Take a look at the Recent articles

Medicinal plants and their bioactive compounds in the control of hyperlipidemia: A comprehensive review

Mohammadpour D

Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran

E-mail : aa

Küpeli Akkol E

Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Turkey

Al-Gazally ME

Department of Biochemistry, Faculty of Medicine, Al-Ameed University, Karbala, Iraq

Bozorg-Ghalati F

Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran

Mohammadpour I

Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran

Basic Sciences in Infectious Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran

DOI: 10.15761/IFNM.1000309

Article
Article Info
Author Info
Figures & Data

Abstract

Hyperlipidemia is one of the chronic diseases that is caused by various metabolic disorders. Although lipid-lowering drugs are available, it is still an important factor in exacerbating cardiovascular disease in patients with diabetes, hypertension and obesity. Factors such as patients’ dissatisfaction with the use of common lipid-lowering drugs, the occurrence of side effects due to excessive and long-term use of these drugs, diseases caused by hyperlipidemia and the costs imposed on patients have led to an increase in the desire for alternative and traditional therapies. Epidemiological observations show that adherence to diet, use of alternative therapies, consumption of medicinal plants and fruits, in addition to reducing treatment costs, have had satisfactory results in the side effects of hyperlipidemia in many communities. It should be noted that the tendency to use medicinal plants to lower blood lipids in most societies, even in developed countries has become widespread. Consumption of these plants is especially significant when conventional therapies are not able to control the disease. Although arbitrary use of medicinal plants by patients with hyperlipidemia may improve the disease, because they are taken irregularly and at an unknown dose, they may interfere with conventional drugs and disease control. In this review, the role of common medicinal plants in the control and reduction of hyperlipidemia and the possibility of their toxicity and drug interactions are investigated.

Keywords

 Hyperlipidemia; Medicinal plants; Cholesterol; Low-density lipoprotein; High-density lipoprotein; Triglyceride

Abbreviations

LDL: Low-Density Lipoprotein; HDL: High-Density Lipoprotein; VLDL: Very Low-Density Lipoprotein; ROS: Reactive Oxygen Species; NO: Nitric Oxide; iNOS: inducible Nitric Oxide Synthase; HMG-CoA: Hydroxy Methylglutaryl Coenzyme A; NF‒κB: Nuclear Factor Kappa B; IL: Interleukin; TNF: Tumor Necrosis Factor; DV: Daily Value; PKA: Protein Kinase A; PKB: Protein Kinase B; ERK: Extracellular Signal‒Regulated Kinase; CREB: cAMP-Response Element Binding Protein; AST: Aspartate Transaminase; ALT: Alanine Transaminase;   

Introduction

Hyperlipidemia is a well-known metabolic derangement that predisposes to atherosclerosis and cardiovascular, cerebrovascular, and peripheral vascular diseases [1]. Hypercholesterolemia is an important factor in exacerbating these diseases. Globally, the prevalence of hyperlipidemia is reported 39%. Recent estimates have shown that around 28.5 million people from the adult population have high levels of total serum lipids, with the reported prevalence being 11.9% [2]. Serum lipid levels are reported to be influenced by anthropometric, demographic, environmental and genetic factors [3‒5]. In the Middle East, the dyslipidemia level is as high as 70% [6]. In Iran, the prevalence for hypercholesterolemia, hypertriglyceridemia, high levels of low-density lipoprotein cholesterol (LDL), and low levels of high-density lipoprotein cholesterol (HDL) are estimated as 41.6%, 46.0%, 35.5% and 43.9%, respectively among both sexes and in both rural and urban areas. Hypercholesterolemia, high LDL and low HDL are more prevalent in women, whereas hypertriglyceridemia is more prevalent in men. All types of lipid component abnormalities are more prevalent in urban residents [7].

Atherosclerosis is a lipoprotein-driven, multifocal, smoldering, immunoinflammatory disease that leads to plaque formation at specific sites of the arterial tree through intimal inflammation, necrosis, fibrosis, and calcification. Atherogenesis can be divided into five key steps, which are i) LDL oxidation caused by free radicals, ii) remodeling of extracellular matrix, iii) upregulation of adhesion molecules, iv) collection and formation of fibrous plaques, and v) endothelial dysfunction [8,9]. Reactive oxygen species (ROS) and nitric oxide (NO) can increase the LDL oxidation and may hasten the atherosclerotic process and plaque rupture [10]. 

Atherosclerotic plaques are typically described as containing a large lipid-rich necrotic core representing more than half of the plaque volume, a thin fibrous cap and a heavy infiltrate of macrophages and lymphocytes, which then turn into foamy cells that exude more fat and cause more inflammation. The coronary arteries, the large branches of the aortic arch, and the abdominal aorta and its visceral and major lower extremity branches are locations specifically susceptible to the atherosclerotic process. Plaque localization in these sites accounts for most of the clinical manifestations of the disease [8,9].

If the plaque shatters open, they’ll initiate the clotting process. Blood clot commonly improves on the irregular surfaces of arteries, which then may become separated, thus blocking the downstream blood flow. Most heart attacks and strokes happen when a plaque in heart or brain suddenly bursts. When a coronary artery is at least 60–70% blocked and stable, most usually angina is caused by physical activity or stress or emotional stress which conducts to myocardial ischemia. The best therapy for atherosclerosis/arteriosclerosis is prevention [11,12]. Hence, routine medical accesses mostly pay attention to lifestyle alterations, such as lessening in the intake of saturated fatty acids, ceasing smoking, and rhythmic exercise. Drugs are also used to diminish lipid levels or hypertension; however, most of them possess substantial side effects [13,14]. In recent years, the use of alternative therapies, especially herbal medicine and their supplements has increased in most countries to treat a variety of diseases, including hyperlipidemia, hypertension, diabetes, and cardiovascular diseases [15‒17]. One of the major problems facing physicians as well as consumers of medicinal plants is the lack of sufficient data about the safety of the drug and its effect on the disease [18,19].

Fortunately, comprehensive research have been conducted on the impressiveness of medicinal herbs used in traditional medicine over the recent years to prove their efficiencies and/or deficiencies. In this review, we aimed to acquaint some encouraging medicinal plants effective in the control and/or treatment of hyperlipidemia, in addition to presenting the probability of their poisoning and drug interaction.  

Medicinal plants that reduce blood lipids

In traditional medicine, there are various medications used to treat hyperlipidemia in which the role of medicinal plants is consequential. Recent studies executed on medicinal herbs and dietary supplements used in traditional medicine signify that the compounds demonstrate in them, including food fibers, vitamins, flavonoids, sterols, and other antioxidant compounds can decrease lipids, inhibit LDL oxidation, and eliminate oxygen free radicals. Therefore, they are effective in recovery of this disease by affecting the immune system and improving the body’s metabolic disorders [20‒22].

Cynara cardunculus var. scolymus (Artichoke)

The globe artichoke (Cynara cardunculus var. scolymus), also known by the name’s French artichoke and green artichoke in the U.S., is a variety of flowering plants characterized by leaves with sharp prickles on the margins, mostly in the family Asteraceae that cultivated as a food. Another variety of the same species is the wild cardoon, a perpetual plant native to the Mediterranean region [23,24]. This vegetable grows to 1.4–2 m tall, with intensely lobed, silvery, glaucous-green leaves. The flowers mature in a large head from an edible bud about 8–15 cm diameter with multiple triangular scales; the individual florets are purple [23,24].

The leaf extracts of C. cardunculus are greatly used alone or in connection with other herbs for embittering alcoholic and soft drinks; they are also used to prepare herbal teas or herbal medicinal products [23,24]. In Persian medicine, the plant parts are usually used for managing cardiovascular system disorders, functional anxiety and agitation [16,25].

Artichoke contains the bioactive agents apigenin, luteolin, anthocyanidins (peonidin, delphinidin and cyanidin), in conjunction with the soluble fibers inulin and pectin, and cynarin (1,3-O-dicaffeoylquinic acid) [26,27]. Apigenin and luteolin are flavones, a type of flavonoids, with a yellow crystalline appearance. Apigenin-7-rutinoside and narirutin has antioxidant and antimicrobial activities [27]. Peonidin has shown potent inhibitory and apoptotic effects on cancer cells in vitro, notably metastatic human breast cancer cells [28,29]. Cynarin, luteolin, and chlorogenic acid (5-O-caffeoylquinic acid) possesses potential effects on choleretic and cholesterol lowering, anti-LDL oxidation, anti-atherosclerotic, hepatoprotective, nephroprotective, antiviral, antioxidant, antibacterial, anti-diabetic, anti-carcinogenic, and immunomodulatory activities [28‒30]. Luteolin and chlorogenic acid are significant players in reducing the synthesis of cholesterol as well as its total level [31]. In addition, cynarin and cyanidin downregulate the expression of inducible nitric oxide synthase (iNOS) in human coronary smooth muscle cells [32]. Luteolin interfere with the biosynthesis of cholesterol and facilitate biliary secretion from the liver [28,29]. These compounds diminish cholesterol by inhibiting HMG-CoA reductase and having a hypolipidemic influence [33‒36].

Furthermore, this plant is a rich source of polyphenolic compounds, mainly caffeoylquinic acids and its derivatives (caffeic acid, dihydrocaffeic acid, ferulic acid, and dihydroferulic acid), flavonoids (apigenin and luteolin), cooperatively with the polysaccharide inulin. Caffeoylquinic acids and flavonoids present in artichoke leaf extract causes a significant decrease in the levels of total cholesterol, triglyceride, LDL and VLDL [37]. Meanwhile, artichoke leaf extract is rich in antioxidants and has a cholesterol-lowering effect; therefore, interferes with atherosclerosis [21,36]. Additionally, it is composed of fatty acids, triterpenes, and sesquiterpenes, as major metabolites [38]. Lipid-lowering sesquiterpenes, such as cynaropicrin, aguerine, and grosheimin suppress serum triglyceride level in 2 h after artichoke administration [39]. Moreover, cynaropicrin has therapeutic potential on hepatitis C virus and overpowers photo-aging of the skin by inhibiting the transcription activity of NF-κB signaling [40,41]. The main phenolic compounds encompass scavenging activity against free radicals and act as a shield against oxidative damage to proteins, lipids, and DNA [20].

Other compounds discovered in artichoke that could contribute to its lipid-lowering properties comprise the family of phytosterols. Phytosterols have been reported to lower cholesterol and LDL plasma levels and may have clinical application for the prevention of non-alcoholic fatty liver disease and cardiovascular diseases [42,43]. The physiological functions of phytosterols can be contemplated as an antioxidant, anti-inflammatory and antipyretic effects. Phytosterols will compete with cholesterol in the intestine and impede the absorption of cholesterol, thereby reducing the level of LDL in the plasma [43]. β-sitosterol and stigmasterol are the major phytosterols discovered in artichoke, which are believed to reduce cholesterol absorption from the intestine [33]. At least 1–2 g/day of isolated phytosterols are necessary to decrease LDL and cholesterol concentrations.

In a 100-gram reference serving, cooked artichoke supplies 74 calories, is a rich source of folate, and is a moderate source of vitamin K, vitamin C, magnesium, sodium, potassium and phosphorus. Six g of the dried herb is the optimal dose to reduce dyspepsia [44]. However, the best dose for hyperlipidemia supposed to be effective ranges from 2 to 3 g/day [37]. No side effect has been perceived in utilizing leaves of artichoke. However, pregnant or nursing women, young children, individuals with severe liver or kidney disease and gallstones should use the leaves with caution [45].

Medicago sativa (Alfalfa)

Medicago sativa (alfalfa, the father of all foods), also called lucerne, is a leguminous flowering plant with high contents of phytoestrogens (including spinasterol, coumestrol, and coumestan) and saponin. It has bundles of tiny purple flowers followed by fruits twisted in 2 to 3 turns containing 10–20 seeds. Dehydrated alfalfa leaf is commercially available as a dietary complement in various forms, such as tablets, powders and tea.

This plant has long been used as traditional herbal medicine and possesses cerebroprotective, cardioprotective, hepatoprotective, hypolipidemic, antioxidant, anticancer, antimicrobial, anthelminthic, estrogenic, anti-inflammatory, antiviral, and antidiabetic effects [46,47]. It is used in the treatment of heart disease, stroke, cancer, diabetes, indigestion, halitosis, constipation, and menopausal disorders in women [46,47]. Many studies proved that saponins are the main active compounds in alfalfa [48‒52].  

The whole plant material contains many important substances, including saponins (soyasapogenols, hederagenins, bayogenins, medicagenic acids, zahnic acids); sterols (β-sitosterol, α-spinasterol, stigmasterol, cycloartenol, and campesterol); coumarins (myrsellinol, scopoletin, esculetin, and 4-coumaric acid); flavones and isoflavones (quercetin, myricetin, luteolin, apigenin, chrysoeriol, tricin, medicarpin, sativan, vestitol, formononetin); phytoestrogens (coumestrol, genistein, daidzein, and biochanin A); L-canaverine; alkaloids (trigoneline, stachydrine, and homostachydrine); plant acids (malic, oxalic, malonic, maleic, and quinic); enzymes (isoflavone reductase, vestitone reductase, iminopeptidase, and two aminopeptidases); vitamins and growth factors (vitamins A, B1, B6, B12, C, E, and K1; niacin; pantothenic acid; biotin; folic acid); amino acids (valine, lysine, arginine, leucine, isoleucine, tryptophan, phenylalanine, methionine, asparagine and threonine); sugars (sucrose, fructose, arabinose, xylose, galactose, ribose, mannoheptulose); polyamines (norspermidine, and norspermine); proteins; minerals; trace elements; and other nutrients [46,47,49,50]. The alfalfa saponins of the aerial parts and roots have been identified as mono-, bis-, or tridesmosides of medicagenic acid, hederagenin, zahnic acid and soysapogenol B. Among them, medicagenic acid and hederagenin glycosides have been recognized as the biologically active saponins [46,49,50]. Saponins are responsible for the lessening of cholesterol absorption and prevention of atherosclerotic plaque formation in experimental animals. Alfalfa meals prevented hypercholestrolemia, triglyceridemia and atherogenesis [51,52].

Alfalfa top saponins have been shown to decrease cholesterolemia without changing the levels of HDL-cholesterol; hence, they reduced the total cholesterol/HDL-cholesterol ratio. Furthermore, they decreased intestinal absorption of cholesterol, increased fecal excretion of endogenous and exogenous neutral steroids and bile acids, and decreased the percentage distribution of fecal deoxycholic and lithocholic acids [49‒51]. Heat-treated M. sativa seeds decrease significantly total serum cholesterol concentrations, LDL and apolipoprotein B in type-II hyperlipoproteinemic patients. Cholestaid, a product available in the USA containing 900 mg of alfalfa extract with 100 mg citric acid, is said to neutralize the cholesterol in the stomach before it reaches the liver, thus facilitating the excretion of cholesterol from the body with no side effects or toxicity [46,49].

Health beverage manufactured from alfalfa buds was found beneficial in maintaining normal digestive function and nutrition balance in the human body, reducing cholesterol, and preventing osteoporosis, arteriosclerosis and aging. Extracts prepared from alfalfa roots may be used to prepare medical preparations like powder, pill, or decoction for lowering the levels of cholesterol and lipid in blood, improving the liver function and the control and transmission of nerve tissue, and treating calculus [46,49].   

L-Canaverine isolated from the plant has been shown to have antineoplastic activity against particular types of leukemia cells in mice and specific toxicity in dog cancer cells grown in vitro. In addition, alfalfa has been reported to be beneficial in the treatment of peptic ulcers, polycystic ovaries, prolactin excess, hemorrhage, dysuria, bowel disturbances, scurvy, secondary hypothyroidism, herpes simplex virus (HSV) infections, and arthritis [46,49,50].   

Raw alfalfa seeds and sprouts are a source of the amino acid L-canavanine. Canavanine competes with arginine, resulting in the synthesis of dysfunctional proteins [53]. Raw unsprouted alfalfa has toxic effects in primates, including humans, which can result in lupus-like syndrome or lupus flares. The lupus-like effects may include muscle pain, skin sores with inflammation and scarring, fatigue, abnormal blood test results, changes in how the immune system functions, and kidney problems [53,54]. Coumestrol, apigenin and quercetin exhibit strong estrogenic activity and have potential for use in treatment of hormone-related cancers [55]. Coumestrol lead to pathological side effects and is suspected of causing infertility in livestock [56]. Taking off coumestrol and canavanine from alfalfa leaf provided an extremely potent form of saponins, which reduce serum cholesterol levels without serious adverse effects.

Raw alfalfa seed sprouts are comprised of 93% water, 2% carbohydrates, 4% protein, and contain negligible fat. In a 100-gram reference amount, raw alfalfa sprouts supply 23 kilocalories of food energy and 29% of the daily value of vitamin K. They are a moderate source of vitamin C, some B vitamins, phosphorus, and zinc [57].

For curing high cholesterol, a dose of 40 g of heated seeds taken by mouth 3 times daily has been suggested. A dose of two tablets (1 g each) of Cholestaid taken by mouth 3 times daily for up to two months, then one tablet 3 times daily, has been recommended. Pregnant and/or lactating women, and people with a history of lupus-like disease should avoid alfalfa supplements. Meanwhile, this plant is a rich source of vitamin K. Therefore, it is contraindicated in people consuming warfarin [58]. 

Trigonella foenum-graecum L (Fenugreek)

Fenugreek (Trigonella foenum-graecum) is a medicinal clover-like plant in the family Fabaceae. This plant has a single stem that is smooth, or with dispersed tomentum. Leaves are oval, serrated, comprising of 3 small obovate to oblong leaflets. Flowers are pale yellow or whitey purple. Seeds are curved pods and have bitter and aromatic taste. This plant primarily was indigenous to Iran and then was transferred to other regions of the world [59,60].

In recent years, some health beneficial physiological attributes of fenugreek seeds have been seen in animal studies as well as human trials. These include cardioprotective, DNA-protective, gastroprotective, haemato-protective, hepatoprotective, immunomodulatory, hypocholesterolemic, antidiabetic, antihypertensive, antioxidant, antineoplastic, anti-breast cancer, anti-arthritis, anti-allergic, anti-cataract, antibacterial, endothelial dysfunction correction, thyroid dysfunction repair, antinociceptive and anti-inflammatory effects [60‒62].  

Constituents of fenugreek seeds include flavonoids (orientin, isoorientin, vitexin, epigenin, and quercetin), alkaloids (trigonelline, gentanin and carpaine choline), polyphenols, coumarins (cinnamic acid and scopoletin), vitamins (C, A, D, and B1), niacin, iron, phosphate, calcium, potassium, amino acids (4-hydroxyisoleucine, lysine, arginine, and L-tryptophan), soluble dietary fiber, gum and saponins (diosgenin, yamogenin, tigogenin, gitogenin, sarsapogenin, yuccagenin, and smilagenin) [60‒62]. Trigonelline is the major alkaloid of fenugreek and has hypoglycemic, hypolipidemic, neuroprotective, antimigraine, sedative, memory-improving, antibacterial, antiviral, and anti-tumor activities, and it has been shown to reduce diabetic auditory neuropathy and platelet aggregation [63]. It acts by affecting β‒cell regeneration, insulin secretion, activities of enzymes related to glucose metabolism, reactive oxygen species, axonal extension, and neuron excitability [63,64]. Trigonelline is transformed into niacin when the seed is grilled. Sotolon is a powerful aromatic component responsible for the distinctive maple syrup smell of fenugreek [63]. Polyphenols and germacrene-D are the most abundant components in fenugreek that could improve hyperlipidemia and the cardiovascular system [65‒67].  

4-Hydroxyisoleucine has been shown to cause an increase in pancreatic insulin secretion, and inhibit α-amylase activity, and diminish the elevated plasma triglyceride and total cholesterol levels [68‒70]. Furthermore, 4-hydroxyisoleucine administered orally (50 mg/kg/day) for 4 weeks not only lowered the blood glucose level but also significantly lowered the elevated lipids (total cholesterol, triglycerides, and LDL) level in rats [71‒73]. In experimentally hyperlipidemic rabbits, the aqueous emulsified fenugreek seeds powder reduced significantly the serum total cholesterol, LDL, and the atheriogenic index, with significant increase in the HDL [74].

Diosgenin, the main sapogenin, is an estrogen precursor and may help in managing menopause and hypolipidemia [75]. Diosgenin plays an anti-atherosclerosis role by improving endothelial dysfunction, improving the lipid profile through lowering the level of total cholesterol, triglyceride, and LDL and increasing the HDL level, inhibiting macrophage proliferation, migration and differentiation, and inhibiting vascular smooth muscle cell proliferation, migration and mineralization [76]. Moreover, diosgenin can inhibit the accumulation of triglyceride and the expression of lipogenic genes in HepG2 cells. Furthermore, diosgenin inhibited the transactivation of Liver-X-receptor-α [77,78]. Diosgenin can be used to manufacture many pharmaceuticals such as progesterone [69,76].

One more principal ingredient of fenugreek which has been stated to have modulating effect on dyslipidemia is saponin [79]. Saponins can impressively hasten cholesterol metabolism and process of reverse cholesterol transport, meanwhile interdict cholesterol synthesis and therefore effectively lessen total cholesterol level [79]. Additionally, saponins induces release of testosterone in males, increases secretory functions and induces uterine contractions in females [80,81]. Therefore, fenugreek extract and leaf must be contraindicated in women during early pregnancy to avoid risk of abortion, and during menstruation to reduce risk of excessive bleeding [81]. Furthermore, fenugreek contain a gel-like liquefiable fiber which react with bile acids and produces too big micelles which cannot be absorbed absolutely by alimentary canal and brings down the triglyceride, cholesterol and LDL levels [82]. Plus, fenugreek seed may reduce the amount of primary substrate for triglyceride synthesis [82].    

The galactomannan of fenugreek gum and the crude saponins exhibited the lipid-lowering effect [62]. Atherogenic lipids, such as triglyceride, cholesterol and LDL, were found to decrease significantly in soluble dietary fiber fraction of fenugreek-fed rats [72,73,82]. The consumption of fiber-rich fenugreek seed powder resulting in reduction of cholesterol content of liver cells coupled with upregulation of hepatic apo-B, apo-E receptors and increased clearance of circulating LDL and VLDL [83,84]. Fenugreek seeds resulted in an increase in HDL and a decrease in LDL levels in type 2 diabetic patients [85,86].  

Fenugreek was disclosed to bring about the hypocholesterolemic effect through heightened excretion of fecal bile acids and neutral sterols; depletion of cholesterol supplies in the liver was also included. Dietary fenugreek also has the property of stimulating bile formation in the liver and of converting cholesterol to bile salts [66]. Moreover, fenugreek considerably diminish fasting blood sugar, postprandial glucose and HbA1c in clinical trial studies [87,88]. 

In a double-blind clinical trial study that was performed on borderline hyperlipidemic people, total cholesterol, triglyceride, and LDL decreased significantly after 2 months of fenugreek use [89]. Moreover, hypolipidemic activity of fenugreek on type 2 diabetic patients revealed significant total cholesterol and triglyceride reduction in a meta-analysis [88]. In addition, significant effect for fenugreek was reported on total cholesterol, triglyceride, LDL reduction and HDL elevation in type 2 diabetic patients [65,67,86].

Fenugreek reduces the levels of lipid peroxidation and concomitantly increase the levels of superoxide dismutase and glutathion in the cartilage. Fenugreek also cause a decrease in the levels of pro-inflammatory cytokines such as IL-1α, IL-1β, IL-2, IL-6, and TNF-α in blood [90].

In a 100-gram reference amount, fenugreek seeds provide 323 kcal of food energy and contain 9% water, 58% carbohydrates, 23% protein, and 6% fat, with calcium at 40% of the Daily Value (DV). Fenugreek seeds (per 100 grams) are a rich source of protein (46% DV), dietary fiber, B vitamins, and dietary minerals, particularly manganese (59% DV) and iron (262% DV) [59,60].

The normal consumption of fenugreek seeds by the population in Iran is reported to be 40 g/day/adult. Some individuals who received 25 g of powdered fenugreek leaf showed digestive problems like diarrhea and cramp that were eliminated after 3 to 4 days. No important change happened in blood variables and no renal or liver side effects were observed. No special side effects have been reported for fenugreek [59].

Allium sativum L (Garlic)

Garlic (Allium sativum) is a species of bulbous flowering plant in the family Alliaceae. It is local to Iran and Central Asia. Garlic has a tall, erect flowering stem that grows up to 1 m. The leaf blade is flat, linear, and solid with an acute apex. The bulb is aromatic and includes outer layers of thin sheathing leaves encircling an inner sheath that encloses the clove. It produces hermaphrodite flowers [91].  

Some studies have demonstrated the beneficial effects of garlic consumption, such as immunomodulatory, antioxidant, cardioprotective, hepatoprotective, renoprotective, neuroprotective, DNA-protective, anti-inflammatory, antibacterial, antiviral, antifungal, anti-parasitic, anticancer, anti-hypertensive, anti-atherosclerotic, anti-hyperlipidemic, anti-obesity, antidiabetic, anti-platelet aggregation, and endothelial-dysfunction repair [92‒94].

Garlic contains 65% water, 28% carbohydrates, 2.3% organosulfur compounds, 2% proteins, 1.2% free amino-acids, and 1.5% fiber. It also contains fat-soluble vitamins (K, E, D, A), water-soluble vitamins (C, B1, B2, B3, B6, and B8), and minerals (Fe, Mg, Ca, P, K, Na, and Zn). Moreover, garlic is rich in several sulfur-containing compounds such as alliin, allicin, ajoenes, vinyldithiins, diallyl polysulfides, and S-allyl cysteine; as well as enzymes, saponins, flavonoids (quercetin), and Millard reaction products, which are not sulfur-containing compounds [93,94].

A great number of sulfur compounds participate in the smell and taste of garlic. Allicin has been discovered to be the compound most liable for the hot feeling of raw garlic. Allicin opens heat-temporary receptor potential channels that are responsible for the scorching feel of heat in foods. Cooking garlic eliminates allicin, therefore softening its piquancy. In addition, sulfur compounds in garlic are responsible for changing garlic green or blue during pickling and cooking. Under acidifying and heating conditions, the sulfoxide alliin reacts with common amino acids to make pyrroles. Alliin is a by-product of the amino acid cysteine. When fresh garlic is chopped or crushed, the enzyme alliinase converts alliin into allicin, which is responsible for the aroma of fresh garlic [93,94]. Allicin and other thiosulfinates in garlic are unsteady and create some other compounds, such as diallyl sulfide (DAS), diallyl disulfide (DADS) and diallyl trisulfide (DAT), dithiins and ajoene. Allicin, along with its decomposition products DADS and DAT, are major contributors to the characteristic odor of garlic, with other allicin-derived compounds, such as vinyldithiins and ajoene [95,96]. Aged garlic lacks allicin but may have some activity because of the presence of S-allyl cysteine. S-allyl mercaptocysteine from aged garlic has anti-proliferative, anti-metastatic and pro-apoptotic effects in various cancer models [97]. DADS is a major bioactive component of garlic and has several beneficial biological functions including anti-inflammatory, antioxidant, antimicrobial, cardioprotective, neuroprotective, and anti-carcinogenic activities [96,98,99]. The great mechanisms of action of DADS in disease prevention and/or treatment comprise inhibition of inflammation, oxidative stress, and cellular apoptosis. Mechanisms, including the activation of protein kinase B (PKB), extracellular signal-regulated kinase 1/2 (ERK1/2), protein kinase A (PKA), and cAMP-response element binding protein (CREB) and the inhibition of histone deacetylases (HDACs), can also mediate the cellular protective effects of DADS in different tissues and organs [96,98]. 

Garlic oil administration in streptozotocin- and alloxan-diabetic animals for 3 weeks at a dose of 100 mg/kg body weight diminishes the glucose, total cholesterol, triglycerides, urea, uric acid, creatinine, AST, and ALT levels and also increased serum concentration of insulin [100,101]. Moreover, a purified NO-generating protein from garlic reduces pro-inflammatory cytokine TNF-α and the stress responsive NF-κB-expression in human blood and liver cell of diabetic mice [101,102]. Fresh garlic homogenate administration reduces the serum level of glucose, total triglyceride, and total cholesterol and prevents streptozotocin-induced diabetic nephropathy possibly through the inhibition of kidney oxidative damage and increase of NO bioavailability [103]. The metabolic effects of time-released garlic powder tablets in patients with type 2 diabetes indicated that garlic reduces cardiovascular disease risks [104]. 

Additionally, hypolipidemic and hypoglycemic activity of garlic on type 2 diabetic and non-alcoholic fatty liver patients revealed significant total cholesterol, triglyceride and LDL reduction and HDL elevation [105,106]. In women with polycystic ovary syndrome, garlic supplementation significantly reduced serum total cholesterol, triglyceride, and LDL levels and systolic blood pressure [107]. In patients with metabolic syndrome, treatment with 100 mg/kg body weight raw crushed garlic 2 times a day with standard diet for 4 weeks significantly reduced components of metabolic syndrome including waist circumference, systolic and diastolic blood pressure, triglycerides, fasting blood glucose and significantly increased serum HDL [108]. Oral treatment of aqueous garlic extract on heavy metal-induced changes in serum lipid profile showed a significant decrease in serum LDL, VLDL and triglyceride level as well as increase in serum HDL level [109]. Garlic tablet administration in patients with type 2 diabetes for 12 weeks at a dose of 300 mg/kg body weight significantly reduced total cholesterol, LDL, while significantly increased HDL level [106,110]. In various studies, the hyperlipidemic patients received garlic tablets at doses of 300, 600, 900, 1200, and 1500 mg per day, respectively, for 24 weeks. The results showed a significant reduction in fasting blood glucose, HbA1c, serum cholesterol, triglyceride, and LDL levels while increased the HDL level [111‒113]. Moreover, garlic significantly improved superoxide dismutase, catalase and glutathione peroxidase in erythrocytes of diabetic patients [114]. Therapy of hyperlipidemic patients with garlic at a dose of 20 mg/kg body weight daily with 1 tablespoon lemon juice significantly decreased the total cholesterol, LDL, and fibrinogen levels [111].  

Aged black garlic is a form of raw garlic obtained via Millard reaction under high temperature and humidity for a period of time. Comprehensive in vitro and in vivo studies have demonstrated that aged black garlic has a variety of biological activities such as antioxidant, anti-inflammatory, anti-cancer, anti-obesity, anti-diabetic, anti-allergic, hypolipidemia, nephroprotective, cardioprotective, and hepatoprotective effects [115‒117]. Several studies reported higher contents of water-soluble antioxidants compounds (S-allyl cysteine, S-allyl mercaptocysteine), organosulfur compounds, 5-hydroxymethylfurfural, polyphenol, volatile compounds, and other products compared to fresh garlic after the thermal processing [115,117]. It has been reported that S-allyl cysteine sulphoxide has significant antidiabetic and hypolipidemic effects in type 2 diabetic and atherogenic rats [118‒120]. Administration of black garlic at a dose of 200 mg/kg body weight decreased the concentration of blood glucose and ameliorated diabetic condition and increased liver and intestinal HMG‒CoA reductase activity and liver hexokinase activity [116]. Intake of aged garlic extract also reduced hydroperoxide as an indicator of oxidative stress [116].  

The side effects of long period garlic consumption are greatly unknown. Possible side effects include gastrointestinal discomfort, sweating, dizziness, allergic reactions, bleeding, and menstrual irregularities [94].

If greater-than-suggested doses of garlic are taken with anticoagulant medications, this can conduct to a higher risk of bleeding. Garlic may interact with warfarin, saquinavir, antihypertensives, calcium channel blockers, the quinolone family of antibiotics such as ciprofloxacin, and hypoglycemic drugs, as well as other medications [94].

Glycine max L. (Soybean)

Glycine max, commonly called soybean, is an annual legume that belongs to Fabaceae family. It characterizes white to purple-pink flowers and trifoliate leaves. Flowers are small and ornamentally insignificant, somewhat resembling those of peas. Fruit may be a hairy pod that grows in clusters of 3‒5. Each pod contains 2‒4 seeds.

Particular studies have demonstrated the advantageous effects of soybean consumption, such as cardioprotective, neuroprotective, anti-atherosclerotic, antidiabetic, antihypertensive, anti-hyperlipidemic, anti-geriatric dementia, anti-osteoporotic, antimutagenic, anticancer, and antibacterial [121‒124].

Soybean consists of proteins (40%), carbohydrates (30%), lipids (18%), vitamins and minerals and moisture (12%). Moreover, it contains phytic acid, glyceollins, triterpenes, phenolics, isoflavones, lignans, carotenoids, coumarins, protease inhibitors, oligosaccharides, and dietary fibers [125]. Soybean oligosaccharides have the capability to advance the rapid growth of probiotic bacteria [126]. Phenolics have exhibited antioxidant properties to inhibit DNA damage caused by reactive oxygen species [127]. Glyceollins have biological activities including cholesterol-lowering, antibacterial, antifungal, antiestrogenic, insulinotropic and lessening of vascular contraction [123]. 

The major isoflavones found in soybeans are aglycons including daidzein, genistein, and glycitein; and glycosides including daidzin, genistin, and glycitin. Isoflavones have the power to activate estrogen receptors in the vagina, oocytes, and mammary glands. Moreover, isoflavones with an impact on the menstrual cycle, reduces the risk of breast cancer [128,129]. Besides, isoflavones diminishes the severity of symptoms in women with menopausal syndrome [130,131]. Isoflavones have been highlighted as phytoestrogens that lessens the risk of osteoporosis by promoting vitamin D activity and increasing calcium absorption [132]. Isoflavones have potential effects on the prevention of metabolic syndrome [133]. Amongst the soybean isoflavones, genistein inhibits the growth and development of cells that cause breast, colon, lung, prostate, and skin cancers in vitro. Furthermore, genistein restrains the formation of blisters by hindering vasculogenesis [134]. Besides, isoflavones reduce blood cholesterol by as much as 35% [134].

Soybean seeds contain 2.6% phytic acid. Phytic acid is considered a non-nutritional compound as it influences the use of bivalent ions and minerals in the body by binding to them and reducing their absorption. However, it has beneficial effects such as hypoglycemic, antioxidant, anticancer, lipid-lowering and antibacterial [135]. Phytic acid has many functions, including storage of phosphorus and cations, preventing the oxidation of cells, and lowering incidence of colorectal cancer. Plus, phytic acid displays antitumor activity [135]. Protease inhibitors of soybeans function as antioxidant and anticancer agents that restrain the production of superoxide radicals or H2O2 by tumor promotor factors. In addition, protease inhibitors in soybeans promote insulin secretion [128,129].

Lignans have characteristics similar to estrogen. Lignans suppress the biosynthesis and metabolism of estrogen, and the formation of bile acids from cholesterol. Therefore, reducing the risk of cancers. Lignans have synergistic effects with flavonoids and can intensify anticancer properties. Lignin downregulates the proliferative potential of breast cancer cells [136]. Saponin is a bipolar, heat-stable sugar complex. It possesses functions such as lowering cholesterol, stimulating immune responses, and anticancer effects. Saponins have essential roles in the inhibition of DNA synthesis in tumor cells, and in the reduction of cervical and epidermal cancer cell growth [128,129]. Soybean dietary fibers can be categorized as water–soluble (pectin and gum) and –insoluble (cellulose). Specifically, one of the most important effects of soybean dietary fiber is the lessening of cholesterol levels [129]. Soybean-derived peptides diminish hyperlipidemia by regulating trans-intestinal cholesterol excretion and bile acid synthesis [137].         

Several investigations have stated that soy protein consumption can reduce cholesterol, triglyceride, and LDL levels in hyperlipidemic and peritoneal dialysis individuals [138‒143]. Furthermore, several meta-analysis studies have reported that soy protein intake can decrease blood cholesterol and LDL level and improve the LDL/HDL ratio in hyperlipidemic patients. Intake of soy protein was allied with mean changes in serum lipid profile as a decrease of 9.3% in cholesterol, 12.9% in LDL, 10.5% in triglyceride, and an increase of 2.4% in HDL [144‒152].

Principal studies recommend 40 g of soybean protein daily. In individuals with iodine deficiency, soybean can restrain the performance of the thyroid gland. Besides, soy can decrease absorption of calcium, iron, and zinc. Clinical studies suggest that neither soy nor isoflavone intake affects male reproductive hormones [153].      

Silybum marianum (Milk thistle)

Silybum marianum is a member of Asteraceae family, which encompasses daisies and thistles. It has large purple flowering heads and grows as a stout thistle in rocky soils. The leaves are characterized by milky veins. It is one of the most thoroughly researched plants in the treatment of the liver diseases [154].

S. marianum has been found to exhibit antioxidant, anti‐inflammatory, immunomodulatory, lipid‐lowering, anti‐obesity, antihypertensive, antidiabetic, anti-cancer, anti-Parkinson’s, skin-protective, cardioprotective, renoprotective, neuroprotective, and hepatoprotective effects [155‒158].

Silymarin, derived from fruits and seeds of S. marianum, is a mixture of flavonolignans containing silibinin (silybin); isosilybin; silychristin; and silydianin. There are also small fragments of other flavonols such as dehydrosilybin, quercetin, taxifolin, and kaempferol, and phospholipids, which contains linoleic, oleic, and palmitic acids, and tocopherol [155‒158].

Silybin is the more important active ingredient of silymarin. Silybin can lessen de novo synthesis of cholesterol by inhibition of HMG-CoA reductase, which is a rate‐limiting enzyme in cholesterol biosynthesis. Moreover, silymarin can hinder resorption of cholesterol from the intestine. Besides, it decreases the synthesis of triglyceride and activates β‐oxidation of fatty acids in liver [157,158]. Also, silymarin is an ally against insulin resistance [159]. 

Several animal studies have stated that silymarin consumption can reduce cholesterol, triglyceride, and LDL levels in hyperlipidemic and hyperglycemic rats [160‒164]. Several meta‐analysis studies have indicated that silymarin supplementation in combination with other supportive therapies can reduce total cholesterol, LDL, and triglyceride and improve HDL concentration [165‒169]. Combination of silymarin with other treatments boosted its efficacy [170‒174].

Milk thistle is believed safe in dosages of 420 mg/day orally in separated doses for up to 40 months. Silymarin would possibly exert abdominal discomfort and somehow laxative effects in some patients.

Red Yeast Rice

Red yeast rice is the result of rice fermented with a type of yeast called Monascus purpureus. It contains different chemicals including monacolins (polyketides), azaphilone pigments (monascin, monascinol, ankaflavin and rubropunctamine), protein, fiber, sterols, fatty acids, flavonoids, lignans, coumarins, terpenoids, and polysaccharides [175,176]. These chemicals affect cholesterol levels. Monacolin K, which is identical to the drug lovastatin, is the most plentiful in red yeast rice. Monacolin K, inhibits production of cholesterol via blocking the activity of the HMG-CoA reductase [177,178]. Moreover, recent studies show that monacolin L, monascinol, and monascodilone also have HMG-CoA reductase activity [178]. Red yeast rice is most commonly used for hyperlipidemia, heart attack, heart disease, hypertension, diabetes, and cancer [175,176]. Red yeast rice is known to have antibacterial, anti-cancer, antioxidant, anti-inflammatory, anti-hypertensive, and hypolipidemic effects [179,180].

Red yeast rice could reduce the risk of vascular diseases associated with hyperlipidemia, through repression of atheroma formation and enhance blood fluidity, by lowering levels of chylomicrons and VLDL [180,181]. Monacolin K suppresses atheroma formation in atherosclerosis by reducing phospholipase A2 activity, improving effects on blood circulation, and pleiotropic effects on nitric oxide production. Moreover, monascin and ankaflavin inhibit oxidation of LDL, and this improves the vascular endothelial function in dyslipidemic subjects [180,181]. 

Studies have shown that certain red yeast rice products that contain statin can significantly decrease levels of total cholesterol, triglyceride, and specifically LDL [182‒188]. Long-term intake of a processed food product containing red yeast rice at 2.4 g/day significantly reduced LDL levels by 22% and total cholesterol by 16% in 12 weeks [189].

Red yeast rice might cause the same side effects as lovastatin, including liver damage, severe muscle pain, and muscle damage. People with liver problems and pregnant and/or breast-feeding women should not use red yeast rice. Cyclosporine, gemfibrozil, hepatotoxic drugs, antifungal azole drugs, cytochrome P450 3A4 (CYP3A4) inhibitors, protease inhibitors (used to treat HIV), statins, and niacin interacts with red yeast rice [189,190].

Commiphora mukul (Guggul, Guggulipid)

Guggul is made from the gum resin of the Commiphora mukul, a small thorny tree that is known as the tree of myrrh. Commiphora mukul is a seed plant within the Burseraceae. The plant is natural to southern Pakistan and western India. This tree has been employed in Ayurvedic medicine for centuries, and ancient Hindu texts dating back to 600 BC recommend it for treating atherosclerosis [191].

Guggul has antioxidant, antimicrobial, anti-inflammatory, anti-platelet aggregation, anti-infertility, antihyperglycemic, antiarthritic, antiatherosclerotic, anti-cancer, hypolipidemic, fibrinolytic, cytotoxic, thyroid stimulatory, and cardioprotective properties [192,193]. Today guggul gum resin is used for arthritis, acne, eczema, psoriasis, hyperlipidemia, and weight loss [194].

Guggul contains terpenoidal constituents (monoterpenoids, diterpenoids, triterpenoids, and sesquiterpenoids), steroids (E-guggulsterone, Z-guggulsterone, and guggulsterols), flavonoids (muscanone, quercetin, naringenin), cembranoids, guggultetrols, lignans (sesamin and diayangambin), sugars (arabinose, galactose, fructose), and amino acids [195].  

The extract of gum guggul, called guggulipid. The stereoisomers E- and Z-guggulsterone have been identified as the active agents in this extract [196]. These compounds are antagonist ligands for the bile acid receptor farnesoid X receptor (FXR), which is an important regulator of cholesterol homeostasis in the liver [197‒199]. The cembranoids did not display a considerable effect on FXR, but lowered the cholate-activated rate of human pancreatic IB phospholipase A2, which controls intestinal absorption of cholesterol [200].   

Several clinical studies have demonstrated that administration of guggulipid, the ethyl acetate extract of gum guggul, significantly lowers cholesterol, LDL, and triglyceride levels in patients with hyperlipidemia. These studies have generally reported a 10 to 20% decrease in triglyceride levels and a 20 to 30% decrease in cholesterol levels [201‒204].

Taking 3 to 6 g of guggul daily seem to lower total cholesterol, LDL, triglycerides, and raise HDL in people with hyperlipidemia who eat a normal diet. Taking guggul 3 g/daily for 4 months can improve symptoms of rheumatoid arthritis [204].

Guggul can cause side effects like dyspepsia, headaches, nausea, vomiting, loose stools, diarrhea, belching, and hiccups. Guggul can even cause hypersensitivity reactions like skin rash and itching. These adverse reactions are more common with higher doses, such as 6 g/day [204].

Guggul might act like estrogen in the body. Guggul is likely unsafe during pregnancy and breast-feeding. It appears to support menstrual flow and stimulates the uterus. Guggul can slow blood clotting and might cause bleeding or bruising in people with bleeding disorders [193,194]. Table 1 summarized the bioactive components, effects, and mechanisms of guggul and other medicinal plants on hyperlipidemia treatment.

Discussion

Hyperlipidemia has been proved to influence the antioxidant situation of various organs and their lipoprotein levels, as well [22]. Lipid-decreasing medicinal herbs may lessen hyperlipidemia, avoiding atherosclerosis and cardiovascular endothelium destruction [21]. Herbal medicine is based on the theory that plants contain natural substances that can advance health and relieve disease. Beneficial effects of medicinal herbs have been ascribed partially to their antioxidant properties [22]. The present review emphasizes the phytochemical, pharmacological, clinical and toxicological reports on medicinal plants effective on hyperlipidemia.

Hyperlipidemia enhances the yield of free radicals, which in line raises oxidative stress and LDL oxidation [20,205]. While this course, LDL converts to oxidized-LDL, causes the expression of adhesion molecules, motivates activation of T-lymphocytes and macrophages, intensifies the production of foam cells, and pulls macrophages toward the sarcoplasmic network. Oxidation of LDL has a critical role in the establishment of atherosclerotic plaques [9,10]. Free radicals are involved in a broad diversity of diseases, including diabetes, atherosclerosis, cardiovascular diseases, neurological disorders, and mental disabilities [20].

Inflammation is also related to cardiovascular disease. For instance, C-reactive protein is increased in atherosclerosis. It has been shown that patients with high C-reactive protein levels are also more tending to hypertension and type 2 diabetes, and both of these diseases are related to atherosclerosis [179]. Reduction of free radicals by plant’s antioxidants, apart from diminishing hyperlipidemia, can decrease the feasibility of atherosclerosis [20]. Several experimental and clinical studies have shown that plants that have antioxidant activity can battle pathologic conditions particularly for the treatment and prevention of life-intimidating diseases such as brain injury, diabetes, hypertension and cardiovascular diseases [118,127,206].    

Antioxidants such as selenium, and vitamins C and E were believed as being able to hinder cell membrane oxidation. Recently their common use has been limited because of their incompetent effects. Therefore, natural compounds with antioxidant and bioactive components appear to have beneficial effects. Essentially, medicinal herbs have bioactive constituents [43,93,129,207]. Hence, they can be very impressive in modulating oxidative stress, so sheltering various organs such as heart, kidney, and liver from oxidative damages. Furthermore, they have been shown to negate cardiovascular disease risk factors and are capable of providing drug supply in complementary and alternative medicine [208,209]. 

Saponins, flavonoids, phenolics, phytoestrogens, coumarins, alkaloids, amino acids, phytosterols, vitamins and terpenes constitute major classes of phyto-ingredients of medicinal plants [27,46]. Recent accomplished studies indicate their uses such as neuroprotective, renoprotective, hepatoprotective, cardioprotective, hypolipidemic, antidiabetic, antioxidant, anti-atherosclerotic, anti-ulcer, anticancer, antimicrobial, immunomodulatory, estrogenic, and nutritive for human health [43,49,94,156,158]. Despite the fact that different ingredients of medicinal plants can have antioxidant properties, the leading part of such effects is ascribed to polyphenolic compounds [38,124,161]. A lot of medicinal herbs such as artichoke, alfalfa, fenugreek, garlic, soybean, and so forth possesses antioxidant activities because of their polyphenolic compounds [38,124,125,207]. 

Conclusion

Nowadays, hyperlipidemia and its side effects are recognized as one of the health problems in most communities. In addition to intensifying the metabolic disorders, hyperlipidemia exacerbates cardiovascular disease, especially in patients with diabetes and hypertension. Ingredients in dietary supplements and medicinal herbs, including saponins, glycosides, polyphenols, flavonoids, triterpenoids, sterols, fibers and other antioxidant compounds, can affect the metabolism of lipids by influencing the metabolic reactions of various tissues. In most cases, the lipid-lowering effects of medicinal plants are due to their antioxidant properties. Finally, although promising, these results warrant further investigation before extrapolating the use of cannabis oil as a complementary nutraceutical that could help in the treatment of some metabolic disorders present in humans MS.

Author contributions

D.M., E.K.A., M.E.A. and F.B.G. conceived, designed, and collected the data and literature for the manuscript and assisted in the data analysis. I.M. supervised the study and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Acknowledgements

Thanks to all the authors for their collaboration in this study.

Funding

The authors received no financial support for the research, authorship, and/or publication of this article.

Conflicts of interest

The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Data availability

The datasets supporting the conclusions of this article are included within the article.

Ethical approval

Not applicable.

References

  1. Durrington P (2003) Dyslipidaemia. Lancet 362: 717‒731. [Crossref]
  2. Pirillo A, Casula M, Olmastroni E, Norata GD, Catapano AL (2021) Global epidemiology of dyslipidaemias. Nat Rev Cardiol 18: 689‒700. [Crossref]
  3. Dumitrescu L, Carty CL, Taylor K, Schumacher FR, Hindorff LA, et al. (2011) Genetic determinants of lipid traits in diverse populations from the population architecture using genomics and epidemiology (PAGE) study. PLoS Genet 7: e1002138. [Crossref]
  4. Bentley AR, Chen G, Shriner D, Doumatey AP, Zhou J, et al. (2014) Gene-based sequencing identifies lipid-influencing variants with ethnicity-specific effects in African Americans. PLoS Genet 10: e1004190. [Crossref]
  5. Liao PJ, Xie RB, Yin RX, Wei DX, Huang J, et al. (2015) Serum lipid profiles, the prevalence of dyslipidemia and the risk factors in two isolated Chinese minorities. Int J Clin Exp Med 8: 19200‒19211. [Crossref]
  6. Raal FJ, Alsheikh-Ali AA, Omar MI, Rashed W, Hamoui O, et al. (2018) Cardiovascular risk factor burden in Africa and the Middle East across country income categories: A post hoc analysis of the cross-sectional Africa Middle East Cardiovascular Epidemiological (ACE) study. Arch Public Health 76: 15. [Crossref]
  7. Tabatabaei-Malazy O, Qorbani M, Samavat T, Sharifi F, Larijani B, et al. (2014) Prevalence of dyslipidemia in Iran: a systematic review and meta-analysis study. Int J Prev Med 5: 373‒393. [Crossref]
  8. Insull W Jr (2009) The pathology of atherosclerosis: plaque development and plaque responses to medical treatment. Am J Med 122: S3–S14. [Crossref]
  9. Bentzon JF, Otsuka F, Virmani R, Falk E (2014) Mechanisms of plaque formation and rupture. Circ Res 114(12): 1852‒66. doi:10.1161/CIRCRESAHA.114.302721 [Crossref]
  10. Förstermann U, Xia N, Li H (2017) Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ Res 120: 713‒735. [Crossref]
  11. Badimon L, Vilahur G (2014) Thrombosis formation on atherosclerotic lesions and plaque rupture. J Intern Med 276: 618‒632. [Crossref]
  12. Duncker DJ, Koller A, Merkus D, Canty JM Jr (2015) Regulation of coronary blood flow in health and ischemic heart disease. Prog Cardiovasc Dis 57: 409‒422. [Crossref]
  13. Sundström J, Gulliksson G, Wirén M (2018) Synergistic effects of blood pressure-lowering drugs and statins: systematic review and meta-analysis. BMJ Evid Based Med 23: 64‒69. [Crossref]
  14. Hegele RA, Tsimikas S (2019) Lipid-lowering agents. Circ Res 124: 386–404.
  15. Karou SD, Tchacondo T, Djikpo Tchibozo MA, Abdoul-Rahaman S, Anani K, Koudouvo K, et al. (2011) Ethnobotanical study of medicinal plants used in the management of diabetes mellitus and hypertension in the Central Region of Togo. Pharm Biol 49: 1286‒1297. [Crossref]
  16. Baharvand-Ahmadi B, Bahmani M, Eftekhari Z, Jelodari M, Mirhoseini M (2016) Overview of medicinal plants used for cardiovascular system disorders and diseases in ethnobotany of different areas in Iran. J HerbMed Pharmacol 5: 39‒44.
  17. Shaito A, Thuan DTB, Phu HT, Nguyen THD, Hasan H, et al. (2020) Herbal medicine for cardiovascular diseases: efficacy, mechanisms, and safety. Front Pharmacol 11: 422. [Crossref]
  18. Ekor M (2014) The growing use of herbal medicines: issues relating to adverse reactions and challenges in monitoring safety. Front Pharmacol 4: 177. [Crossref]
  19. Fitzgerald M, Heinrich M, Booker A (2020) Medicinal plant analysis: a historical and regional discussion of emergent complex techniques. Front Pharmacol 10: 1480. [Crossref]
  20. Lobo V, Patil A, Phatak A, Chandra N (2010) Free radicals, antioxidants and functional foods: impact on human health. Pharmacogn Rev 4: 118‒126. [Crossref]
  21. Kirichenko TV, Sukhorukov VN, Markin AM, Nikiforov NG, Liu PY, et al. (2020) Medicinal plants as a potential and successful treatment option in the context of atherosclerosis. Front Pharmacol 11: 403. [Crossref]
  22. Song L, Zhang J, Lai R, Li Q, Ju J, Xu H (2021) Chinese herbal medicines and active metabolites: potential antioxidant treatments for atherosclerosis. Front Pharmacol 12: 675999. [Crossref]
  23. Lattanzio V, Kroon PA, Linsalata V, Cardinali A (2009) Globe artichoke: a functional food and source of nutraceutical ingredients. J Funct Foods 1: 131–144.
  24. Ceccarelli N, Curadi M, Picciarelli P, Martelloni L, Sbrana C, et al. (2010) Globe artichoke as a functional food. Mediterr J Nutr Metab 3: 197–201.
  25. Bahmani M, Mirhoseini M, Shirzad H, Sedighi M, Shahinfard N, et al.  (2015) A review on promising natural agents effective on hyperlipidemia. J Evid Based Complementary Altern Med 20: 228‒238. [Crossref]
  26. Gonçalves AC, Nunes AR, Falcão A, Alves G, Silva LR (2021) Dietary effects of anthocyanins in human health: a comprehensive review. Pharmaceuticals 14: 690. [Crossref]
  27. Sharma P, Verma PK, Pankaj NK, Agarwal S (2021) The phytochemical ingredients and therapeutic potential of Cynara scolymus L. Pharm Biomed Res 7: 141‒60.
  28. Zayed A, Serag A, Farag MA (2020) Cynara cardunculus L.: outgoing and potential trends of phytochemical, industrial, nutritive and medicinal merits. J Funct Foods 69: 103937.
  29. de Falco B, Incerti G, Amato M, Lanzotti V (2015) Artichoke: botanical, agronomical, phytochemical, and pharmacological overview. Phytochem Rev 14: 993–1018.
  30. Jalili C, Moradi S, Babaei A, Boozari B, Asbaghi O, et al. (2020) Effects of Cynara scolymus L. on glycemic indices: a systematic review and meta-analysis of randomized clinical trials. Complement Ther Med 52: 102496.  [Crossref]
  31. Shahinfar H, Bazshahi E, Amini MR, Payandeh N, Pourreza S, et al. (2021) Effects of artichoke leaf extract supplementation or artichoke juice consumption on lipid profile: a systematic review and dose-response meta-analysis of randomized controlled trials. Phytother Res 35: 6607‒6623. [Crossref]
  32. Xia N, Pautz A, Wollscheid U, Reifenberg G, Förstermann U, et al. (2014) Artichoke, cynarin and cyanidin downregulate the expression of inducible nitric oxide synthase in human coronary smooth muscle cells. Molecules 19: 3654‒3668. [Crossref]
  33. Santos HO, Bueno AA, Mota JF (2018) The effect of artichoke on lipid profile: a review of possible mechanisms of action. Pharmacol Res 137: 170‒8. [Crossref]
  34. Riva A, Petrangolini G, Allegrini P, Perna S, Giacosa A, Peroni G, et al. (2021) Artichoke and bergamot phytosome alliance: a randomized double blind clinical trial in mild hypercholesterolemia. Nutrients 14: 108. [Crossref]
  35. Frigerio J, Tedesco E, Benetti F, Insolia V, Nicotra G, et al. (2021) Anticholesterolemic activity of three vegetal extracts (artichoke, caigua, and fenugreek) and their unique blend. Front Pharmacol 12: 726199. [Crossref]
  36. Sahebkar A, Pirro M, Banach M, Mikhailidis DP, Atkin SL, et al. (2018) Lipid-lowering activity of artichoke extracts: a systematic review and meta-analysis. Crit Rev Food Sci Nutr 58: 2549‒2556. [Crossref]
  37. Wittemer SM, Ploch M, Windeck T, Müller SC, Drewelow B, et al. (2005) Bioavailability and pharmacokinetics of caffeoylquinic acids and flavonoids after oral administration of artichoke leaf extracts in humans. Phytomedicine 12: 28‒38. [Crossref]
  38. Rocchetti G, Giuberti G, Lucchini F, Lucini L (2020) Polyphenols and sesquiterpene lactones from artichoke heads: modulation of starch digestion, gut bioaccessibility, and bioavailability following in vitro digestion and large intestine fermentation. Antioxidants 9: 306. [Crossref]
  39. Shimoda H, Ninomiya K, Nishida N, Yoshino T, Morikawa T, et al. (2003) Anti-hyperlipidemic sesquiterpenes and new sesquiterpene glycosides from the leaves of artichoke (Cynara scolymus L.): structure requirement and mode of action. Bioorg Med Chem Lett 13: 223‒228. [Crossref]
  40. Elsebai MF, Mocan A, Atanasov AG (2016) Cynaropicrin: a comprehensive research review and therapeutic potential as an anti-hepatitis C virus agent. Front Pharmacol 7: 472. [Crossref]
  41. Tanaka YT, Tanaka K, Kojima H, Hamada T, Masutani T, et al. (2013) Cynaropicrin from Cynara scolymus L. suppresses photo aging of skin by inhibiting the transcription activity of nuclear factor-kappa B. Bioorg Med Chem Lett 23: 518‒523. [Crossref]
  42. Javanmardi MA, Mohammad Shahi M, Seyedian SS, Haghighizadeh MH (2018) Effects of phytosterol supplementation on serum levels of lipid profiles, liver enzymes, inflammatory markers, adiponectin, and leptin in patients affected by nonalcoholic fatty liver disease: a double-blind, placebo-controlled, randomized clinical trial. J Am Coll Nutr 16:1‒8. [Crossref]
  43. Li X, Xin Y, Mo Y, Marozik P, He T, et al. (2022) The bioavailability and biological activities of phytosterols as modulators of cholesterol metabolism. Molecules 27: 523. [Crossref]
  44. Giacosa A, Guido D, Grassi M, Riva A, Morazzoni P, et al. (2015) The effect of ginger (Zingiber officinalis) and artichoke (Cynara cardunculus) extract supplementation on functional dyspepsia: a randomised, double-blind, and placebo-controlled clinical trial. Evid Based Complement Alternat Med 2015: 915087. [Crossref]
  45. Bent S, Ko R (2004) Commonly used herbal medicines in the United States: a review. Am J Med 116: 478‒485. [Crossref]
  46. Bora KS, Sharma A (2011) Phytochemical and pharmacological potential of Medicago sativa: a review. Pharm Biol 49: 211‒220. [Crossref]
  47. Karimi E, Oskoueian E, Oskoueian A, Omidvar V, Hendra R, et al. (2013) Insight into the functional and medicinal properties of Medicago sativa (Alfalfa) leaves extract. J Med Plant Res 7: 290‒297.
  48. Cui Y, Li F, Zhu X, Xu J, Muhammad A, et al. (2022) Alfalfa saponins inhibit oxidative stress-induced cell apoptosis through the MAPK signaling pathway. Redox Rep 27: 1‒8. [Crossref]
  49. Wang G, Wang J, Liu W, Nisar MF, El-Esawi MA, et al. (2021) Biological activities and chemistry of triterpene saponins from Medicago species: an update review. Evid Based Complement Alternat Med 2021: 6617916. [Crossref]
  50. Tava A, Avato P (2006) Chemical and biological activity of triterpene saponins from Medicago species. Nat Prod Commun 1: 1159‒1180.
  51. Yu CH, Xie G, He RR, Zhai YJ, Li YF, et al. (2011) Effects of a purified saponin mixture from alfalfa on plasma lipid metabolism in hyperlipidemic mice. J Health Sci 57: 401–405.
  52. Shi Y, Guo R, Wang X, Yuan D, Zhang S, et al. (2014) The regulation of alfalfa saponin extract on key genes involved in hepatic cholesterol metabolism in hyperlipidemic rats. PLoS One 9: e88282. [Crossref]
  53. Akaogi J, Barker T, Kuroda Y, Nacionales DC, Yamasaki Y, et al. (2006) Role of non-protein amino acid L-canavanine in autoimmunity. Autoimmun Rev 5: 429‒435. [Crossref]
  54. Bax CE, Chakka S, Concha JSS, Zeidi M, Werth VP (2021) The effects of immunostimulatory herbal supplements on autoimmune skin diseases. J Am Acad Dermatol 84: 1051‒1058. [Crossref]
  55. Domínguez-López I, Yago-Aragón M, Salas-Huetos A, Tresserra-Rimbau A, Hurtado-Barroso S (2020) Effects of dietary phytoestrogens on hormones throughout a human lifespan: a review. Nutrients 12: 2456. [Crossref]
  56. Ferreira-Dias G, Botelho M, Zagrajczuk A, Rebordão MR, Galvão AM, et al. (2013) Coumestrol and its metabolite in mares’ plasma after ingestion of phytoestrogen-rich plants: potent endocrine disruptors inducing infertility. Theriogenology 80: 684‒692. [Crossref]
  57. He C, Fu P, Zhang K, Xia Q, Yang Y, et al. (2018) Chinese herbal medicine for dyslipidemia: protocol for a systematic review and meta-analysis. Medicine 97: e13048. [Crossref]
  58. Liu Y, Liu Y, Yang J, Wang X, Xiu C, et al. (2021) Chinese herbal medicine for hypertension complicated with hyperlipidemia: a protocol for a systematic review and meta-analysis. Medicine 100: e24345. [Crossref]
  59. Bahmani M, Shirzad H, Mirhosseini M, Mesripour A, Rafieian-Kopaei M (2016) A review on ethnobotanical and therapeutic uses of fenugreek (Trigonella foenum-graceum L). J Evid Based Complementary Altern Med 21: 53‒62. [Crossref]
  60. Nagulapalli Venkata KC, Swaroop A, Bagchi D, Bishayee A (2017) A small plant with big benefits: fenugreek (Trigonella foenum-graecum Linn.) for disease prevention and health promotion. Mol Nutr Food Res 61: 950. [Crossref]
  61. Visuvanathan T, Than LTL, Stanslas J, Chew SY, Vellasamy S (2022) Revisiting Trigonella foenum-graecum L.: pharmacology and therapeutic potentialities. Plants 11: 1450. [Crossref]
  62. Yadav UCS, Baquer NZ (2014) Pharmacological effects of Trigonella foenum-graecum L. in health and disease. Pharm Biol 52: 243–254. [Crossref]
  63. Zhou J, Chan L, Zhou S (2012) Trigonelline: a plant alkaloid with therapeutic potential for diabetes and central nervous system disease. Curr Med Chem 19: 3523‒3531. [Crossref]
  64. Subramanian SP, Prasath GS (2014) Antidiabetic and antidyslipidemic nature of trigonelline, a major alkaloid of fenugreek seeds studied in high-fat-fed and low-dose streptozotocin-induced experimental diabetic rats. Biomed Prev Nutr 4: 475‒480.
  65. Geberemeskel GA, Debebe YG, Nguse NA (2019) Antidiabetic effect of fenugreek seed powder solution (Trigonella foenum-graecum L.) on hyperlipidemia in diabetic patients. J Diabetes Res 2019: 8507453. [Crossref]
  66. Askarpour M, Alami F, Campbell MS, Venkatakrishnan K, Hadi A, et al. (2020) Effect of fenugreek supplementation on blood lipids and body weight: a systematic review and meta-analysis of randomized controlled trials. J Ethnopharmacol 253: 112538. [Crossref]
  67. Heshmat-Ghahdarijani K, Mashayekhiasl N, Amerizadeh A, Teimouri Jervekani Z, Sadeghi M (2020) Effect of fenugreek consumption on serum lipid profile: a systematic review and meta-analysis. Phytother Res 34: 2230‒2245. [Crossref]
  68. Narender T, Puri A, Shweta, Khaliq T, Saxena R, Bhatia G, Chandra R (2006) 4-Hydroxyisoleucine an unusual amino acid as antidyslipidemic and antihyperglycemic agent. Bioorg Med Chem Lett 16: 293‒296. [Crossref]
  69. Fuller S, Stephens JM (2015) Diosgenin, 4-hydroxyisoleucine, and fiber from fenugreek: mechanisms of actions and potential effects on metabolic syndrome. Adv Nutr 6: 189‒197. [Crossref]
  70. Avalos-Soriano A, De la Cruz-Cordero R, Rosado JL, Garcia-Gasca T (2016) 4-Hydroxyisoleucine from fenugreek (Trigonella foenum-graecum): effects on insulin resistance associated with obesity. Molecules 21: 1596. [Crossref]
  71. Annida B, Stanely Mainzen Prince P (2004) Supplementation of fenugreek leaves lower lipid profile in streptozotocin-induced diabetic rats. J Med Food 7: 153‒156. [Crossref]
  72. Belguith-Hadriche O, Bouaziz M, Jamoussi K, El Feki A, Sayadi S, et al. (2010) Lipid-lowering and antioxidant effects of an ethyl acetate extract of fenugreek seeds in high-cholesterol-fed rats. J Agric Food Chem 58: 2116‒2122. [Crossref]
  73. Mohammad-Sadeghipour M, Afsharinasab M, Mohamadi M, Mahmoodi M, Falahati-Pour SK, et al. (2020) The effects of hydro-alcoholic extract of fenugreek seeds on the lipid profile and oxidative stress in fructose-fed rats. J Obes Metab Syndr 29: 198‒207. [Crossref]
  74. Sharma MS, Choudhary PR (2017) Effect of fenugreek seeds powder (Trigonella foenum-graecum L.) on experimental induced hyperlipidemia in rabbits. J Diet Suppl 14: 1‒8. [Crossref]
  75. Sun F, Yang X, Ma C, Zhang S, Yu L, et al. (2021) The effects of diosgenin on hypolipidemia and its underlying mechanism: a review. Diabetes Metab Syndr Obes 14: 4015‒4030. [Crossref]
  76. Wu FC, Jiang JG (2019) Effects of diosgenin and its derivatives on atherosclerosis. Food Funct 10: 7022‒7036. [Crossref]
  77. Uemura T, Goto T, Kang MS, Mizoguchi N, Hirai S, et al. (2011) Diosgenin, the main aglycon of fenugreek, inhibits LXRα activity in HepG2 cells and decreases plasma and hepatic triglycerides in obese diabetic mice. J Nutr 141: 17‒23. [Crossref]
  78. Khateeb S, Albalawi A, Alkhedaide A (2021) Regulatory effect of diosgenin on lipogenic genes expression in high-fat diet-induced obesity in mice. Saudi J Biol Sci 28: 1026‒1032. [Crossref]
  79. Chen Z, Lei YL, Wang WP, Lei YY, Liu YH, Hei J, et al. (2017) Effects of saponin from Trigonella foenum-graecum seeds on dyslipidemia. Iran J Med Sci 42: 577‒585. [Crossref]
  80. Maheshwari A, Verma N, Swaroop A, Bagchi M, Preuss HG, et al. (2017) Efficacy of FurosapTM, a novel Trigonella foenum-graecum seed extract, in enhancing testosterone level and improving sperm profile in male volunteers. Int J Med Sci 14: 58‒66. [Crossref]
  81. Park HJ, Lee KS, Lee EK, Park NC (2018) Efficacy and safety of a mixed extract of Trigonella foenum-graecum seed and Lespedeza cuneata in the treatment of testosterone deficiency syndrome: a randomized, double-blind, placebo-controlled clinical trial. World J Men’s Health 36: 230‒238. [Crossref]
  82. Hannan JM, Ali L, Rokeya B, Khaleque J, Akhter M, et al. (2007) Soluble dietary fibre fraction of Trigonella foenum-graecum (fenugreek) seed improves glucose homeostasis in animal models of type 1 and type 2 diabetes by delaying carbohydrate digestion and absorption, and enhancing insulin action. Br J Nutr 97: 514‒521. [Crossref]
  83. Venkatesan N, Devaraj SN, Devaraj H (2003) Increased binding of LDL and VLDL to apo B, E receptors of hepatic plasma membrane of rats treated with Fibernat. Eur J Nutr 42: 262‒271. [Crossref]
  84. Vijayakumar MV, Pandey V, Mishra GC, Bhat MK (2010) Hypolipidemic effect of fenugreek seeds is mediated through inhibition of fat accumulation and upregulation of LDL receptor. Obesity 18: 667‒674. [Crossref]
  85. Kassaian N, Azadbakht L, Forghani B, Amini M (2009) Effect of fenugreek seeds on blood glucose and lipid profiles in type 2 diabetic patients. Int J Vitam Nutr Res 79: 34‒39. [Crossref]
  86. Verma N, Usman K, Patel N, Jain A, Dhakre S, et al. (2016) A multicenter clinical study to determine the efficacy of a novel fenugreek seed (Trigonella foenum-graecum) extract (Fenfuro™) in patients with type 2 diabetes. Food Nutr Res 60: 32382. [Crossref]
  87. Neelakantan N, Narayanan M, de Souza RJ, van Dam RM (2014) Effect of fenugreek (Trigonella foenum-graecum L.) intake on glycemia: a meta-analysis of clinical trials. Nutr J 13: 7. [Crossref]
  88. Gong J, Fang K, Dong H, Wang D, Hu M, et al. (2016) Effect of fenugreek on hyperglycaemia and hyperlipidemia in diabetes and prediabetes: a meta-analysis. J Ethnopharmacol 194: 260‒268. [Crossref]
  89. Yousefi E, Zareiy S, Zavoshy R, Noroozi M, Jahanihashemi H, et al. (2017) Fenugreek: a therapeutic complement for patients with borderline hyperlipidemia: a randomised, double-blind, placebo-controlled, clinical trial. Adv Integr Med 4: 31‒35.
  90. Sindhu G, Shyni GL, Pushpan CK, Nambisan B, Helen A (2018) Evaluation of anti-arthritic potential of Trigonella foenum graecum L. (fenugreek) mucilage against rheumatoid arthritis. Prostaglandins Other Lipid Mediat 138: 48‒53. [Crossref]
  91. Stavělíková H (2008) Morphological characteristics of garlic (Allium sativum L.) genetic resources collection – information. Hort Sci 35: 130‒135.
  92. Ansary J, Forbes-Hernández TY, Gil E, Cianciosi D, Zhang J, et al. (2020) Potential health benefit of garlic based on human intervention studies: a brief overview. Antioxidants 9: 619. [Crossref]
  93. Shang A, Cao SY, Xu XY, Gan RY, Tang GY, et al. (2019) Bioactive compounds and biological functions of garlic (Allium sativum L.). Foods 8: 246. [Crossref]
  94. Melguizo-Rodríguez L, García-Recio E, Ruiz C, De Luna-Bertos E, Illescas-Montes R, et al. (2022) Biological properties and therapeutic applications of garlic and its components. Food Funct 13: 2415‒2426. [Crossref]
  95. Nakamoto M, Kunimura K, Suzuki JI, Kodera Y (2020) Antimicrobial properties of hydrophobic compounds in garlic: allicin, vinyldithiin, ajoene and diallyl polysulfides. Exp Ther Med 19: 1550‒1553. [Crossref]
  96. He H, Ma Y, Huang H, Huang C, Chen Z, et al. (2021) A comprehensive understanding about the pharmacological effect of diallyl disulfide other than its anti-carcinogenic activities. Eur J Pharmacol 893: 173803. [Crossref]
  97. Lv Y, So KF, Wong NK, Xiao J (2019) Anti-cancer activities of S-allylmercaptocysteine from aged garlic. Chin J Nat Med 17: 43‒49. [Crossref]
  98. Song X, Yue Z, Nie L, Zhao P, Zhu K, et al. (2021) Biological functions of diallyl disulfide, a garlic-derived natural organic sulfur compound. Evid Based Complement Alternat Med 2021: 5103626. [Crossref]
  99. Alrumaihi F, Khan MA, Babiker AY, Alsaweed M, Azam F, et al. (2022) The effect of liposomal diallyl disulfide and oxaliplatin on proliferation of colorectal cancer cells: in vitro and in silico analysis. Pharmaceutics 14: 236. [Crossref]
  100. Madkor HR, Mansour SW, Ramadan G (2011) Modulatory effects of garlic, ginger, turmeric and their mixture on hyperglycaemia, dyslipidaemia and oxidative stress in streptozotocin-nicotinamide diabetic rats. Br J Nutr 105: 1210‒1217. [Crossref]
  101. Bhattacharya S, Maji U, Khan GA, Das R, Sinha AK, et al. (2019) Antidiabetic role of a novel protein from garlic via NO in expression of Glut-4/insulin in liver of alloxan induced diabetic mice. Biomed Pharmacother 111: 1302‒1314. [Crossref]
  102. Keiss HP, Dirsch VM, Hartung T, Haffner T, Trueman L, et al. (2003) Garlic (Allium sativum L.) modulates cytokine expression in lipopolysaccharide-activated human blood thereby inhibiting NF-κB activity. J Nutr 133: 2171‒2175. [Crossref]
  103. Mariee AD, Abd-Allah GM, El-Yamany MF (2009) Renal oxidative stress and nitric oxide production in streptozotocin-induced diabetic nephropathy in rats: the possible modulatory effects of garlic (Allium sativum L.). Biotechnol Appl Biochem 52: 227‒232.
  104. Sobenin IA, Myasoedova VA, Iltchuk MI, Zhang DW, Orekhov AN (2019) Therapeutic effects of garlic in cardiovascular atherosclerotic disease. Chin J Nat Med 17: 721‒728. [Crossref]
  105. Sangouni AA, Mohammad Hosseini Azar MR, Alizadeh M (2020) Effect of garlic powder supplementation on hepatic steatosis, liver enzymes and lipid profile in patients with non-alcoholic fatty liver disease: a double-blind randomised controlled clinical trial. Br J Nutr 124: 450‒456. [Crossref]
  106. Shabani E, Sayemiri K, Mohammadpour M (2019) The effect of garlic on lipid profile and glucose parameters in diabetic patients: a systematic review and meta-analysis. Prim Care Diabetes 13: 28‒42. [Crossref]
  107. Zadhoush R, Alavi-Naeini A, Feizi A, Naghshineh E, Ghazvini MR (2021) The effect of garlic (Allium sativum) supplementation on the lipid parameters and blood pressure levels in women with polycystic ovary syndrome: a randomized controlled trial. Phytother Res 35: 6335‒6342. [Crossref]
  108. Choudhary PR, Jani RD, Sharma MS (2018) Effect of raw crushed garlic (Allium sativum L.) on components of metabolic syndrome. J Diet Suppl 15: 499‒506. [Crossref]
  109. Das Gupta A, Das SN, Dhundasi SA, Das KK (2008) Effect of garlic (Allium sativum) on heavy metal (nickel II and chromium VI) induced alteration of serum lipid profile in male albino rats. Int J Environ Res Public Health 5: 147‒151. [Crossref]
  110. Wang J, Zhang X, Lan H, Wang W (2017) Effect of garlic supplement in the management of type 2 diabetes mellitus (T2DM): a meta-analysis of randomized controlled trials. Food Nutr Res 61: 1377571. [Crossref]
  111. Aslani N, Entezari MH, Askari G, Maghsoudi Z, Maracy MR (2016) Effect of garlic and lemon juice mixture on lipid profile and some cardiovascular risk factors in people 30‒60 years old with moderate hyperlipidaemia: a randomized clinical trial. Int J Prev Med 7: 95. [Crossref]
  112. Sun YE, Wang W, Qin J (2018) Anti-hyperlipidemia of garlic by reducing the level of total cholesterol and low-density lipoprotein: a meta-analysis. Medicine 97: e0255. [Crossref]
  113. Chan WJJ, McLachlan AJ, Luca EJ, Harnett JE (2020) Garlic (Allium sativum L.) in the management of hypertension and dyslipidemia – a systematic review. J Herb Med 19: 100292.
  114. Askari M, Mozaffari H, Darooghegi Mofrad M, Jafari A, Surkan PJ, et al. (2021) Effects of garlic supplementation on oxidative stress and antioxidative capacity biomarkers: a systematic review and meta-analysis of randomized controlled trials. Phytother Res 35: 3032‒3045. [Crossref]
  115. Ryu JH, Kang D (2017) Physicochemical properties, biological activity, health benefits, and general limitations of aged black garlic: a review. Molecules 22: 919. [Crossref]
  116. Ahmed T, Wang CK (2021) Black garlic and its bioactive compounds on human health diseases: a review. Molecules 26: 5028. [Crossref]
  117. Afzaal M, Saeed F, Rasheed R, Hussain M, Aamir M, et al. (2021) Nutritional, biological, and therapeutic properties of black garlic: a critical review. Int J Food Prop 24: 1387‒1402.
  118. Lee YM, Gweon OC, Seo YJ, Im J, Kang MJ, et al. (2009) Antioxidant effect of garlic and aged black garlic in animal model of type 2 diabetes mellitus. Nutr Res Pract 3: 156‒161. [Crossref]
  119. Seo YJ, Gweon OC, Im J, Lee YM, Kang MJ, et al. (2009) Effect of garlic and aged black garlic on hyperglycemia and dyslipidemia in animal model of type 2 diabetes mellitus. J Food Sci Nutr 14: 1‒7.
  120. Najman K, Sadowska A, Buczak K, Leontowicz H, Leontowicz M (2022) Effect of heat-treated garlic (Allium sativum L.) on growth parameters, plasma lipid profile and histological changes in the ileum of atherogenic rats. Nutrients 14: 336. [Crossref]
  121. Ahmad A, Hayat I, Arif S, Masud T, Khalid N, et al. (2014) Mechanisms involved in the therapeutic effects of soybean (Glycine max). Int J Food Prop 17: 1332‒1354.
  122. Pham TH, Lecomte S, Efstathiou T, Ferriere F, Pakdel F (2019) An update on the effects of glyceollins on human health: possible anticancer effects and underlying mechanisms. Nutrients 11: 79. [Crossref]
  123. Wu H, Zhang Z, Huang H, Li Z (2017) Health benefits of soy and soy phytochemicals. AME Med J 2: 162.
  124. Khosravi A, Razavi SH (2021) Therapeutic effects of polyphenols in fermented soybean and black soybean products. J Funct Foods 81: 104467.
  125. Chatterjee C, Gleddie S, Xiao CW (2018) Soybean bioactive peptides and their functional properties. Nutrients 10: 1211. [Crossref]
  126. Zhang C, Zhang Y, Liu G, Li W, Xia S, et al. (2021) Effects of soybean protein isolates and peptides on the growth and metabolism of Lactobacillus rhamnosus. J Funct Foods 77: 104335.
  127. Król-Grzymała A, Amarowicz R (2020) Phenolic compounds of soybean seeds from two European countries and their antioxidant properties. Molecules 25: 2075. [Crossref
  128. Kim IS, Kim CH, Yang WS (2021) Physiologically active molecules and functional properties of soybeans in human health‒a current perspective. Int J Mol Sci 22: 4054. [Crossref]
  129. Kim IS, Yang WS, Kim CH (2021) Beneficial effects of soybean-derived bioactive peptides. Int J Mol Sci 22: 8570. [Crossref]
  130. Chen LR, Chen KH (2021) Utilization of isoflavones in soybeans for women with menopausal syndrome: an overview. Int J Mol Sci 22: 3212. [Crossref]
  131. Rizzo G, Feraco A, Storz MA, Lombardo M (2022) The role of soy and soy isoflavones on women’s fertility and related outcomes: an update. J Nutr Sci 11: e17. [Crossref]
  132. Gómez-Zorita S, González-Arceo M, Fernández-Quintela A, Eseberri I, Trepiana J, et al. (2020) Scientific evidence supporting the beneficial effects of isoflavones on human health. Nutrients 12: 3853. [Crossref]
  133. Yamagata K, Yamori Y (2021) Potential effects of soy isoflavones on the prevention of metabolic syndrome. Molecules 26: 5863. [Crossref]
  134. Garbiec E, Cielecka-Piontek J, Kowalówka M, Hołubiec M, Zalewski P (2022) Genistein-opportunities related to an interesting molecule of natural origin. Molecules 27: 815. [Crossref]
  135. Kumar A, Singh B, Raigond P, Sahu C, Mishra UN, et al. (2021) Phytic acid: blessing in disguise, a prime compound required for both plant and human nutrition. Food Res Int 142: 110193. [Crossref]
  136. Vinardell MP, Mitjans M (2017) Lignins and their derivatives with beneficial effects on human health. Int J Mol Sci 18: 1219. [Crossref]
  137. Lee H, Shin E, Kang H, Youn H, Youn B (2021) Soybean-derived peptides attenuate hyperlipidemia by regulating trans-intestinal cholesterol excretion and bile acid synthesis. Nutrients 14: 95. [Crossref]
  138. Wang Y, Jones PJH, Ausman LM, Lichtenstein AH (2004) Soy protein reduces triglyceride levels and triglyceride fatty acid fractional synthesis rate in hypercholesterolemic subjects. Atherosclerosis 173: 269–275. [Crossref]
  139. Chen ST, Chen JR, Yang CS, Peng SJ, Ferng SH (2006) Effect of soya protein on serum lipid profile and lipoprotein concentrations in patients undergoing hypercholesterolaemic haemodialysis. Br J Nutr 95: 366‒371. [Crossref]
  140. Maki KC, Butteiger DN, Rains TM, Lawless A, Reeves MS, et al. (2010) Effects of soy protein on lipoprotein lipids and fecal bile acid excretion in men and women with moderate hypercholesterolemia. J Clin Lipidol 4: 531‒542. [Crossref]
  141. Tabibi H, Imani H, Hedayati M, Atabak S, Rahmani L (2010) Effects of soy consumption on serum lipids and apoproteins in peritoneal dialysis patients: a randomized controlled trial. Perit Dial Int 30: 611‒618. [Crossref]
  142. Wofford MR, Rebholz CM, Reynolds K, Chen J, Chen CS, Myers L, et al. (2012) Effect of soy and milk protein supplementation on serum lipid levels: a randomized controlled trial. Eur J Clin Nutr 66: 419‒425. [Crossref]
  143. Ramdath DD, Padhi EM, Sarfaraz S, Renwick S, Duncan AM (2017) Beyond the cholesterol-lowering effect of soy protein: a review of the effects of dietary soy and its constituents on risk factors for cardiovascular disease. Nutrients 9: 324. [Crossref]
  144. Yeung J, Yu TF (2003) Effects of isoflavones (soy phyto-estrogens) on serum lipids: a meta-analysis of randomized controlled trials. Nutr J 2: 15. [Crossref]
  145. Zhuo XG, Melby MK, Watanabe S (2004) Soy isoflavone intake lowers serum LDL cholesterol: a meta-analysis of 8 randomized controlled trials in humans. J Nutr 134: 2395‒2400. [Crossref]
  146. Zhan S, Ho SC (2005) Meta-analysis of the effects of soy protein containing isoflavones on the lipid profile. Am J Clin Nutr 81: 397‒408. [Crossref]
  147. Reynolds K, Chin A, Lees KA, Nguyen A, Bujnowski D, He J (2006) A meta-analysis of the effect of soy protein supplementation on serum lipids. Am J Cardiol 98: 633‒640. [Crossref]
  148. Sirtori CR, Eberini I, Arnoldi A (2007) Hypocholesterolaemic effects of soya proteins: results of recent studies are predictable from the Anderson meta-analysis data. Br J Nutr 97: 816–822. [Crossref]
  149. Taku K, Umegaki K, Sato Y, Taki Y, Endoh K, et al. (2007) Soy isoflavones lower serum total and LDL cholesterol in humans: a meta-analysis of 11 randomized controlled trials. Am J Clin Nutr 85: 1148‒1156. [Crossref]
  150. Tokede OA, Onabanjo TA, Yansane A, Gaziano JM, Djoussé L (2015) Soya products and serum lipids: a meta-analysis of randomised controlled trials. Br J Nutr 114: 831‒843. [Crossref]
  151. Moradi M, Daneshzad E, Azadbakht L (2020) The effects of isolated soy protein, isolated soy isoflavones and soy protein containing isoflavones on serum lipids in postmenopausal women: a systematic review and meta-analysis. Crit Rev Food Sci Nutr 60: 3414‒3428. [Crossref]
  152. Barańska A, Błaszczuk A, Kanadys W, Baczewska B, Jędrych M, et al. (2021) Effects of soy protein containing of isoflavones and isoflavones extract on plasma lipid profile in postmenopausal women as a potential prevention factor in cardiovascular diseases: systematic review and meta-analysis of randomized controlled trials. Nutrients 13: 2531. [Crossref]
  153. Reed KE, Camargo J, Hamilton-Reeves J, Kurzer M, Messina M (2021) Neither soy nor isoflavone intake affects male reproductive hormones: an expanded and updated meta-analysis of clinical studies. Reprod Toxicol 100: 60‒67. [Crossref]
  154. Gillessen A, Schmidt HHJ (2020) Silymarin as supportive treatment in liver diseases: a narrative review. Adv Ther 37: 1279–1301. [Crossref]
  155. Porwal O, Mohammed Ameen MS, Anwer ET, Uthirapathy S, Ahamad J, et al. (2019) Silybum marianum (milk thistle): review on its chemistry, morphology, ethno medical uses, phytochemistry and pharmacological activities. J Drug Del Ther 9: 199‒206.
  156. Qadir A, Sahoo PK (2021) Silymarin: an account of phytochemistry and pharmacology. J Phytopharmacol 10: 262‒265.
  157. Abenavoli L, Izzo AA, Milić N, Cicala C, Santini A, et al. (2018) Milk thistle (Silybum marianum): a concise overview on its chemistry, pharmacological, and nutraceutical uses in liver diseases. Phytother Res 32: 2202‒2213. [Crossref]
  158. Wang X, Zhang Z, Wu SC (2020) Health benefits of Silybum marianum: phytochemistry, pharmacology, and applications. J Agric Food Chem 68: 11644‒11664. [Crossref]
  159. MacDonald-Ramos K, Michán L, Martínez-Ibarra A, Cerbón M (2021) Silymarin is an ally against insulin resistance: a review. Ann Hepatol 23: 100255. [Crossref]
  160. Škottová N, Večeřa R, Urbánek K, Vána P, Walterová D, et al. (2003) Effects of polyphenolic fraction of silymarin on lipoprotein profile in rats fed cholesterol-rich diets. Pharmacol Res 47: 17‒26. [Crossref]
  161. Sobolová L, Škottová N, Večeřa R, Urbánek K (2006) Effect of silymarin and its polyphenolic fraction on cholesterol absorption in rats. Pharmacol Res 53: 104‒112. [Crossref]
  162. Gobalakrishnan S, Asirvatham SS, Janarthanam V (2016) Effect of silybin on lipid profile in hypercholesterolaemic rats. J Clin Diagn Res 10: FF01‒FF05. [Crossref]
  163. Saadh MJ (2020) Hypoglycemic and hypolipidemic activity of combined milk thistle and fenugreek seeds in alloxan-induced diabetic albino rats. Vet World 13: 1732‒1736. [Crossref]
  164. Khazaei R, Seidavi A, Bouyeh M (2022) A review on the mechanisms of the effect of silymarin in milk thistle (Silybum marianum) on some laboratory animals. Vet Med Sci 8: 289–301. [Crossref]
  165. Tajmohammadi A, Razavi BM, Hosseinzadeh H (2018) Silybum marianum (milk thistle) and its main constituent, silymarin, as a potential therapeutic plant in metabolic syndrome: a review. Phytother Res 32: 1933‒1949. [Crossref]
  166. Hadi A, Pourmasoumi M, Mohammadi H, Symonds M, Miraghajani M (2018) The effects of silymarin supplementation on metabolic status and oxidative stress in patients with type 2 diabetes mellitus: a systematic review and meta-analysis of clinical trials. Complement Ther Med 41: 311‒319.
  167. Mohammadi H, Hadi A, Arab A, Moradi S, Rouhani MH (2019) Effects of silymarin supplementation on blood lipids: a systematic review and meta-analysis of clinical trials. Phytother Res 33: 871‒880. [Crossref]
  168. Xiao F, Gao F, Zhou S, Wang L (2020) The therapeutic effects of silymarin for patients with glucose/lipid metabolic dysfunction: a meta-analysis. Medicine 99: e22249. [Crossref]
  169. Soleymani S, Ayati MH, Mansourzadeh MJ, Namazi N, Zargaran A (2022) The effects of silymarin on the features of cardiometabolic syndrome in adults: a systematic review and meta-analysis. Phytother Res 36: 842‒856. [Crossref]
  170. Di Pierro F, Bellone I, Rapacioli G, Putignano P (2015) Clinical role of a fixed combination of standardized Berberis aristata and Silybum marianum extracts in diabetic and hypercholesterolemic patients intolerant to statins. Diabetes Metab Syndr Obes 8: 89‒96. [Crossref]
  171. Derosa G, Bonaventura A, Bianchi L, Romano D, D’Angelo A, et al. (2013) Berberis aristata/Silybum marianum fixed combination on lipid profile and insulin secretion in dyslipidemic patients. Expert Opin Biol Ther 13: 1495‒506. [Crossref]
  172. Derosa G, Romano D, D’Angelo A, Maffioli P (2015) Berberis aristata/Silybum marianum fixed combination (Berberol®) effects on lipid profile in dyslipidemic patients intolerant to statins at high dosages: a randomized, placebo-controlled, clinical trial. Phytomedicine 22: 231‒237.
  173. Derosa G, D’Angelo A, Romano D, Maffioli P (2017) Effects of a combination of Berberis aristata, Silybum marianum and monacolin on lipid profile in subjects at low cardiovascular risk; a double-blind, randomized, placebo-controlled trial. Int J Mol Sci 18: 343. [Crossref]
  174. Marková I, Malínská H, Hüttl M, Miklánková D, Oliyarnyk O, et al. (2021) The combination of atorvastatin with silymarin enhances hypolipidemic, antioxidant and anti-inflammatory effects in a rat model of metabolic syndrome. Physiol Res 70: 33‒43. [Crossref]
  175. Zhu B, Qi F, Wu J, Yin G, Hua J, et al. (2019) Red yeast rice: a systematic review of the traditional uses, chemistry, pharmacology, and quality control of an important Chinese folk medicine. Front Pharmacol 10: 1449. [Crossref]
  176. Yanli F, Xiang Y (2020) Perspectives on functional red mold rice: functional ingredients, production, and application. Front Microbiol 11: 606959. [Crossref]
  177. Xiong Z, Cao X, Wen Q, Chen Z, Cheng Z, et al. (2019) An overview of the bioactivity of monacolin K / lovastatin. Food Chem Toxicol 131: 110585. [Crossref]
  178. Xu G, Lin M, Dai X, Hu J (2022) Comparing the effectiveness of Chinese patent medicines containing red yeast rice on hyperlipidaemia: a network meta-analysis of randomized controlled trials. Endocrinol Diabetes Metab 5: e00314.
  179. Xiong X, Wang P, Li X, Zhang Y, Li S (2017) The effects of red yeast rice dietary supplement on blood pressure, lipid profile, and C-reactive protein in hypertension: a systematic review. Crit Rev Food Sci Nutr 57: 1831‒1851. [Crossref]
  180. Fukami H, Higa Y, Hisano T, Asano K, Hirata T, et al. (2021) A review of red yeast rice, a traditional fermented food in Japan and East Asia: its characteristic ingredients and application in the maintenance and improvement of health in lipid metabolism and the circulatory system. Molecules 26: 1619. [Crossref]
  181. Puato M, Zambon A, Nardin C, Faggin E, Pesavento R, et al. (2021) Lipid profile and vascular remodeling in young dyslipidemic subjects treated with nutraceuticals derived from red yeast rice. Cardiovasc Ther 2021: 5546800. [Crossref]
  182. Li Y, Jiang L, Jia Z, Xin W, Yang S, et al. (2014) A meta-analysis of red yeast rice: an effective and relatively safe alternative approach for dyslipidemia. PLoS One 9: e98611. [Crossref]
  183. Li P, Wang Q, Chen K, Zou S, Shu S, et al. (2022) Red yeast rice for hyperlipidemia: a meta-analysis of 15 high-quality randomized controlled trials. Front Pharmacol 12: 819482. [Crossref]
  184. Ong YC, Aziz Z (2016) Systematic review of red yeast rice compared with simvastatin in dyslipidaemia. J Clin Pharm Ther 41: 170‒179. [Crossref]
  185. Gerards MC, Terlou RJ, Yu H, Koks CH, Gerdes VE (2015) Traditional Chinese lipid-lowering agent red yeast rice results in significant LDL reduction but safety is uncertain - a systematic review and meta-analysis. Atherosclerosis 240: 415‒423. [Crossref]
  186. Heinz T, Schuchardt JP, Möller K, Hadji P, Hahn A (2016) Low daily dose of 3 mg monacolin K from RYR reduces the concentration of LDL-C in a randomized, placebo-controlled intervention. Nutr Res 36: 1162‒1170. [Crossref]
  187. Minamizuka T, Koshizaka M, Shoji M, Yamaga M, Hayashi A, et al. (2021) Low dose red yeast rice with monacolin K lowers LDL cholesterol and blood pressure in Japanese with mild dyslipidemia: a multicenter, randomized trial. Asia Pac J Clin Nutr 30: 424‒435. [Crossref]
  188. Cicero AFG, Fogacci F, Banach M (2019) Red yeast rice for hypercholesterolemia. Methodist Debakey Cardiovasc J 15: 192‒199. [Crossref]
  189. Farkouh A, Baumgärtel C (2019) Mini-review: medication safety of red yeast rice products. Int J Gen Med 12: 167‒171. [Crossref]
  190. Righetti L, Dall’Asta C, Bruni R (2021) Risk assessment of RYR food supplements: perception vs. reality. Front Nutr 8: 792529. [Crossref]
  191. Gyawali D, Vohra R, Orme-Johnson D, Ramaratnam S, Schneider RH (2021) A systematic review and meta-analysis of Ayurvedic herbal preparations for hypercholesterolemia. Medicina 57: 546. [Crossref]
  192. Deng R (2007) Therapeutic effects of guggul and its constituent guggulsterone: cardiovascular benefits. Cardiovasc Drug Rev 25: 375‒390. [Crossref]
  193. Kunnumakkara AB, Banik K, Bordoloi D, Harsha C, Sailo BL, et al. (2018) Googling the guggul (Commiphora and Boswellia) for prevention of chronic diseases. Front Pharmacol 9: 686. [Crossref]
  194. Yamada T, Sugimoto K (2016) Guggulsterone and its role in chronic diseases. Adv Exp Med Biol 929: 329‒361. [Crossref]
  195. Sarup P, Bala S, Kamboj S (2015) Pharmacology and phytochemistry of oleo-gum resin of Commiphora wightii (Guggulu). Scientifica 2015: 138039. [Crossref]
  196. Yang D, Yang J, Shi D, Xiao D, Chen YT, et al. (2012) Hypolipidemic agent Z-guggulsterone: metabolism interplays with induction of carboxylesterase and bile salt export pump. J Lipid Res 53: 529‒539. [Crossref]
  197. Cui J, Huang L, Zhao A, Lew JL, Yu J, et al. (2003) Guggulsterone is a farnesoid X receptor antagonist in coactivator association assays but acts to enhance transcription of bile salt export pump. J Biol Chem 278: 10214‒10220. [Crossref]
  198. Urizar NL, Moore DD (2003) GUGULIPID: a natural cholesterol-lowering agent. Annu Rev Nutr 23: 303‒313. [Crossref]
  199. She J, Gu T, Pang X, Liu Y, Tang L, et al. (2022) Natural products targeting liver X receptors or farnesoid X receptor. Front Pharmacol 12: 772435. [Crossref]
  200. Yu BZ, Kaimal R, Bai S, El Sayed KA, Tatulian SA, et al. (2009) Effect of guggulsterone and cembranoids of Commiphora mukul on pancreatic phospholipase A2: role in hypocholesterolemia. J Nat Prod 72: 24‒28. [Crossref]
  201. Szapary PO, Wolfe ML, Bloedon LT, Cucchiara AJ, DerMarderosian AH, et al. (2003) Guggulipid for the treatment of hypercholesterolemia: a randomized controlled trial. JAMA 290: 765‒772. [Crossref]
  202. Ulbricht C, Basch E, Szapary P, Hammerness P, Axentsev S, et al. (2005) Guggul for hyperlipidemia: a review by the Natural Standard Research Collaboration. Complement Ther Med 13: 279‒290. [Crossref]
  203. Nohr LA, Rasmussen LB, Straand J (2009) Resin from the mukul myrrh tree, guggul, can it be used for treating hypercholesterolemia? a randomized, controlled study. Complement Ther Med 17: 16‒22. [Crossref]
  204. Donato F, Raffetti E, Toninelli G, Festa A, Scarcella C, et al. (2021) Guggulu and Triphala for the treatment of hypercholesterolaemia: a placebo-controlled, double-blind, randomised trial. Complement Med Res 28: 216‒225.
  205. Chen X, Pang S, Lin J, Xia J, Wang Y (2016) Allicin prevents oxidized low-density lipoprotein-induced endothelial cell injury by inhibiting apoptosis and oxidative stress pathway. BMC Complement Altern Med 16: 133. [Crossref]
  206. Taleb A, Ahmad KA, Ihsan AU, Qu J, Lin N, et al. (2018) Antioxidant effects and mechanism of silymarin in oxidative stress induced cardiovascular diseases. Biomed Pharmacother 102: 689‒698. [Crossref]
  207. Wrona O, Rafińska K, Walczak-Skierska J, Możeński C, Buszewski B (2019) Extraction and determination of polar bioactive compounds from alfalfa (Medicago sativa L.) using supercritical techniques. Molecules 24: 4608. [Crossref]
  208. Teo TY, Yap J, Shen T, Yeo KK (2016) Complementary and alternative medicine use amongst patients with cardiovascular disease in Singapore. BMC Complement Altern Med 16: 446. [Crossref]
  209. Salah AO, Salameh AD, Bitar MA, Zyoud SH, Alkaiyat AS, et al. (2020) Complementary and alternative medicine use in coronary heart disease patients: a cross-sectional study from Palestine. BMC Complement Med Ther 20: 231. [Crossref]

Editorial Information

Editor-in-Chief

Renee Dufault

Article Type

Research Article

Publication History

Received: June 13, 2023
Accepted: June 30, 2023
Published: July 07, 2023

Copyright

©2023 Degrave V. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Degrave V, Battisti M, Vega Joubert MB, Ingaramo P, Vaccarini C, et al. (2023) Cannabis oil: Effects on Cannabinoid-Induced Tetrad, blood pressure and metabolic parameters in female rats fed a sucrose-rich diet. Integr Food Nutr Metab 10: DOI: 10.15761/IFNM.1000309

Corresponding author

M.E. Oliva y M.E.

M.E. Oliva y M.E. D’Alessandro, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral. Ciudad Universitaria, cc242 (3000) Santa Fe. Argentina

Table 1: Effects and mechanisms of medicinal plants in the control of hyperlipidemia.

No

Medicinal Plants

Active ingredients

Lipid metabolism

Mechanism of action

References

1

Cynara cardunculus var. scolymus

Polyphenols, cynarin, chlorogenic acid

↓TC, TG, LDL, VLDL;

↑HDL

1) Inhibit cholesterol and triglyceride synthesis;

2) Inhibit cholesterol

esterification and

absorption;

3) Promote fatty acid oxidation;

4) Improve lipid transport protein system

[27‒29,33,37]

Flavonoids, luteolin, apigenin

↓TC, TG, LDL, Apo B;

↑Apo A1

1) Inhibit lipid synthesis;

2) Promote bile acids biosynthesis;

3) Promote fatty acid oxidation and lipolysis

[37]

Sesquiterpenes, cynaropicrin, aguerine

↓TG

Suppress serum triglyceride

level

[38,39]

Phytosterols

↓TC, TG, LDL;

↑HDL

1) Inhibit intestinal lipid absorption;

2) Promote lipids transport and decomposition

[42,43]

Soluble fibers, pectin, inulin

↓TC, TG, LDL

1) Promote bile acids biosynthesis;

2) Inhibit intestinal bile acid reabsorption

[27‒29]

2

Medicago sativa

Saponin

↓TC, TG, LDL;

↑HDL

1) Inhibit intestinal cholesterol absorption;

2) Promote fecal bile acids excretion;

3) Regulate gut microbiota

[46,47,49‒52]

Sterols

↓TC, TG, LDL;

↑HDL

Promote lipids transport and decomposition

[46,47]

Flavonoids, quercetin

↓TC, TG, LDL;

↑HDL

1) Promote mitochondrial

fatty acid oxidation;

2) Improve insulin

resistance;

3) Inhibit cholesterol

biosynthesis;

4) Promote cholesterol change to bile acid

[46,47]

3

Trigonella foenum-graecum

Flavonoids, Vitexin

↓TC, TG

Inhibit de novo lipogenesis

[60‒62]

 

Alkaloids, Trigonelline

↓TC, TG, LDL

1) Inhibit cholesterol esterification and

absorption;

2) Promote fatty acid oxidation;

3) Increase insulin sensitivity

[63,64]

Polyphenols

↓TC, TG, LDL, VLDL;

↑HDL

1) Inhibit cholesterol and triglyceride

synthesis;

2) Promote fatty acid oxidation;

3) Improve lipid transport protein system

[60‒62]

4‒Hydroxyisoleucine

↓TC, TG, LDL, FFA;

↑HDL

1) Inhibit cholesterol and triglyceride synthesis;

2) Promote fatty acid oxidation and lipolysis;

3) Improve insulin

resistance

[68‒70]

Soluble fibers

↓TC, TG, LDL, VLDL;

↑HDL

1) Reduce cholesterol synthesis;

2) Decrease lipids and bile acids reabsorption;

3) Increase fecal cholesterol and bile acids excretion

[69]

Diosgenin

↓TC, TG, LDL;

↑HDL

1) Inhibit cholesterol absorption;

2) Decrease liver cholesterol concentration;

3) Facilitate cholesterol excretion

[69,75‒77]

Saponin

↓TC, TG, LDL;

↑HDL

1) Inhibit cholesterol synthesis;

2) Increase reverse cholesterol transport;

3) Regulate gut microbiota

[79]

4

Allium sativum

Alliin; Allicin

↓TC, TG, LDL;

↑HDL

1) Modulate cholesterol and triglyceride metabolism;

2) Reduce hepatic lipid level

[93‒95,106,107]

S-allyl cysteine

↓TC, TG, LDL;

↑HDL

1) Inhibit lipid deposition;

2) Decrease myeloperoxidase activity and lipid hydroperoxide in serum

3) Decrease plasma concentration of F2-isoprostanes

[93,94]

Polyphenols

↓TC, TG, LDL;

↑HDL

1) Inhibit triglyceride and cholesterol synthesis;

2) Promote fatty acid oxidation;

3) Inhibit cholesterol esterification and absorption;

4) Improve lipid transport protein system

[93,94,106,107]

Saponin

↓TC, TG, LDL;

↑HDL

Modulate the composition, abundance and diversity of gut microbiota

[93,94,106,107]

Flavonoids, quercetin

↓TC, TG, LDL;

↑HDL

1) Inhibit lipid synthesis;

2) Promote fatty acid

oxidation;

3) Promote bile acids synthesis and excretion;

4) Promote cholesterol convert to bile acid;

5) Improve insulin

resistance

[93,94,106,107]

DAS; DADS; DATS

↓TC, TG, LDL

1) Increase superoxide dismutase activity;

2) Inhibit Lipid peroxidation

[95,96,98]

5

Glycine max

Phytoalexins, glyceollins

↓TC, TG, LDL, VLDL

1) Inhibit cholesterol

esterification and

absorption;

2) Inhibit lipid peroxidation in liver;

3) Modulate triglyceride and cholesterol metabolism;

4) Decrease triglyceride storage;

5) Alter expression of the genes related to cholesterol in liver

[121‒123]

 

Phytic acid

↓TC, TG, LDL

1) Decrease triglyceride and cholesterol synthesis;

2) Increase fecal cholesterol and bile acids excretion

[121,123,135]

 

Isoflavones, genistein, daidzein

↓TC, TG, LDL

1) Reduce hepatic lipid synthesis;

2) Decrease bile acids biosynthesis; 3) Decrease cholesterol reabsorption

[124,132‒134]

 

Soy lipids; Lecithins and phospholipids

↓TC, TG, LDL

1) Inhibit intestinal cholesterol absorption;

2) Promote biliary cholesterol excretion

[121,123]

 

Pectin

↓TC, TG, LDL

1) Promote bile acids biosynthesis;

2) Inhibit intestinal bile acid reabsorption

[121,123,128]

 

Peptides

↓TC, TG, LDL

1) Promote cholesterol efflux;

2) Decrease triglyceride fatty acid fractional synthesis;

3) Motivate bile acid biosynthesis and excretion

[125,129,137,138]

 

Saponin

↓TC, TG, LDL;

↑HDL

Regulate gut microbiota

[121,123,128]

 

Soy fibers

↓TC, TG, LDL

1) Improve intestinal

flora diversity;

2) Regulate gut microbiota

[121,123,128]

 

Lignans

↓TC, TG, LDL

Inhibit bile acids biosynthesis

[136]

6

Silybum marianum

Silybin (Silibinin)

↓TC, TG

1) Decrease de novo cholesterol biosynthesis;

2) Inhibit HMG-CoA reductase activity

[155,157]

 

Silymarin

↓TC, TG, LDL

1) Inhibit lipids biosynthesis;

2) Inhibit intestinal cholesterol absorption;

3) Increase cholesterol efflux;

4) Promote fatty acid oxidation;

4) Improve insulin resistance

[155‒159]

 

Kaempferol

↓TG

Inhibit fatty acid synthesis and adipogenesis

[155,157,158]

 

Quercetin

↓TC, TG, LDL;

↑HDL

1) Inhibit cholesterol biosynthesis;

2) Promote cholesterol convert to bile acid;

3) Promote fatty acid oxidation;

4) Improve insulin

resistance

[155‒159]

7

Red Yeast Rice

Monacolin K, Monacolin L

↓TC, TG, LDL, VLDL

1) Inhibit lipids biosynthesis;

2) Inhibit HMG-CoA reductase activity;

3) Decrease phospholipase A2 activity

[175‒177,180]

 

Polysaccharides

↓TC, TG, LDL;

↑HDL

1) Inhibit lipid synthesis;

2) Inhibit lipids absorption;

3) Promote fatty acid oxidation and lipolysis;

4) Promote bile acids synthesis and excretion;

4) Regulate gut microbiota

[175‒177,180]

8

Commiphora mukul

Guggulsterone (E- and Z)

↓TC, TG, LDL;

↑HDL

1) Decrease hepatic steroid production;

2) Increase plasma LDL catabolism;

3) Increase hepatic LDL clearance;

4) Increase cholesterol catabolism and excretion;

5) Regulate cholesterol metabolism;

6) Increase bile salts excretion

[192,194‒196]

 

Cembranoids

↓TC, TG, LDL,

1) Decrease phospholipase A2 activity;

2) Inhibit cholesterol and triglyceride absorption

[200]

 

Quercetin; Naringenin

↓TC, TG, LDL;

↑HDL

1) Inhibit lipids biosynthesis;

2) Promote cholesterol metabolism;

3) Promote fatty acid oxidation

[193,195]