Abstract
Dyslipidemia is a multifactorial disorder that is a causative factor and risk factor for cardiovascular disease. The incidence of dyslipidemia is expected to increase because of the presence of comorbidities. Although several lipid-lowering drugs have been developed and approved, they are not completely effective and are associated with side effects. Traditional herbal medicine (THM) represents an alternative and complementary approach for managing dyslipidemia because of its low toxicity and beneficial effects, such as anti-inflammatory and antioxidant effects. This review focuses on our current understanding of the antidyslipidemic effect of THMs and discusses the associated regulatory mechanisms. The current findings indicate that THM may lead to the development of novel therapeutic regimens for dyslipidemia.
Keywords: Dyslipidemia, traditional herbal medicine, lipid-lowering drugs, Scutellariae Radix, Alismatis Rhizoma, Atractylodis Rhizoma Alba
Introduction
Dyslipidemia refers to the abnormal levels of lipid substances in plasma. It is characterized by elevated levels of serum triglycerides (TGs), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-c) and reduced levels of high-density lipoprotein cholesterol (HDL-c) [1]. Dyslipidemia includes hypertriglyceridemia, hypercholesterolemia, hyper-LDL cholesterolemia, and hypo-HDL cholesterolemia [2]. These conditions can result from poor diet, tobacco exposure, genetic factors, or cardiovascular disease with severe complications. The prevalence of dyslipidemia has increased worldwide with modern lifestyle changes and is markedly increased in older individuals and those with a high body mass index [3]. The World Health Organization estimated that the prevalence of hypercholesterolemia in adults increased from 9.0% to 20.7% between 2007 and 2018 [4]. Moreover, the prevalence of dyslipidemia has continued to increase in Korea-from one in four adults to two in five adults in 2022 [5].
A serious concern regarding dyslipidemia is its associated comorbidities. Dyslipidemia is a multifactorial disorder that is accompanied by metabolic syndromes, such as obesity, diabetes, fatty liver, hypertension, coronary artery disease, hyperinsulinemia, type 2 diabetes mellitus, and hyperglycemia [6]. According to epidemiological studies, the most common combination in middle-aged and older patients is hyperlipidemia coupled with hypertension [7]. Over 70% of individuals treated for hypertension received concurrent treatment for dyslipidemia (hyper-LDL-c ≥ 130 mg/dL, hyper-TG ≥ 200 mg/dL, or hypo-HDL-c < 40 mg/dL) [8]. Patients with diabetes have a higher risk of having dyslipidemia compared with those without diabetes. A substantial proportion (87%) of individuals with diabetes have dyslipidemia (hyper-LDL-c ≥ 100 mg/dL, hyper-TG ≥ 200 mg/dL, or hypo-HDL-c < 40 mg/dL), and the incidence is expected to increase with the rapidly growing older population. Dyslipidemias are associated with severe diseases of other organ systems, including acute pancreatitis [9], nonalcoholic fatty liver disease [10], allergic rhinitis [11], and COVID-19 [12] as well as endocrine and metabolic disorders.
Traditional herbal medicine (THM) constitutes a major part of alternative and complementary medicine for the management of metabolic disorders [13,14]. It has the potential to enhance therapeutic effects with fewer side effects. The role of herbal medicines in inflammation and oxidative stress is well known [15]. THM may also exhibit activity in many other diseases including dyslipidemia [16,17]. In this review, we focus on our current understanding of the antidyslipidemic effect of THMs and discuss the associated regulatory mechanisms based on single herbal medicines and prescriptions frequently used to treat dyslipidemia.
Conventional therapy for dyslipidemia
The most commonly prescribed lipid-lowering drugs include statins, ezetimibe, cholestyramine, nicotinic acid, fibrates, omega-3, and their combinations [18]. Statins, which inhibit 3-hydroxy-3-methylglutarylcoenzyme A (HMG-CoA) reductase, are considered the first-line therapy for reducing plasma cholesterol levels by inhibiting the mevalonate pathway and preventing cholesterol biosynthesis [19]. Blocking the activity of this enzyme decreases intrahepatic de novo level of cholesterol, upregulates the normal expression of LDL receptor (LDLR), and increases the hepatic clearance of LDL particles, resulting in reduced LDL-c levels in the bloodstream [20]. Ezetimibe is a selective inhibitor of cholesterol absorption that binds and blocks Niemann-Pick C1-like 1 (NPC1L1) protein, which is responsible for the absorption of biliary and dietary cholesterol from the small intestine, resulting in an incremental reduction in LDL-c levels [21]. Cholestyramine is used as an adjunct to reduce elevated serum cholesterol levels by sequestering intestinal bile acids. This results in the enhanced conversion of cholesterol to bile acids in the liver, which is normally regulated by the negative feedback of bile acids [22]. Nicotinic acid reduces circulating lipid levels including TGs, very-low-density lipoprotein (VLDL) cholesterol, and LDL-c, by reducing the synthesis of the LDL precursor and VLDL, while increasing HDL-c levels [23]. Fibrates are PPAR-α agonists that reduce plasma TG levels and increase HDL-c levels by initiating catabolism through β-oxidation, stimulating cellular fatty acid uptake, reducing TG levels, promoting lipoprotein lipase (LPL) activity, and reversing cholesterol transport [24]. Omega-3 (n-3) also substantially reduces blood TG levels by increasing fatty acid oxidation, which suppresses hepatic lipogenesis and acts as a poor substrate for TG-synthesizing enzymes [25].
Several approaches have been used to develop targeted agents for lipid-lowering therapies. Lomitapide inhibits microsomal TG transfer protein, which is responsible for the formation of lipoproteins, such as chylomicron (CM) precursors in the small intestine and VLDL precursors in the liver [26]. Mipomersen is an antisense oligonucleotide inhibitor that targets apolipoprotein B-100 (apoB-100) packaged into VLDLs in the liver [27]. RGX-501 is a novel gene therapy that corrects defective hepatic LDLR in patients with familial hypercholesterolemia (HoFH) [28,29]. Sebelipase restores the levels of lysosomal acid lipase (LIPA), which hydrolyzes TG and cholesteryl ester in the lysosomes of the liver in patients with LIPA deficiency [30]. In plasma, Glybera (alipogene tiparvovec) restores the LPL activity required to hydrolyze TGs within CMs and VLDL in patients with LPL deficiency [31]. Evolocumab, alirocumab, and bococizumab are proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitors and markedly lower LDL-c levels [32]. Volanesorsen is an antisense inhibitor of Apo C-III formation that can inhibit LPL and reduce TG levels [33]. Evinacumab is an anti-angiopoietin-like protein 3 (ANGPTL3) inhibitor that is effective at reducing LDL-c levels in patients with HoFH [34]. TThe antisense oligonucleotide Apo(a)-LRx inhibits LP(a) synthesis by disrupting apo(a) synthesis [35]. In peripheral cells, MEDI6012 is a recombinant human lecithin cholesterol acyltransferase (LCAT) that restores LCAT activity in patients with LCAT deficiency [36]. CSL112 is an infused apoA-I peptide that increases cholesterol efflux [37]. Continued drug development and new strategies for lipid reduction are currently underway. Combining medications is occasionally necessary for the effective treatment of lipid abnormalities in patients with mixed dyslipidemia with comorbidities [38]. Although statin monotherapy does not always achieve the desired LDL-c level, combination therapy with other lipid-reducing agents is underutilized. For example, ezetimibe may be used in combination with statins to lower elevated levels of LDL-c, TC, TG, and ApoB in patients with hypercholesterolemia with additive and synergistic effects [39].
However, combination therapy is not completely effective and may be limited because of adverse effects [40]. Although statins are generally well-tolerated in most patients with dyslipidemia, some adverse effects may occur, including myopathy, muscle pain, stiffness, myalgia, and a severe form of rhabdomyolysis. These effects are caused by coenzyme Q deficiency, impaired protein prenylation, oxidative stress, mitochondrial dysfunction, abnormal protein glycosylation, and apoptosis [41,42]. Rhabdomyolysis, a rare but potentially life-threatening side effect of statins, is caused by the breakdown of skeletal muscle, resulting in the release of the sarcoplasmic proteins AST, ALT, myoglobin, lactate dehydrogenase, aldolase, and creatine kinase as well as electrolytes. It typically presents as myalgia and muscle weakness of the proximal musculature, causing contractile dysfunction and leading to cell damage and muscle cell death [43]. Because of its association with an increased risk of statin-induced myopathy and rhabdomyolysis, the United States Food and Drug Administration does not recommend administering statins at high doses (80 mg/daily) [44]. However, the combined use of statins with other drugs can also increase the risk of rhabdomyolysis because of drug-drug interactions. For example, concurrent therapy with fibrates, primarily fenofibrate, induces rhabdomyolysis compared with monotherapy [45]. Combined therapy with an inhibitor of the enzyme cytochrome P450 3A4 (CYP3A4), which is involved in the oxidation and metabolism of statins, maintains statins longer in the body, thus increasing the risk of adverse effects [46]. Statins are also linked to an increased risk of hepatic dysfunction associated with elevated hepatic transaminase levels, peripheral neuropathy, hyperglycemia, and a risk of incident diabetes [47].
Despite combined therapy, it is difficult to fully engage all therapeutic targets. The pathophysiology of dyslipidemia is likely multifactorial, including heterogeneity and coexisting risk factors [48]. There is still no strategy to evaluate the differential effects of an individual’s predisposition to dyslipidemia or comorbidity. In this regard, novel therapeutic modalities as well as appropriate medications and recommendations are needed for patients with dyslipidemia.
Use of THM as a fundamental treatment strategy
THM can serve as an alternative to conventional medicine for the improved management of dyslipidemia owing to its low toxicity and beneficial effects on reducing lipid levels [49,50]. An important approach to the use of THM is to trace a disease back to the cause of the disease, that is, the internal cause, the external cause, or the cause that is neither internal nor external. Dyslipidemia is a causative factor and risk factor for cardiovascular disease [51]; thus, it is important to prevent and treat dyslipidemia strategically. A discussion of dyslipidemia from the perspective of THM and its fundamental treatment is needed, rather than conventional therapy to simply address the temporary symptoms. According to traditional medicine theory, metabolic syndromes may be categorized into spleen qi (the physical substrate and dynamic force of the functional activities of the spleen) deficiency, qi stagnation (depressed and stagnant flow of qi), dampness obstruction, phlegm, and blood stasis. Depending on the underlying mechanisms of the syndromes, the decoction or treatment method may be varied accordingly [52].
Although the term “dyslipidemia” has not been used in the context of traditional medicine, it is a category of dampness, phlegm, and blood stasis based on the current traditional medicine theory [49] (Figure 1), which refers to pathological causes and products resulting from pathologic conditions, such as overeating high-fat diet, dysfunction of transportation, and transformation, impaired digestion, and stagnation [6,53]. Dampness is an overly wet or moist condition characterized by stickiness, turbidity, heaviness, and a downward trend, which obstructs vital energy. Consequently, it causes symptoms such as high cholesterol, metabolic disorders, chronic fatigue syndrome, cancer, achy joints and limbs, headaches, fibromyalgia, allergies, and various digestive issues. Organically, dampness floods as water and is gathered into phlegm [54]. Phlegm is an endogenous pathological factor that is defined as a viscous, turbid pathological product (such as thick mucus) that accumulates in certain parts of the body, resulting in various diseases [55]. Dampness and phlegm result from an imbalance in body fluid concentration when digestion, transformation, and fluid metabolism are not functioning properly. A constitution study using genomics revealed that phlegm-dampness constitution is susceptible to hypertension, obesity, diabetes, and metabolic syndrome [56]. Another study found altered gut microbiota in obese individuals with phlegm-dampness [57]. Blood stasis is a pathological state of abnormal blood flow in which the circulation is not smooth or becomes stagnant and forms stasis [58]. In a preclinical study, blood stasis was investigated in terms of the early stages of atherosclerosis [59], coronary heart disease [60], hyperviscosity, hyperlipidemia, inflammation, neoplasm, and ischemic brain injury [61].
Figure 1.
Pathological causes and products of dyslipidemia in traditional medicine theory.
A deficiency in spleen and kidney function is the basis of dyslipidemia, following disorders of digestion, absorption, transport, and excretion of food, which results in pathological products, such as phlegm, dampness, and blood stasis [6]. It is aggravated by overeating greasy meals, being overweight, a lack of physical activity, and metabolic diseases [49]. Therefore, the principal treatment for dyslipidemia is removal of the accumulated phlegm-dimness and blood stasis and improvement of blood circulation in the body, similar to clearing a pipe to improve clean water flow.
Advantages of THMs for the treatment of dyslipidemia
Phlegm-dampness and blood stasis may be associated with several clinical manifestations, including fever, inflammation, and swelling. There are many options in traditional medicine, including prescriptions, and/or herbal medicine to address these problems. For example, in clinical practice, interior heat-clearing medicines are usually effective in eliminating heat and dampness, which indicates that an interior heat pattern of dyslipidemia may be treated using THM. Plant-derived bioactive compounds with antioxidant and anti-inflammatory properties have been used to protect the vascular endothelium, prevent lipid oxidation, and lower lipid levels. As representative compounds, phenolics and flavonoids from medicinal plants have been reported to be effective [62]. Mitochondria-targeting antioxidants represent a potential strategy for treating nonalcoholic fatty liver disease (NAFLD)-associated liver diseases [15]. Thus, antioxidants may serve as a therapeutic framework for disease treatment.
Another strength of THM is its multicomponent nature. The diversity and complexity of multicomponent THM enable the targeting of alternative pathways and biological processes to treat complex diseases [63]. As mentioned above, dyslipidemia is the consequence of multiple factors; therefore, its clinical manifestations are also complicated, not only in terms of elevated serum lipid levels but also multifarious disorders. In this regard, a holistic approach to treating different conditions is needed; thus, THM can play a key role in the treatment of dyslipidemia [64]. When medicinal herbs are prescribed based on the combination principle, combined herbs can exert beneficial effects as a result of the synergistic interactions of the herb components. Well-established multiherb combination therapies not only achieve the desired therapeutic outcome but may also decrease undesirable and harmful effects; however, the pharmacokinetic interactions of single or combined medicines remain a major concern [65].
Screening of clinical prescriptions used frequently
We conducted a screening study of 89 clinical prescriptions used frequently on dyslipidemia to identify applicable THMs for dyslipidemia treatment. We found that 17 prescriptions had an inhibitory effect on lipid accumulation when they were administered prior to lipid induction, and 15 prescriptions had an inhibitory effect when they were administered after lipid induction in HepG2 cells compared with atorvastatin-treated cells (Table 1). Based on screening for the prescriptions’ preventative or curative effects on lipid accumulation, the top three effective prescriptions were selected and are discussed below.
Table 1.
Lipid-lowering effect of 89 clinical prescriptions used frequently on HepG2 cell lines
NO. | Prescriptions | Prevention strategy | Treatment strategy |
---|---|---|---|
| |||
Reduction rate (%) | Reduction rate (%) | ||
1 | Antae-eum | -9 | 57 |
2 | Baenongsangeup-san | 30 | 42 |
3 | Bangkeehwangkee-tang | -12 | 47 |
4 | Banhabakchulcheonma-tang | 40 | 33 |
5 | Banhahoo Granule | 24 | 26 |
6 | Banhahubak-tang | 28 | 53 |
7 | Banhasasim-tang | -28 | 21 |
8 | Bojungikgi-tang | 19 | 22 |
9 | Buhnsimgieum | 19 | 35 |
10 | Bulhwangeumjeonggi-san | 15 | 39 |
11 | Chengsimyeunjaeum | -3 | 27 |
12 | Cheongsangbangpung-san | 49 | 52 |
13 | Cheongsanggyeontong-tang | 34 | 51 |
14 | Cheonwangbosim-dan | -15 | 34 |
15 | Chokyungjongok-tang | 32 | 44 |
16 | Chungseoikgi-tang | 36 | 29 |
17 | Daegunjoong-tang | 28 | 50 |
18 | Daehwajung-eum | 29 | 30 |
19 | Daehwangmokdan-tang | 27 | 18 |
20 | Daeshiho-tang | 27 | 35 |
21 | Daeyoung-jeon | 18 | 28 |
22 | Dangguijakyak-san | 17 | 47 |
23 | Dangguiniantong-tang | 30 | 3 |
24 | Dangguisu-san | -18 | 14 |
25 | Doinseunggi-tang | 28 | 19 |
26 | Dokhwalgisaeng-tang | 28 | 43 |
27 | Eunkyo-san | 19 | 37 |
28 | Galgeun-tang | 17 | 5 |
29 | Gamiondam-tang | 24 | 45 |
30 | Gamisoyo-san | 13 | 10 |
31 | Gumiganghwal-tang | 17 | 19 |
32 | Gungha-tang | 17 | 36 |
33 | Gwakhyangjeonggi-san | -13 | -1 |
34 | Gyejibokryeong-hwan | -20 | -2 |
35 | Hwanggigunjung-tang | 39 | 28 |
36 | Hwanglyeonhaedok-tang | 11 | 56 |
37 | Hyangsapyeongwi-san | 24 | 16 |
38 | Hyangsayukgunja-tang | -21 | 16 |
39 | Hyangso-san | 15 | 19 |
40 | Hyeonggaeyeongyo-tang | 28 | 43 |
41 | Hyulbuchukeo-tang | 25 | 48 |
42 | Insamyangyoung-tang | 23 | 51 |
43 | Jaeumganghwa-tang | 27 | 33 |
44 | Jagamcho-tang | 32 | 42 |
45 | Jakyak gamcho-tang | 6 | 12 |
46 | Jihwangeum-ja | -16 | 53 |
47 | Jodeung-san | 25 | 56 |
48 | Kamiguibitang | 26 | -3 |
49 | Kumikanghal-tang | 10 | 24 |
50 | Kungkwikyoae-tang | 29 | 11 |
51 | Kyeji-tang | 11 | 24 |
52 | Maekmundong-tang | -41 | -3 |
53 | Mahwang-tang | 6 | 27 |
54 | Majain-hwan | -9 | 29 |
55 | Naeso-san | 17 | 16 |
56 | Ojeok-san | 40 | 33 |
57 | Oncheong-eum | 33 | 53 |
58 | Onkyung-tang | 2 | -10 |
59 | Oryeong-san | 36 | 26 |
60 | Paljung-san | 10 | 44 |
61 | Palmijihwang-tang | -17 | 15 |
62 | Palmul-tang | 41 | 27 |
63 | Pyeongwi-san | 40 | 16 |
64 | Saengkangsasim-tang | 36 | 34 |
65 | Saengmaek-san | 3 | 0 |
66 | Samchulgeonbi-tang | 31 | 24 |
67 | Samryeongbaekchul-san | -23 | -12 |
68 | Samul-tang | 23 | 15 |
69 | Sanjoin-tang | 28 | 50 |
70 | Sayeoksan | 5 | 23 |
71 | Sikyungbanha-tang | 35 | 28 |
72 | Sipjeondaebo-tang | 25 | 42 |
73 | Siryung-tang | 30 | 51 |
74 | Socheongryong-tang | 14 | 17 |
75 | Sogunjung-tang | 28 | 44 |
76 | Sokyunghwalhul-tang | 20 | 52 |
77 | Sopung-san | 26 | 46 |
78 | Ssanghwa-tang | 28 | 39 |
79 | Taglisodog-eum | 34 | 43 |
80 | Ukgan-sangajinphibanha | 31 | 45 |
81 | Wiryeong-tang | 38 | 30 |
82 | Yeongyechulgam-tang | -2 | 18 |
83 | Yeonkyopaedok-san | 24 | 35 |
84 | Yijung-tang | 38 | 35 |
85 | Yongdamsagan-tang | 17 | 28 |
86 | Youngseonjaetong-eum | 23 | 52 |
87 | Yukgunja-tang | -12 | 55 |
88 | Yukmijihwang-tang | 40 | 30 |
89 | Zeolyung-tang | 38 | 22 |
- | Atorvastatin (20 µM) | 32 | 49 |
HepG2 cells were cultured in 96-well plates at a density of 5 × 104 cells/well for 24 h. To investigate the preventive effect, cells were incubated with each of the above prescriptions (100 µg/mL) or atorvastatin (20 µM) as a positive control in the presence of a free fatty acid (FFA) mixture (oleic acid/palmitic acid, 2:1 ratio) for 24 h. The following day, the intracellular lipid content was fluorescently measured using AdipoRed reagent at excitation/emission wavelengths of 485/572 nm. For the treatment effect experiment, lipid accumulation was induced in the cells with an FFA mixture for 24 h and then the cells were incubated with each prescription (100 µg/mL) or atorvastatin (20 µM) for a further 24 h. The lipid content was analyzed using AdipoRed reagent according to the manufacturer’s protocol.
Cheong-sang-bang-pung-san
Cheong-sang-bang-pung-san (CSBPS; “Qing-shang-fang-feng-san” in Chinese and “Seijo-bofu-san” in Japanese) is a well-known traditional herbal prescription for the treatment of purulent inflammation, such as acne, and urticaria. CSBPS was first mentioned in Wanbinhuichun and it is also mentioned in Dongeuibogam by Heo Jun of the Joseon Dynasty of Korea [66]. It consists of Saposhnikoviae Radix, Forsythiae Fructus, Platycodonis Radix, Angelicae Dahuricae Radix, Scutellariae Radix, Schizonepetae Spica, Cnidii Rhizoma, Gardeniae Fructus, Coptidis Rhizoma, Menthae Herba, Aurantii Fructus Immaturus, and Glycyrrhizae Radix et Rhizoma. Studies have demonstrated the pharmacological effects of CSBPS including antibacterial [67] and anti-lipase activities [68] as well as skin rush in acne patients [69]. CSBPS cleans the heat of the upper energizer (the chest cavity), which is considered the major cause of not only acne, furuncles, and sores but also inflammation. CSBPS resolves toxicity and expels wind, as implied by the name of the prescription “Clear the Upper and Guard the Wind Decoction” [70]. As indicated by the composition of the formulation, CSBPS focuses on the “heat” pattern as a therapeutic target for treating inflammatory skin disease by cooling heat [71] as well as antioxidative stress activity [72].
On-cheong-eum
On-cheong-eum (OCE; “Wen-qing-yin” in Chinese and “Unsei-in” in Japanese), also known as “Cleanse the Top and Relieve Headache Decoction”, is a traditional herbal prescription that combines Sa-mul-tang and Hwan-ryun-hae-dok-tang. It consists of Coptidis Rhizoma, Phellodendri Cortex, Scutellariae Radix, Gardeniae Fructus, Rehmanniae Radix, Cnidii Rhizoma, Angelicae Gigantis Radix, and Paeoniae Radix [73]. This prescription was first introduced in Wanbinhuichun and has been used to treat abnormal uterine bleeding, restore the menstrual cycle, clear heat, purge pathogenic fire, tonify the blood, and resolve toxins [74]. It is used to treat uterine bleeding resulting from heat and gynecological disorders characterized by heat and blood deficiency. Recently, its use has been extended to other diseases, such as incurable skin diseases [75], atopic dermatitis, eczema, psoriasis vulgaris [76], recurrent aphthous stomatitis, Behcet’s disease, and diabetes as well as neurological symptoms [77].
Cheong-sang-gyeon-tong-tang
Cheong-sang-gyeon-tong-tang (CSGTT; “Qing-shang-juan-tong-tang” in Chinese and “Seijo-ken-tsu-to” in Japanese), literally meaning “Cleanse the Top and Pain-alleviating Decoction” was first mentioned in Jejungshinpyun as a major prescription for headaches and facial pain [78]. CSGTT dispels wind-heat, ameliorates wind-cold dampness, relieves pain, and purges liver fire against headaches caused by cold, migraine, and tension. It consists of Angelicae Gigantis Radix, Cnidii Rhizoma, Araliae Continentalis Radix, Osterici seu Notopterygii Radix et Rhizoma, Angelicae Dahuricae Radix, Saposhnikoviae Radix, Atractylodis Rhizoma, Liriopis seu Ophiopogonis Tuber, Scutellariae Radix, Chrysanthemi Indici Flos, Viticis Fructus, Asiasari Radix et Rhizoma, Glycyrrhizae Radix et Rhizoma, and Zingiberis Rhizoma Recens. CSGTT plays a role in analgesia, anticonvulsion, and antihyperlipidemia [79]; however, its pharmacological activity has not been studied extensively.
Screening of single herbal medicines
THMs commonly used to treat dyslipidemia include Salviae Miltiorrhizae Radix, Crataegi Fructus, Alismatis Rhizoma, Atractylodis Rhizoma Alba, Ginkgo, Ganoderma, and Curcumae Longae Rhizoma, which are effective at reducing serum lipid levels [80]. We discovered that most of the listed prescriptions used clinically contain Scutellariae Radix, Alismatis Rhizoma, and Atractylodis Rhizoma Alba. Based on this finding, an optimized herbal medicine was developed and is considered effective for thrombotic disease [81]. Herein, we discuss the properties of each herbal medicine that exhibits antidyslipidemia activity.
Scutellariae Radix
Scutellariae Radix (the dried root of Scutellariae baicalensis Georgi) is a perennial herb belonging to the family Lamiaceae. It has a long history of use in traditional medicinal formulations because of its activity in clearing away heat, purging fire, draining dampness, and removing toxins [50]. Scutellariae Radix exhibits a wide range of pharmacological activities, including effects on the nervous and immune systems; liver protection; and antitumor, anti-inflammatory [82-85], antibacterial, and antiviral effects, lipid-lowering, antiobesity [86-88], and antioxidant [89] effects. These therapeutic effects are predominantly induced by flavonoids, the most abundant components of Scutellariae Radix.
The six major bioactive flavones in Scutellariae Radix have been identified. The existing forms include aglycones, such as baicalein, wogonin, and oroxylin A, and glycosides, such as baicalin, wogonoside, and oroxylin A-7-glucuronide. All are pharmacologically active flavones that are useful for treating inflammation, virus-related diseases, and cancers [90]. As predicted, the lipid-lowering component of Scutellariae Radix with the highest activity is the flavonoid compound baicalin and its aglycone baicalein. Baicalin exerts anti-lipidemic [87] and anti-inflammatory activities [91,92]. Baicalein has strong anti-inflammatory [93], antioxidative [94], antitumor [95], antiobesity [96] activities. It exhibits potent antioxidant, antithrombotic, and anti-inflammatory activities in endothelial cells and antihypertensive effects in vivo [97]. Huang et al. reported the protective effect of baicalein on cardiovascular disease, suggesting an associated mechanism that can protect against oxidative stress by interfering with apoptosis induced by endoplasmic reticulum stress in cardiomyocytes [98].
Alismatis Rhizoma
Alismatis Rhizoma is derived from the dried stem tuber of Alisma orientale (Sam.) Juzep. (Alismataceae). It is used to treat dysuria, edema, hyperlipidemia, nephropathy, and diabetes by promoting diuresis with mechanisms of resolving dampness and expelling heat. Alismatis Rhizoma exhibits multiple pharmacological effects, including antihyperlipidemic [80], antihyperglycemic [99], anti-inflammatory [100-102], hepatoprotective [103], antiobesity [104], and anticancer [105] activities.
A wide range of pharmacologically active compounds, primarily triterpenoids, sesquiterpenoids, and diterpenoids, have been isolated from Alismatis Rhizoma, of which alisols, the protostane-type triterpenoids, are well known for their unique chemical structures with numerous derivatives and various biological activities [106]. Triterpenes are the main active components of Alismatis Rhizoma, which exert hypolipidemic effects by inhibiting cholesterol absorption and synthesis and improving lipid metabolism [50].
Atractylodis Rhizoma Alba
Atractylodis Rhizoma primarily contains Atractylodes japonica Koidzumi and Atractylodes macrocephala Koidzumi is commonly used as a traditional medicine in East Asia. It is used for the treatment of digestive disorders, diarrhea, rheumatic diseases, systemic edema, bloating, night blindness, and influenza. Previous studies have indicated that Atractylodis Rhizoma Alba exhibits a range of pharmacological activities, including antioxidant [107], antiobesity [108,109], antidiabetes [110,111], and anti-inflammatory [112,113] effects as well as lowering serum lipid levels [80].
It contains a series of sesquiterpenoids, oligosaccharides, polysaccharides, monoterpenes, polyacetylenes, phenolic acids, and steroids that contributes to its beneficial effects [114]. Atractylenolide exhibits various pharmacological effects, including anti-inflammatory [115,116], antioxidant [117-120], and anticancer [117] activities. In terms of metabolism, atractylenolide enhances energy metabolism [121], modulates farnesoid X receptor receptor [122], improves NAFLD [123], and stimulates the browning of adipocytes [124]. Atractyloside, a diterpenoid glycoside, was shown to exert a therapeutic effect on type 2 diabetes [125] and hepatic steatosis [126] through metabolic regulation.
Discussion
The three selected prescriptions CSBPS, OCE, and CSGTT have been approved by The Korean Ministry of Food and Drug Safety for clinical use. Interestingly, these THM prescriptions have not been traditionally used for obesity or dyslipidemia. This review highlights the role of THM as a potent lipid-lowering treatment with expanded clinical indications. THM may serve as an alternative to Western medications for dyslipidemia treatment. The current literature indicates that a battery of compounds, including terpenoids, saponins, alkaloids, and polyphenols, are derived from THM and have been shown to exert therapeutic effects in metabolic systems. Of these, polyphenols, including flavonoids and tannins, are abundant in many THMs used for treating dyslipidemia. Notably, their protective effects are primarily attributed to their anti-inflammatory, antioxidant, antiobesity, hypolipidemic, and antitumor effects (Figure 2) as has been described earlier [80].
Figure 2.
Pharmacological effects of THM related to a potential mechanism of anti-dyslipidemia activity.
Antioxidants are considered potential therapies for treating many illnesses. A significant positive correlation was observed between oxidative stress and dyslipidemia [127]; therefore, enhancing antioxidant supply may prevent or reverse the course of dyslipidemia development. For example, resveratrol is well known for its antioxidant activity in grapes [128], Polygoni Cuspidati Rhizoma et Radix [129], and Mori Fructus [130]. Furthermore, resveratrol inhibits the transcription of PCSK9, including sterol regulatory element-binding protein-2 (SREBP2) and hepatocyte nuclear factor 1α (HNF1α), the PCSK9/LDLR interaction, maturation of PCSK9 in the endoplasmic reticulum, and the secretion of PCSK9 from hepatic cells [131].
Considering the traditional indications of the three prescriptions, they share a mechanism for the control of inflammation and fever by ameliorating heat and pain. Because metabolic disease is associated with low-grade chronic inflammation [132], managing the inflammatory response using THM represents a promising therapeutic strategy for dyslipidemia.
The THM formula consists of a wide variety of ingredients with innumerous chemical molecules, making it possible for THM treatment to exhibit “multiple herbs, multiple target” pharmacological effects [72]. This results in add-on effects not only for certain indications but also for symptoms of the disease. Hence, THMs are expected to have a synergistic effect on dyslipidemia as well as other conditions that are associated with dyslipidemia, although a single herbal medicine also has potential effects on the disease. This suggests an emerging picture of prevention and treatment for a series of diseases, such as hyperlipidemia, hypertension, and arteriosclerosis, through the development of THM preparations that target metabolic diseases.
Based on current data, it is difficult to identify individual compounds and their molecular targets and completely interpret the observed therapeutic benefits and molecular actions of THM. However, several approaches have been developed, such as systems biology for understanding effects on the cell, tissue, or organism as well as network pharmacology, which is a powerful tool for revealing the complex mechanisms associated with THM [133]. Further studies are needed to elucidate the molecular mechanism underlying the therapeutic effects of THM and to take advantage of the multitarget nature of THM, which may be superior to commercial agents. The precise study of the mechanisms associated with THM will aid in the development of individualized treatment strategies for patients with dyslipidemia.
Acknowledgements
This research was supported by the National Research Foundation of Korea Grant supported by the Korean Government (NRF-2021R1I1A2040301).
Disclosure of conflict of interest
None.
References
- 1.Liu C, Huang Y. Chinese herbal medicine on cardiovascular diseases and the mechanisms of action. Front Pharmacol. 2016;7:469. doi: 10.3389/fphar.2016.00469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Ha KH, Kwon HS, Kim DJ. Epidemiologic characteristics of dyslipidemia in Korea. J Lipid Atheroscler. 2015;4:91–99. [Google Scholar]
- 3.Gebreegziabiher G, Belachew T, Mehari K, Tamiru D. Prevalence of dyslipidemia and associated risk factors among adult residents of Mekelle City, Northern Ethiopia. PLoS One. 2021;16:e0243103. doi: 10.1371/journal.pone.0243103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cho SMJ, Lee H, Lee HH, Baek J, Heo JE, Joo HJ, Hong SJ, Kim HC Korean Society of Lipid and Atherosclerosis (KSoLA) Public Relations Committee. Dyslipidemia fact sheets in Korea 2020: an analysis of nationwide population-based data. J Lipid Atheroscler. 2021;10:202–209. doi: 10.12997/jla.2021.10.2.202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Choi D. Dyslipidemia fact sheet in Korea, 2022. Seoul, Republic of Korea: The Korean Society of Lipid and Atherosclerosis (KSoLA); 2022. [Google Scholar]
- 6.Ma YL, Yao H, Yang WJ, Ren XX, Teng L, Yang MC. Correlation between traditional Chinese medicine constitution and dyslipidemia: a systematic review and meta-analysis. Evid Based Complement Alternat Med. 2017;2017:1896746. doi: 10.1155/2017/1896746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Castano G, Mas R, Fernandez JC, Fernandez L, Illnait J, Lopez E. Effects of policosanol on older patients with hypertension and type II hypercholesterolaemia. Drugs R D. 2002;3:159–172. doi: 10.2165/00126839-200203030-00004. [DOI] [PubMed] [Google Scholar]
- 8.Lalonde L, Goudreau J, Hudon E, Lussier MT, Bareil C, Duhamel F, Levesque L, Turcotte A, Lalonde G Group for TRANSIT to Best Practices in Cardiovascular Disease Prevention in Primary Care. Development of an interprofessional program for cardiovascular prevention in primary care: a participatory research approach. SAGE Open Med. 2014;2:2050312114522788. doi: 10.1177/2050312114522788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Yang AL, McNabb-Baltar J. Hypertriglyceridemia and acute pancreatitis. Pancreatology. 2020;20:795–800. doi: 10.1016/j.pan.2020.06.005. [DOI] [PubMed] [Google Scholar]
- 10.Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty liver disease and dyslipidemia: an update. Metabolism. 2016;65:1109–1123. doi: 10.1016/j.metabol.2016.05.003. [DOI] [PubMed] [Google Scholar]
- 11.Sheha D, El-Korashi L, AbdAllah AM, El Begermy MM, Elzoghby DM, Elmahdi A. Lipid profile and IL-17A in allergic rhinitis: correlation with disease severity and quality of life. J Asthma Allergy. 2021;14:109–117. doi: 10.2147/JAA.S290813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Liu Y, Pan Y, Yin Y, Chen W, Li X. Association of dyslipidemia with the severity and mortality of coronavirus disease 2019 (COVID-19): a meta-analysis. Virol J. 2021;18:157. doi: 10.1186/s12985-021-01604-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wang J, Ke W, Bao R, Hu X, Chen F. Beneficial effects of ginger Zingiber officinale Roscoe on obesity and metabolic syndrome: a review. Ann N Y Acad Sci. 2017;1398:83–98. doi: 10.1111/nyas.13375. [DOI] [PubMed] [Google Scholar]
- 14.Fan W, Huang Y, Zheng H, Li S, Li Z, Yuan L, Cheng X, He C, Sun J. Ginsenosides for the treatment of metabolic syndrome and cardiovascular diseases: pharmacology and mechanisms. Biomed Pharmacother. 2020;132:110915. doi: 10.1016/j.biopha.2020.110915. [DOI] [PubMed] [Google Scholar]
- 15.Park MN, Rahman MA, Rahman MH, Kim JW, Choi M, Kim JW, Choi J, Moon M, Ahmed KR, Kim B. Potential therapeutic implication of herbal medicine in mitochondria-mediated oxidative stress-related liver diseases. Antioxidants (Basel) 2022;11:2041. doi: 10.3390/antiox11102041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hong N, Lin Y, Ye Z, Yang C, Huang Y, Duan Q, Xie S. The relationship between dyslipidemia and inflammation among adults in east coast China: a cross-sectional study. Front Immunol. 2022;13:937201. doi: 10.3389/fimmu.2022.937201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Medina-Vera I, Gomez-de-Regil L, Gutierrez-Solis AL, Lugo R, Guevara-Cruz M, Pedraza-Chaverri J, Avila-Nava A. Dietary strategies by foods with antioxidant effect on nutritional management of dyslipidemias: a systematic review. Antioxidants (Basel) 2021;10:225. doi: 10.3390/antiox10020225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zodda D, Giammona R, Schifilliti S. Treatment strategy for dyslipidemia in cardiovascular disease prevention: focus on old and new drugs. Pharmacy (Basel) 2018;6:10. doi: 10.3390/pharmacy6010010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Stancu C, Sima A. Statins: mechanism of action and effects. J Cell Mol Med. 2001;5:378–387. doi: 10.1111/j.1582-4934.2001.tb00172.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell. 2015;161:161–172. doi: 10.1016/j.cell.2015.01.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ge L, Wang J, Qi W, Miao HH, Cao J, Qu YX, Li BL, Song BL. The cholesterol absorption inhibitor ezetimibe acts by blocking the sterol-induced internalization of NPC1L1. Cell Metab. 2008;7:508–519. doi: 10.1016/j.cmet.2008.04.001. [DOI] [PubMed] [Google Scholar]
- 22.Suckling KE, Benson GM, Bond B, Gee A, Glen A, Haynes C, Jackson B. Cholesterol lowering and bile acid excretion in the hamster with cholestyramine treatment. Atherosclerosis. 1991;89:183–190. doi: 10.1016/0021-9150(91)90059-c. [DOI] [PubMed] [Google Scholar]
- 23.Lukasova M, Malaval C, Gille A, Kero J, Offermanns S. Nicotinic acid inhibits progression of atherosclerosis in mice through its receptor GPR109A expressed by immune cells. J Clin Invest. 2011;121:1163–1173. doi: 10.1172/JCI41651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Gryn SE, Hegele RA. Novel therapeutics in hypertriglyceridemia. Curr Opin Lipidol. 2015;26:484–491. doi: 10.1097/MOL.0000000000000231. [DOI] [PubMed] [Google Scholar]
- 25.Davidson MH, Benes LB. The future of n-3 polyunsaturated fatty acid therapy. Curr Opin Lipidol. 2016;27:570–578. doi: 10.1097/MOL.0000000000000353. [DOI] [PubMed] [Google Scholar]
- 26.Goulooze SC, Cohen AF, Rissmann R. Lomitapide. Br J Clin Pharmacol. 2015;80:179–181. doi: 10.1111/bcp.12612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bell DA, Hooper AJ, Burnett JR. Mipomersen, an antisense apolipoprotein B synthesis inhibitor. Expert Opin Investig Drugs. 2011;20:265–272. doi: 10.1517/13543784.2011.547471. [DOI] [PubMed] [Google Scholar]
- 28.Ajufo E, Cuchel M. Recent developments in gene therapy for homozygous familial hypercholesterolemia. Curr Atheroscler Rep. 2016;18:22. doi: 10.1007/s11883-016-0579-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hegele RA, Tsimikas S. Lipid-lowering agents. Circ Res. 2019;124:386–404. doi: 10.1161/CIRCRESAHA.118.313171. [DOI] [PubMed] [Google Scholar]
- 30.Frampton JE. Sebelipase Alfa: a review in lysosomal acid lipase deficiency. Am J Cardiovasc Drugs. 2016;16:461–468. doi: 10.1007/s40256-016-0203-2. [DOI] [PubMed] [Google Scholar]
- 31.Ferreira V, Petry H, Salmon F. Immune responses to AAV-vectors, the Glybera example from bench to bedside. Front Immunol. 2014;5:82. doi: 10.3389/fimmu.2014.00082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chaudhary R, Garg J, Shah N, Sumner A. PCSK9 inhibitors: a new era of lipid lowering therapy. World J Cardiol. 2017;9:76–91. doi: 10.4330/wjc.v9.i2.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, Yang Q, Hughes SG, Geary RS, Arca M, Stroes ESG, Bergeron J, Soran H, Civeira F, Hemphill L, Tsimikas S, Blom DJ, O’Dea L, Bruckert E. Volanesorsen and triglyceride levels in familial chylomicronemia syndrome. N Engl J Med. 2019;381:531–542. doi: 10.1056/NEJMoa1715944. [DOI] [PubMed] [Google Scholar]
- 34.Reeskamp LF, Millar JS, Wu L, Jansen H, van Harskamp D, Schierbeek H, Gipe DA, Rader DJ, Dallinga-Thie GM, Hovingh GK, Cuchel M. ANGPTL3 inhibition with evinacumab results in faster clearance of IDL and LDL apoB in patients with homozygous familial hypercholesterolemia-brief report. Arterioscler Thromb Vasc Biol. 2021;41:1753–1759. doi: 10.1161/ATVBAHA.120.315204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Korneva VA, Kuznetsova TY, Julius U. Modern approaches to lower lipoprotein(a) concentrations and consequences for cardiovascular diseases. Biomedicines. 2021;9:1271. doi: 10.3390/biomedicines9091271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.George RT, Abuhatzira L, Stoughton SM, Karathanasis SK, She D, Jin C, Buss NAPS, Bakker-Arkema R, Ongstad EL, Koren M, Hirshberg B. MEDI6012: recombinant human lecithin cholesterol acyltransferase, high-density lipoprotein, and low-density lipoprotein receptor-mediated reverse cholesterol transport. J Am Heart Assoc. 2021;10:e014572. doi: 10.1161/JAHA.119.014572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Capodanno D, Mehran R, Gibson CM, Angiolillo DJ. CSL112, a reconstituted, infusible, plasma-derived apolipoprotein A-I: safety and tolerability profiles and implications for management in patients with myocardial infarction. Expert Opin Investig Drugs. 2018;27:997–1005. doi: 10.1080/13543784.2018.1543399. [DOI] [PubMed] [Google Scholar]
- 38.Strain JD, Farver DK, Clem JR. A review on the rationale and clinical use of concomitant rosuvastatin and fenofibrate/fenofibric acid therapy. Clin Pharmacol. 2010;2:95–104. doi: 10.2147/CPAA.S7375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Genest J. Combination of statin and ezetimibe for the treatment of dyslipidemias and the prevention of coronary artery disease. Can J Cardiol. 2006;22:863–868. doi: 10.1016/s0828-282x(06)70305-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li Y, Wang X, Shen Z. Traditional Chinese medicine for lipid metabolism disorders. Am J Transl Res. 2017;9:2038–2049. [PMC free article] [PubMed] [Google Scholar]
- 41.Beltowski J, Wojcicka G, Jamroz-Wisniewska A. Adverse effects of statins - mechanisms and consequences. Curr Drug Saf. 2009;4:209–228. doi: 10.2174/157488609789006949. [DOI] [PubMed] [Google Scholar]
- 42.Bouitbir J, Sanvee GM, Panajatovic MV, Singh F, Krahenbuhl S. Mechanisms of statin-associated skeletal muscle-associated symptoms. Pharmacol Res. 2020;154:104201. doi: 10.1016/j.phrs.2019.03.010. [DOI] [PubMed] [Google Scholar]
- 43.Safitri N, Alaina MF, Pitaloka DAE, Abdulah R. A narrative review of statin-induced rhabdomyolysis: molecular mechanism, risk factors, and management. Drug Healthc Patient Saf. 2021;13:211–219. doi: 10.2147/DHPS.S333738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.USFDA. FDA drug safety communication: new restrictions, contraindications, and dose limitations of Zocor (simvastatin) to reduce the risk of muscle injury. 2011 [Google Scholar]
- 45.Enger C, Gately R, Ming EE, Niemcryk SJ, Williams L, McAfee AT. Pharmacoepidemiology safety study of fibrate and statin concomitant therapy. Am J Cardiol. 2010;106:1594–1601. doi: 10.1016/j.amjcard.2010.07.041. [DOI] [PubMed] [Google Scholar]
- 46.Rowan CG, Brunelli SM, Munson J, Flory J, Reese PP, Hennessy S, Lewis J, Mines D, Barrett JS, Bilker W, Strom BL. Clinical importance of the drug interaction between statins and CYP3A4 inhibitors: a retrospective cohort study in The Health Improvement Network. Pharmacoepidemiol Drug Saf. 2012;21:494–506. doi: 10.1002/pds.3199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ramkumar S, Raghunath A, Raghunath S. Statin therapy: review of safety and potential side effects. Acta Cardiol Sin. 2016;32:631–639. doi: 10.6515/ACS20160611A. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fabbian F, De Giorgi A, Pala M, Tiseo R, Manfredini R, Portaluppi F. Evidence-based statin prescription for cardiovascular protection in renal impairment. Clin Exp Nephrol. 2011;15:456–463. doi: 10.1007/s10157-011-0454-9. [DOI] [PubMed] [Google Scholar]
- 49.Ba Tuyen P, Huyen TT, Hang DTT, Thi Van Anh P. A novel herbal medicine for dyslipidemia: assessments in experimental models. Evid Based Complement Alternat Med. 2021;2021:5529744. doi: 10.1155/2021/5529744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Guo M, Liu Y, Gao ZY, Shi DZ. Chinese herbal medicine on dyslipidemia: progress and perspective. Evid Based Complement Alternat Med. 2014;2014:163036. doi: 10.1155/2014/163036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kuo PT. Dyslipidemia and coronary artery disease. Clin Cardiol. 1994;17:519–527. doi: 10.1002/clc.4960171003. [DOI] [PubMed] [Google Scholar]
- 52.Sun F, Ren Z, Jiang Y, Fang X, Wang N, Jin W. A placebo-controlled study on the treatment of metabolic syndrome of Qi stagnation and dampness obstruction related to atypical antipsychotics with Traditional Chinese Medicine (TCM) Evid Based Complement Alternat Med. 2020;2020:5103046. doi: 10.1155/2020/5103046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Shang H, Zhang L, Xiao T, Zhang L, Ruan J, Zhang Q, Liu K, Yu Z, Ni Y, Wang B. Study on the differences of gut microbiota composition between phlegm-dampness syndrome and qi-yin deficiency syndrome in patients with metabolic syndrome. Front Endocrinol (Lausanne) 2022;13:1063579. doi: 10.3389/fendo.2022.1063579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hao EW, Su ZX, Gong YL, Du ZC, Yang X, Huang CT, Hou XT, Deng JG. Analysis on application law of dampness-removing traditional Chinese medicines in treatment of coronavirus disease 2019. Chin Herb Med. 2021;13:518–524. doi: 10.1016/j.chmed.2021.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhang QY, Liang H, Gong HW, Huang HY, Zhou XQ, Sun X. Correlations between Phlegm syndrome of Chinese medicine and coronary angiography: a systematic review and meta-analysis. Evid Based Complement Alternat Med. 2015;2015:751743. doi: 10.1155/2015/751743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wang J, Wang Q, Li L, Li Y, Zhang H, Zheng L, Yang L, Zheng Y, Yang Y, Peng G, Zhang Y, Han Y. Phlegm-dampness constitution: genomics, susceptibility, adjustment and treatment with traditional Chinese medicine. Am J Chin Med. 2013;41:253–262. doi: 10.1142/S0192415X13500183. [DOI] [PubMed] [Google Scholar]
- 57.Shin J, Li T, Zhu L, Wang Q, Liang X, Li Y, Wang X, Zhao S, Li L, Li Y. Obese individuals with and without phlegm-dampness constitution show different gut microbial composition associated with risk of metabolic disorders. Front Cell Infect Microbiol. 2022;12:859708. doi: 10.3389/fcimb.2022.859708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Li HQ, Wei JJ, Xia W, Li JH, Liu AJ, Yin SB, Wang C, Song L, Wang Y, Zheng GQ, Fan JP. Promoting blood circulation for removing blood stasis therapy for acute intracerebral hemorrhage: a systematic review and meta-analysis. Acta Pharmacol Sin. 2015;36:659–675. doi: 10.1038/aps.2014.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Morita A, Namiki T, Nakaguchi T, Murai K, Watanabe Y, Nakamura M, Kawasaki Y, Shiko Y, Tamura Y, Suganami A, Murakami A, Yagi A, Okamoto H, Hirasaki Y. Role of blood stasis syndrome of kampo medicine in the early pathogenic stage of atherosclerosis: a retrospective cross-sectional study. Evid Based Complement Alternat Med. 2021;2021:5557392. doi: 10.1155/2021/5557392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Yu G, Wang J. Blood stasis syndrome of coronary heart disease: a perspective of modern medicine. Chin J Integr Med. 2014;20:300–306. doi: 10.1007/s11655-013-1332-3. [DOI] [PubMed] [Google Scholar]
- 61.Park B, You S, Jung J, Lee JA, Yun KJ, Lee MS. Korean studies on blood stasis: an overview. Evid Based Complement Alternat Med. 2015;2015:316872. doi: 10.1155/2015/316872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Adegbola P, Aderibigbe I, Hammed W, Omotayo T. Antioxidant and anti-inflammatory medicinal plants have potential role in the treatment of cardiovascular disease: a review. Am J Cardiovasc Dis. 2017;7:19–32. [PMC free article] [PubMed] [Google Scholar]
- 63.Zhang W, Huai Y, Miao Z, Qian A, Wang Y. Systems pharmacology for investigation of the mechanisms of action of traditional Chinese medicine in drug discovery. Front Pharmacol. 2019;10:743. doi: 10.3389/fphar.2019.00743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Adel Mehraban MS, Tabatabaei-Malazy O, Rahimi R, Daniali M, Khashayar P, Larijani B. Targeting dyslipidemia by herbal medicines: a systematic review of meta-analyses. J Ethnopharmacol. 2021;280:114407. doi: 10.1016/j.jep.2021.114407. [DOI] [PubMed] [Google Scholar]
- 65.Che CT, Wang ZJ, Chow MS, Lam CW. Herb-herb combination for therapeutic enhancement and advancement: theory, practice and future perspectives. Molecules. 2013;18:5125–5141. doi: 10.3390/molecules18055125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Chen HY, Lin YH, Chen YC. Identifying Chinese herbal medicine network for treating acne: implications from a nationwide database. J Ethnopharmacol. 2016;179:1–8. doi: 10.1016/j.jep.2015.12.032. [DOI] [PubMed] [Google Scholar]
- 67.Higaki S, Hasegawa Y, Morohashi M, Takayoshi Y. The correlation of Kampo formulations and their ingredients on anti-bacterial activities against Propionibacterium acnes. J Dermatol. 1995;22:4–9. doi: 10.1111/j.1346-8138.1995.tb03332.x. [DOI] [PubMed] [Google Scholar]
- 68.Higaki S, Nakamura M, Morohashi M, Hasegawa Y, Yamagishi T. Anti-lipase activity of Kampo formulations, coptidis rhizoma and its alkaloids against Propionibacterium acnes. J Dermatol. 1996;23:310–314. doi: 10.1111/j.1346-8138.1996.tb04021.x. [DOI] [PubMed] [Google Scholar]
- 69.Higaki S, Toyomoto T, Morohashi M. Seijo-bofu-to, Jumi-haidoku-to and Toki-shakuyaku-san suppress rashes and incidental symptoms in acne patients. Drugs Exp Clin Res. 2002;28:193–196. [PubMed] [Google Scholar]
- 70.Kim K, Kim KI, Lee J. Inhibitory effects of Cheongsangbangpoong-tang on both inflammatory acne lesions and facial heat in patients with acne vulgaris: a randomized controlled trial protocol. BMC Complement Altern Med. 2016;16:21. doi: 10.1186/s12906-016-1000-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kim B, Kim KI, Lee J, Kim K. Inhibitory effects of Cheongsangbangpoong-tang on both inflammatory acne lesion and facial heat in patients with acne vulgaris: a double-blinded randomized controlled trial. Complement Ther Med. 2019;44:110–115. doi: 10.1016/j.ctim.2019.03.018. [DOI] [PubMed] [Google Scholar]
- 72.Yang SH, Lin YH, Lin JR, Chen HY, Hu S, Yang YH, Yang YH, Yang YS, Fang YF. The efficacy and safety of a fixed combination of Chinese herbal medicine in chronic urticaria: a randomized, double-blind, placebo-controlled pilot study. Front Pharmacol. 2018;9:1474. doi: 10.3389/fphar.2018.01474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Seo CS, Shin HK. Simultaneous analysis of 19 marker components for quality control of Oncheong-Eum using HPLC-DAD. Molecules. 2022;27:2992. doi: 10.3390/molecules27092992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Yeh CC, Huang SS, Liu PY, Wang BC, Tsai CF, Wang DY, Cheng HF. Simultaneous quantification of six indicator compounds in Wen-Qing-Yin by high-performance liquid chromatography-diode array detection. J Food Drug Anal. 2019;27:749–757. doi: 10.1016/j.jfda.2018.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Andoh T, Al-Akeel A, Tsujii K, Nojima H, Kuraishi Y. Repeated treatment with the traditional medicine Unsei-in inhibits substance P-induced itch-associated responses through downregulation of the expression of nitric oxide synthase 1 in mice. J Pharmacol Sci. 2004;94:207–210. doi: 10.1254/jphs.94.207. [DOI] [PubMed] [Google Scholar]
- 76.Iijima S, Otsuka F, Kikuchi Y. Unsei-in inhibits rheological activity of leukocytes: mechanism of action in neutrophil-related skin diseases. Am J Chin Med. 1995;23:81–90. doi: 10.1142/S0192415X95000110. [DOI] [PubMed] [Google Scholar]
- 77.Higaki S, Morohashi M, Yamagishi T. Anti-lipase activity of Unsei-in against Propionibacterium avidum in the human axilla. Int J Antimicrob Agents. 2003;21:597–599. doi: 10.1016/s0924-8579(03)00089-x. [DOI] [PubMed] [Google Scholar]
- 78.Chang YY, Tsai YT, Lai JN, Yeh CH, Lin SK. The traditional Chinese medicine prescription patterns for migraine patients in Taiwan: a population-based study. J Ethnopharmacol. 2014;151:1209–1217. doi: 10.1016/j.jep.2013.12.040. [DOI] [PubMed] [Google Scholar]
- 79.Eun-Ju Lee S-yG, Hyung-Su Kwon, Hyun-Soo Keum, Seoung-Jun Kang, Chi-Sang Park, Chang-Gook Park. The experimental study on the effect of Chungsanggyuntong-tang on hyperlipidemia. The Journal of Internal Korean Medicine. 2000;21:409–416. [Google Scholar]
- 80.Hao P, Jiang F, Cheng J, Ma L, Zhang Y, Zhao Y. Traditional Chinese medicine for cardiovascular disease: evidence and potential mechanisms. J Am Coll Cardiol. 2017;69:2952–2966. doi: 10.1016/j.jacc.2017.04.041. [DOI] [PubMed] [Google Scholar]
- 81.Kim YJ, Kim TI, Kim K. An optimized herbal medicine containing Scutellaria baicalensis Georgi, Alisma orientale Juzepzuk, and Atractylodes japonica Koidzumi has potent antiplatelet and antithrombotic activities. J Tradit Complement Med. 2023;13:285–296. doi: 10.1016/j.jtcme.2023.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Chen QY, Wang CQ, Yang ZW, Tang Q, Tan HR, Wang X, Cai SQ. Differences in anti-inflammatory effects between two specifications of Scutellariae Radix in LPS-induced macrophages in vitro. Chin J Nat Med. 2017;15:515–524. doi: 10.1016/S1875-5364(17)30077-8. [DOI] [PubMed] [Google Scholar]
- 83.Liao H, Ye J, Gao L, Liu Y. The main bioactive compounds of Scutellaria baicalensis Georgi. for alleviation of inflammatory cytokines: a comprehensive review. Biomed Pharmacother. 2021;133:110917. doi: 10.1016/j.biopha.2020.110917. [DOI] [PubMed] [Google Scholar]
- 84.Khan S, Zhang D, Zhang Y, Li M, Wang C. Wogonin attenuates diabetic cardiomyopathy through its anti-inflammatory and anti-oxidative properties. Mol Cell Endocrinol. 2016;428:101–108. doi: 10.1016/j.mce.2016.03.025. [DOI] [PubMed] [Google Scholar]
- 85.Guo LT, Wang SQ, Su J, Xu LX, Ji ZY, Zhang RY, Zhao QW, Ma ZQ, Deng XY, Ma SP. Baicalin ameliorates neuroinflammation-induced depressive-like behavior through inhibition of toll-like receptor 4 expression via the PI3K/AKT/FoxO1 pathway. J Neuroinflammation. 2019;16:95. doi: 10.1186/s12974-019-1474-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Dai J, Liang K, Zhao S, Jia W, Liu Y, Wu H, Lv J, Cao C, Chen T, Zhuang S, Hou X, Zhou S, Zhang X, Chen XW, Huang Y, Xiao RP, Wang YL, Luo T, Xiao J, Wang C. Chemoproteomics reveals baicalin activates hepatic CPT1 to ameliorate diet-induced obesity and hepatic steatosis. Proc Natl Acad Sci U S A. 2018;115:E5896–E5905. doi: 10.1073/pnas.1801745115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wang ZY, Jiang ZM, Xiao PT, Jiang YQ, Liu WJ, Liu EH. The mechanisms of baicalin ameliorate obesity and hyperlipidemia through a network pharmacology approach. Eur J Pharmacol. 2020;878:173103. doi: 10.1016/j.ejphar.2020.173103. [DOI] [PubMed] [Google Scholar]
- 88.Na HY, Lee BC. Scutellaria baicalensis alleviates insulin resistance in diet-induced obese mice by modulating inflammation. Int J Mol Sci. 2019;20:727. doi: 10.3390/ijms20030727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Zhao T, Tang H, Xie L, Zheng Y, Ma Z, Sun Q, Li X. Scutellaria baicalensis Georgi. (Lamiaceae): a review of its traditional uses, botany, phytochemistry, pharmacology and toxicology. J Pharm Pharmacol. 2019;71:1353–1369. doi: 10.1111/jphp.13129. [DOI] [PubMed] [Google Scholar]
- 90.Li C, Lin G, Zuo Z. Pharmacological effects and pharmacokinetics properties of Radix Scutellariae and its bioactive flavones. Biopharm Drug Dispos. 2011;32:427–445. doi: 10.1002/bdd.771. [DOI] [PubMed] [Google Scholar]
- 91.An HJ, Lee JY, Park W. Baicalin modulates inflammatory response of macrophages activated by LPS via Calcium-CHOP pathway. Cells. 2022;11:3076. doi: 10.3390/cells11193076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ku SK, Bae JS. Baicalin, baicalein and wogonin inhibits high glucose-induced vascular inflammation in vitro and in vivo. BMB Rep. 2015;48:519–524. doi: 10.5483/BMBRep.2015.48.9.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Dinda B, Dinda S, DasSharma S, Banik R, Chakraborty A, Dinda M. Therapeutic potentials of baicalin and its aglycone, baicalein against inflammatory disorders. Eur J Med Chem. 2017;131:68–80. doi: 10.1016/j.ejmech.2017.03.004. [DOI] [PubMed] [Google Scholar]
- 94.Liu BY, Li L, Liu GL, Ding W, Chang WG, Xu T, Ji XY, Zheng XX, Zhang J, Wang JX. Baicalein attenuates cardiac hypertrophy in mice via suppressing oxidative stress and activating autophagy in cardiomyocytes. Acta Pharmacol Sin. 2021;42:701–714. doi: 10.1038/s41401-020-0496-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Chandrashekar N, Pandi A. Baicalein: a review on its anti-cancer effects and mechanisms in lung carcinoma. J Food Biochem. 2022;46:e14230. doi: 10.1111/jfbc.14230. [DOI] [PubMed] [Google Scholar]
- 96.Tsai FJ, Li TM, Cheng CF, Wu YC, Lai CH, Ho TJ, Liu X, Tsang H, Lin TH, Liao CC, Huang SM, Li JP, Lin JC, Lin CC, Liang WM, Lin YJ. Effects of Chinese herbal medicine on hyperlipidemia and the risk of cardiovascular disease in HIV-infected patients in Taiwan. J Ethnopharmacol. 2018;219:71–80. doi: 10.1016/j.jep.2018.03.006. [DOI] [PubMed] [Google Scholar]
- 97.Shen M, Wang L, Yang G, Gao L, Wang B, Guo X, Zeng C, Xu Y, Shen L, Cheng K, Xia Y, Li X, Wang H, Fan L, Wang X. Baicalin protects the cardiomyocytes from ER stress-induced apoptosis: inhibition of CHOP through induction of endothelial nitric oxide synthase. PLoS One. 2014;9:e88389. doi: 10.1371/journal.pone.0088389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Huang Y, Tsang SY, Yao X, Chen ZY. Biological properties of baicalein in cardiovascular system. Curr Drug Targets Cardiovasc Haematol Disord. 2005;5:177–184. doi: 10.2174/1568006043586206. [DOI] [PubMed] [Google Scholar]
- 99.Li Q, Qu H. Study on the hypoglycemic activities and metabolism of alcohol extract of Alismatis Rhizoma. Fitoterapia. 2012;83:1046–1053. doi: 10.1016/j.fitote.2012.05.009. [DOI] [PubMed] [Google Scholar]
- 100.Liu SS, Sheng WL, Li Y, Zhang SS, Zhu JJ, Gao HM, Yan LH, Wang ZM, Gao L, Zhang M. Chemical constituents from Alismatis Rhizoma and their anti-inflammatory activities in vitro and in vivo. Bioorg Chem. 2019;92:103226. doi: 10.1016/j.bioorg.2019.103226. [DOI] [PubMed] [Google Scholar]
- 101.Han CW, Kwun MJ, Kim KH, Choi JY, Oh SR, Ahn KS, Lee JH, Joo M. Ethanol extract of Alismatis Rhizoma reduces acute lung inflammation by suppressing NF-kappaB and activating Nrf2. J Ethnopharmacol. 2013;146:402–410. doi: 10.1016/j.jep.2013.01.010. [DOI] [PubMed] [Google Scholar]
- 102.Wang K, Zhang B, Song D, Xi J, Hao W, Yuan J, Gao C, Cui Z, Cheng Z. Alisol A alleviates arterial plaque by activating AMPK/SIRT1 signaling pathway in apoE-deficient mice. Front Pharmacol. 2020;11:580073. doi: 10.3389/fphar.2020.580073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Meng Q, Chen X, Wang C, Liu Q, Sun H, Sun P, Huo X, Liu Z, Liu K. Protective effects of alisol B 23-acetate from edible botanical Rhizoma alismatis against carbon tetrachloride-induced hepatotoxicity in mice. Food Funct. 2015;6:1241–1250. doi: 10.1039/c5fo00082c. [DOI] [PubMed] [Google Scholar]
- 104.Ho C, Gao Y, Zheng D, Liu Y, Shan S, Fang B, Zhao Y, Song D, Zhang Y, Li Q. Alisol A attenuates high-fat-diet-induced obesity and metabolic disorders via the AMPK/ACC/SREBP-1c pathway. J Cell Mol Med. 2019;23:5108–5118. doi: 10.1111/jcmm.14380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Chen X, Liu H. Alisol A inhibited the proliferation, migration, and invasion of nasopharyngeal carcinoma cells by inhibiting the Hippo signaling pathway. Yonsei Med J. 2021;62:895–902. doi: 10.3349/ymj.2021.62.10.895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Zhang LL, Xu W, Xu YL, Chen X, Huang M, Lu JJ. Therapeutic potential of Rhizoma Alismatis: a review on ethnomedicinal application, phytochemistry, pharmacology, and toxicology. Ann N Y Acad Sci. 2017;1401:90–101. doi: 10.1111/nyas.13381. [DOI] [PubMed] [Google Scholar]
- 107.Li X, Lin J, Han W, Mai W, Wang L, Li Q, Lin M, Bai M, Zhang L, Chen D. Antioxidant ability and mechanism of rhizoma Atractylodes macrocephala. Molecules. 2012;17:13457–13472. doi: 10.3390/molecules171113457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Song MY, Lim SK, Wang JH, Kim H. The root of Atractylodes macrocephala Koidzumi prevents obesity and glucose intolerance and increases energy metabolism in mice. Int J Mol Sci. 2018;19:278. doi: 10.3390/ijms19010278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Kim CK, Kim M, Oh SD, Lee SM, Sun B, Choi GS, Kim SK, Bae H, Kang C, Min BI. Effects of Atractylodes macrocephala Koidzumi rhizome on 3T3-L1 adipogenesis and an animal model of obesity. J Ethnopharmacol. 2011;137:396–402. doi: 10.1016/j.jep.2011.05.036. [DOI] [PubMed] [Google Scholar]
- 110.Song MY, Kang SY, Oh TW, Kumar RV, Jung HW, Park YK. The roots of Atractylodes macrocephala Koidzumi enhanced glucose and lipid metabolism in C2C12 myotubes via mitochondrial regulation. Evid Based Complement Alternat Med. 2015;2015:643654. doi: 10.1155/2015/643654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Han Y, Jung HW, Park YK. The roots of Atractylodes japonica Koidzumi promote adipogenic differentiation via activation of the insulin signaling pathway in 3T3-L1 cells. BMC Complement Altern Med. 2012;12:154. doi: 10.1186/1472-6882-12-154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Zhou Y, Tao H, Wang A, Zhong Z, Wu X, Wang M, Bian Z, Wang S, Wang Y. Chinese herb pair Paeoniae Radix Alba and Atractylodis Macrocephalae Rhizoma suppresses LPS-induced inflammatory response through inhibiting MAPK and NF-kappaB pathway. Chin Med. 2019;14:2. doi: 10.1186/s13020-019-0224-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Jeong YH, Li W, Go Y, Oh YC. Atractylodis Rhizoma Alba attenuates neuroinflammation in BV2 microglia upon LPS stimulation by inducing HO-1 activity and inhibiting NF-kappaB and MAPK. Int J Mol Sci. 2019;20:4015. doi: 10.3390/ijms20164015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zhang WJ, Zhao ZY, Chang LK, Cao Y, Wang S, Kang CZ, Wang HY, Zhou L, Huang LQ, Guo LP. Atractylodis Rhizoma: a review of its traditional uses, phytochemistry, pharmacology, toxicology and quality control. J Ethnopharmacol. 2021;266:113415. doi: 10.1016/j.jep.2020.113415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Li CQ, He LC, Jin JQ. Atractylenolide I and atractylenolide III inhibit Lipopolysaccharide-induced TNF-alpha and NO production in macrophages. Phytother Res. 2007;21:347–353. doi: 10.1002/ptr.2040. [DOI] [PubMed] [Google Scholar]
- 116.Ren Y, Jiang W, Luo C, Zhang X, Huang M. Atractylenolide III ameliorates TNBS-induced intestinal inflammation in mice by reducing oxidative stress and regulating intestinal flora. Chem Biodivers. 2021;18:e2001001. doi: 10.1002/cbdv.202001001. [DOI] [PubMed] [Google Scholar]
- 117.Bailly C. Atractylenolides, essential components of Atractylodes-based traditional herbal medicines: antioxidant, anti-inflammatory and anticancer properties. Eur J Pharmacol. 2021;891:173735. doi: 10.1016/j.ejphar.2020.173735. [DOI] [PubMed] [Google Scholar]
- 118.Wang M, Hu R, Wang Y, Liu L, You H, Zhang J, Wu X, Pei T, Wang F, Lu L, Xiao W, Wei L. Atractylenolide III attenuates muscle wasting in chronic kidney disease via the oxidative stress-mediated PI3K/AKT/mTOR pathway. Oxid Med Cell Longev. 2019;2019:1875471. doi: 10.1155/2019/1875471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Liu M, Wang RB, Xing JH, Tang YX. Atractylenolide inhibits apoptosis and oxidative stress of HTR-8/SVneo cells by activating MAPK/ERK signalling in preeclampsia. Phytomedicine. 2021;93:153773. doi: 10.1016/j.phymed.2021.153773. [DOI] [PubMed] [Google Scholar]
- 120.Wang P, Zhao YN, Xu RZ, Zhang XW, Sun YR, Feng QM, Li ZH, Xu JY, Xie ZS, Zhang ZQ, E HC. Sesquiterpene lactams and lactones with antioxidant potentials from atractylodes macrocephala discovered by molecular networking strategy. Front Nutr. 2022;9:865257. doi: 10.3389/fnut.2022.865257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Song MY, Jung HW, Kang SY, Park YK. Atractylenolide III enhances energy metabolism by increasing the SIRT-1 and PGC1alpha expression with AMPK phosphorylation in C2C12 mouse skeletal muscle cells. Biol Pharm Bull. 2017;40:339–344. doi: 10.1248/bpb.b16-00853. [DOI] [PubMed] [Google Scholar]
- 122.Tsai CJ, Liang JW, Lin HR. Sesquiterpenoids from Atractylodes macrocephala act as farnesoid X receptor and progesterone receptor modulators. Bioorg Med Chem Lett. 2012;22:2326–2329. doi: 10.1016/j.bmcl.2012.01.048. [DOI] [PubMed] [Google Scholar]
- 123.Li Q, Tan JX, He Y, Bai F, Li SW, Hou YW, Ji LS, Gao YT, Zhang X, Zhou ZH, Yu Z, Fang M, Gao YQ, Li M. Atractylenolide III ameliorates non-alcoholic fatty liver disease by activating hepatic adiponectin receptor 1-mediated AMPK pathway. Int J Biol Sci. 2022;18:1594–1611. doi: 10.7150/ijbs.68873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Liu X, Huang Y, Liang X, Wu Q, Wang N, Zhou LJ, Liu WW, Ma Q, Hu B, Gao H, Cui YL, Li X, Zhao QC. Atractylenolide III from Atractylodes macrocephala Koidz promotes the activation of brown and white adipose tissue through SIRT1/PGC-1alpha signaling pathway. Phytomedicine. 2022;104:154289. doi: 10.1016/j.phymed.2022.154289. [DOI] [PubMed] [Google Scholar]
- 125.Li H, Shi X, Jiang H, Kang J, Yu M, Li Q, Yu K, Chen Z, Pan H, Chen W. CMap analysis identifies Atractyloside as a potential drug candidate for type 2 diabetes based on integration of metabolomics and transcriptomics. J Cell Mol Med. 2020;24:7417–7426. doi: 10.1111/jcmm.15357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Zhang P, Li L, Sun H, Zhang Y, Zhang G, Zhang T, Zeng C. Mitochondrial energy-regulating effect of atractyloside inhibits hepatocellular steatosis through the activation of autophagy. Front Pharmacol. 2020;11:575695. doi: 10.3389/fphar.2020.575695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Miljkovic M, Stefanovic A, Simic-Ogrizovic S, Vekic J, Bogavac-Stanojevic N, Cerne D, Kocbek P, Marc J, Jelic-Ivanovic Z, Spasojevic-Kalimanovska V, Kotur-Stevuljevic J. Association of dyslipidemia, oxidative stress, and inflammation with redox status in VLDL, LDL, and HDL lipoproteins in patients with renal disease. Angiology. 2018;69:861–870. doi: 10.1177/0003319718780041. [DOI] [PubMed] [Google Scholar]
- 128.Liu Q, Tang GY, Zhao CN, Feng XL, Xu XY, Cao SY, Meng X, Li S, Gan RY, Li HB. Comparison of antioxidant activities of different grape varieties. Molecules. 2018;23:2432. doi: 10.3390/molecules23102432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Huang Z, Wang J, Li C, Zheng W, He J, Wu Z, Tang J. Application of natural antioxidants from traditional Chinese medicine in the treatment of spinal cord injury. Front Pharmacol. 2022;13:976757. doi: 10.3389/fphar.2022.976757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Yu-Hao D, Chun C, Qiang H, Xiong F. Study on a novel spherical polysaccharide from Fructus Mori with good antioxidant activity. Carbohydr Polym. 2021;256:117516. doi: 10.1016/j.carbpol.2020.117516. [DOI] [PubMed] [Google Scholar]
- 131.Adorni MP, Zimetti F, Lupo MG, Ruscica M, Ferri N. Naturally occurring PCSK9 inhibitors. Nutrients. 2020;12:1440. doi: 10.3390/nu12051440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Russo S, Kwiatkowski M, Govorukhina N, Bischoff R, Melgert BN. Meta-inflammation and metabolic reprogramming of macrophages in diabetes and obesity: the importance of metabolites. Front Immunol. 2021;12:746151. doi: 10.3389/fimmu.2021.746151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Huang Y, Jin X, Liu J, Wu W, Wang H. Systems pharmacology approach to investigate the mechanism of Artemisia argyi in treating rheumatic diseases. Sci Rep. 2022;12:18786. doi: 10.1038/s41598-022-23635-6. [DOI] [PMC free article] [PubMed] [Google Scholar]