The Role of Ginsenosides in Myocardial Ischemic Injury-A Systematic Review

Authors

  • Song Wenhao School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, antai University, Yantai, China
  • Wang Quanwei Departments of Cardiovascular Medicine, First Hospital, Jilin University, Changchun, China
  • Fu Wenwen Department of Pharmacology, School of Pharmacy, Jilin University, Changchun, China
  • Zhang Ce School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education,  Yantai University, Yantai, China
  • Wang Tian School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education,  Yantai University, Yantai, China

DOI:

https://doi.org/10.22270/ajprd.v10i6.1191

Keywords:

Curcuma longa, wound healing, topical preparations, clinical trials, randomized control trials

Abstract

Topical herbal formulations are gaining more attention in wound healing due to their safety and efficacy. This is a systematic review of the effects of Curcuma longatopical formulations on wound healing. C. longa is a traditional medicinal plant used due to its antimicrobial, antioxidant, anti-inflammatory, and wound healing properties. Studies carried outas randomized control trials and clinical trials were included for the analysis of this review. Searches were conducted in PubMed, Scopus, Cochrane Central Register of Controlled Trials, and Google scholar (up to March 2022) with 04 studies meeting the inclusion criteria. All randomized control trials and clinical trials have shown a significant improvement in wound healing. The topical formulations containing C. longa as an ingredient have reportedly improved wound healing in cases of eczema, radiodermatitis, acne vulgaris, atopic dermatitis, ichthyosis vulgaris, senile pruritis, and xerotic skin conditions, according to the studies covered in this review.This review identified some evidence that C. longa extract has good effects for wound healing, in accordance with its use in traditional medicine. To evaluate the impact of C. longa on wound healing, including complications, larger, well-designed randomized control trials are required.

 

Downloads

Download data is not yet available.

Author Biographies

Song Wenhao, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, antai University, Yantai, China

School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, antai University, Yantai, China

Wang Quanwei, Departments of Cardiovascular Medicine, First Hospital, Jilin University, Changchun, China

Departments of Cardiovascular Medicine, First Hospital, Jilin University, Changchun, China

Fu Wenwen, Department of Pharmacology, School of Pharmacy, Jilin University, Changchun, China

Department of Pharmacology, School of Pharmacy, Jilin University, Changchun, China

Zhang Ce, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education,  Yantai University, Yantai, China

School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education,  Yantai University, Yantai, China

Wang Tian, School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education,  Yantai University, Yantai, China

School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education,  Yantai University, Yantai, China

References

1. Ginks WR, Sybers HD, Maroko PR, Covell JW, Sobel BE, Ross J, Jr. Coronary artery reperfusion. II. Reduction of myocardial infarct size at 1 week after the coronary occlusion. The Journal of clinical investigation. 1972;51(10):2717-23.
2. Ibáñez B, Heusch G, Ovize M, Van de Werf F. Evolving therapies for myocardial ischemia/reperfusion injury. Journal of the American College of Cardiology. 2015;65(14):1454-71.
3. Zhao ZQ. Oxidative stress-elicited myocardial apoptosis during reperfusion. Current opinion in pharmacology. 2004;4(2):159-65.
4. Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. Cell death. The New England journal of medicine. 2009;361(16):1570-83.
5. Dixon SJ, Stockwell BR. The role of iron and reactive oxygen species in cell death. Nature chemical biology. 2014;10(1):9-17.
6. Duilio C, Ambrosio G, Kuppusamy P, DiPaula A, Becker LC, Zweier JL. Neutrophils are primary source of O2 radicals during reperfusion after prolonged myocardial ischemia. American journal of physiology Heart and circulatory physiology. 2001;280(6):H2649-57.
7. Vinten-Johansen J, Jiang R, Reeves JG, Mykytenko J, Deneve J, Jobe LJ. Inflammation, proinflammatory mediators and myocardial ischemia-reperfusion Injury. Hematology/oncology clinics of North America. 2007;21(1):123-45.
8. Palaniyandi SA, Suh JW, Yang SH. Preparation of Ginseng Extract with Enhanced Levels of Ginsenosides Rg1 and Rb1 Using High Hydrostatic Pressure and Polysaccharide Hydrolases. Pharmacognosy magazine. 2017;13(Suppl 1):S142-s7.
9. Leung KW, Cheung LW, Pon YL, Wong RN, Mak NK, Fan TP, et al. Ginsenoside Rb1 inhibits tube-like structure formation of endothelial cells by regulating pigment epithelium-derived factor through the oestrogen beta receptor. British journal of pharmacology. 2007;152(2):207-15.
10. Lü JM, Yao Q, Chen C. Ginseng compounds: an update on their molecular mechanisms and medical applications. Current vascular pharmacology. 2009;7(3):293-302.
11. Kwok HH, Chan LS, Poon PY, Yue PY, Wong RN. Ginsenoside-Rg1 induces angiogenesis by the inverse regulation of MET tyrosine kinase receptor expression through miR-23a. Toxicology and applied pharmacology. 2015;287(3):276-83.
12. Li H, Xu J, Wang X, Yuan G. Protective effect of ginsenoside Rg1 on lidocaine-induced apoptosis. Molecular medicine reports. 2014;9(2):395-400.
13. Li CY, Deng W, Liao XQ, Deng J, Zhang YK, Wang DX. The effects and mechanism of ginsenoside Rg1 on myocardial remodeling in an animal model of chronic thromboembolic pulmonary hypertension. European journal of medical research. 2013;18(1):16.
14. Zheng Q, Bao XY, Zhu PC, Tong Q, Zheng GQ, Wang Y. Ginsenoside Rb1 for Myocardial Ischemia/Reperfusion Injury: Preclinical Evidence and Possible Mechanisms. Oxidative medicine and cellular longevity. 2017;2017:6313625.
15. Buettner C, Yeh GY, Phillips RS, Mittleman MA, Kaptchuk TJ. Systematic review of the effects of ginseng on cardiovascular risk factors. The Annals of pharmacotherapy. 2006;40(1):83-95.
16. Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. The New England journal of medicine. 2007;357(11):1121-35.
17. Kalogeris T, Baines CP, Krenz M, Korthuis RJ. Cell biology of ischemia/reperfusion injury. International review of cell and molecular biology. 2012;298:229-317.
18. Sabharwal SS, Schumacker PT. Mitochondrial ROS in cancer: initiators, amplifiers or an Achilles' heel? Nature reviews Cancer. 2014;14(11):709-21.
19. Sugamura K, Keaney JF, Jr. Reactive oxygen species in cardiovascular disease. Free radical biology & medicine. 2011;51(5):978-92.
20. Viña J, Borras C, Abdelaziz KM, Garcia-Valles R, Gomez-Cabrera MC. The free radical theory of aging revisited: the cell signaling disruption theory of aging. Antioxidants & redox signaling. 2013;19(8):779-87.
21. Cadenas S. ROS and redox signaling in myocardial ischemia-reperfusion injury and cardioprotection. Free radical biology & medicine. 2018;117:76-89.
22. Raedschelders K, Ansley DM, Chen DD. The cellular and molecular origin of reactive oxygen species generation during myocardial ischemia and reperfusion. Pharmacology & therapeutics. 2012;133(2):230-55.
23. Chen YR, Zweier JL. Cardiac mitochondria and reactive oxygen species generation. Circulation research. 2014;114(3):524-37.
24. Algoet M, Janssens S, Himmelreich U, Gsell W, Pusovnik M, Van den Eynde J, et al. Myocardial ischemia-reperfusion injury and the influence of inflammation. Trends in cardiovascular medicine. 2022.
25. Wallert M, Ziegler M, Wang X, Maluenda A, Xu X, Yap ML, et al. α-Tocopherol preserves cardiac function by reducing oxidative stress and inflammation in ischemia/reperfusion injury. Redox biology. 2019;26:101292.
26. Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 2011;123(6):594-604.
27. Ziegler M, Wang X, Peter K. Platelets in cardiac ischaemia/reperfusion injury: a promising therapeutic target. Cardiovascular research. 2019;115(7):1178-88.
28. Wang J, Wang H, Mou X, Luan M, Zhang X, He X, et al. The Advances on the Protective Effects of Ginsenosides on Myocardial Ischemia and Ischemia-Reperfusion Injury. Mini reviews in medicinal chemistry. 2020;20(16):1610-8.
29. He B, Chen D, Zhang X, Yang R, Yang Y, Chen P, et al. Oxidative Stress and Ginsenosides: An Update on the Molecular Mechanisms. Oxidative medicine and cellular longevity. 2022;2022:9299574.
30. Kim YJ, Zhang D, Yang DC. Biosynthesis and biotechnological production of ginsenosides. Biotechnology advances. 2015;33(6 Pt 1):717-35.
31. Im DS. Pro-Resolving Effect of Ginsenosides as an Anti-Inflammatory Mechanism of Panax ginseng. Biomolecules. 2020;10(3).
32. Ratan ZA, Haidere MF, Hong YH, Park SH, Lee JO, Lee J, et al. Pharmacological potential of ginseng and its major component ginsenosides. Journal of ginseng research. 2021;45(2):199-210.
33. Lu H, Zhou X, Kwok HH, Dong M, Liu Z, Poon PY, et al. Ginsenoside-Rb1-Mediated Anti-angiogenesis via Regulating PEDF and miR-33a through the Activation of PPAR-γ Pathway. Frontiers in pharmacology. 2017;8:783.
34. Jiang L, Yin X, Chen YH, Chen Y, Jiang W, Zheng H, et al. Proteomic analysis reveals ginsenoside Rb1 attenuates myocardial ischemia/reperfusion injury through inhibiting ROS production from mitochondrial complex I. Theranostics. 2021;11(4):1703-20.
35. Zhang C, Han M, Zhang X, Tong H, Sun X, Sun G. Ginsenoside Rb1 Protects Against Diabetic Cardiomyopathy by Regulating the Adipocytokine Pathway. Journal of inflammation research. 2022;15:71-83.
36. Li CY, Yang P, Jiang YL, Lin Z, Pu YW, Xie LQ, et al. Ginsenoside Rb1 attenuates cardiomyocyte apoptosis induced by myocardial ischemia reperfusion injury through mTOR signal pathway. Biomedicine & pharmacotherapy. 2020;125:109913.
37. Qin L, Wang J, Zhao R, Zhang X, Mei Y. Ginsenoside-Rb1 Improved Diabetic Cardiomyopathy through Regulating Calcium Signaling by Alleviating Protein O-GlcNAcylation. Journal of agricultural and food chemistry. 2019;67(51):14074-85.
38. Liu X, Jiang Y, Fu W, Yu X, Sui D. Combination of the ginsenosides Rb3 and Rb2 exerts protective effects against myocardial ischemia reperfusion injury in rats. International journal of molecular medicine. 2020;45(2):519-31.
39. Sun J, Yu X, Huangpu H, Yao F. Ginsenoside Rb3 protects cardiomyocytes against hypoxia/reoxygenation injury via activating the antioxidation signaling pathway of PERK/Nrf2/HMOX1. Biomedicine & pharmacotherapy. 2019;109:254-61.
40. Shi Y, Han B, Yu X, Qu S, Sui D. Ginsenoside Rb3 ameliorates myocardial ischemia-reperfusion injury in rats. Pharmaceutical biology. 2011;49(9):900-6.
41. Wang T, Yu X, Qu S, Xu H, Han B, Sui D. Effect of ginsenoside Rb3 on myocardial injury and heart function impairment induced by isoproterenol in rats. European journal of pharmacology. 2010;636(1-3):121-5.
42. Xue Y, Fu W, Liu Y, Yu P, Sun M, Li X, et al. Ginsenoside Rb2 alleviates myocardial ischemia/reperfusion injury in rats through SIRT1 activation. Journal of food science. 2020;85(11):4039-49.
43. Ma L, Liu H, Xie Z, Yang S, Xu W, Hou J, et al. Ginsenoside Rb3 protects cardiomyocytes against ischemia-reperfusion injury via the inhibition of JNK-mediated NF-κB pathway: a mouse cardiomyocyte model. PloS one. 2014;9(8):e103628.
44. Chen X, Wang Q, Shao M, Ma L, Guo D, Wu Y, et al. Ginsenoside Rb3 regulates energy metabolism and apoptosis in cardiomyocytes via activating PPARα pathway. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2019;120:109487.
45. Lee CH, Kim JH. A review on the medicinal potentials of ginseng and ginsenosides on cardiovascular diseases. Journal of ginseng research. 2014;38(3):161-6.
46. Li L, Wang Y, Guo R, Li S, Ni J, Gao S, et al. Ginsenoside Rg3-loaded, reactive oxygen species-responsive polymeric nanoparticles for alleviating myocardial ischemia-reperfusion injury. Journal of controlled release : official journal of the Controlled Release Society. 2020;317:259-72.
47. Ren B, Feng J, Yang N, Guo Y, Chen C, Qin Q. Ginsenoside Rg3 attenuates angiotensin II-induced myocardial hypertrophy through repressing NLRP3 inflammasome and oxidative stress via modulating SIRT1/NF-κB pathway. International immunopharmacology. 2021;98:107841.
48. Zhang LP, Jiang YC, Yu XF, Xu HL, Li M, Zhao XZ, et al. Ginsenoside Rg3 Improves Cardiac Function after Myocardial Ischemia/Reperfusion via Attenuating Apoptosis and Inflammation. Evidence-based complementary and alternative medicine : eCAM. 2016;2016:6967853.
49. Shi L, Fu W, Xu H, Li S, Yang X, Yang W, et al. Ginsenoside Rc attenuates myocardial ischaemic injury through antioxidative and anti-inflammatory effects. Pharmaceutical biology. 2022;60(1):1038-46.
50. Huang Q, Su H, Qi B, Wang Y, Yan K, Wang X, et al. A SIRT1 Activator, Ginsenoside Rc, Promotes Energy Metabolism in Cardiomyocytes and Neurons. Journal of the American Chemical Society. 2021;143(3):1416-27.
51. Yu HT, Zhen J, Pang B, Gu JN, Wu SS. Ginsenoside Rg1 ameliorates oxidative stress and myocardial apoptosis in streptozotocin-induced diabetic rats. Journal of Zhejiang University Science B. 2015;16(5):344-54.
52. Wang Y, Li X, Wang X, Lau W, Wang Y, Xing Y, et al. Ginsenoside Rd attenuates myocardial ischemia/reperfusion injury via Akt/GSK-3β signaling and inhibition of the mitochondria-dependent apoptotic pathway. PloS one. 2013;8(8):e70956.
53. Lu C, Sun Z, Wang L. Inhibition of L-type Ca(2+) current by ginsenoside Rd in rat ventricular myocytes. Journal of ginseng research. 2015;39(2):169-77.
54. Gai Y, Ma Z, Yu X, Qu S, Sui D. Effect of ginsenoside Rh1 on myocardial injury and heart function in isoproterenol-induced cardiotoxicity in rats. Toxicology mechanisms and methods. 2012;22(8):584-91.
55. Zhu D, Wu L, Li CR, Wang XW, Ma YJ, Zhong ZY, et al. Ginsenoside Rg1 protects rat cardiomyocyte from hypoxia/reoxygenation oxidative injury via antioxidant and intracellular calcium homeostasis. Journal of cellular biochemistry. 2009;108(1):117-24.
56. Li L, Pan CS, Yan L, Cui YC, Liu YY, Mu HN, et al. Ginsenoside Rg1 Ameliorates Rat Myocardial Ischemia-Reperfusion Injury by Modulating Energy Metabolism Pathways. Frontiers in physiology. 2018;9:78.
57. Wei HJ, Yang HH, Chen CH, Lin WW, Chen SC, Lai PH, et al. Gelatin microspheres encapsulated with a nonpeptide angiogenic agent, ginsenoside Rg1, for intramyocardial injection in a rat model with infarcted myocardium. Journal of controlled release : official journal of the Controlled Release Society. 2007;120(1-2):27-34.
58. Fu W, Xu H, Yu X, Lyu C, Tian Y, Guo M, et al. 20(S)-Ginsenoside Rg2 attenuates myocardial ischemia/reperfusion injury by reducing oxidative stress and inflammation: role of SIRT1. RSC advances. 2018;8(42):23947-62.
59. Li Y, Hao H, Yu H, Yu L, Ma H, Zhang H. Ginsenoside Rg2 Ameliorates Myocardial Ischemia/Reperfusion Injury by Regulating TAK1 to Inhibit Necroptosis. Frontiers in cardiovascular medicine. 2022;9:824657.
60. Lim KH, Lim DJ, Kim JH. Ginsenoside-Re ameliorates ischemia and reperfusion injury in the heart: a hemodynamics approach. Journal of ginseng research. 2013;37(3):283-92.
61. Sun H, Ling S, Zhao D, Li J, Li Y, Qu H, et al. Ginsenoside Re Treatment Attenuates Myocardial Hypoxia/Reoxygenation Injury by Inhibiting HIF-1α Ubiquitination. Frontiers in pharmacology. 2020;11:532041.
62. Zhang XH, Xu XX, Xu T. Ginsenoside Ro suppresses interleukin-1β-induced apoptosis and inflammation in rat chondrocytes by inhibiting NF-κB. Chinese journal of natural medicines. 2015;13(4):283-9.
63. Kim S, Oh MH, Kim BS, Kim WI, Cho HS, Park BY, et al. Upregulation of heme oxygenase-1 by ginsenoside Ro attenuates lipopolysaccharide-induced inflammation in macrophage cells. Journal of ginseng research. 2015;39(4):365-70.
64. Xu HL, Chen GH, Wu YT, Xie LP, Tan ZB, Liu B, et al. Ginsenoside Ro, an oleanolic saponin of Panax ginseng, exerts anti-inflammatory effect by direct inhibiting toll like receptor 4 signaling pathway. Journal of ginseng research. 2022;46(1):156-66.

Published

2022-12-14

How to Cite

Wenhao, S., Quanwei, W., Wenwen, F., Ce, Z., & Tian, W. (2022). The Role of Ginsenosides in Myocardial Ischemic Injury-A Systematic Review. Asian Journal of Pharmaceutical Research and Development, 10(6), 61–70. https://doi.org/10.22270/ajprd.v10i6.1191