PHYTOCHEMICAL CONSTITUENTS AND ANTI-ATHEROSCLEROTIC ACTIVITIES OF A POLYHERBAL FORMULATION –GSTC
HTML Full TextPHYTOCHEMICAL CONSTITUENTS AND ANTI-ATHEROSCLEROTIC ACTIVITIES OF A POLYHERBAL FORMULATION –GSTC
G. Sathyapooja *, S. K. Arun, J. Aswin, N. Hariharan and K. Loganathan
Department of Pharmaceutical Chemistry, KMCH College of Pharmacy, Coimbatore, Tamil Nadu, India.
ABSTRACT: GSTC is a polyherbal formulation comprising Commiphora mukul, Salacia reticulata, Terminalia arjuna, and Curcuma longa, developed to address atherosclerosis through multi-targeted mechanisms. Its phytochemical constituents include guggulsterones, mangiferin, arjunolic acid, and curcuminoids, each offering diverse pharmacological actions. Guggulsterones from C. mukul are recognized for their hypolipidemic and anti-inflammatory effects, while mangiferin and salacinol from S. reticulata provide antidiabetic, anti-obesity, and cholesterol-lowering benefits. T. arjuna contributes arjunolic acid and phytosterols, known for cardioprotective, antioxidant, and anti-inflammatory properties. Curcuminoids and ar-turmerone from C. longa exhibit potent antioxidant, anti-inflammatory, and hypolipidemic actions. Pharmacological studies demonstrate GSTC’s efficacy in reducing cholesterol, triglycerides, LDL, and VLDL levels while elevating HDL in high-fat diet models. Its mechanisms include inhibition of lipid peroxidation, regulation of HMG-CoA reductase, and modulation of inflammatory pathways such as NF-κB and COX-2. The formulation also prevents fatty liver accumulation and enhances lipid metabolism, GSTC exhibits significant antioxidant activity and platelet peroxidation inhibition. GSTC’s unique composition and pharmacological profile suggest its potential as a safe and effective therapeutic agent for managing atherosclerosis and related cardiovascular conditions.
Keywords: GSTC, Atherosclerosis, Phytoconstituents, Hypolipidemic, Polyherbals
INTRODUCTION: Global health challenges are becoming increasingly urgent, with complex diseases like cancer, diabetes and atherosclerosis often caused by a complete regulatory network malfunction rather than a single gene malfunction 1. To diagnose and treat these disorders, innovative approaches must be developed to target the entire biological networks underlying the disease. Understanding the molecular pathways governing disease prognosis is critical in the fight against complicated diseases 2.
Natural products, such as Ayurveda, Unani, and Chinese, are used in pharmaceutical agents, particularly in disease treatments 3. High-throughput techniques have improved the screening of herbal medicines in drug discovery. The concept of developing multi-target drugs against complex diseases is rapidly growing in drug discovery 4. In an attempt to develop an anti-atherosclerotic drug candidate, a polyherbal formulation containing four different herbal plant materials were prepared, based on their scientifically proven efficacy in reducing atherosclerosis risks 5.
Atherosclerosis is a chronic degenerative disease that causes high morbidity and mortality due to its clinical repercussions, including angina pectoris, acute myocardial infarction, stroke, and peripheral vascular insufficiency 6.
In 2020, 28% of people aged 30–79 had abnormal carotid intima-media thickness (≥1.0 mm), over 21% had carotid plaque, and 1.5% had carotid stenosis affecting more than one billion, 816 million, and 58 million individuals, respectively. These conditions were more common in older adults and men 7. The disease is characterized by the accumulation of lipid materials in the arterial wall due to autoimmune and inflammation mechanisms. Hyperlipidemia, hypertension, and diabetes mellitus are important risk factors in atherogenesis. Current treatment strategies focus on lowering cholesterol using statins, but high doses present side effects such as muscle tissue breakdown 8.
The polyherbal formulation GSTC is a suspension made up of extracts from gum resin of Commiphora mukul, root bark of Salacia reticulata, bark of Terminalia arjuna and rhizome of Curcuma longa. Experimental evidence has revealed the antioxidant, hypercholesterolemic effect of C. mukul, which is used in medoroga and inflammatory conditions. T. arjuna has been well documented for its antithrombotic efficacy, with phytosterols and glycosides being the active component. S. reticulata is known as an antidiabetic plant, preventing intestinal absorption of sugars and interfering in sugar metabolism.
C. longa is a natural ingredient in culinary preparations, and the rhizome is known for its antioxidant, anti-inflammatory, and hypolipidemic properties 9.
GSTC has been found to be a good antioxidant and ant-inflammatory agent, as well as anti-atherosclerotic potential, preventing serum oxidation, platelet peroxidation, and hypolipidemic activity in high fat-diet (hfd) fed rats. Based on these anti-atherosclerotic activities and nontoxic nature, GSTC could prove to be a fruitful drug candidate against atherosclerosis. GSTC has showed the most potent activity, with an IC50 of 50 µg/mL, indicating strong inhibition of platelet peroxidation and the cyclooxygenase pathway. GSTC was effective in preventing serum lipid oxidation and lowering cholesterol, triglycerides, LDL, and VLDL levels in rats fed a high-fat diet, similar to atorvastatin. Unlike atorvastatin, GSTC also reduced lipid peroxidation and fatty liver accumulation, suggesting enhanced lipid metabolism. Additionally, GSTC improved HMG-CoA reductase regulation, indicating cholesterol biosynthesis inhibition. It exhibited antioxidant, anti-inflammatory, and non-toxic properties, making it a promising candidate for future atherosclerotic drug development 10.
FIG. 1: POLYHERBAL FORMULATION COMPRISING COMMIPHORA MUKUL, SALACIA RETICULATA, TERMINALIA ARJUNA AND CURCUMA LONGA
Phytoconstituents and its Therapeutic Potential:
Commiphora mukul: Guggul, derived from the resin of Commiphora wightii (also known as Commiphora mukul) belonging to the Burseraceae family, characterized by its prostrate growth, whitish bark, and serrated, non-hairy, trifoliate leaves, is highly valued in Ayurvedic medicine 11, 12. Indigenous to India, it also thrives in the wild across various states of India, Afghanistan, Arabia, and northeastern Africa. The Atharva Veda, refers to Guggul as 'Krimighna,' praised for its fragrant properties that repel parasites. This ancient text, along with other significant Ayurvedic works such as the Charaka Samhita, Sushruta Samhita, and writings by Vagbhata, detail the drug's applications and benefits 13. The gum resin of Guggul, obtained by drying the white sap of the Balsamodendron mukul tree, is utilized in Indian folk medicine for alleviating inflammation 14, arthritis 15, reducing fat, mending bone fractures 16, atherosclerosis, obesity, and hyperlipidemia 17. It is traditionally administered as 'Yog,' combined with other substances and often accompanied by castor oil or Indian spices 18. The sesquiterpene elements present in Myrrh (akin to Guggul), furanoeudesma-1,3-diene and curzarene, exhibit analgesic properties 19. However, due to its toxicity, myrrh is seldom used in modern medicine, except as a mouthwash in India. The Ethiopian resin "agarsu" is used to protect livestock from ticks and has medicinal properties like antimalarial, cold prevention, and wound healing 20. Guggulsterone, a component of Guggul, is known to stimulate fat-breaking enzymes, inhibits the cholesterol production in the liver, and lowers the serum LDL and cholesterol levels 21. The major chemical compounds of Commiphora mukul are listed in Table 1 22-26.
TABLE 1: PHYTOCONSTITUENTS OF COMMIPHORA MUKUL
Compound Name | PubChem CID | Compound Name | PubChem CID |
Dehydroguggulsterone-M | 73088872 | 3,4-dihyroxybenzoic acid | 72 |
Verbenone | 29025 | (20r)-20-Hydroxypregn-4-En-3-One | 249866 |
Diasesartemin | 3732009 | Eicosane-1-2-3-4-tetraol | 14352754 |
Diayangambin | 99091 | Docosane-1-2-3-4-tetraol | 14352765 |
Dihydro guggulsterone-M | 5316451 | Curzerene | 572766 |
D-limonene | 22311 | ∆3 -carene | 26049 |
Eicosan-1,2,3,4-tetrol | 14352754 | 16-α-hydroxy-pregn-4-en-3-one | 69232409 |
Ellagic acid | 5281855 | 1-triacontanol | 68972 |
Epiexcelsin | 14707487 | Phelligridin-D | 85115053 |
Epi-magnolin | 5319210 | Phellinstatin | 76212065 |
Epoxyprogesterone | 538463 | Picropolygamain | 78171395 |
Ergosterol peroxide | 633877 | Eugenol | 3314 |
Ferulic acid | 709 | 20,22-dihydroxycholest-4-en-3-one | 73744000 |
Furanodien-6-one | 6506548 | Furanoeudesma-1,3,-diene | 13874240 |
Geraniol | 4458 | 3,7,7-trimethylcyclohepta-1,3,5-triene | 576718 |
Guggulsterol I | 5250524 | 3α-Acetoxy-5α- Pregnan-16-One | 86182527 |
Guggulsterol Y | 5317852 | 3α-Acetyloxy-5α-Pregnan-16-One | 15767893 |
Guggulsterol-IV | 73149915 | 3-o-(1"8"14"trimethylhexadecanyl)-naringenin | 5319966 |
Guggulsterone - E | 6439929 | 4-o-methyl-d-glucuronic acid | 18186221 |
Guggulsterone - M | 643658 | 4-pregnene-3,16-dione | 163099085 |
Guggultetrol-18 | 13964481 | Adenosine | 191 |
Hexadecane-1-2-3-4-tetraol | 554098 | Aldobiouronic acid | 157009990 |
Hispidin | 54722180 | α -camphorene | 101750 |
Hypholomine-B | 76212349 | α -copaene | 19725 |
Interfungin-A | 76211575 | α -humulene | 23204 |
L-arabinose | 229 | α -phellandren-8-ol | 519323 |
L-fructose | 1101 | α -pinene | 6654 |
Linalool | 6549 | α -terpineol | 17100 |
Lindestrene | 12311269 | α -terpinyl acetate | 111037 |
Longifolene | 289151 | α -thujene | 17868 |
Mansumbinoic acid | 53462065 | β -bisabolene | 403919 |
Mansumbinone | 53420917 | β -caryophyllene | 26318 |
Meta-cymen-8-ol | 255195 | β -elemene | 10583 |
Methyldavallialactone | 76211550 | β -pinene | 14896 |
Methylheptanone | 246728 | β -sitosterol | 86821 |
Myrcene | 31253 | Bornyl acetate | 6448 |
Myrrhanol A | 42608309 | Cadinene | 78298939 |
Guggulsterol-V | 633464 | Caffeic acid | 2518 |
Myrrhanol-B | 74961077 | Campesterol | 312822 |
Myrrhanolide C | 74831241 | Cembrene-A | 328947 |
Myrrhanolide-A | 74831239 | Cholesterol | 304 |
Myrrhone | 78061679 | Cineole | 2758 |
Myrtenol | 10582 | Commiferin | 5316022 |
Naringenin | 932 | Commiphorin | 85037448 |
Nonadecan-1,2,3,4-tetrol | 14352756 | Commiphotetrol | 162977317 |
Octadecane-1-2-3-4-tetraol | 13964481 | Curzerenone | 5315433 |
Para-cymene | 7463 | Cycloartane | 633926 |
Phellifuropyranone-A | 91539906 | D-α-phellandrene | 7460 |
Phelligridin-C | 162875977 | Protocatechualdehyde | 8768 |
Pluviatilol | 130679 | Sabinene | 18818 |
Pregna-1,4-diene-3,16-dione | 67237052 | Stigmasterol | 122544 |
Trans-pinocarveol | 102667 | Terpinen-4-ol | 11230 |
FIG. 2: CHEMICAL CONSTITUENTS OF COMMIPHORA MUKUL
TABLE 2: POTENTIAL ACTIVITIES OF COMMIPHORA MUKUL
Compound | Activity | Study model | Observation |
Naringenin | Antihyperlipidemic, Hepatoprotective | Ldlr deficient homozygous mice | Prevent hypercholesterolemia, hypertriglyceridemia, and hyperinsulinemia; and reduced hepatic steatosis [27]. |
Anti-atherosclerotic | HUVEC cell line | Alleviates the adhesion of THP-1 monocytes by inducing NF-κB signaling pathway [28]. | |
Anti-atherosclerotic | Ldlr deficient homozygous mice | Alleviate cholesterol levels and TG level and plaques due to macrophages. Enhanced the metabolic correction of obesity, steatosis, and insulin resistance [29]. | |
Terpinen-4-ol | Ameliorative effect | In-vitro, In-vivo - Vascular Calcification mice model | Ameliorates the Vascular calcification by upregulating the SIRT1 expression [30]. |
Guggulsterone | Antioxidant | Male Sprague dawley rats | Inhibits the stress due to ROS and inflammatory mediators release in Anaerobic respiration [31]. |
Cardioprotective & Antioxidant | Isoproterenol induced mycocardial Ischemia in rats | Ameliorated the oxidative lipid degradation in human LDL and rat hepatic microsomes [32]. | |
Farnesoid receptor Antagonist | In-silico model | high ligand receptor interaction with Farnesoid receptor [33]. | |
Diayangambin | Myeloperoxidase activity | Murine macrophage cell line | Reduces the leukocyte infiltration and prostaglandin level E2 [34]. |
Campesterol | Cholesterol absorption inhibitor | Male spraguedawley Rats | Activates PPARα and reduces the uptake of cholesterol in liver by reducing SREBP-1 expression [35]. |
Myrrhanol A | Anti-inflammatory effect | Adjuvant induced air pouch by mice granuloma and angiogenesis. | More potent than hydrocortisone [36]. |
Guggulsterol Y | Anti-inflammatory | In-vitro-LPS induced macrophages cell line | Inhibits the NO Formation [37]. |
Protocatechualdehyde | Anti-apoptotic effect, Cardioprotective | In-vitro, In-vivo- myocardial fibrosis mice model | inhibits cardiomyocyte apoptosis via blocking ER stress [38]. |
β-elemene | Improves enothelial dysfunction | Human umbilical artery endothelial cell line | Reduce the ROS, NO level, phosphorylates ERK, and Akt [39]. |
Anti-inflammatory & Cardioprotective | In-vitro OGD/R - induced H9C2 mouse model | Improves the heart function and decreases lipid disposition [40]. | |
Quercetin | Endothelial protection | In-vivo - Postmenopausal women | Protects the LDL against oxidation [41]. |
Anti-inflammatory | HUVEC cell line | Attenuates caveolin-1 expression in HUVEC cell line [42]. | |
Anti-inflammatory | HUVEC cell line | Downregulates MCP-1 expression and diminishes NF-κB p65 subunit translocation by attenuating TLR-NF-κB signaling pathway [43]. | |
Anti-apoptosis | EA. hy926cells | Regulate Akt/GSK3β signalling pathway [44]. | |
Antihyperlipidemic | Human Hep G2 cell line | Increased selective influx of HDL by enhanced SR-BI expression by stimulating the PPARγ/LXRα pathway [45]. | |
Antihyperlipidemic | THP-1 macrophages cell line | Increases ABCA1 expression and cholesterol release through LXRα pathway [46]. |
Salacia reticulata: Kothala himbatu (Salacia reticulata Wight) is a substantial woody climbing shrub characterised by greenish-brown bark, belonging to the Hippocrateaceae family. It is indigenous to Sri Lanka and the southern region of India, with other species such as S. chinensis and S. oblonga also distributed across Asia and various global regions 47, 48. In Ayurvedic medicine, it is sometimes referred to as "Ponkoranti". Species of Salacia, including S. oblonga, S. prinoides, and S. reticulata, have been utilised for millennia in traditional medicine, especially for diabetes management 49, 50. Salacia species have recently been utilised in Japan, the United States, and other nations as a dietary supplement for the prevention of obesity and diabetes 51, 52. In the Ayurvedic system of traditional medicine, the roots and stems of S. reticulata and S. oblonga have been extensively employed to treat rheumatism, gonorrhoea, skin maladies, and as a specific remedy for the initial stages of diabetes. Decoctions of S. reticulata and extracts from other Salacia species have been utilised for centuries to treat asthma, rheumatism, haemorrhoids, pruritus and oedema, gonorrhoea, dermatological conditions, and amenorrhoea 53, 54, 55. The roots possess acrid, bitter, thermogenic, diuretic, astringent, analgesic, and anti-inflammatory properties 56, 57. The interest in Salacia extracts has surged recently due to the escalating prevalence of diabetes and pre-diabetes, the demand for safe and effective pharmaceuticals and functional foods that aid in regulating blood sugar and lipid levels, and the diverse mechanisms of action exhibited by Salacia extracts 58. The principal chemical elements of Salacia reticulata root are shown in Table 3 59-66.
TABLE 3: PHYTOCONSTITUENTS OF SALACIA RETICULATA
Compound | PubChem CID | Compound | PubChem CID |
Triptotriterpenic Acid A | 5257562 | Beta-Amyrin | 225689 |
Galactitol | 453 | Isoiguesterin | 157614 |
Glycerin | 753 | Celastrol | 4274774 |
Mangiferin | 5358385 | Pristimerin | 264268 |
Leucopelargonidin | 3286789 | Isoiguesterol | 72962773 |
Anthocyanidins | 145858 | Netzahualcoyene | 188842 |
Neokotalanol | 44514358 | Iguesterin | 162727 |
Kotalanol | 18423720 | 19-Hydroxyferruginol | 240051 |
Neoponkoranol | 46187831 | Lambertic Acid | 241938 |
Ponkoranol | 6918817 | 29-Hydroxyfriedelan-3-One | 588284 |
Salacinol | 18730125 | Macquarimicin C | 72966725 |
Neosalaprinol | 52938732 | 22-Hydroxytingenone | 500289 |
Alaprinol | 25110936 | Tingenone | 3527193 |
FIG. 3: CHEMICAL CONSTITUENTS OF SALACIA RETICULATA
TABLE 4: POTENTIAL ACTIVITIES OF SALACIA RETICULATA
Compounds | Activity | Study design | Observation |
Mangiferin | Cardioprotective | In- vitro, in -vivo-mice fed with HFD | Diminishes the size of atherosclerotic plaques, lowers LDL, TG and total cholesterol level, while improving reverse cholesterol transport efficiency and enhances HDL level [67]. |
Anti-inflammatory and cholesterol-lowering | Atherogenic mice model fed a high-choline diet | diminished inflammation and decreased plasma total cholesterol levels, leading to a diminution of aortic plaque.[68]. | |
Anti-inflammatory | Mangiferin‑stimulated PVAT-derived exosomes on endothelial function | Alleviates inflammation‑induced endothelial dysfunction via altering NF‑κb signalling pathway [69] | |
(-)-Epicatechins | Anti-atherogenic | Blood epigenetic profiles in male smokers | Promote vascular function by reprogramming endothelial-immune cell signalling and reversing low-grade inflammation [70]. |
Anticoagulant and pro-fibrinolytic | In- vitro global assays mimicking the complex
In -vivo haemostasis systems |
Reduces platelet function [71]. | |
Anti-atherogenic | In-vitro and In-vivo aging model, | Alters age-related decline in eNOS functionality and enhances endothelial function.[72]. | |
Cardioprotective | In-vivo male healthy albino wistar rats | Suppressed tachycardia, cardiac hypertrophy, and the NF-κb inflammatory pathway, thereby safeguarding the heart.[73] | |
Lowers cholestrol | In vivo rat fed with HFHC diet | Alleviates LDL & TAG level and an increase in HDL levels [74]. | |
Celastrol | Lowers cholestrol | In -vitro analysis | Suppresses lipid accumulation, inhibits lipid storage induced by autophagy in VSMCS [75]. |
Anti-inflammatory | In -vivo female mice | Reduced inflammatory cells in plaque area [76]. | |
Anti-inflammatory and anticoagulant | Thrombogenic mice model with HFD-fed mice | Attenuates HFD-induced inflammation, platelet clustering and thrombus formation [77]. | |
Anti-atherogenic | In-vitro, in-vivo rabbit model | Alleviate calcific aortic valve disease [78]. | |
Anti-inflammatory | In -vitro, macrophage inflammation model | Regulates mitochondrial homeostasis and inhibiting of inflammatory responses [79]. | |
Anti-atherogenic | In -vitro, in -vivo mice | Prevent restenosis by inhibition of the intimal hyperplasia and hyperproliferation of vsmcs [80]. | |
Pristimerin | Anti-inflammatory and anti-atherogenic and | Acute lung inflammation model – in-vitro, in-vivo | Inhibits monocyte adherence on endothelium and leukocyte transmigration by downregulating the expression of ICAM-1, VCAM-1 and the pro-inflammatory cytokine [81]. |
Terminalia arjuna: T. arjuna is often described as Arjuna, Indradru, Partha, and Veeravriksha. It belongs to Combretaceae family, having 200 species found all around the world 82. The arjuna tree, which reaches a height of 60 to 80 feet, is found in the Indo-Himalayan regions. Although it can thrive in about any kind of soil, it prefers red lateritic and damp loam soils. It is propagated by seeds 83. Indigenous medical systems employ plant components including T. arjuna's fruits, leaves, and stem bark to address a range of ailments. It has been discovered that the powdered bark has the following benefits: it is hypocholesterolemic, hypo-ischemic, antioxidant, antibacterial, anti-inflammatory, immunomodulatory, and antinociceptive 84.
Terminalia arjuna's bark is smooth, pinkish-grey on the exterior, curved, roughly flat. Each piece can vary in length by up to 15 cm, in breadth by up to 10 cm, and in thickness by up to 10 mm. Heartwood is brown, while sapwood is reddish-white. The bark has a uni-layered epidermis with hair-like projections and a few dispersed lenticels, as revealed by histopathological analysis. Epidermis is covered with a thin layered cortex. The bark contains secondary phloem and periderm 85. Traditional methods of using the stem bark (asava) for cardiac diseases involve either making an alcoholic preparation of it or giving it along with purified boiled milk (kshirpak) or butter (ghrita). There are other formulations on the market with suggested dose of bark juice, notably Arjunarishta, Shankara vati, and Kakubhadi kshira 86.
When T. arjuna bark was initially recorded, it had 34% ash value and composed of calcium carbonate. While only colouring matter and tannins were present in the alcoholic extract, the aqueous extract showed calcium salts (23%) and tannins (16%). Subsequent analysis of the bark revealed the presence of sugar, colouring materials, a glycoside and calcium carbonates, and trace amounts of alkali metal chloride. Subsequently, the existence of a glycoside and an alkaloid was verified. Glycoside isolation produced a high melting point organic acid, phytosterols, 12% tannins, calcium concentrations, trace amounts of magnesium and aluminium salts, sugars 87. The primary chemical components of T.arjuna are listed in Table 5 88-95.
TABLE 5: PHYTOCONSTITUENTS OF TERMINALIA ARJUNA
Compound | PubChem CID | Compound | PubChem CID |
Gallic Acid | 370 | 9,12,15-Octadecatrienoic Acid, Methyl -Ester | 5319706 |
Oxalic-Acid | 971 | Pyrocatechol | 289 |
Catechin | 1203 | Quadranoside VIII | 10675744 |
(+)-Gallocatechol | 1249 | Arjungenin | 12444386 |
Protriptyline | 4976 | Arjunolitin | 13518118 |
Butanoic Acid, 2,3 Dihydroxypropyl Ester | 11188 | Terminoic-Acid | 69569061 |
Ethyl Gallate | 13250 | Kajiichigoside F1 | 14019178 |
Pelargonidin | 67249 | Arjunone | 14034821 |
(+)-Leucocyanidin | 71629 | Casuariin | 14035442 |
Arjunolic-Acid | 73641 | Arjunglucoside I | 14658050 |
2-Naphthalene Methanol | 74128 | Arjunic-Acid | 15385516 |
1-Methoxyhexane | 78484 | Punicalagin | 16129869 |
Friedelin | 91472 | Terchebulin | 16175789 |
Castalagin | 168165 | Arjunetin | 21152828 |
Beta-Sitosterol | 222284 | Ellagic-Acid | 5281855 |
Heptadecane, 9-Hexyl | 296566 | Terminic Acid | 132568257 |
Casuarinin | 442673 | 3-O-Methyl-Ellagic Acid 4-O-Β-D-Xylopyranoside | 25156981 |
Arabinitol, Pentakis-O-(Trimethylsilyl) | 518901 | Arjunglucoside-Ii | 52951052 |
D-Xylose, Tetrakis(Trimethylsilyl) | 529416 | Arjunolone | 71625126 |
9-Oximino 2,7-Diethoxyfluorene | 547102 | Punicalin | 92131301 |
3-Hydroxyspirost-8-En-11-One | 628694 | Terflavin-C | 101589227 |
Quercetin | 5280343 | Arjunglucoside-Iii | 102117122 |
Luteolin | 5280445 | Arjunglucoside V | 102272757 |
Kampferol | 5280863 | Arjunin | 102316370 |
Baicalein | 5281605 | Psidinin-C | 131752695 |
FIG. 4: CHEMICAL CONSTITUENTS OF TERMINALIA ARJUNA
TABLE 6: POTENTIAL ACTIVITIES OF TERMINALIA ARJUNA
Compound | Activity | Study Model | Observation |
Arjunolic Acid | Hypocholesterolaemic effect | Myocardial necrosis in Rats induced By Isoproterenol | Reducing ischemic myocardial damage, suppressing free radical formation, decreasing enzymatic activities linked to heart injury, and exhibiting antiplatelet and anticoagulant property [96]. |
Reducing cardiac fibrosis | Ligation of the right renal artery in male wistar rats. | Act as a PPAR agonist to block the activation of TAK1 in the non-canonical TGF-β pathway, reducing the pro-fibrotic signals and collagen deposition associated with cardiac hypertrophy [97]. | |
Cardio protective | LPS-treated male albino mice | Reduced cardiac injury markers, enhanced antioxidants, decreased lipid peroxidation and inflammation, lowered apoptotic caspase activity, and improved histopathological heart changes [98]. | |
Cardio protective effect | Hundred patients with CAD | Improved lipid profile and decreases in blood pressure, pulse rate, and total platelet count [99]. | |
Arjungenin | FXR agonistic and insulin sensitization activity | In-silico and in-vitro models | Promote adipogenesis and adipocyte differentiation [100]. |
Cardio protective | DPPH assay, NGT reduction assay | Moderate free radical scavenging activity [101]. | |
Arjunic Acid | Antioxidant and anti-apoptotic | In-vitro cell line study. H9c2 (rat myoblast cells) | Improved cell viability against CoCl2 induced cytotoxicity in h9c2 myoblast [102]. |
Terminic Acid | Myocardial infarction | Cox-2 target protein. Docking study | Formed a stable complex with the cox-2 receptor, showing encouraging inhibition, with binding energy of -7.79 kcal/mol [103]. |
Terminoside A | Anti atherogenic | HFD fed male wistar rat | Reductions in body weight, total cholesterol, triglycerides, phospholipids, LDL-C, and VLDL-C levels, while increasing HDL-C levels in HFD fed rats [104]. |
Inhibit NO production | Thioglycollate-induced peritoneal macrophages of rats | Potently inhibits the generation of NO and reduces inducible nitric oxide synthase [105]. | |
Arjunaphthanoloside | Antioxidant activity | Rat peritoneal macrophages | Shown strong antioxidant activity as measured by the prevention of LDL oxidation and radical scavenging [106]. |
Baicalein | Hepatoprotective activity | Goat liver slice culture model | Reduced LDH and liver marker enzyme levels, indicating hepatoprotective effect [107]. |
Antilipoperoxidative and radical scavenging effect | Rat liver mitochondria and cardiac homogenate | Inhibit lipid peroxidation process, ROS generation and plasma oxidation [108]. |
Curcuma longa: A ubiquitous spice used in Asian cuisine; turmeric (Curcuma longa L.) belongs to the Zingiberaceae family 109. The yellow-orange, oblong, aromatic, coarsely segmented rhizomes of turmeric plants are 2.5–7.0 cm in length and about 2.5 cm in diameter 110. Originating in Southeast Asia, this plant is widely cultivated in tropical and subtropical climates worldwide. In India and numerous other nations, it stands as one of the most significant spices. It is also utilized in herbal medicine and as a natural yellow food colour 111. Owing to its striking yellow coloration, it is frequently named as "Indian Saffron" 112. Active ingredients found in turmeric include polyphenols, sterols, alkaloids, sesquiterpenes, diterpenes, and triterpenoids and its yellow colour is due to the presence curcuminoids 113. Ayurvedic and traditional Chinese medicine have utilized Curcuma longa extract, which contains Biological attributes including the inhibition of platelet aggregation, management of diabetes, tumor suppression, reduction of inflammation, antioxidant properties, protection of the gastrointestinal tract, lowering of lipid levels, effects associated with Alzheimer's, and more 114,115. Among them curcumin shows greater biological activity and its capacity to interact with diverse proteins enables the specific control of many cellular signaling pathways linked to a range of chronic illnesses 116. The main chemical constituents of the rhizome of Cucuma longa are displayed in Table 7 117-131.
TABLE 7: PHYTOCONSTITUENTS OF CURCUMA LONGA
Compound | PubChem CID | Compound | PubChem CID |
Dodecanoic Acid | 3893 | Curcumenone | 14632994 |
(-)-Isolongifolol | 572865 | Curcumin | 2889 |
1,8-Cineole | 2758 | Curcumin-O-Glucuronide | 92024088 |
10-Epi-Gamma-Eudesmol | 518516 | Curcuminol | 101148924 |
1-Bisabolene | 62346 | Curcumol | 3559861 |
2-Decanol | 14254 | Curcuphenol | 122836 |
2-Ethenyl-1,1-Dimethyl-3-Methylene-Cyclohexane | 550088 | Curdione | 518796 |
2-Heptanol | 10976 | Curlone | 196216 |
2-Hydroxy-Methyl-Anthraquinone | 87014 | Curzerene | 572766 |
2-Isopropylidene-3-Methylhexa-3,5-Dienal | 562478 | Curzerenone | 5315433 |
2-Methoxy- 4-Vinyl Phenol | 332 | Cyclocurcumin | 77736151 |
2-Nonanol | 12367 | Dehydrocurdione | 78173043 |
2-Octanol | 20083 | Dehydrosaussurea Lactone | 556920 |
3,7,11-Trimethyl-1,3,6,10-Dodecatetraene | 5362889 | Delta-Cadinene | 10223 |
3,7-Dimethyl-1,3,7-Octatriene | 5320249 | Demethoxycurcumin | 146723 |
3-Carene | 26049 | Dicinnamoylmethane | 390472 |
4(S)-5(S)-Epoxy-Germacrone | 73037839 | Dicyclohexyl-Propanedinitrile | 557872 |
4-Hydroxy-Cinnamoyl-(Feruloyl)-Methane | 146723 | Diferuloyl-Methane | 969516 |
4-Hydroxy-Cinnamoyl-Methane | 95648 | Dihydro-Ar-Turmerone | 10921984 |
4-Terpineol | 11230 | Dihydrocostunolide | 102769 |
5-Hydroxy-1,7-Bis(4-Hydroxyphenyl) Hept-1-En-3-One | 78144189 | Dihydrocurcumin | 85140635 |
5-Isopropenyl-1,2-Dimethylcyclohexan-2-Enol | 536558 | Di-P-Coumaroyl-Methane | 147439 |
5'-Methoxy-Curcumin | 90788261 | Dl-2,3-Butanediol | 262 |
7-Epi-Sesquithujene | 53439065 | Elimicin | 10248 |
8-2-Carene | 79044 | Ferrulic Acid | 709 |
8-P-Cimenol | 95376 | Furanodiene | 171597 |
Agarospirol | 289964 | Furanodienone | 179413 |
Alpha-Atlantone | 3013901 | Gallic Acid | 370 |
Alpha-Bergatomene | 86608 | Gamma-Atlantone | 54223152 |
Alpha-Bisabolene | 86597 | Gamma-Curcumene | 6428861 |
Alpha-Bisabolol | 10586 | Gamma-Terpinene | 7461 |
Alpha-Bisabolol Acetate | 524246 | Geraniol | 4458 |
Alpha-Cadinene | 101708 | Geranyl Acetate | 7780 |
Alpha-Copen-11-Ol | 14807655 | Geranyl Butyrate | 7796 |
Alpha-Cubebene | 86609 | Geranyl Formate | 7779 |
Alpha-Fenchol | 15406 | Geranyl Hexanoate | 24837 |
Alpha-Guainene | 6949 | Germacrene | 3470 |
Alpha-Patchoulene | 521710 | Germacrene B | 177602 |
Alpha-Phellandrene | 7460 | Germacrene-D | 91104 |
Alpha-Santalene | 94164 | Germacrone | 81323 |
Alpha-Selinene | 10123 | Guaiacol | 460 |
Alpha-Terpinene | 7462 | Hemellitol | 10686 |
Alpha-Terpineol | 17100 | Iso-Bornyl Acetate | 6448 |
Alpha-Thujene | 17868 | Isobutyl Acetate | 8038 |
Alpha-Thujone | 11027 | Isocurcumenol | 5255901 |
Alpha-Turmerone | 14632996 | Isoprocurcumenol | 14543197 |
Alpha-Ylangene | 19725 | Isorhamnetin | 5281654 |
Arabinose | 229 | Isoshyobunone | 12304470 |
Azulene | 9231 | Limonene | 22311 |
Benzene-2-Methyl-1,4-Bis(1-Methylethyl) | 143557 | Linalool | 6549 |
Bergamotol | 564395 | Linalool Oxide | 22310 |
Beta-Acorenol | 14105905 | Linalyl Acetate | 8294 |
Beta-Bisabolene | 403919 | L-Trans-Chrysanthenyl Acetate | 162747 |
Beta-Bisabolol | 27208 | Methyl Eugenol | 7127 |
Beta-Cedrene | 102432 | Myrcene | 31253 |
Beta-Curcumene | 6428461 | Myrtenal | 61130 |
Beta-Farnesene | 10407 | Myrtenol | 10582 |
Beta-Germacene | 71404157 | Neral | 8843 |
Beta-Himachalene | 15095 | N-Heptane | 8900 |
Beta-Longipinene | 25203064 | N-Nonene | 8141 |
Beta-Phellandrene | 11142 | N-Octane | 356 |
Beta-Pinene | 14896 | Octahydrobisdemethoxycurcumin | 14427394 |
Beta-Santalene | 10534 | O-Cymene | 10703 |
Beta-Selinene | 519361 | P-Coumaroyl-Feruloyl-Methane | 4436278 |
Beta-Sesquiphellandrene | 519764 | P-Cymene | 7463 |
Beta-Sitosterol | 86821 | P-Cymene-8-Ol | 14529 |
Beta-Vatirenene | 608753 | Perilla Ketone | 68381 |
Bis-(Para-Hydroxy-Cinnamoyl)-Methane | 147439 | Pinene | 6654 |
Bisabola-3,10-Dien-2-One | 10857025 | P-Methoxy-Cinnamic-Acid | 13245 |
Bisacumol | 5315469 | P-Methyl Acetophenone | 8500 |
Bisacurone | 14287395 | Procurcumenol | 5320710 |
Bis-Demethoxycurcumin | 147439 | P-Tolymethylcarbinol | 10817 |
Borneol | 64685 | Quercetin | 5280343 |
Caffeic Acid Hexoside | 4484225 | Sabinene | 18818 |
Caffeic-Acid | 2518 | Sabinyl Acetate | 94266 |
Calebin-A | 78200755 | Sesquicineole | 341779 |
Campesterol | 312822 | Sesquisabinene | 25202482 |
Camphene | 6616 | Sesquisabinene Hydrate | 20055539 |
Caprylic-Acid | 379 | Sinapic Acid | 10743 |
Carvacrol | 10364 | ß-Eudesmol | 521215 |
Carvone | 7439 | ß-Ocimene | 18756 |
Caryophyllene | 26318 | ß-Patchouline | 101731 |
Caryophyllene Oxide | 14350 | Syringic Acid | 10742 |
Casuarinin | 157395 | T-Cadinol | 519662 |
Catechin | 1203 | Terpinolene | 11463 |
Cedrene | 521207 | Tetradecane | 12389 |
Chamigran-9-One-2,10-Dibromo-3-Chloro | 558220 | Tetra hydrobisdemethoxy curcumin | 9796792 |
Cinnamic-Acid | 8784 | Thymol | 6989 |
Cinnamyl Cinnamate | 31224 | Thymol Acetate | 68252 |
Cis-Beta-Elemenone | 519762 | Tolyl-Methylcarbinol | 110953 |
Cis-Carveol | 7438 | Trans-Nerolidol | 8888 |
Cis-Carvotanacetol | 534485 | Trans-P-Menth-2-En-1-Ol | 526657 |
Cis-P-Menth-2,8-Dienol | 155626 | Tumerone | 558173 |
Cis-Sabinol | 564260 | Turmerone | 558221 |
Cis-ß-Elemene | 10583 | Turmeronol-A | 15858385 |
Cis-Z-Alpha-Bisabolene Epoxide | 6429142 | Turmeronol-B | 10955433 |
Corymbolone | 535226 | Undecane | 14257 |
Coumaric Acid | 322 | Undecanol | 8184 |
Cuminyl-Alcohol | 325 | Valoneic Acid Bilactone | 10151874 |
Curcumalongin A | 73507489 | Vanillic Acid | 8468 |
Curcumalongin C | 73507491 | Viridiflorol | 101716 |
Curcumene | 92139 | Zedoarondiol | 14632997 |
Curcumenol | 387977 | Zingiberene | 521253 |
FIG. 5: CHEMICAL CONSTITUENTS OF CURCUMA LONGA
TABLE 8: POTENTIAL ACTIVITIES OF CURCUMA LONGA
Compound name | Activity | Study model | Observation |
α-Bisabolol | Cardio protective | In -vivo, ISO-induced rats | Reduced size of the myocardial infract and serum LDH activity [132]. |
Anti inflammatory | In- vivo, ISO-induced rats | Reduced expressions of inflammatory mediators and pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6) [132]. | |
Cyclocurcumin | Anti platelet | In -vitro, human blood | Prevented the activation of platelets caused by shear stress. [133]. |
Anti-oxidant | In -silico | Serve as an OH and OOH free radical scavenger [134]. | |
p-cymene | Anti-oxidant | In vivo, mice | significantly reduced the level of lipid peroxidation, nitrite content [135]. |
Vaso relaxant | In -vitro, Aorta of male Wistar rats | Showed that aortic rings were loosened by p-cymene [136]. | |
Curcumin | Anti atherosclerosis | In -vivo, mice | Significantly lower levels of macrophage infiltration and TLR4 expression in atherosclerosis plaque [137]. |
Hypo lipidemic effect | In -vivo, hyper-cholesterolemic Albino rats | Improve the lipid profile, endothelial function, and serum biochemical markers. [138]. | |
Immunomodulatory effect | In -vivo, mice | Binding to different receptors such as TLR, PAMPS, NF-kB, STAT [139]. | |
Antioxidant | Randomized Clinical Trials | Elimination of reactive oxygen and nitrogen, and control of several enzymes [140]. | |
ar-turmerone | Antiplatelet | In -vitro, rabbit platelets | ar-turmerone was significantly more potent platelet inhibitor than aspirin [141]. |
Anti angiogenic | Zebra fish model | Down-regulation of Angiopoietin-2 and Tie-2 expressions [142]. | |
Zingiberene | Cardio protective | Isoproterenol-induced cardiotoxicity in rats | Protected against cardiotoxicity caused by isoproterenol by reducing oxidative stress and hyperlipidemia [143]. |
CONCLUSION: GSTC is a unique poly-herbal formulation with multi-targeted activity based on the scientific data mentioned above. Together, they control several stages of atherogenesis.
A multicentric clinical study of GSTC is necessary, even if these individual plants are already being used clinically. A deeper understanding of its harmful or useful constituents may be obtained by bioactive screening, raising GSTC's present quality standard and offering fresh perspectives on designing bioactive lead compounds.
ACKNOWLEDGEMENT: Nil
Data Availability Statement: Data sharing is not applicable to this article as it is a review of previously published studies. All data cited are available in the original articles referenced within the manuscript.
CONFLICT OF INTEREST: None of the authors of the above manuscript has declared any conflict of interest which may arise from being named as an author on the manuscript.
REFERENCES:
- Noor F, Tahir ul Qamar M, Ashfaq UA, Albutti A, Alwashmi AS and Aljasir MA: Network pharmacology approach for medicinal plants: review and assessment. Pharmaceuticals 2022; 15(5): 572.
- Makhoba XH, Viegas C, Mosa RA, Viegas FP and Pooe OJ: Potential impact of the multi-target drug approach in the treatment of some complex diseases. Drug Design, Development and Therapy 2020; 3235-49.
- Akram, Muhammad, Adetunji, Charles, Laila, Umme, Michael, Olugbenga, Olerimi, Samson, Kadiri, Oseni, Ansari, Rumaisa, Adetunji, Juliana, Ozolua, Phebean, Mtewa, Andrew, Egbuna and Chukwuebuka: Overview of the traditional systems of medicine in different continents during postwar recovery 2021; 10.1016/B978-0-12-821556-2.00009-8.
- Singh DB, Pathak RK and Rai D: From traditional herbal medicine to rational drug discovery: strategies, challenges, and future perspectives. Revista Brasileira de Farmacognosia 2022; 32(2): 147-59.
- Manalil JJ, Puthanveettil J, Babu TD and Raghavamenon AC: Antioxidant and Anti-inflammatory Properties of a Nontoxic Herbal formulation–GSTC. Int J Pharmtech Res 2014; 6(1): 924-32.
- Nedkoff L, Briffa T, Zemedikun D, Herrington S and Wright FL: Global trends in atherosclerotic cardiovascular disease. Clinical Therapeutics 2023.
- Song P, Fang Z, Wang H, Cai Y, Rahimi K, Zhu Y, Fowkes FG, Fowkes FJ and Rudan I: Global and regional prevalence, burden, and risk factors for carotid atherosclerosis: a systematic review, meta-analysis, and modelling study. The Lancet Global Health 2020; 8(5): 721-9.
- Gusev E and Sarapultsev A: Atherosclerosis and inflammation: insights from the theory of general pathological processes. International Journal of Molecular Sciences 2023; 24(9): 7910.
- Manalil JJ, Suseela IM, Ramavarma SK, Narayanankutty A and Raghavamenon AC: Component Authentication and Standardisation of an Anti-atherosclerotic Herbal Formulation-GSTC3. Pharmacognosy Journal 2015; 7(6).
- Manalil JJ, Baby M, Ramavarma SK, Suseela IM, Padikkala J and Raghavamenon AC: Development of an anti-atherosclerotic polyherbal formulation: GSTC3. Journal of Environmental Pathology, Toxicology and Oncology 2015; 34(3).
- Pendyala V, Janga RB and Suryadevara V: Phytochemical and pharmacological evaluation of Commiphora mukul for antidepressant activity in albino mice. Asian Journal of Pharmaceutical and Clinical Research. 2017 Jan 1:360-3.
- Thosar SL, Yende MR. Cultivation and conservation of Guggulu (Commiphora mukul). Ancient Science of Life 2009; 29(1): 22-5.
- Kewat ML, Inchulkar SR, Kaushik Y, Chauhan NS and Sahu UK: Applied aspect of ayurvedic fumigation (Dhupana Karma) in epidemic era: a conceptual review. TMR Integr Med 2022; 6: 22003.
- Malik J and Mandal SC: Commiphora spp.(Guggul): A Wonder Ayurvedic Drug. Advances in Medicinal and Aromatic Plants: Production, Processing, and Pharmaceutics 2024; 2: 261.
- Latha S, Selvamani P and Prabha T: Pharmacological uses of the plants belonging to the genus Commiphora. Cardiovascular & Hematological Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Cardiovascular & Hematological Agents) 2021; 19(2): 101-17.
- Sarup P, Pahuja S and Malik J: Chemistry, biological activities, and uses of oleo-gum resin of Commiphora wightii. In Gums, Resins and Latexes of Plant Origin: Chemistry, Biological Activities and Uses 2022; 447-478. Cham: Springer International Publishing.
- Shekhawat M and Sisodia SS: A review on pharmacology of oleo-gum resin of Commiphora wightii. World Journal of Pharmaceutical Research 2021; 10: 735-23.
- Srivastava S. Guggul: Potency and Mechanistic Insight for Tremendous Therapeutic Benefit. Current Traditional Medicine 2024; 10(3): 75-83.
- Ulrich J, Stiltz S, St-Gelais A, El Gaafary M, Simmet T, Syrovets T and Schmiech M: Phytochemical composition of Commiphora oleogum resins and their cytotoxicity against skin cancer cells. Molecules 2022; 27(12): 3903.
- Muhati GL: Ecosystem services of Hurri hills, a montane woodland ecosystem in the arid lands of northern Kenya. Global Ecology and Conservation 2022; 33: 01951.
- Patil S: Guggulu: (Commiphora mukul)–A Medicinal Importance Herb in Ayurveda. Research Journal of Medicinal Plants in Ayurveda 2023; 3(1).
- Verma RK, Ibrahim M, Fursule A, Mitra R, Sastry JL and Ahmad S: Metabolomic profiling of Commiphora wightii (Arn.) Bhandari bark, oleogum-resin, and stem collected from different geographical regions of India. South African Journal of Botany 2022; 149: 211-21.
- Sandeep Kumar SK, Verma RK and Neelu Singh NS: Chemical composition and phytotherapeutic potential of Commiphora wightii.
- Zhu N, Rafi MM, DiPaola RS, Xin J, Chin CK, Badmaev V, Ghai G, Rosen RT and Ho CT: Bioactive constituents from gum guggul (Commiphora wightii). Phytochemistry 2001; 56(7): 723-7.
- Sarup P, Bala S and Kamboj S: Pharmacology and phytochemistry of oleo‐gum resin of Commiphora wightii (Guggulu). Scientifica 2015; 2015(1): 138039.
- Kimura I, Yoshikawa M, Kobayashi S, Sugihara Y, Suzuki M, Oominami H, Murakami T, Matsuda H and Doiphode VV: New triterpenes, myrrhanol A and myrrhanone A, from guggul-gum resins, and their potent anti-inflammatory effect on adjuvant-induced air-pouch granuloma of mice. Bioorganic & Medicinal Chemistry Letters 2001; 11(8): 985-9.
- Mulvihill EE, Assini JM, Sutherland BG, DiMattia AS, Khami M, Koppes JB, Sawyez CG, Whitman SC and Huff MW: Naringenin decreases progression of atherosclerosis by improving dyslipidemia in high-fat–fed low-density lipoprotein receptor–null mice. Arteriosclerosis, Thrombosis, and Vascular Biology 2010; 30(4): 742-8.
- Hsueh TP, Sheen JM, Pang JH, Bi KW, Huang CC, Wu HT and Huang ST: The anti-atherosclerotic effect of naringin is associated with reduced expressions of cell adhesion molecules and chemokines through NF-κB pathway. Molecules 2016; 21(2): 195.
- Burke AC, Sutherland BG, Telford DE, Morrow MR, Sawyez CG, Edwards JY and Huff MW: Naringenin enhances the regression of atherosclerosis induced by a chow diet in Ldlr−/− mice. Atherosclerosis 2019; 286: 60-70.
- Zhang Y, He L, Tu M, Huang M, Chen Y, Pan D, Peng J and Shen X: The ameliorative effect of terpinen-4-ol on ER stress-induced vascular calcification depends on SIRT1-mediated regulation of PERK acetylation. Pharmacological Research 2021; 170: 105629.
- Liu T, Wang W, Liu M, Ma Y, Mu F, Feng X, Zhang Y, Guo C, Ding Y and Wen A: Z-Guggulsterone alleviated oxidative stress and inflammation through inhibiting the TXNIP/NLRP3 axis in ischemic stroke. International Immunopharmacology 2020; 89: 107094.
- Chander R, Rizvi F, Khanna AK and Pratap R: Cardioprotective activity of synthetic guggulsterone (E and Z-isomers) in isoproterenol induced myocardial ischemia in rats: a comparative study. Indian Journal of Clinical Biochemistry 2003; 18: 71-9.
- Parmar P, Bhoir S, Naik PP, Waghulde S, Gorde N and Kale MK: Guggulsterone–Farnesoid X receptor interaction. Journal of Pharmacognosy and Phytochemistry 2018; 7(6): 107-14.
- De León EJ, Olmedo DA, Solís PN, Gupta MP and Terencio MC: Diayangambin exerts immunosuppressive and anti-inflammatory effects in-vitro and in-vivo. Planta Medica 2002; 68(12): 1128-31.
- Ikeda I, Konno R, Shimizu T, Ide T, Takahashi N, Kawada T, Nagao K, Inoue N, Yanagita T, Hamada T and Morinaga Y: Campest-5-en-3-one, an oxidized derivative of campesterol, activates PPARα, promotes energy consumption and reduces visceral fat deposition in rats. Biochimica et Biophysica Acta (BBA)-General Subjects 2006; 1760(5): 800-7.
- Kimura I, Yoshikawa M, Kobayashi S, Sugihara Y, Suzuki M, Oominami H, Murakami T, Matsuda H and Doiphode VV: New triterpenes, myrrhanol A and myrrhanone A, from guggul-gum resins, and their potent anti-inflammatory effect on adjuvant-induced air-pouch granuloma of mice. Bioorganic & Medicinal Chemistry Letters 2001; 11(8): 985-9.
- Meselhy MR: Inhibition of LPS-induced NO production by the oleogum resin of Commiphora wightii and its constituents. Phytochemistry 2003; 62(2): 213-8.
- Wan YJ, Wang YH, Guo Q, Jiang Y, Tu PF and Zeng KW: Protocatechualdehyde protects oxygen-glucose deprivation/reoxygenation-induced myocardial injury via inhibiting PERK/ATF6α/IRE1α pathway. European Journal of Pharmacology 2021; 891: 173723.
- Shao M, Wang M, Ma L, Wang Q, Gao P, Tian X, Li C, Lu L, Li C, Wang W and Wang Y: β-elemene blocks lipid-induced inflammatory pathways via PPARβ activation in heart failure. European Journal of Pharmacology 2021; 910: 174450.
- Lanlan DU, Jing DO, Xiangcheng FA, Junyi ZH, Yifan ZH, Jichun HA and Jing SH: β-Elemene improves endothelial cells dysfunction, and abnormal proliferation and migration of vascular smooth muscle cells. Journal of China Pharmaceutical University 2020; 51(3): 333-9.
- Bhavnani BR, Cecutti A, Gerulath A, Woolever AC and Berco M: Comparison of the antioxidant effects of equine estrogens, red wine components, vitamin E, and probucol on low-density lipoprotein oxidation in postmenopausal women. Menopause 2018; 25(11): 1214-23.
- Kamada C, Mukai R, Kondo A, Sato S and Terao J: Effect of quercetin and its metabolite on caveolin-1 expression induced by oxidized LDL and lysophosphatidylcholine in endothelial cells. Journal of Clinical Biochemistry and Nutrition 2016; 58(3): 193-201.
- Shobha Bhaskar SB, Sudhakaran PR and Helen A: Quercetin attenuates atherosclerotic inflammation and adhesion molecule expression by modulating TLR-NF-κBsignaling pathway.
- Zhu MeiXia ZM, Li JianKuan LJ, Wang Ke WK, Hao XuLiang HX, Ge Rui GR and Li QingShan LQ: Isoquercitrin inhibits hydrogen peroxide-induced apoptosis of EA. hy926 cells via the PI3K/Akt/GSK3β signaling pathway.
- Ren K, Jiang T and Zhao GJ: Quercetin induces the selective uptake of HDL-cholesterol via promoting SR-BI expression and the activation of the PPARγ/LXRα pathway. Food & Function 2018; 9(1): 624-35.
- Lu Y and Jia YP: Quercetin upregulates ABCA1 expression through liver X receptor alpha signaling pathway in THP-1 macrophages. European Review for Medical & Pharmacological Sciences 2016; 20(18).
- Amarathunge SM, Gunaratne LH and Prasada DV: A case study in the utilization of the underutilized crop; Salacia species in lowland wet zone of Sri Lanka. Agrieast 2021; 15(1): 43-54.
- Charkravarty A and Ramasamy G: Profiling of volatiles in tissues of Salacia reticulata with anti-diabetic potential using GC-MS And Molecular Docking. arXiv preprint arXiv:2307.04109. 2023 Jul 9.
- Morikawa T, Ninomiya K, Tanabe G, Matsuda H, Yoshikawa M and Muraoka O: A review of antidiabetic active thiosugar sulfoniums, salacinol and neokotalanol, from plants of the genus Salacia. Journal of Natural Medicines 2021; 75(3): 449-66.
- Nimbekar T, Jain A and Mohanty PK: Effects of Salacia reticulata root bark on blood glucose levels of normal and alloxan-monohydrate induced diabetic mice. J Cardiovasc Dis Res 2021; 12(3): 854-60.
- Premakumara GA and Abeysekera WK: Anti-protein glycation and free-radical scavenging properties of Sri Lankan antidiabetic medicinal plant Salacia reticulata l.(Kothala Himbutu). BMC Complementary Medicine and Therapies 2023; 23(1): 394.
- Amarathunge SM and Pushpakumara DK: Morphological and Qualitative Phytochemical Comparison of Selected Salacia Species in Lowland Wet Zone of Sri Lanka. Sabaragamuwa University Journal 2021; 19(1).
- Asija R, Swami A and Sharma KR: Evaluation of anti-urolithiatic activity of ethanolic extract of Salacia reticulata against ethylene glycol induced urolithiasis in wistar albino rats. Asian Journal of Pharmaceutical Research and Development 2024; 12(3): 58-65.
- Zhu S, Liu Q, He J, Nakajima N, Samarakoon SP, Swe S, Zaw K and Komatsu K: Genetic identification of medicinally used Salacia species by nrDNA ITS sequences and a PCR-RFLP assay for authentication of Salacia-related health foods. Journal of Ethnopharmacology 2021; 274: 113909.
- Patra S, Naskar A, Das DR and Maity A: Medicinal Plants in the Treatment of Diseases Caused by Influenza B Virus and Parainfluenza Virus Type 3. InAnti-Viral Metabolites from Medicinal Plants 2023; 489-508 Cham: Springer International Publishing.
- Chavan J, Patil P, Patil A, Deshmukh A, Panari P, Mohite A, Lawand P, Yadav P, Bodhe M, Kadam A and Namdas D: Salacia spp.: recent insights on biotechnological interventions and future perspectives. Applied Microbiology and Biotechnology 2024; 108(1): 200.
- Raveendran R, Sundararajan S, Victoria S, Balamurugan A and Muthukumaran M: Medicinal plants used in neerilivu (diabetes mellitus) in traditional medicine-review and assessment of scientific evidences. International Journal of Ayurveda and Pharma Research 2021; 7-18.
- Jung J, Park J, Lee M, Kim J, Oh D, Jun W, Kim OK and Lee J: Salacia reticulata extract suppresses fat accumulation by regulating lipid metabolism. Foods 2023; 12(17): 3149.
- Ramakrishna A, Balasundar S, Rao PP, Divya KM, Renu D and Sangu PK: A pharmacognostic and phytopharmacological study of Salacia reticulata (W.) root. Annals of Phytomedicine 2023; 12(2): 1022-31.
- Yoshikawa M, Shimoda H, Matsuda H, Nishida N and Takada M: Salacia reticulata and its polyphenolic constituents with lipase inhibitory and lipolytic activities have mild antiobesity effects in rats. The Journal of Nutrition 2002; 132(7): 1819-24.
- Li Y, Huang TH and Yamahara J: Salacia root, a unique Ayurvedic medicine, meets multiple targets in diabetes and obesity. Life Sciences 2008; 82(21-22): 1045-9.
- Uthirapathy S, Ahamad J and SH Mohammed Ameen M: Safety standards and antimicrobial activity of root of Salacia reticulata. Research Journal of Phytochemistry 2021; 15(1): 30-40.
- Peiris DS, Fernando DT, Senadeera SP and Ranaweera CB: A Comprehensive Review of Salacia reticulata: Botanical, Ethnomedicinal, Phytochemical, and Pharmacological Insights. Asian Plant Research Journal. 2023; 11(6): 27-43.
- Stohs SJ and Ray S: Anti‐diabetic and Anti‐hyperlipidemic Effects and Safety of Salacia reticulata and Related Species. Phytotherapy Research 2015; 29(7): 986-95.
- Yoshikawa M, Ninomiya K, Shimoda H, Nishida N and Matsuda H: Hepatoprotective and antioxidative properties of Salacia reticulata: preventive effects of phenolic constituents on CCl4-induced liver injury in mice. Biological and Pharmaceutical Bulletin 2002; 25(1): 72-6.
- Arunakumara KK and Subasinghe S: Salacia reticulata Wight: A review of botany, phytochemistry and pharmacology. Tropical Agricultural Research and Extension 2011; 13(2).
- Ren K, Li H, Zhou HF, Liang Y, Tong M, Chen L, Zheng XL and Zhao GJ: Mangiferin promotes macrophage cholesterol efflux and protects against atherosclerosis by augmenting the expression of ABCA1 and ABCG1. Aging (Albany NY) 2019; 11(23): 10992.
- He Z, Zhu H, Liu J, Kwek E, Ma KY and Chen ZY: Mangiferin alleviates trimethylamine-N-oxide (TMAO)-induced atherogenesis and modulates gut microbiota in mice. Food & Function 2023; 14(20): 9212-25.
- Zhao Q, Yang J, Liu B, Huang F and Li Y: Exosomes derived from mangiferin stimulated perivascular adipose tissue ameliorate endothelial dysfunction. Molecular Medicine Reports 2019; 19(6): 4797-805.
- Milenkovic D, Declerck K, Guttman Y, Kerem Z, Claude S, Weseler AR, Bast A, Schroeter H and Morand C: Berghe WV. (−)-Epicatechin metabolites promote vascular health through epigenetic reprogramming of endothelial-immune cell signaling and reversing systemic low-grade inflammation. Biochemical Pharmacology 2020; 173: 113699.
- Sinegre T, Teissandier D, Milenkovic D, Morand C and Lebreton A: Epicatechin influences primary hemostasis, coagulation and fibrinolysis. Food & Function 2019; 10(11): 7291-8.
- Garate-Carrillo A, Navarrete-Yañez V, Ortiz-Vilchis P, Guevara G, Castillo C, Mendoza-Lorenzo P, Ceballos G, Ortiz-Flores M, Najera N, Bustamante-Pozo MM and Rubio-Gayosso I: Arginase inhibition by (−)-Epicatechin reverses endothelial cell aging. European Journal of Pharmacology 2020; 885: 173442.
- Ponnian SM: Preventive effects of (−) epicatechin on tachycardia, cardiac hypertrophy, and nuclear factor-κB inflammatory signaling pathway in isoproterenol-induced myocardial infarcted rats. European Journal of Pharmacology 2022; 924: 174909.
- Connolly K, Batacan R, Jackson D and Fenning AS: Effects of epicatechin on cardiovascular function in middle-aged diet-induced obese rat models of metabolic syndrome. British Journal of Nutrition 2024; 131(4): 593-605.
- Shi Y, Jiang S, Zhao T, Gong Y, Liao D and Qin L: Celastrol suppresses lipid accumulation through LXRα/ABCA1 signaling pathway and autophagy in vascular smooth muscle cells. Biochemical and Biophysical Research Communicat 2020; 532(3): 466-74.
- Allen SD, Liu YG, Kim T, Bobbala S, Yi S, Zhang X, Choi J and Scott EA: Celastrol-loaded PEG-b-PPS nanocarriers as an anti-inflammatory treatment for atherosclerosis. Biomaterials Science 2019; 7(2): 657-68.
- Ouyang M, Qin T, Liu H, Lu J, Peng C and Guo Q: Enhanced inflammatory reaction and thrombosis in high-fat diet-fed ApoE-/-mice are attenuated by celastrol. Experimental and Clinical Endocrinology & Diabetes 2021; 129(05): 339-48.
- Liu H, Wang L, Pan Y, Wang X, Ding Y, Zhou C, Shah AM, Zhao G and Zhang M: Celastrol alleviates aortic valve calcification via inhibition of NADPH oxidase 2 in valvular interstitial cells. Basic to Translational Science 2020; 5(1): 35-49.
- Tao Z, Xiao Q, Che X, Zhang H, Geng N and Shao Q: Regulating mitochondrial homeostasis and inhibiting inflammatory responses through Celastrol. Annals of Translational Medicine 2022; 10(7).
- Shi YN, Liu LP, Deng CF, Zhao TJ, Shi Z, Yan JY, Gong YZ, Liao DF and Qin L: Celastrol ameliorates vascular neointimal hyperplasia through Wnt5a-involved autophagy. International Journal of Biological Sciences 2021; 17(10): 2561.
- Liang J, Yuan S, Wang X, Lei Y, Zhang X, Huang M and Ouyang H: Attenuation of pristimerin on TNF-α-induced endothelial inflammation. International Immunopharmacology 2020; 82: 106326.
- Mandal S, Patra A, Samanta A, Roy S, Mandal A, Mahapatra TD, Pradhan S, Das K and Nandi DK: Analysis of phytochemical profile of Terminalia arjuna bark extract with antioxidative and antimicrobial properties. Asian Pacific J of Tropical Biomedicine 2013; 3(12): 960-6.
- Dwivedi S and Chopra D: Revisiting Terminalia arjuna–an ancient cardiovascular drug. Journal of Traditional and Complementary Medicine 2014; 4(4): 224-31.
- Chaudhari GM and Mahajan RT: Comprehensive study on pharmacognostic, physico and phytochemical evaluation of Terminalia arjuna stem bark. Journal of Pharmacognosy and Phytochemistry 2015; 4(3): 186-93.
- Dwivedi S: Terminalia arjuna Wight & Arn.—a useful drug for cardiovascular disorders. Journal of Ethnopharmacology 2007; 114(2): 114-29.
- Kapoor D, Vijayvergiya R and Dhawan V: Terminalia arjuna in coronary artery disease: ethnopharmacology, pre-clinical, clinical & safety evaluation. Journal of Ethnopharmacology 2014; 155(2): 1029-45.
- Gaikwad D and Jadhav N: A review on biogenic properties of stem bark of Terminalia arjuna: An update. Asian Journal of Pharmaceutical and Clinical Research 2018; 11(8): 35-9.
- Jain S, Yadav PP, Gill V, Vasudeva N and Singla N: Terminalia arjuna a sacred medicinal plant: phytochemical and pharmacological profile. Phytochemistry Reviews 2009; 8: 491-502.
- Amalraj A and Gopi S: Medicinal properties of Terminalia arjuna (Roxb.) Wight &Arn.: a review. Journal of Traditional and Complementary Medicine 2017; 7(1): 65-78.
- Khan ZH, Faruquee HM and Shaik MM: Phytochemistry and pharmacological potential of Terminalia arjuna Medicinal Plant Research 2013; 3.
- Wang W, Ali Z, Li XC, Shen Y and Khan IA: Triterpenoids from two Terminalia species. Planta Medica 2010; 76(15): 1751-4.
- Wang W, Ali Z, Li XC, Shen Y and Khan IA: 18, 19-secooleanane type triterpene glycosyl esters from the bark of Terminalia arjuna. Planta Medica 2010; 76(09): 903-8.
- Gupta S, Bishnoi JP, Kumar N, Kumar H and Nidheesh T: Terminalia arjuna (Roxb.) Wight & Arn.: Competent source of bioactive components in functional food and drugs. The Pharma Innovation Journal 2018; 7(3): 223-31.
- Ramesh P and Palaniappan A: Terminalia arjuna, a cardioprotective herbal medicine–relevancy in the modern era of pharmaceuticals and green nanomedicine—a review. Pharmaceuticals 2023 Jan 13; 16(1): 126.
- Saha A, Pawar VM and Jayaraman S: Characterisation of polyphenols in Terminalia arjuna bark extract. Indian Journal of Pharmaceutical Sciences 2012; 74(4): 339.
- Sumitra M, Manikandan P, Kumar DA, Arutselvan N, Balakrishna K, Manohar BM and Puvanakrishnan R: Experimental myocardial necrosis in rats: role of arjunolic acid on platelet aggregation, coagulation and antioxidant status. Molecular and Cellular Biochemistry 2001; 224: 135-42.
- Bansal T, Chatterjee E, Singh J, Ray A, Kundu B, Thankamani V, Sengupta S and Sarkar S: Arjunolic acid, a peroxisome proliferator-activated receptor α agonist, regresses cardiac fibrosis by inhibiting non-canonical TGF-β signaling. Journal of Biological Chemistry 2017; 292(40): 16440-62.
- Elsawy H, Almalki M, Elmenshawy O and Abdel-Moneim A: In-vivo evaluation of the protective effects of arjunolic acid against lipopolysaccharide-induced septic myocardial injury. Peer J 2022; 10: 12986.
- Priya N, Mathur KC, Sharma A, Agrawal RP, Agarwal V and Acharya J: Effect of Terminalia arjuna on total platelet count and lipid profile in patients of coronary artery disease. Advances in Human Biology 2019; 9(1): 98-101.
- Anand T, Fathima A and Khanum F: In-silico therapeutic investigations of arjunic acid and arjungenin as an FXR agonist and validation in 3T3-L1 adipocytes. Computational Biology and Chemistry 2020; 84: 107163.
- Pawar RS and Bhutani KK: Effect of oleanane triterpenoids from Terminalia arjuna a cardioprotective drug on the process of respiratory oxyburst. Phytomedicine 2005; 12(5): 391-3.
- Manu TM, Anand T, Pandareesh MD, Kumar PB and Khanum F: Terminalia arjuna extract and arjunic acid mitigate cobalt chloride–induced hypoxia stress–mediated apoptosis in H9c2 cells. NSAP 2019; 392: 1107-19.
- Chauhan D, Sahoo S and Ahmad N: Molecular Docking Study of Cox-2 Target Protein with Terminic acid and its Role for Myocardial Infarction. Cardiometry 2022; 1(24): 1040-5.
- Jensi VD and Gopu PA: Evaluation of hypolipidemic activity of various phytoconstituents from Terminalia arjuna (Roxb. ex DC.) in rat fed with high fat diet. International Journal of Pharmaceutical Sciences Review and Research 2018; 50(2): 41-8.
- Ali A, Kaur G, Hamid H, Abdullah T, Ali M, Niwa M and Alam MS: Terminoside A, a new triterpene glycoside from the bark of Terminalia arjuna inhibits nitric oxide production in murine macrophages. Journal of Asian Natural Products Research 2003; 5(2): 137-42.
- Ali A, Kaur G, Hayat K, Ali M and Ather M: A novel naphthanol glycoside from Terminalia arjuna with antioxidant and nitric oxide inhibitory activities. Die Pharmazie 2003; 58(12): 932-4.
- Chaudari GM and Mahajan RT: In-vitro hepatoprotective activity of Terminalia arjuna stem bark and its flavonoids against CCl4 induced hepatotoxicity in goat liver slice culture. Asian Journal of Plant Science & Research 2016.
- Tilak JC, Devasagayam TP, Adhikari S, Lele RD, Kon T, Handa O, Naito Y and Yoshikawa T: Cellular membrane protection against reactive oxygen species by Terminalia arjuna and its active component baicalein. Journal of Clinical Biochemistry and Nutrition 2006; 39(2): 75-87.
- Chumroenphat T, Somboonwatthanakul I, Saensouk S and Siriamornpun S: Changes in curcuminoids and chemical components of turmeric (Curcuma longa) under freeze-drying and low-temperature drying methods. Food Chemistry 2021; 339: 128121.
- Sontsa-Donhoung AM, Bahdjolbe M, Nekou GN, Tadjouo IK and Nwaga D: Growing Curcuma longa for rhizome production on diverse arable soil types in Cameroon: agronomic and microbial parameters. Agricultural Sciences 2021; 12(5): 464-80.
- Guimarães AF, Vinhas AC, Gomes AF, Souza LH and Krepsky PB: Essential oil of Curcuma longa rhizomes chemical composition, yield variation and stability. Química Nova 2020; 43: 909-13.
- Agrawal S, Nair R, Thomas M, Anjana G, Patel SK, Uikey P, Birla S, Singh J and Tripathi N: Morphological characterization of turmeric (Curcuma spp.) genotypes. Journal of Eco-friendly Agriculture 2024; 19(1): 67-72.
- Iweala EJ, Uche ME, Dike ED, Etumnu LR, Dokunmu TM, Oluwapelumi AE, Okoro BC, Dania OE, Adebayo AH and Ugbogu EA: Curcuma longa (Turmeric): Ethnomedicinal uses, phytochemistry, pharmacological activities and toxicity profiles A review. Pharmacological Research-Modern Chinese Medicine 2023; 6: 100222.
- Wang Z, Jones G, Winzenberg T, Cai G, Laslett LL, Aitken D, Hopper I, Singh A, Jones R, Fripp J and Ding C: Effectiveness of Curcuma longa extract for the treatment of symptoms and effusion–synovitis of knee osteoarthritis: A randomized trial. Annals of Internal Medicine 2020; 173(11): 861-9.
- Rajagopal K, Varakumar P, Baliwada A and Byran G: Activity of phytochemical constituents of Curcuma longa (turmeric) and Andrographis paniculata against coronavirus (COVID-19): an in-silico Future Journal of Pharmaceutical Sciences 2020; 6: 1-0.
- Urošević M, Nikolić L, Gajić I, Nikolić V, Dinić A and Miljković V: Curcumin: Biological activities and modern pharmaceutical forms. Antibiotics 2022; 11(2): 135.
- Awasthi PK and Dixit SC: Chemical composition of Curcuma Longa leaves and rhizome oil from the plains of Northern India. Journal of Young Pharmacists 2009; 1(4): 322.
- Naz S, Ilyas S, Parveen Z and Javed S: Chemical analysis of essential oils from turmeric (Curcuma longa) rhizome through GC-MS. Asian Journal of Chemistry 2010; 22(4): 3153.
- Gounder DK and Lingamallu J: Comparison of chemical composition and antioxidant potential of volatile oil from fresh, dried and cured turmeric (Curcuma longa) rhizomes. Industrial Crops and Products 2012; 38: 124-31.
- Raina VK, Srivastava SK and Syamsundar KV: Rhizome and leaf oil composition of Curcuma longa from the lower Himalayan region of northern India. Journal of Essential Oil Research 2005; 17(5): 556-9.
- Abdel-Lateef E, Mahmoud F, Hammam O, El-Ahwany E, El-Wakil E, Kandil S, Abu Taleb H, El-Sayed M and Hassenein H: Bioactive chemical constituents of Curcuma longa rhizomes extract inhibit the growth of human hepatoma cell line (HepG2). Acta Pharmaceutica 2016; 66(3): 387-98.
- Quirós-Fallas MI, Vargas-Huertas F, Quesada-Mora S, Azofeifa-Cordero G, Wilhelm-Romero K, Vásquez-Castro F, Alvarado-Corella D, Sánchez-Kopper A and Navarro-Hoyos M: Polyphenolic HRMS characterization, contents and antioxidant activity of Curcuma longa rhizomes from Costa Rica. Antioxidants 2022; 11(4): 620.
- El Demerdash A, Dawidar AM, Keshk EM, Abdel-Mogib M. Gingerdione from the rhizomes of Curcuma longa. Chemistry of Natural Compounds 2012; 48: 646-8.
- Ajaiyeoba EO, Sama W, Essien EE, Olayemi JO, Ekundayo O, Walker TM and Setzer WN: Larvicidal Activity of Turmerone-Rich Essential Oils of Curcuma longa. Leaf and Rhizome from Nigeria on Anopheles gambiae. Pharmaceutical Biology 2008; 46(4): 279-82.
- Bansal RP, Bahl JR, Garg SN, Naqvi AA and Kumar S: Differential chemical compositions of the essential oils of the shoot organs, rhizomes and rhizoids in the tumeric Curcuma longa grown in Indo-Gangetic plains. Pharmaceutical Biology 2002; 40(5): 384-9.
- Tsai SY, Huang SJ, Chyau CC, Tsai CH, Weng CC and Mau JL: Composition and antioxidant properties of essential oils from Curcuma rhizome. Asian J. Arts Sci 2011; 2(1): 57-66.
- Sahoo A, Kar B, Jena S, Dash B, Ray A, Sahoo S and Nayak S: Qualitative and quantitative evaluation of rhizome essential oil of eight different cultivars of Curcuma longa(Turmeric). Journal of Essential Oil Bearing Plants 2019; 22(1): 239-47.
- Pal K, Chowdhury S, Dutta SK, Chakraborty S, Chakraborty M, Pandit GK, Dutta S, Paul PK, Choudhury A, Majumder B and Sahana N: Analysis of rhizome colour content, bioactive compound profiling and ex-situ conservation of turmeric genotypes (Curcuma longa) from sub-Himalayan terai region of India. Industrial Crops and Products 2020; 150: 112401.
- Chassagnez-Mendez AL, Machado NT, Araujo ME, Maia JG and Meireles MA: Supercritical CO2 extraction of curcumins and essential oil from the rhizomes of turmeric (Curcuma longa). Industrial & Engineering Chemistry Research 2000; 39(12): 4729-33.
- Sabir SM, Zeb A, Mahmood M, Abbas SR, Ahmad Z and Iqbal N: Phytochemical analysis and biological activities of ethanolic extract of Curcuma longa Brazilian Journal of Biology 2020; 81(3): 737-40.
- Singh G, Kapoor IP, Singh P, de Heluani CS, de Lampasona MP and Catalan CA: Comparative study of chemical composition and antioxidant activity of fresh and dry rhizomes of turmeric (Curcuma longa). Food and Chemical Toxicology 2010; 48(4): 1026-31.
- Meeran MN, Azimullah S, Laham F, Tariq S, Goyal SN, Adeghate E and Ojha S: α-Bisabolol protects against β-adrenergic agonist-induced myocardial infarction in rats by attenuating inflammation, lysosomal dysfunction, NLRP3 inflammasome activation and modulating autophagic flux. Food & Function 2020; 11(1): 965-76.
- Ngo T, Kim K, Bian Y, An GJ, Bae ON, Lim KM and Chung JH: Cyclocurcumin from Curcuma longa selectively inhibits shear stress-induced platelet aggregation. Journal of Functional Foods 2019; 61: 103462.
- Li Y, Toscano M, Mazzone G and Russo N: Antioxidant properties and free radical scavenging mechanisms of cyclocurcumin. New Journal of Chemistry 2018; 42(15): 12698-705.
- de Oliveira TM, de Carvalho RB, da Costa IH, de Oliveira GA, de Souza AA, de Lima SG and de Freitas RM: Evaluation of p-cymene, a natural antioxidant. Pharmaceutical Biology 2015; 53(3): 423-8.
- Silva MT, Ribeiro FP, Medeiros MA, Sampaio PA, Silva YM, Silva MT, Quintans JS, Quintans-Júnior LJ and Ribeiro LA: The vasorelaxant effect of p‐Cymene in rat aorta involves potassium channels. The Scientific World Journal 2015; 2015(1): 458080.
- Zhang S, Zou J, Li P, Zheng X and Feng D: Curcumin protects against atherosclerosis in apolipoprotein E-knockout mice by inhibiting toll-like receptor 4 expression. Journal of Agricultural and Food Chemistry 2018; 66(2): 449-56.
- Hussein SA, El-Senosi YA, Ragab MR, Hammad MM and Apo A: Hypolipidemic effect of curcumin in hyper-cholesterolemic rats. Benha Vet Med J 2014; 27(2): 277-89.
- Abdollahi E, Momtazi AA, Johnston TP and Sahebkar A: Therapeutic effects of curcumin in inflammatory and immune‐mediated diseases: A nature‐made jack‐of‐all‐trades?. Journal of Cellular Physiology 2018; 233(2): 830-48.
- Jakubczyk K, Drużga A, Katarzyna J and Skonieczna-Żydecka K: Antioxidant potential of curcumin A meta-analysis of randomized clinical trials. Antioxidants 2020; 9(11): 1092.
- Lee HS: Antiplatelet property of Curcuma longa rhizome-derived ar-turmerone. Bioresource Technology 2006; 97(12): 1372-6.
- Yue GG, Kwok HF, Lee JK, Jiang L, Chan KM, Cheng L, Wong EC, Leung PC, Fung KP and Bik-San Lau C: Novel anti-angiogenic effects of aromatic-turmerone, essential oil isolated from spice turmeric. Journal of Functional Foods 2015; 15: 243-53.
- Asokan BR, Jaikumar S and Sengottuvelu S: Zingiberene protects from isoproterenol-induced cardiotoxicity via attenuation of hyperlipidemia and oxidative stress in rats. Revista Brasileira de Farmacognosia 2022; 32(1): 74-80.
How to cite this article:
Sathyapooja G, Arun SK, Aswin J, Hariharan N and Loganathan K: Phytochemical constituents and anti-atherosclerotic activities of a polyherbal formulation –GSTC. Int J Pharmacognosy 2025; 12(2): 58-74. doi link: http://dx.doi.org/10.13040/IJPSR.0975-8232.IJP.12(2).58-74.
This Journal licensed under a Creative Commons Attribution-Non-commercial-Share Alike 3.0 Unported License.
Article Information
2
58-74
1185 KB
45
English
IJP
G. Sathyapooja *, S. K. Arun, J. Aswin, N. Hariharan and K. Loganathan
Department of Pharmaceutical Chemistry, KMCH College of Pharmacy, Coimbatore, Tamil Nadu, India.
sathyapooja.g@kmchcop.ac.in
30 January 2025
26 February 2025
27 February 2025
10.13040/IJPSR.0975-8232.IJP.12(2).58-74
28 February 2025