated group.
Figure 5. Effect of diosmetin on histopathological changes. Photomicrographs of histopathological examination of heart tissue (H and E, 200×) of the control and ISO-treated experimental rats. The heart section from (A) shows normal cardiomyocytes indicated by arrow; the heart section from (B) shows marked inflammation indicated by the arrow, edema indicated by the arrow head, and confluent necrosis indicated by the asterix; the heart section from (C) revealed moderate reduction in inflammation, edema and necrosis; and the heart section from (D,E) shows normal cardiac fibers with maximal reduction of inflammation edema and without any necrosis. (F) Qualitative analysis of different groups showing the preventive effects of diosmetin and atenolol on the degree of histological changes in comparison to ISO-alone-treated group.
Cimb 45 00297 g005aCimb 45 00297 g005b
Figure 6. Schematic diagram showing the cardioprotective response “θ” of diosmetin against the isoproterenol (ISO)-induced myocardial infarction. Key: ALT, alanine transaminase; AST, aspartate transferase; β1R, β1 adrenergic receptors; CPK, Creatine phosphokinase; CK-MB, Creatine Kinase MB; cTnI, cardiac troponin I; LDH, lactate dehydrogenase; ROS, reactive oxygen species; SR, sarcoplasmic reticulum.
Figure 6. Schematic diagram showing the cardioprotective response “θ” of diosmetin against the isoproterenol (ISO)-induced myocardial infarction. Key: ALT, alanine transaminase; AST, aspartate transferase; β1R, β1 adrenergic receptors; CPK, Creatine phosphokinase; CK-MB, Creatine Kinase MB; cTnI, cardiac troponin I; LDH, lactate dehydrogenase; ROS, reactive oxygen species; SR, sarcoplasmic reticulum.
Cimb 45 00297 g006
Table 1. Distribution of experiment rats in different groups for diosmetin cardioprotective study.
Table 1. Distribution of experiment rats in different groups for diosmetin cardioprotective study.
S.NO.GroupsProcedure
1Control
1 to 5% DMSO (1 mL/kg, i.p., 5 days);
Normal saline (1 mL/kg, s.c.) on 4th and 5th day.
2ISO
1 to 5% DMSO (1 mL/kg) for 5 consecutive days ISO, 80 mg/kg, s.c. on 4th and 5th day, at 24 h gap.
3Atenolol + ISO
This group received atenolol (1 mg/kg i.p) for 5 consecutive days and ISO at 4th and 5th days.
4Diosmetin + ISO
Diosmetin at 1 and 3 mg/kg, i.p. for 5 days and also ISO at 4th and 5th.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ahmad, T.; Khan, T.; Kirabo, A.; Shah, A.J. Antioxidant Flavonoid Diosmetin Is Cardioprotective in a Rat Model of Myocardial Infarction Induced by Beta 1-Adrenergic Receptors Activation. Curr. Issues Mol. Biol. 2023, 45, 4675-4686. https://doi.org/10.3390/cimb45060297

AMA Style

Ahmad T, Khan T, Kirabo A, Shah AJ. Antioxidant Flavonoid Diosmetin Is Cardioprotective in a Rat Model of Myocardial Infarction Induced by Beta 1-Adrenergic Receptors Activation. Current Issues in Molecular Biology. 2023; 45(6):4675-4686. https://doi.org/10.3390/cimb45060297

Chicago/Turabian Style

Ahmad, Taseer, Taous Khan, Annet Kirabo, and Abdul Jabbar Shah. 2023. "Antioxidant Flavonoid Diosmetin Is Cardioprotective in a Rat Model of Myocardial Infarction Induced by Beta 1-Adrenergic Receptors Activation" Current Issues in Molecular Biology 45, no. 6: 4675-4686. https://doi.org/10.3390/cimb45060297

Article Metrics

Back to TopTop