Next Article in Journal
Antiradical and Antioxidant Activity of Compounds Containing 1,3-Dicarbonyl Moiety: An Overview
Previous Article in Journal
Field-Flow Fractionation in Molecular Biology and Biotechnology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Micro-Executor of Natural Products in Metabolic Diseases

by
Jinxin Liu
1,
Huanwen Chen
2,
Xiaoli Li
3,
Chunmei Song
1,
Li Wang
4,* and
Deguo Wang
1,5,*
1
Food and Pharmacy College, Xuchang University, Xuchang 461000, China
2
Center for Agricultural and Rural Development, Zhangdian District, Zibo 255000, China
3
Zibo Digital Agriculture and Rural Development Center, Zibo 255000, China
4
School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
5
Key Laboratory of Biomarker Based Rapid-Detection Technology for Food Safety of Henan Province, Xuchang University, Xuchang 461000, China
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(17), 6202; https://doi.org/10.3390/molecules28176202
Submission received: 29 June 2023 / Revised: 18 August 2023 / Accepted: 22 August 2023 / Published: 23 August 2023

Abstract

:
Obesity, diabetes, and cardiovascular diseases are the major chronic metabolic diseases that threaten human health. In order to combat these epidemics, there remains a desperate need for effective, safe, and easily available therapeutic strategies. Recently, the development of natural product research has provided new methods and options for these diseases. Numerous studies have demonstrated that microRNAs (miRNAs) are key regulators of metabolic diseases, and natural products can improve lipid and glucose metabolism disorders and cardiovascular diseases by regulating the expression of miRNAs. In this review, we present the recent advances involving the associations between miRNAs and natural products and the current evidence showing the positive effects of miRNAs for natural product treatment in metabolic diseases. We also encourage further research to address the relationship between miRNAs and natural products under physiological and pathological conditions, thus leading to stronger support for drug development from natural products in the future.

1. Introduction

Metabolic diseases, which encompass a variety of risk factors highly associated with obesity, diabetes, and cardiovascular diseases, have come to be regarded as public health challenges [1,2,3,4]. Due to their complex mechanisms of action, effective comprehensive treatments are still lacking. Even worse, the side effects of some curative drugs have been a major concern for their therapeutic usage [5,6,7]. Therefore, it is imperative to provide an effective treatment approach to overcome the aforementioned diseases.
Natural products that are extracted from the source and from concentrated, fractionated, and purified yielding, which are generally defined as bioactive compounds [8,9], have the ability to modulate lipid metabolism, improve insulin signaling, and protect against cardiovascular damage [10,11]. More importantly, natural products are widely distributed and readily available in nature [12]. To date, extensive studies have shown that plentiful drugs are derived from structural modification based on natural products [13]. MicroRNAs (miRNAs) and small noncoding RNAs are characterized by binding to the regulatory sites of 3′UTR of target mRNA, resulting in the inhibition of transcription or the promotion of degradation, accompanied by decreased protein synthesis [14,15]. Natural products could also ameliorate metabolic diseases by targeting abundant miRNAs [16,17,18]. Thus, the possibility for natural products to modify the abnormal patterns of these diseases is, at least in part, possible through a newly defined mechanism: the miRNAs cascade.
In this review, we summarize the positive effects of natural products on lipid and glucose metabolism disorders and cardiovascular diseases, explain the underlying molecular mechanisms, and provide the theoretical basis for metabolic diseases. We also highlight the regulatory effect of natural products on miRNAs in this therapeutic process. In the hope of the preferable utilization of existing data, we thus provide a new route for future drug discovery.

2. Research Methodology

A systematic search of the literature was performed in the PubMed and Web of Science databases (up to December 2022). The following text terms were used to identify any study candidates: (“microRNA(s)” OR “miRNA(s)” OR “miR”) AND (“natural product(s)” OR “plant(s)” OR “Herb(s)” OR “extract(s)”) AND (“metabolic disease(s)” OR “glyco-lipid metabolism disorders” OR “lipid metabolism disorder(s)” OR “Glucose metabolism disorder(s)” OR “cardiovascular disease(s)” OR “obesity” OR “diabetes” OR “glucose” OR “lipid” OR “fat” OR “adipocyte” OR “metabolism”). Moreover, we hand-searched the citation lists of the included studies to identify the relevant literature. As for data extraction, the titles, keywords, abstracts, and full texts of all enrolled articles were screened. Any duplicates and irrelevant studies were excluded; traditional Chinese medicine formulas, extracts, or combinations with unclear functional components were also excluded from the literature. The useful data were extracted from the relevant qualified literature into specifically designed spreadsheets. The following data were included: the first author, country, year of publication, natural products, functional components, experimental models, dosage and duration of treatment, observed effects, associated miRNAs, and target genes.

3. Effects of Natural Products on Lipid Metabolism Disorders

Lipid metabolism is a crucial and complex biochemical reaction in the body, and diseases caused by lipid metabolism disorders are common in modern society, such as obesity and hyperlipidemia [19]. Lipids are known to be important substances in energy storage and energy supply. Hence, the proper amount of adipose tissue is necessary for the human body. In general, however, patients have difficulty sticking to a long-term diet and physical activity regimen to combat these metabolic disorders. Therefore, food components that ameliorate the risk factors associated with these diseases can facilitate dietary-based therapies [16]. Dietary natural products have long been of great interest for improving lipid metabolism by modulating miRNA expression.

3.1. Regulatory Effects on Fatty Acid Synthesis and Decomposition

It is well known that fatty acids are the simplest type of lipids and are the building blocks of many more complex fats. Furthermore, they are also perceived as one of the main sources of energy on account of releasing a lot of energy during oxidation into CO2 and H2O in the case of a sufficient oxygen supply. Therefore, the role of fatty acids in the processes of lipogenesis and lipodieresis cannot be ignored. Recently, investigators have examined the regulatory effects of natural products on lipogenesis and lipodieresis, resulting in improved lipid metabolism through the management of diverse miRNAs (Table 1 and Table 2).
Specifically, miR-122 and miR-33 are two of the best-studied miRNAs involved in the regulation of lipid metabolism [53]. As is shown in Table 1 and Table 2, numerous pieces of evidence have revealed that grape seed proanthocyanidin extract treatments reduced fatty acid synthesis and de novo lipogenesis, increased liver cholesterol efflux to high-density lipoprotein (HDL) formation by decreasing the expression of miR-122 and miR-33, which could regulate several genes that control fatty acid and transcriptional regulatory factors, such as fatty acid synthase (FAS) and peroxisome proliferator-activated receptor beta/delta (PPARβ/δ), as well as genes that regulate fatty acid β-oxidation, such as ATP-binding cassette transporter A1 (ABCA1) and carnitine palmitoyltransferase 1a (CPT1a), respectively [16,25,26,27,29,54]. Further detection revealed that the levels of total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-C) were reduced while the level of high-density lipoproteins cholesterol (HDL-C) was enhanced in a dose-dependent manner [16,25,26,27]. Averrhoa carambola-free phenolic extract, citrus peel flavonoids, lychee pulp phenolics, mulberry fruit extract, and portulaca oleracea extract treatments could also improve lipid metabolism in in vitro and in vivo studies; the underlying mechanism was miR-33 or miR-122-mediated changes in the signaling pathways [20,22,33,34,37,38]. However, the opposite expression of miR-122 was reflected in a natural product experiment using coffee polyphenols, which could enhance energy metabolism and reduce lipogenesis by targeting sterol regulatory element binding protein (SREBP) 1c mediated by miR-122 [23]. Accountably, SREBP1c, one of the three isoforms of SREBPs, comes into play in fatty acid synthesis and metabolism [55,56]. It potentially illustrates the point that the same type of miRNAs can act on a variety of target genes with different expressions. Similarly, the same target gene may also be regulated by multiple miRNAs. Recent studies have shown that miR-103 and miR-107 reduced obesogenic diet-induced hepatic steatosis via decreasing the protein expression of SREBP1 in resveratrol-treated rats [49], and pseudoprotodioscin promoted cholesterol effluxion through targeting SREBP1c and SREBP2 mediated by miR-33a/b in an in vitro experiment [48]. In addition, distinctively, the overexpression of hepatic miR-98 induced by oleanolic acid, an active component of the traditional Chinese herb olea europaea, increased the degradation of peroxisome proliferator-activated receptor gamma coactivator-1beta (PGC1β), known as a transcriptional co-activator of SREBP-1 and the master regulator of hepatic lipogenesis [46,57].
Intuitively, both FAS and SREBP1 are involved in the process of fatty acid synthesis, and the connection between them is found in the following experiments. SP1 transcription factor (SP1), an important member of the ubiquitously expressed SP/KLF transcription factor family, acts together with SREBP1 to synergistically activate the promoter of the FAS gene and is involved in de novo lipogenesis [58]. Hence, resveratrol reduced the expression of SP1 through upregulating miR-539, along with decreasing the expression of the SREBP1 protein and FAS gene in vivo and in vitro [50]. Nevertheless, the correlation between them still needs to be systematically and intensively investigated beyond all doubt.
Lipid metabolism is a complex process, and natural products can regulate lipogenesis and lipodieresis in a variety of ways. Zerumbone is a cyclic sesquiterpene isolated from the wild ginger Zingiber zerumbet smith. It has been proved that zerumbone could improve lipid metabolism disorder by reducing lipogenesis and increasing fatty acid oxidation [52]. For one thing, zerumbone acted as a miR-146b inhibitor and downregulated miR-146b, leading to the activation of sirtuin type 1 (Sirt1), which induced the de-acetylation of forkhead box O1 (FOXO1) and peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC1α); for another, zerumbone induced the phosphorylation of AMP-activated protein kinase (AMPK), which could limit fatty acid efflux from adipocytes and favor fatty acid oxidation, as well as decrease de novo fatty acid synthesis through the phosphorylation-mediated inhibition of acetyl-CoA carboxylase (ACC) [59,60,61] and also activated Sirt1 indirectly [52]. With these similar natural product experiments, miR-27a/b, miR-96, miR-34a, miR-194, and miR-355 also participated in the process of lipid metabolism by targeting Sirt1 or FOXO1, which could both increase energy expenditure and the clearance of lipid accumulation [21,28,30,31,35,36]. And ginger extract could enhance AMPK activity and ameliorate obesity and inflammation by regulating miRNAs expressions in high-fat diet (HFD)-fed rats [24].
Based on the present studies, regulating miRNAs is potentially becoming a dominant feature in terms of natural products regulating lipid metabolism (Figure 1). On the one hand, it can inhibit fatty synthesis by reducing fatty acid synthesis and increasing fatty acids mobilization. On the other hand, it can also accelerate lipodieresis by enhancing the oxidation and phosphorylation of fatty acids.

3.2. Inhibitory Effects on Adipocyte Differentiation and Accumulation

From the perspective of the cellular level, however, the growth of adipose tissue is the result of an increase in the number of adipocytes and the volume of individual cells [51]. The former contributes to promoting pre-adipocyte differentiation into mature adipocytes, whereas the latter is due to lipid accumulation. Here, we summarized the functional role of natural products in this regard, as well as their potential mechanisms of action (Figure 1 and Table 1 and Table 2).
Adipocyte differentiation is a highly precisely regulated cellular process. Ahead of terminal differentiation, the mitotic clonal expansion (MCE) of stimulated pre-adipocytes is an essential procedure in adipocyte differentiation. Moreover, the transcriptional activation of adipocyte-specific functional genes is closely related to their differentiation [62]. 3T3-L1 pre-adipocytes have long been considered as the “gold standard” for investigating pre-adipocyte differentiation in vitro [63,64]. There has been evidence that the MCE process could be delayed by persimmon tannin by enhancing the expression of miR-27 in 3T3-L1 pre-adipocytes [47]. Furthermore, multiple transcriptional factors, including peroxisome proliferator-activated receptor-gamma (PPARγ) and CCAAT/enhancer-binding protein alpha (C/EBPα) were also attenuated by miR-27, resulting in a decrease in adipocyte-specific genes, such as adipocyte fatty acid binding protein (aP2) and lipoprotein lipase (LPL). Similarly, the MCE process was blocked by miR-27a/b in the study of a-type ECG and EGCG dimers [40]. As we all know, lipids are important structural components in cell membranes [65]. Notably, with different molecular structures, a-type ECG and EGCG dimers strongly disturbed the structures of cell membranes by decreasing fluidity and hydrophobicity and increasing the permeability of the membrane of 3T3-L1 pre-adipocyte cells, thus displaying significant inhibition on differentiation [40]. EGCG also suppressed 3T3-L1 cell growth via miR-143/MAPK7 pathways [43]. Nonivamide-induced reduction in lipid accumulation was mediated by transient receptor potential cation channel subfamily V member 1 (TRPV1) activation [45]. Although miRNAs are involved in the adipocyte differentiation process, whether they affect membrane structure remains to be intensively studied in natural product therapy.
The activation of C/EBPα and PPARγ is not only necessary for adipocyte differentiation in the early stage but is also crucial for terminal adipocyte differentiation [66]. The evidence suggests that grape seed procyanidin B2 could inhibit pre-adipocyte differentiation and reduce intracellular lipid accumulation by modulating the miR-483/PPARγ axis [42]. Resveratrol reduced the expression of CEBP/α by boosting miR-155, resulting in decreasing lipogenesis [51]. Consistent with these, as shown in Table 2, similar results were also obtained in the research of lycopene by regulating the expression of miR-21 [44]. What is noteworthy is that accompanied with the involvement of multiple miRNAs, Rosmarinus officinalis extract significantly reduced triglyceride incorporation during pre-adipocyte maturation in a dose-dependent manner and decreased the expression of cell cycle genes, such as cyclin-dependent kinase 4, cyclin D1, and cyclin-dependent kinase inhibitor 1A [39]. The final and most studied phase of adipocyte differentiation involves terminal differentiation and the induction of a signaling cascade to promote the expression of the genes necessary for adipocyte function [67,68]. The canonical Wnt signaling cascade is an effective approach to suppress adipogenesis [69,70,71]. Recently, investigators found that curcumin repressed 3T3-L1 pre-adipocyte cell adipogenic differentiation by inhibiting the expression of miR-17 and stimulating transcription factor 7-like 2 (TCF7L2), which is the Wnt signaling pathway effector and a direct downstream target of miR-17 [41]. And guarana extract also exerted an anti-adipogenic effect by regulating the Wnt signaling pathway, mediated by miRNAs [32]. In summary, natural products may inhibit adipocyte differentiation and accumulation by regulating miRNAs, which play a crucial role in the process of lipogenesis.

4. Effects of Natural Products on Glucose Metabolism Disorders

Glucose metabolism is the basis of metabolism in the body. Metabolic diseases caused by abnormal glucose metabolism, such as diabetes, are the focus and difficulty of current social research due to their progressively expanding populations, complex pathogenesis, drug-maintained recovery, and high expenditure [72,73]. Diabetes is a heterogeneous group of disorders characterized by hyperglycemia due to an absolute or relative deficit in insulin production or action [74]. Under hyperglycemic conditions, reactive oxygen species (ROS) increase, causing cells to activate various abnormal metabolic pathways and inducing oxidative stress [75]. Now that the prevention and treatment of diabetes offers a new avenue, natural products are an increasingly significant area of product development for anti-diabetic drugs. Here, we revealed the effects of natural products on the action of hypoglycemia and the inhibition of oxidative stress by regulating miRNAs (Table 3 and Table 4).

4.1. Hypoglycemic Action

At the heart of glucose metabolism is maintaining an equilibrium of glucose concentrations in the blood. So, blood glucose concentration is used as an important indicator of glucose metabolism in the body [91]. Insulin, secreted by β-cells in the pancreas, is the hormone currently known to lower blood glucose in the body. However, it has been now well established from a variety of studies that natural products can reduce blood glucose by acting on insulin (Table 3 and Table 4), while the potential mechanisms of action are diverse (Figure 2).
Protein tyrosine phosphatase 1B (PTP1B) is a major negative regulator of the insulin signaling pathway in metabolism and dephosphorylates insulin receptor (IR) and insulin receptor substrate 1 (IRS1) at tyrosine residues to inhibit the activation of downstream Akt and ERK1/2 signaling cascades [92,93]. Curcumin, however, could induce miR-206 expression, which, in turn, decreased fructose-induced PTP1B overexpression to improve glucose intolerance and insulin sensitivity in fructose-fed rats [83]. Interestingly, the same results were also found in a study on licorice flavonoid, which reversed the decrease of miR-122 induced by the overexpression of PTP1B and abrogated the hepatic insulin resistance induced by an HFD diet [80]. Gypenoside A attenuated the dysfunction of pancreatic β cells by activating pancreatic duodenal homeobox-1 (PDX1) signal transduction via the inhibition of miR-150 in HFD-fed mice [86]. Vaccarin, an active flavonoid glycoside extracted from vaccariae semen, reduced blood glucose, increased glucose and insulin tolerance, and relieved glucose metabolism disturbances in STZ/HFD-induced type 2 diabetes mellitus (T2DM) mice by regulating miR-34a expression [90]. Coreopsis tinctoria nutt extract treatment also showed the effect of lowering fasting blood by inhibiting the expression of miR-192 and miR-200b [78]. From the above, insulin plays an important role in blood glucose stability, and importantly, both low-insulin secretion and insulin resistance can lead to blood glucose disorders. This can, however, be reversed, at least in part, by natural products through miRNA cascades.
In addition, by using hierarchical clustering analysis, the miRNA expression patterns, as well as miRNA microarray analysis, are shown in Table 3. It was macroscopically discovered that various miRNAs participated in the hypoglycemic process in Alpinia oxyphylla extract treatment [76]; however, the underlying mechanism of action is not yet clear.

4.2. Restraining Effects on Oxidative Stress

Insulin signaling has been one of the most important and highly studied metabolic hormones for glucose metabolism homeostasis. ROS are usually produced in the process of biological oxidation and energy conversion in mitochondria; however, the enhancement of ROS induced by a hyperglycemic environment disrupts the balance between ROS and the antioxidant system, resulting in oxidative stress, which subsequently induces insulin resistance and pancreatic β cell dysfunction via their ability to activate stress-sensitive signaling pathways [94]. Therefore, oxidative stress has been defined as a disturbance in the dynamic balance between ROS generation and antioxidant capacity [95], and ROS generation is also regarded as a marker of oxidative stress, which can lead to pancreatic β cell dysfunction and peripheral insulin resistance, hence, resulting in glucose metabolic disorders [95,96]. A growing number of studies have shown that natural products can inhibit oxidative stress by monitoring various miRNAs (Table 3 and Table 4).
Insulin resistance in the brain is a specific form of T2DM; however, Nigella sativa oil has a possible benefit as a disease-modifying agent for insulin resistance in the brain by suppressing oxidative stress and enhancing the brain insulin signaling pathway; multiple miRNAs are involved in this process, especially miR-34a and miR-26b [81]. This also supports the view that insulin resistance partly originates from oxidative stress [97]. In addition, in experiments on blueberry anthocyanin extract, Crataegus persica extract, polydatin, and sodium tanshinone IIA sulfonate treatments all decreased the ROS level and alleviated the oxidative stress induced by different high-concentration glucose environments [77,79,88,89]. Pieces of evidence have been accumulating regarding Sirt1 playing an important role in the cellular redox balance and resistance to oxidative stress [98,99]. Furthermore, Sirt1 can regulate nuclear factor erythroid 2-related factor 2 (Nrf2) to regulate the transcription of pro- and anti-oxidant enzymes, subsequently affecting the cellular redox state [100]. Dioscin, a natural steroid saponin isolated from various herbs [101], significantly decreased the formation of ROS and suppressed oxidative stress by regulating the miR-34a/Sirt1/Nrf2-mediated pathway in vivo and in vitro [84]. These results were consistent with the data obtained from a study on genistein which could raise anti-oxidative ability through the upregulation of Sirt1 via inhibiting miR-34a in in vitro experiments [85]. Additionally, astragaloside IV also increased cellular antioxidant capacity and alleviated high-glucose-induced cell damage, the potential mechanism of which probably owes credit to the enhanced Sirt1/Nrf2 activity induced by miR-138 [82]. Oridonin, a diterpenoid isolated from Rabdosia rubescens, attenuated hydrogen peroxide-induced oxidative stress by altering miRNAs expression; statistically, six miRNAs were upregulated, and 15 miRNAs were downregulated by using microarray analysis [87].
In general, as a negative effect produced by excess ROS in the body, oxidative stress is an important common pathogenesis of pancreatic β cell injury, which, in turn, affects insulin secretion. As antioxidants, natural products are effective at removing excess free radicals from the body and regulating oxidative stress (Figure 2). From the perspective of drug development, the study of oxidative stress can help to shed further light on the pathogenesis of abnormal glucose metabolism and provide a theoretical basis for the prevention and treatment of glucose metabolism disorders and their complications.

5. Effects of Natural Products on Cardiovascular Diseases

Cardiovascular disease is one of the major causes of death worldwide, with morbidity and mortality rising year by year [102]. Cardiovascular disease, also known as circulatory disease, is a series of diseases that involve the circulatory systems [103]. Moreover, abnormal lipid metabolism and glucose metabolism are important factors in the process of cardiovascular disease [104]. The changes experienced by using natural products in recent years are still unprecedented. Existing natural product studies have shown therapeutic effects on myocardial cell injury and protective effects on vascular endothelial cells via miRNA-mediated signaling pathways (Table 5 and Table 6).

5.1. Therapeutic Effects on Myocardial Cell Injury

Cardiomyocyte injury is closely related to the development of cardiovascular diseases, such as myocardial failure, myocardial ischemia, cardiac fibrosis, and myocardial infarction [105,106,107,108]. Increasing evidence suggests that natural products protect cardiomyocytes from various injuries by managing the expression of miRNAs.
Tanshinone IIA, the active ingredient isolated from the rhizome of the Chinese herb Salvia miltiorrhiza (also known as “Danshen” in Chinese), is an effective cardioprotective agent. Latterly, it was indicated that tanshinone IIA could protect cardiomyocytes from ischemic and hypoxic damage, which was based on downregulating the expression of miR-1 and upregulating the expression of miR-133 by activating the P38 MAPK and ERK1/2 signal pathway, respectively [109,110]. It could also modulate the overexpressed miR-1 by regulating serum response factor (SRF) [111], a transcriptional regulator of muscle-specific and growth-regulated genes, which may lower the risk of sudden cardiac death [112]. As we mentioned in the context of glucose metabolism disorders, ROS accumulation is not only an indicator of oxidative stress injury but also a marker of cardiomyocyte damage. The interventional treatment of gypenoside A, resveratrol, and portulaca oleracea extract significantly reduced ROS production and attenuated myocardial injuries; meanwhile, it exerted cardio-protective effects via miRNA-mediated signaling pathways [113,114,115], whereas dioscin inhibited myocardial oxidative insult and alleviated doxorubicin-induced cardiotoxicity via the miR-140/Sirt2/Nrf2 signaling pathway [116].
There is evidence that apoptosis is involved in the development of myocardial infarction and heart failure [117]. A test study indicated that resina draconis treatment inhibited the endoplasmic reticulum-induced apoptosis of myocardial cells via regulating the miR-423/ERK signaling pathway in a tree shrew myocardial IR model [118], whereas salvianolate treatment blocked apoptosis during myocardial infarction by downregulating miR-122 [119]. As a heart-healthy compound, it was uncovered for the first time that resveratrol (100 mg/kg/day) treatment could suppress the apoptosis of myocardium in cold-treated mice by inhibiting miR-328 expression [120]. Furthermore, curcumin could also protect cardiomyocytes against hypoxia-induced apoptosis by modulating specific protein 1, which participated in co-ordinating the transactivation of survivin, a crucial gene in regulating cell apoptosis [121], which is regulated by miR-7a/b [122]. Some other natural products or their extracts, such as puerarin, ginsenoside Rb1, theaflavin, astragalus root dry extract, and Crataegus persica extract, also contributed to the protection of various types of myocardial injury and exhibited cardio-protective effects by controlling miRNA cascades, respectively [79,123,124,125,126].
Myocardial fibrosis, a common cardiac response in a variety of forms of damage, is characterized by excessive collagen deposition and extra-cellular matrix accumulation [127]. However, celastrol, a quinone methide triterpene isolated from the root extracts of Tripterygium wilfordii (Thunder god vine) [128], could reverse these undesirable phenomena induced by downregulating miR-21 expression and inhibiting MAPK/ERK signaling in transverse aortic constriction mice [129]. Similarly, Luteolin-7-diglucuronide, a naturally occurring flavonoid glycoside found in the leaves of Basil or Verbena officinalis, also attenuated isoproterenol-induced myocardial fibrosis both at the histo-pathological and molecular levels, accompanied by regulating the expression of miRNAs, including miR-29c, miR-39c, miR-133b, and miR-21 via the TGF-β signaling pathway [130]. Astragaloside IV inhibited cardiac fibrosis by targeting the miR-135a-TRPM7-TGF-β/Smads signaling pathway [131]. Identical results were also detected in the study of panax notoginseng saponins [18]. Myocardial damage and the consequent fibrotic alterations impair the normal heart architecture and cause cardiac dysfunction (Figure 3). Fortunately, these studies provide new insight into the molecular mechanisms of natural products in the studies of cardiovascular diseases.
Table 5. The effects of natural products (extracts) on cardiovascular diseases.
Table 5. The effects of natural products (extracts) on cardiovascular diseases.
Natural Products (Extracts)Relevant miRNAsDoseAdministration MethodsExperimental ModelsTargetsObserved EffectsReferences
Açaí and red muscadine grape polyphenolics miR-126↑5–20 mg GAE/L for 30 minCell cultureHUVECsVCAM-1● Protected HUVEC against glucose-induced oxidative stress and inflammation;
● Inhibited gene expression of adhesion molecules and NF-κB activation.
[132]
Astragalus root dry extractmiR-1↓20 mg/kg/d for 7 daysIntraperitoneal injectionCVB3-treated miceCx43● Prevented the increase of immune cell infiltration and arrhythmia.[90]
Crataegus persica extractmiR-126↑300 mg/kg/d for 10 weeksGavageDiabetic rats/● Decreased elevated levels of renal oxidative stress, glomerular filtration rate, insulin sensitivity, and pathological score;
● Ameliorated myocardial ischemia-reperfusion-induced renal injury.
[79]
Panax notoginseng saponins miR-29c↑150 mg/kg/d for 20 days Intraperitoneal injectionISO-treated miceCols, Fbn1● Alleviated ISO-induced myocardial injury and fibrotic alterations;
● Cardioprotective effects.
[18]
Portulaca oleracea extractmiR-146↑
miR-let-7↑
300 mg/kg/d for 35 days GavageLipopolysaccharide treated mice/● Protected from LPS-induced neuroinflammation and memory decline through antioxidant and anti-inflammatory effects.[115]
Resina draconismiR-423↑0.25, 0.5 and 1.0 mg/mLIntramuscular injectionIschemia-reperfusion tree shrewERK● Reduced the infarct size, enhanced the superoxide dismutase expression, and downregulated the malondialdehyde concentration;
● Suppressed the ischemia-reperfusion-induced apoptosis.
[118]
SalvianolatemiR-122↓12, 24 and 48 mg/kg/d for 2 weeksIntraperitoneal injectionMyocardial infarction rats/● Induced the anti-apoptosis mechanism of cardiomyocytes.[119]
XiaoxianggoumiR-203↓10, 20, and 40 g/kg, two times one week for 16 weeksGavageEndogenous high Ang II ApoE −/− miceEts2● Reduced the atherosclerotic plaque area and serum autoantibodies against oxLDL.[133]
The up arrow means an increase, and the down arrow means a decrease.
Table 6. The effects of natural products (compounds) on cardiovascular diseases.
Table 6. The effects of natural products (compounds) on cardiovascular diseases.
Natural Products (Compounds)Relevant miRNAsDoseAdministration MethodsExperimental ModelsTargets Observed EffectsReferences
Astragaloside IV
Molecules 28 06202 i021
miR-135a↑10 mg/kg/d for 9 daysGavageIsoproterenol-treated ratsTPRM7● Inhibited cardiac fibrosis by targeting the miR-135a-TRPM7-TGF-β/Smads pathway.[131]
Berberine
Molecules 28 06202 i022
miR-133a↓1.0 g/kg/d for 8 weeksGavageSTZ-induced diabetic rats/● Improved vascular dementia;
● Improved impairments of learning and memory.
[134]
Celastrol
Molecules 28 06202 i023
miR-21↓1 mg/kg/d for 3 and 12 weeksIntraperitoneal injectionTransverse aortic constriction miceERK1/2● Attenuated TAC-induced cardiac hypertrophy;
● Reduced the increased collagen deposition and downregulated α-smooth muscle actin;
● Attenuated pathological myocardial fibrosis.
[129]
Curcumin
Molecules 28 06202 i024
miR-7a/b↑50 mg/kg/d for 1 weekGavageMyocardial infarction miceSP1● Reduced the infarct size;
● Protected against hypoxia-induced cardiac myocytes apoptosis.
[122]
Dioscin
Molecules 28 06202 i025
miR-140↓Rats treated with 60, 30, and 15 mg/kg/d for 7 days;
mice treated with 80, 40, and 20 mg/kg/d for 7 days
GavageDoxorubicin-treated rats and miceSirt2, Nrf2● Improved histopathological and electrocardiogram changes;
● Inhibited myocardial oxidative insult;
● Alleviated doxorubicin-induced cardiotoxicity.
[116]
Emodin
Molecules 28 06202 i026
miR-126↑40 mg/kg/d for 7 weeksGavageBalloon-injured carotid artery ratsWnt4, Dvl-1, β-catenin● Prevented intimal thickening via Wnt4/Dvl-1/β-catenin signaling pathway.[135]
Ginsenoside Rb1
Molecules 28 06202 i027
miR-208↓40 μM for 24 hCell cultureNeonatal rat cardiomyocytesNLK● Ameliorated cardiomyocytes apoptosis;
● Protected cardiomyocytes injuries.
[124]
Ginsenoside Rg1
Molecules 28 06202 i028
miR-23a↓150 nM for 24 hCell cultureHUVECsMET● Increased MET protein expression in a time-dependent manner;
● Induced angiogenesis by the inverse regulation of MET tyrosine kinase receptor expression.
[136]
Gypenoside A
Molecules 28 06202 i029
miR-143↓100 mg/kg 1 h before
I/R administration
GavageMyocardial I/R injured rats/● Cardio-protective effect;
● Attenuated I/R-induced injures;
● Activated AMPK signaling.
[114]
Luteolin-7-diglucuronide
Molecules 28 06202 i030
miR-29c, miR-30c, miR-133b: ↑;
miR-21↓
40 mg/kg/d for 11 daysIntraperitoneal injectionIsoproterenol-treated miceCols, elastin and Fbn1;
CTGF
● Attenuated ISO-induced myocardial fibrosis;
● Suppresses ISO-induced oxidative stress and upregulation of NADPH oxidase;
● Reduced myocardial fibrotic lesions.
[130]
Puerarin
Molecules 28 06202 i031
miR-22↓100 μM for 20 daysCell cultureThe mES cell line D3 and its transgenic cell line αPIG (clone 44)Cav3● Improved the myofibrillar alignment and sarcomere development;
● Promoted the development of t-tubules;
● Upregulated the t-tubules biogenesis-related genes.
[123]
Resveratrol
Molecules 28 06202 i032
miR-34a↓20, 50 or 100 mΜ for 48 h Cell cultureRat heart-derived H9c2 cells/● Enhanced cell viability;
● Reduced cell apoptosis;
● Protective effect on cardiomyocytes.
[113]
miR-328↓100 mg/kg/d for 8 weeksGavageCold-treated mice/● Inhibited alteration of cardiac structure;
● Improved ultrastructure of myocardium;
● Improved cardiac function;
● Suppressed cold-induced hypertension;
● Suppressed apoptosis of myocardium.
[120]
miR-29b↓0.1 mg/mL for 2 months DrinkingFbn1C1039G/+ Marfan miceBcl-2● Promoted elastin integrity and smooth muscle cell survival;
● Inhibited aortic root dilatation.
[137]
Sodium Tanshinone IIA Sulfonate
Molecules 28 06202 i033
miR-133a↓10 mg/kg/d for 3 weeksGavagePAD mice/● Improved perfusion recovery, increased capillary densities, decreased ROS level in the ischemic hindlimb in diabetic mice;
● Improved angiogenesis via inhibiting miR-133a expression and increasing GCH-1 protein levels.
[89]
Tanshinone IIA
Molecules 28 06202 i034
miR-375↓10 mg/kg for 20 weeksIntraperitoneal injectionHFD-fed ApoE-/- miceKLF4● Attenuated atherosclerosis.[138]
miR-1↓20 mg/kg/d for 3 months GavageMyocardial infarction ratsCx43● Improved the hemodynamic parameters;
● Regulated P38 MAPK pathway;
● Relieved ischemia-induced injury.
[109].
miR-1↓10 mg/kg/d for 3 months GavageMyocardial infarction ratsKCNJ2, SRF● Raised survival rates;
● Ameliorated dysfunction of IK1;
● Suppressed ischemic arrhythmias and cardiac mortality.
[111]
miR-133↑10 µM for 30 minCell cultureNeonatal rat cardiomyocytesERK1/2● Increased cell viability;
● Protected cell against apoptosis;
● Activated MAPK ERK1/2 signaling.
[110]
Theaflavin
Molecules 28 06202 i035
miR-24↑5, 10 mg/kg/d for 12 weeksGavageHFD-fed ApoE−/− miceNrf2, HO-1● Promoted the activities of antioxidant enzymes (SOD, CAT, and GSH-Px);
● Inhibited the formation of atherosclerotic plaque and the process of histological alterations in the aorta.
[125]
The up arrow means an increase, and the down arrow means a decrease.

5.2. Protective Effects on Vascular Endothelial Cells

Vascular endothelial cells form the interface between blood and tissues and are involved in physiological and pathological processes, including cardiovascular diseases [139,140]. Massive cardiovascular diseases lead to various degrees of vascular endothelial injury, which, in turn, exacerbates cardiovascular diseases. Vascular endothelial dysfunction is closely related to the development of cardiovascular diseases [141]. Vascular endothelial cells are not only the target organs of cardiovascular diseases but also the new target organs of many drugs [142,143]. Therefore, improving vascular endothelial function has been an important aspect of anti-cardiovascular drug development in recent years. Obviously, as shown in Table 5 and Table 6, natural products that regulate the expressions of miRNAs may be a better choice.
Polyphenolics from açaí and red muscadine grape ameliorated human umbilical vascular endothelial cell (HUVEC) injury by inhibiting the gene expression of adhesion molecules, including vascular cell adhesionmolecule-1 (VCAM-1), which is targeted by miR-126 [132]. Ginsenoside-Rg1, which is derived from Ginseng, was considered an agent that promotes angiogenesis because the decreased expression of miR-23a negatively regulates the angiogenic activities of HUVEC in vitro [136]. Sodium tanshinone IIA sulfonate treatment improved angiogenesis by regulating the miR-133a/GCH-1 signaling pathway in experimental peripheral arterial disease (PAD) in diabetes [89].
In addition, vascular endothelial cell injury is the initial stage of atherosclerosis [144]. According to the research, xiaoxianggou, the dried root and stem of Ficus pandurata hance var. angustifolia Cheng, Ficus panduram hane var. hoiophylla Migo, and Ficus erecta thunb. var. bcecheyana King, could reduce the area of atherosclerotic plaque by elevating miR-203 expression and reducing the expression of E26 oncogene homolog 2 (Ets2) [133], which could promote further lesion destabilization by directly affecting endothelial cell function, promoting vessel leakage and expansive neovascular growth from the adventitia into the intimal area [145]. Intimal hyperplasia has long been a major problem plaguing vascular surgery. The proliferation of vascular smooth muscle cells (VSMCs) is an important factor that causes intimal thickening [146]. Nevertheless, resveratrol improved atherosclerosis by reducing higher collagen deposition and promoting elastin integrity and VSMC survival mediated by reducing the expression of miR-29b in the Fbn1C1039G/+ Marfan mouse model [137]. There were similar results in the study on emodin; interestingly, miR-126 participated in this process by mediating the Wnt4/Dvl-1/β-catenin signaling pathway in balloon-injured carotid artery rats [135]. Berberine improved vascular dementia in diabetes, which is possibly related to the suppression of miR-133a ectopic expression in endothelial cells [134]. A key regulatory role for Krüppel-like factor 4 (KLF4) in vascular function has been shown in vitro and in vivo, and KLF4 deficiency is associated with atherothrombosis [147,148,149,150]. Tanshinone IIA harmonized the crosstalk of autophagy and polarization in macrophages via activating KLF4 mediated by miR-375 to attenuate atherosclerosis [138]. These studies set out our vision of the protective effect of natural products on vascular endothelial cells by the regulation of multiple miRNAs (Figure 3), and provide molecular evidence for further studies on natural products as novel anti-cardiovascular therapies.

6. Conclusions

In all the studies reviewed here, while natural products have provided new insights into the treatment of metabolic diseases by regulating miRNA cascades and have revealed anti-obesity, anti-diabetes, and anti-cardiovascular disease functions, as well as demonstrating a rich source of therapeutic agents, there are still some pressing issues that need to be addressed. Primarily, the mechanisms of metabolic diseases and the correlations between them are complex and still require systematic and in-depth studies beyond all doubt. Moreover, the plentiful miRNAs existing in our bodies often act together with their cluster members or other miRNAs [151]; hence, the complicated regulatory network of miRNAs can also not be ignored in natural product treatments. In addition, with a view of providing better clues for drug development, there are many natural products that have not yet been discovered in nature, of which the active substances and their effects on miRNAs still need to be further investigated.

Author Contributions

Conceptualization, J.L., L.W. and D.W.; Methodology, J.L., L.W. and D.W.; Software, H.C. and X.L.; Validation, H.C. and X.L.; Formal Analysis, J.L. and C.S.; Investigation, J.L. and C.S.; Resources, L.W. and D.W.; Data Curation, J.L.; Writing—Original Draft Preparation, J.L. and C.S.; Writing—Review and Editing, L.W. and D.W.; Visualization, J.L., H.C. and X.L.; Supervision, L.W. and D.W.; Project Administration, J.L., L.W. and D.W.; Funding Acquisition, J.L. and D.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Key R&D and Promotion Special Project of Henan Province (Technology Research, 232102320291); the Key Project of Research Fund of Xuchang University (2023ZD006); and the Talent introduction and scientific research start-up funded project of Xuchang University (20221002).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank Yan Li from School of Food Science and Technology of Jiangnan University for useful advice regarding writing the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

Abbreviations

ABCA1: ATP-binding cassette transporter A1; ABCG1, adenosine triphosphate binding cassette transporter G1; ACC, acetyl-CoA carboxylase; ACLY, ATP citrate lyase; ALT, alanine aminotransferase; AMPK, AMP-activated protein kinase; aP2, adipocyte fatty acid binding protein; APOA5, apolipoprotein A5; AST, aspartate aminotransferase; Bcl-2, B cell lymphoma 2; C/EBPα, CCAAT/enhancer-binding protein alpha; CAT, catalase; Cav3, caveolin-3; Cols, collagens; CPT1a, carnitine palmitoyltransferase 1a; CTGF, connective tissue growth factor; CVB3, coxsackievirus B3; Cx43, connexin 43; Dvl-1, dishevelled 1; ECG, (-)-Epicatechin-3-gallate; EGCG, Epigallocatechin-3-gallate; ER, endoplasmic reticulum; ERK, extracellular signal-regulated kinases; Ets2, E26 oncogene homolog 2; FABP7, fatty acid-binding protein 7; FAS, fatty acid synthase; Fbn1, fibrillin 1; FOXO1, forkhead box O1; GCH-1, GTP cyclohydrolase-1; GSH-Px, glutathione peroxidase; GSK3β, glycogen synthase kinase 3β; HDL, high density lipoproteins; HDL-C, high density lipoproteins cholesterol; HFD, high-fat diet; HMGCS, 3-hydroxy-3-methylglutaryl-CoA synthase; HO-1, heme oxygenase-1; HUVECs, human umbilical vascular endothelial cells; I/R, ischemia reperfusion injury; INSIG2, insulin-induced gene 2; IR, insulin receptor; IRS1, insulin receptor substrate 1; ISO, isoproterenol; ITT, insulin tolerance test; KCNJ2, Kir2.1 mRNA; Keap1, Kelch-like ECH-associated protein 1; KLF4, Krüppel-like factor 4; LDL-C, low density lipoproteins cholesterol; LPL, lipoprotein lipase; MAPK7, mitogen-activated protein kinase 7; MCE, mitotic clonal expansion; MCL-1, myeloid cell leukemia 1; MDA, malondialdehyde; MET, hepatocyte growth factor receptor; NASH, nonalcoholic steatohepatitis; NPC1, Niemann-Pick C1; Nrf2, nuclear factor erythroid 2-related factor 2; OGG1, 8-oxoguanine-DNA glycosylase; oxLDL, low density lipoprotein; PAD, peripheral arterial disease; PDX1, pancreatic duodenal homeobox-1; PGC1α, peroxisome proliferator-activated receptor gamma coactivator-1alpha; PGC1β, peroxisome proliferator-activated receptor gamma coactivator-1beta; PPARβ/δ, peroxisome proliferator-activated receptor beta/delta; PPARγ, peroxisome proliferator-activated receptor-gamma; PTEN, phosphatase Phosphatase and tensin homologue deleted on chromosome 10; PTP1B, protein tyrosine phosphatase 1B; ROS, reactive oxygen species; SCD1, stearoyl-coenzyme A desaturase 1; Sirt1, sirtuin type 1; SOD, superoxide dismutase; SP1, Sp1 transcription factor; SREBP1c, sterol regulatory element binding protein 1c; SRF, serum response factor; STZ, streptozotocin; T2DM, type 2 diabetes mellitus; TC, total cholesterol; TCF7L2, transcription factor 7-like 2; TG, triglyceride; TGF-β, transforming growth factor β; TRPM7, transient receptor potential melastatin 7; TRPV1, transient receptor potential cation channel subfamily V member 1; VCAM-1, vascular cell adhesionmolecule-1; VSMCs, vascular smooth muscle cells; Wnt1, wingless-type MMTV integration site family member 1; Wnt10b, wingless-type MMTV integration site family member 10b; Wnt3a, wingless-type MMTV integration site family member 3a; Wnt4, wingless-type MMTV integration site family member 4; ZEB2, zinc finger E-Box binding homeobox 2.

References

  1. Cawley, J.; Wen, K. Policies to Prevent Obesity and Promote Healthier Diets: A Critical Selective Review. Clin. Chem. 2018, 64, 163–172. [Google Scholar] [CrossRef]
  2. Unnikrishnan, R.; Pradeepa, R.; Joshi, S.R.; Mohan, V. Type 2 Diabetes: Demystifying the Global Epidemic. Diabetes 2017, 66, 1432–1442. [Google Scholar] [CrossRef] [PubMed]
  3. Kan, J.; Velliquette, R.A.; Grann, K.; Burns, C.R.; Scholten, J.; Tian, F.; Zhang, Q.; Gui, M. A novel botanical formula prevents diabetes by improving insulin resistance. BMC Complement. Altern. Med. 2017, 17, 352. [Google Scholar] [CrossRef] [PubMed]
  4. Smilowitz, N.R.; Gupta, N.; Guo, Y.; Beckman, J.A.; Bangalore, S.; Berger, J.S. Trends in cardiovascular risk factor and disease prevalence in patients undergoing non-cardiac surgery. Heart 2018, 104, 1180–1186. [Google Scholar] [CrossRef] [PubMed]
  5. Derosa, G.; Maffioli, P. Anti-obesity drugs: A review about their effects and their safety. Expert Opin. Drug Saf. 2012, 11, 459–471. [Google Scholar] [CrossRef] [PubMed]
  6. Kang, J.G.; Park, C.Y. Anti-Obesity Drugs: A Review about Their Effects and Safety. Diabetes Metab. J. 2012, 36, 13–25. [Google Scholar] [CrossRef]
  7. Dietrich, M.O.; Horvath, T.L. Limitations in anti-obesity drug development: The critical role of hunger-promoting neurons. Nat. Rev. Drug Discov. 2012, 11, 675–691. [Google Scholar] [CrossRef]
  8. Baker, D.D.; Chu, M.; Oza, U.; Rajgarhia, V. The value of natural products to future pharmaceutical discovery. Nat. Prod. Rep. 2007, 24, 1225–1244. [Google Scholar] [CrossRef]
  9. Koehn, F.E.; Carter, G.T. The evolving role of natural products in drug discovery. Nat. Rev. Drug Discov. 2005, 4, 206–220. [Google Scholar] [CrossRef]
  10. Martel, J.; Ojcius, D.M.; Chang, C.J.; Lin, C.S.; Lu, C.C.; Ko, Y.F.; Tseng, S.F.; Lai, H.C.; Young, J.D. Anti-obesogenic and antidiabetic effects of plants and mushrooms. Nat. Rev. Endocrinol. 2017, 13, 149–160. [Google Scholar] [CrossRef]
  11. Shukla, S.K.; Gupta, S.; Ojha, S.K.; Sharma, S.B. Cardiovascular friendly natural products: A promising approach in the management of CVD. Nat. Prod. Res. 2010, 24, 873–898. [Google Scholar] [CrossRef] [PubMed]
  12. Cragg, G.M.; Newman, D.J. Natural products: A continuing source of novel drug leads. Biochim. Biophys. Acta 2013, 1830, 3670–3695. [Google Scholar] [CrossRef] [PubMed]
  13. Yao, H.; Liu, J.; Xu, S.; Zhu, Z.; Xu, J. The structural modification of natural products for novel drug discovery. Expert Opin. Drug Discov. 2017, 12, 121–140. [Google Scholar] [CrossRef] [PubMed]
  14. Martello, G.; Rosato, A.; Ferrari, F.; Manfrin, A.; Cordenonsi, M.; Dupont, S.; Enzo, E.; Guzzardo, V.; Rondina, M.; Spruce, T.; et al. A MicroRNA targeting dicer for metastasis control. Cell 2010, 141, 1195–1207. [Google Scholar] [CrossRef]
  15. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef]
  16. Baselga-Escudero, L.; Pascual-Serrano, A.; Ribas-Latre, A.; Casanova, E.; Salvado, M.J.; Arola, L.; Arola-Arnal, A.; Blade, C. Long-term supplementation with a low dose of proanthocyanidins normalized liver miR-33a and miR-122 levels in high-fat diet-induced obese rats. Nutr. Res. 2015, 35, 337–345. [Google Scholar] [CrossRef]
  17. Zhang, L.; He, S.; Yang, F.; Yu, H.; Xie, W.; Dai, Q.; Zhang, D.; Liu, X.; Zhou, S.; Zhang, K. Hyperoside ameliorates glomerulosclerosis in diabetic nephropathy by downregulating miR-21. Can. J. Physiol. Pharmacol. 2016, 94, 1249–1256. [Google Scholar] [CrossRef]
  18. Liu, L.; Ning, B.; Cui, J.; Zhang, T.; Chen, Y. miR-29c is implicated in the cardioprotective activity of Panax notoginseng saponins against isoproterenol-induced myocardial fibrogenesis. J. Ethnopharmacol. 2017, 198, 1–4. [Google Scholar] [CrossRef]
  19. Tung, Y.T.; Chen, H.L.; Wu, H.S.; Ho, M.H.; Chong, K.Y.; Chen, C.M. Kefir Peptides Prevent Hyperlipidemia and Obesity in High-Fat-Diet-Induced Obese Rats via Lipid Metabolism Modulation. Mol. Nutr. Food Res. 2017, 62, 1700505. [Google Scholar] [CrossRef]
  20. Pang, D.; You, L.; Zhou, L.; Li, T.; Zheng, B.; Liu, R.H. Averrhoa carambola free phenolic extract ameliorates nonalcoholic hepatic steatosis by modulating mircoRNA-34a, mircoRNA-33 and AMPK pathways in leptin receptor-deficient db/db mice. Food Funct. 2017, 8, 4496–4507. [Google Scholar] [CrossRef]
  21. Liu, S.; Chang, X.; Yu, J.; Xu, W. Cerasus humilis Cherry Polyphenol Reduces High-Fat Diet-Induced Obesity in C57BL/6 Mice by Mitigating Fat Deposition, Inflammation, and Oxidation. J. Agric. Food Chem. 2020, 68, 4424–4436. [Google Scholar] [CrossRef] [PubMed]
  22. Su, D.; Liu, H.; Qi, X.; Dong, L.; Zhang, R.; Zhang, J. Citrus peel flavonoids improve lipid metabolism by inhibiting miR-33 and miR-122 expression in HepG2 cells. Biosci. Biotechnol. Biochem. 2019, 83, 1747–1755. [Google Scholar] [CrossRef] [PubMed]
  23. Murase, T.; Misawa, K.; Minegishi, Y.; Aoki, M.; Ominami, H.; Suzuki, Y.; Shibuya, Y.; Hase, T. Coffee polyphenols suppress diet-induced body fat accumulation by downregulating SREBP-1c and related molecules in C57BL/6J mice. Am. J. Physiol. Endocrinol. Metab. 2011, 300, E122–E133. [Google Scholar] [CrossRef] [PubMed]
  24. Kim, S.; Lee, M.S.; Jung, S.; Son, H.Y.; Park, S.; Kang, B.; Kim, S.Y.; Kim, I.H.; Kim, C.T.; Kim, Y. Ginger Extract Ameliorates Obesity and Inflammation via Regulating MicroRNA-21/132 Expression and AMPK Activation in White Adipose Tissue. Nutrients 2018, 10, 1567. [Google Scholar] [CrossRef]
  25. Baselga-Escudero, L.; Blade, C.; Ribas-Latre, A.; Casanova, E.; Salvado, M.J.; Arola, L.; Arola-Arnal, A. Chronic supplementation of proanthocyanidins reduces postprandial lipemia and liver miR-33a and miR-122 levels in a dose-dependent manner in healthy rats. J. Nutr. Biochem. 2014, 25, 151–156. [Google Scholar] [CrossRef]
  26. Baselga-Escudero, L.; Blade, C.; Ribas-Latre, A.; Casanova, E.; Salvado, M.J.; Arola, L.; Arola-Arnal, A. Grape seed proanthocyanidins repress the hepatic lipid regulators miR-33 and miR-122 in rats. Mol. Nutr. Food Res. 2012, 56, 1636–1646. [Google Scholar] [CrossRef]
  27. Baselga-Escudero, L.; Arola-Arnal, A.; Pascual-Serrano, A.; Ribas-Latre, A.; Casanova, E.; Salvado, M.J.; Arola, L.; Blade, C. Chronic administration of proanthocyanidins or docosahexaenoic acid reverses the increase of miR-33a and miR-122 in dyslipidemic obese rats. PLoS ONE 2013, 8, e69817. [Google Scholar] [CrossRef]
  28. Shi, Y.; Jia, M.; Xu, L.; Fang, Z.; Wu, W.; Zhang, Q.; Chung, P.; Lin, Y.; Wang, S.; Zhang, Y. miR-96 and autophagy are involved in the beneficial effect of grape seed proanthocyanidins against high-fat-diet-induced dyslipidemia in mice. Phytother. Res. 2019, 33, 1222–1232. [Google Scholar] [CrossRef]
  29. Lu, R.H.; Qin, C.B.; Yang, F.; Zhang, W.Y.; Zhang, Y.R.; Yang, G.K.; Yang, L.P.; Meng, X.L.; Yan, X.; Nie, G.X. Grape seed proanthocyanidin extract ameliorates hepatic lipid accumulation and inflammation in grass carp (Ctenopharyngodon idella). Fish Physiol. Biochem. 2020, 46, 1665–1677. [Google Scholar] [CrossRef]
  30. Torres, L.F.; Cogliati, B.; Otton, R. Green Tea Prevents NAFLD by Modulation of miR-34a and miR-194 Expression in a High-Fat Diet Mouse Model. Oxid. Med. Cell. Longev. 2019, 2019, 4168380. [Google Scholar] [CrossRef]
  31. Otton, R.; Bolin, A.P.; Ferreira, L.T.; Marinovic, M.P.; Rocha, A.L.S.; Mori, M.A. Polyphenol-rich green tea extract improves adipose tissue metabolism by down-regulating miR-335 expression and mitigating insulin resistance and inflammation. J. Nutr. Biochem. 2018, 57, 170–179. [Google Scholar] [CrossRef]
  32. Lima, N.D.S.; Numata, E.P.; Mesquita, L.M.S.; Dias, P.H.; Vilegas, W.; Gambero, A.; Ribeiro, M.L. Modulatory Effects of Guarana (Paullinia cupana) on Adipogenesis. Nutrients 2017, 9, 635. [Google Scholar] [CrossRef]
  33. Su, D.; Zhang, R.; Hou, F.; Chi, J.; Huang, F.; Yan, S.; Liu, L.; Deng, Y.; Wei, Z.; Zhang, M. Lychee pulp phenolics ameliorate hepatic lipid accumulation by reducing miR-33 and miR-122 expression in mice fed a high-fat diet. Food Funct. 2017, 8, 808–815. [Google Scholar] [CrossRef]
  34. Lee, S.; Lee, M.S.; Chang, E.; Lee, Y.; Lee, J.; Kim, J.; Kim, C.T.; Kim, I.H.; Kim, Y. Mulberry Fruit Extract Promotes Serum HDL-Cholesterol Levels and Suppresses Hepatic microRNA-33 Expression in Rats Fed High Cholesterol/Cholic Acid Diet. Nutrients 2020, 12, 1499. [Google Scholar] [CrossRef] [PubMed]
  35. Yang, T.Y.; Yu, M.H.; Wu, Y.L.; Hong, C.C.; Chen, C.S.; Chan, K.C.; Wang, C.J. Mulberry Leaf (Morus alba L.) Extracts and Its Chlorogenic Acid Isomer Component Improve Glucolipotoxicity-Induced Hepatic Lipid Accumulation via Downregulating miR-34a and Decreased Inflammation. Nutrients 2022, 14, 4808. [Google Scholar] [CrossRef]
  36. Monraz-Méndez, C.A.; Escutia-Gutiérrez, R.; Rodriguez-Sanabria, J.S.; Galicia-Moreno, M.; Monroy-Ramírez, H.C.; Sánchez-Orozco, L.; García-Bañuelos, J.; de la Rosa-Bibiano, R.; Santos, A.; Armendáriz-Borunda, J.; et al. Moringa oleifera Improves MAFLD by Inducing Epigenetic Modifications. Nutrients 2022, 14, 4225. [Google Scholar] [CrossRef]
  37. Qiao, J.Y.; Li, H.W.; Liu, F.G.; Li, Y.C.; Tian, S.; Cao, L.H.; Hu, K.; Wu, X.X.; Miao, M.S. Effects of Portulaca Oleracea Extract on Acute Alcoholic Liver Injury of Rats. Molecules 2019, 24, 2887. [Google Scholar] [CrossRef] [PubMed]
  38. Jang, S.; Lee, M.S.; Kang, S.A.; Kim, C.T.; Kim, Y. Portulaca oleracea L. Extract Regulates Hepatic Cholesterol Metabolism via the AMPK/MicroRNA-33/34a Pathway in Rats Fed a High-Cholesterol Diet. Nutrients 2022, 14, 3330. [Google Scholar] [CrossRef]
  39. Stefanon, B.; Pomari, E.; Colitti, M. Effects of Rosmarinus officinalis extract on human primary omental preadipocytes and adipocytes. Exp. Biol. Med. 2015, 240, 884–895. [Google Scholar] [CrossRef] [PubMed]
  40. Zhu, W.; Zou, B.; Nie, R.; Zhang, Y.; Li, C.M. A-type ECG and EGCG dimers disturb the structure of 3T3-L1 cell membrane and strongly inhibit its differentiation by targeting peroxisome proliferator-activated receptor gamma with miR-27 involved mechanism. J. Nutr. Biochem. 2015, 26, 1124–1135. [Google Scholar] [CrossRef] [PubMed]
  41. Tian, L.; Song, Z.; Shao, W.; Du, W.W.; Zhao, L.R.; Zeng, K.; Yang, B.B.; Jin, T. Curcumin represses mouse 3T3-L1 cell adipogenic differentiation via inhibiting miR-17-5p and stimulating the Wnt signalling pathway effector Tcf7l2. Cell Death Dis. 2017, 8, e2559. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, J.; Huang, Y.; Shao, H.; Bi, Q.; Chen, J.; Ye, Z. Grape seed procyanidin B2 inhibits adipogenesis of 3T3-L1 cells by targeting peroxisome proliferator-activated receptor gamma with miR-483-5p involved mechanism. Biomed. Pharmacother. 2017, 86, 292–296. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, C.P.; Su, T.C.; Yang, M.J.; Chen, W.T.; Siao, A.C.; Huang, L.R.; Lin, Y.Y.; Kuo, Y.C.; Chung, J.F.; Cheng, C.F.; et al. Green tea epigallocatechin gallate suppresses 3T3-L1 cell growth via microRNA-143/MAPK7 pathways. Exp. Biol. Med. 2022, 247, 1670–1679. [Google Scholar] [CrossRef]
  44. Ahn, J.; Lee, H.; Jung, C.H.; Ha, T. Lycopene inhibits hepatic steatosis via microRNA-21-induced downregulation of fatty acid-binding protein 7 in mice fed a high-fat diet. Mol. Nutr. Food Res. 2012, 56, 1665–1674. [Google Scholar] [CrossRef]
  45. Rohm, B.; Holik, A.K.; Kretschy, N.; Somoza, M.M.; Ley, J.P.; Widder, S.; Krammer, G.E.; Marko, D.; Somoza, V. Nonivamide enhances miRNA let-7d expression and decreases adipogenesis PPARgamma expression in 3T3-L1 cells. J. Cell. Biochem. 2015, 116, 1153–1163. [Google Scholar] [CrossRef]
  46. Chen, S.; Wen, X.; Zhang, W.; Wang, C.; Liu, J.; Liu, C. Hypolipidemic effect of oleanolic acid is mediated by the miR-98-5p/PGC-1beta axis in high-fat diet-induced hyperlipidemic mice. FASEB J. 2017, 31, 1085–1096. [Google Scholar] [CrossRef] [PubMed]
  47. Zou, B.; Ge, Z.; Zhu, W.; Xu, Z.; Li, C. Persimmon tannin represses 3T3-L1 preadipocyte differentiation via up-regulating expression of miR-27 and down-regulating expression of peroxisome proliferator-activated receptor-gamma in the early phase of adipogenesis. Eur. J. Nutr. 2015, 54, 1333–1343. [Google Scholar] [CrossRef]
  48. Gai, Y.; Li, Y.; Xu, Z.; Chen, J. Pseudoprotodioscin inhibits SREBPs and microRNA 33a/b levels and reduces the gene expression regarding the synthesis of cholesterol and triglycerides. Fitoterapia 2019, 139, 104393. [Google Scholar] [CrossRef] [PubMed]
  49. Gracia, A.; Fernandez-Quintela, A.; Miranda, J.; Eseberri, I.; Gonzalez, M.; Portillo, M.P. Are miRNA-103, miRNA-107 and miRNA-122 Involved in the Prevention of Liver Steatosis Induced by Resveratrol? Nutrients 2017, 9, 360. [Google Scholar] [CrossRef]
  50. Gracia, A.; Miranda, J.; Fernandez-Quintela, A.; Eseberri, I.; Garcia-Lacarte, M.; Milagro, F.I.; Martinez, J.A.; Aguirre, L.; Portillo, M.P. Involvement of miR-539-5p in the inhibition of de novo lipogenesis induced by resveratrol in white adipose tissue. Food Funct. 2016, 7, 1680–1688. [Google Scholar] [CrossRef]
  51. Eseberri, I.; Lasa, A.; Miranda, J.; Gracia, A.; Portillo, M.P. Potential miRNA involvement in the anti-adipogenic effect of resveratrol and its metabolites. PLoS ONE 2017, 12, e0184875. [Google Scholar] [CrossRef] [PubMed]
  52. Ahn, J.; Lee, H.; Jung, C.H.; Choi, W.H.; Ha, T.Y. Zerumbone ameliorates high-fat diet-induced adiposity by restoring AMPK-regulated lipogenesis and microRNA-146b/SIRT1-mediated adipogenesis. Oncotarget 2017, 8, 36984–36995. [Google Scholar] [CrossRef] [PubMed]
  53. Rottiers, V.; Naar, A.M. MicroRNAs in metabolism and metabolic disorders. Nat. Rev. Mol. Cell Biol. 2012, 13, 239–250. [Google Scholar] [CrossRef]
  54. Moore, K.J.; Rayner, K.J.; Suarez, Y.; Fernandez-Hernando, C. The role of microRNAs in cholesterol efflux and hepatic lipid metabolism. Annu. Rev. Nutr. 2011, 31, 49–63. [Google Scholar] [CrossRef] [PubMed]
  55. Qiu, C.P.; Lv, Q.T.; Dongol, S.; Wang, C.; Jiang, J. Single nucleotide polymorphism of SREBF-1 gene associated with an increased risk of endometrial cancer in Chinese women. PLoS ONE 2014, 9, e90491. [Google Scholar] [CrossRef]
  56. Horton, J.D.; Goldstein, J.L.; Brown, M.S. SREBPs: Activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Investig. 2002, 109, 1125–1131. [Google Scholar] [CrossRef]
  57. Nagai, Y.; Yonemitsu, S.; Erion, D.M.; Iwasaki, T.; Stark, R.; Weismann, D.; Dong, J.; Zhang, D.; Jurczak, M.J.; Loffler, M.G.; et al. The role of peroxisome proliferator-activated receptor gamma coactivator-1 beta in the pathogenesis of fructose-induced insulin resistance. Cell Metab. 2009, 9, 252–264. [Google Scholar] [CrossRef]
  58. Bennett, M.K.; Lopez, J.M.; Sanchez, H.B.; Osborne, T.F. Sterol regulation of fatty acid synthase promoter. Coordinate feedback regulation of two major lipid pathways. J. Biol. Chem. 1995, 270, 25578–25583. [Google Scholar] [CrossRef]
  59. Sullivan, J.E.; Brocklehurst, K.J.; Marley, A.E.; Carey, F.; Carling, D.; Beri, R.K. Inhibition of lipolysis and lipogenesis in isolated rat adipocytes with AICAR, a cell-permeable activator of AMP-activated protein kinase. FEBS Lett. 1994, 353, 33–36. [Google Scholar] [CrossRef]
  60. Daval, M.; Foufelle, F.; Ferre, P. Functions of AMP-activated protein kinase in adipose tissue. J. Physiol. 2006, 574, 55–62. [Google Scholar] [CrossRef]
  61. Saha, A.K.; Schwarsin, A.J.; Roduit, R.; Masse, F.; Kaushik, V.; Tornheim, K.; Prentki, M.; Ruderman, N.B. Activation of malonyl-CoA decarboxylase in rat skeletal muscle by contraction and the AMP-activated protein kinase activator 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside. J. Biol. Chem. 2000, 275, 24279–24283. [Google Scholar] [CrossRef]
  62. Bengestrate, L.; Virtue, S.; Campbell, M.; Vidal-Puig, A.; Hadaschik, D.; Hahn, P.; Bielke, W. Genome-wide profiling of microRNAs in adipose mesenchymal stem cell differentiation and mouse models of obesity. PLoS ONE 2011, 6, e21305. [Google Scholar] [CrossRef] [PubMed]
  63. Tang, Q.Q.; Lane, M.D. Adipogenesis: From stem cell to adipocyte. Annu. Rev. Biochem. 2012, 81, 715–736. [Google Scholar] [CrossRef] [PubMed]
  64. Gregoire, F.M.; Smas, C.M.; Sul, H.S. Understanding adipocyte differentiation. Physiol. Rev. 1998, 78, 783–809. [Google Scholar] [CrossRef]
  65. Nickels, J.D.; Chatterjee, S.; Stanley, C.B.; Qian, S.; Cheng, X.; Myles, D.A.A.; Standaert, R.F.; Elkins, J.G.; Katsaras, J. The in vivo structure of biological membranes and evidence for lipid domains. PLoS Biol. 2017, 15, e2002214. [Google Scholar] [CrossRef] [PubMed]
  66. Rosen, E.D.; MacDougald, O.A. Adipocyte differentiation from the inside out. Nat. Rev. Mol. Cell Biol. 2006, 7, 885–896. [Google Scholar] [CrossRef]
  67. Price, N.L.; Fernandez-Hernando, C. miRNA regulation of white and brown adipose tissue differentiation and function. Biochim. Biophys. Acta 2016, 1861, 2104–2110. [Google Scholar] [CrossRef] [PubMed]
  68. Chen, Q.; Shou, P.; Zheng, C.; Jiang, M.; Cao, G.; Yang, Q.; Cao, J.; Xie, N.; Velletri, T.; Zhang, X.; et al. Fate decision of mesenchymal stem cells: Adipocytes or osteoblasts? Cell Death Differ. 2016, 23, 1128–1139. [Google Scholar] [CrossRef] [PubMed]
  69. Ross, S.E.; Hemati, N.; Longo, K.A.; Bennett, C.N.; Lucas, P.C.; Erickson, R.L.; MacDougald, O.A. Inhibition of adipogenesis by Wnt signaling. Science 2000, 289, 950–953. [Google Scholar] [CrossRef]
  70. Dogan, A.; Demirci, S.; Apdik, H.; Bayrak, O.F.; Gulluoglu, S.; Tuysuz, E.C.; Gusev, O.; Rizvanov, A.A.; Nikerel, E.; Sahin, F. A new hope for obesity management: Boron inhibits adipogenesis in progenitor cells through the Wnt/beta-catenin pathway. Metabolism 2017, 69, 130–142. [Google Scholar] [CrossRef]
  71. Christodoulides, C.; Lagathu, C.; Sethi, J.K.; Vidal-Puig, A. Adipogenesis and WNT signalling. Trends Endocrinol. Metab. 2009, 20, 16–24. [Google Scholar] [CrossRef] [PubMed]
  72. Cuadros, D.F.; Li, J.; Musuka, G.; Awad, S.F. Spatial epidemiology of diabetes: Methods and insights. World J. Diabetes 2021, 12, 1042–1056. [Google Scholar] [CrossRef] [PubMed]
  73. Cho, N.H.; Shaw, J.E.; Karuranga, S.; Huang, Y.; da Rocha Fernandes, J.D.; Ohlrogge, A.W.; Malanda, B. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res. Clin. Pract. 2018, 138, 271–281. [Google Scholar] [CrossRef] [PubMed]
  74. Alam, U.; Asghar, O.; Azmi, S.; Malik, R.A. General aspects of diabetes mellitus. Handb. Clin. Neurol. 2014, 126, 211–222. [Google Scholar] [PubMed]
  75. Kim, A.; Lee, W.; Yun, J.M. Luteolin and fisetin suppress oxidative stress by modulating sirtuins and forkhead box O3a expression under in vitro diabetic conditions. Nutr. Res. Pract. 2017, 11, 430–434. [Google Scholar] [CrossRef]
  76. Du, G.; Xiao, M.; Zhang, X.; Wen, M.; Pang, C.; Jiang, S.; Sang, S.; Xie, Y. Alpinia oxyphylla Miq. extract changes miRNA expression profiles in db-/db- mouse kidney. Biol. Res. 2017, 50, 9. [Google Scholar] [CrossRef]
  77. Wang, C.; Wang, K.; Li, P. Blueberry anthocyanins extract attenuated diabetic retinopathy by inhibiting endoplasmic reticulum stress via the miR-182/OGG1 axis. J. Pharmacol. Sci. 2022, 150, 31–40. [Google Scholar] [CrossRef]
  78. Yu, S.; Zhao, H.; Yang, W.; Amat, R.; Peng, J.; Li, Y.; Deng, K.; Mao, X.; Jiao, Y. The Alcohol Extract of Coreopsis tinctoria Nutt Ameliorates Diabetes and Diabetic Nephropathy in db/db Mice through miR-192/miR-200b and PTEN/AKT and ZEB2/ECM Pathways. Biomed. Res. Int. 2019, 2019, 5280514. [Google Scholar] [CrossRef] [PubMed]
  79. Asgari, M.; Salehi, I.; Ranjbar, K.; Khosravi, M.; Zarrinkalam, E. Interval training and Crataegus persica ameliorate diabetic nephropathy via miR-126/Nrf-2 mediated inhibition of stress oxidative in rats with diabetes after myocardial ischemia-reperfusion injury. Biomed. Pharmacother. 2022, 153, 113411. [Google Scholar] [CrossRef]
  80. Yang, Y.M.; Seo, S.Y.; Kim, T.H.; Kim, S.G. Decrease of microRNA-122 causes hepatic insulin resistance by inducing protein tyrosine phosphatase 1B, which is reversed by licorice flavonoid. Hepatology 2012, 56, 2209–2220. [Google Scholar] [CrossRef]
  81. Balbaa, M.; Abdulmalek, S.A.; Khalil, S. Oxidative stress and expression of insulin signaling proteins in the brain of diabetic rats: Role of Nigella sativa oil and antidiabetic drugs. PLoS ONE 2017, 12, e0172429. [Google Scholar] [CrossRef]
  82. Tang, X.; Li, X.; Zhang, D.; Han, W. Astragaloside-IV alleviates high glucose-induced ferroptosis in retinal pigment epithelial cells by disrupting the expression of miR-138-5p/Sirt1/Nrf2. Bioengineered 2022, 13, 8240–8254. [Google Scholar] [CrossRef] [PubMed]
  83. Ding, X.Q.; Gu, T.T.; Wang, W.; Song, L.; Chen, T.Y.; Xue, Q.C.; Zhou, F.; Li, J.M.; Kong, L.D. Curcumin protects against fructose-induced podocyte insulin signaling impairment through upregulation of miR-206. Mol. Nutr. Food Res. 2015, 59, 2355–2370. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, Y.M.; Tao, X.F.; Yin, L.H.; Xu, L.; Xu, Y.W.; Qi, Y.; Han, X.; Song, S.S.; Zhao, Y.Y.; Lin, Y.; et al. Protective effects of dioscin against cisplatin-induced nephrotoxicity via the microRNA-34a/sirtuin 1 signalling pathway. Br. J. Pharmacol. 2017, 174, 2512–2527. [Google Scholar] [CrossRef]
  85. Zhang, H.; Zhao, Z.; Pang, X.; Yang, J.; Yu, H.; Zhang, Y.; Zhou, H.; Zhao, J. MiR-34a/sirtuin-1/foxo3a is involved in genistein protecting against ox-LDL-induced oxidative damage in HUVECs. Toxicol. Lett. 2017, 277, 115–122. [Google Scholar] [CrossRef]
  86. Li, Y. Gypenoside A attenuates dysfunction of pancreatic β cells by activating PDX1 signal transduction via the inhibition of miR-150-3p both in vivo and in vitro. J. Biochem. Mol. Toxicol. 2022, 36, e23004. [Google Scholar] [CrossRef]
  87. Bae, S.; Lee, E.J.; Lee, J.H.; Park, I.C.; Lee, S.J.; Hahn, H.J.; Ahn, K.J.; An, S.; An, I.S.; Cha, H.J. Oridonin protects HaCaT keratinocytes against hydrogen peroxide-induced oxidative stress by altering microRNA expression. Int. J. Mol. Med. 2014, 33, 185–193. [Google Scholar] [CrossRef] [PubMed]
  88. Zhao, X.J.; Yu, H.W.; Yang, Y.Z.; Wu, W.Y.; Chen, T.Y.; Jia, K.K.; Kang, L.L.; Jiao, R.Q.; Kong, L.D. Polydatin prevents fructose-induced liver inflammation and lipid deposition through increasing miR-200a to regulate Keap1/Nrf2 pathway. Redox Biol. 2018, 18, 124–137. [Google Scholar] [CrossRef] [PubMed]
  89. Chen, L.; He, W.; Peng, B.; Yuan, M.; Wang, N.; Wang, J.; Lu, W.; Wang, T. Sodium Tanshinone IIA sulfonate improves post-ischemic angiogenesis in hyperglycemia. Biochem. Biophys. Res. Commun. 2019, 520, 580–585. [Google Scholar] [CrossRef]
  90. Xu, F.; Liu, Y.; Zhu, X.; Li, S.; Shi, X.; Li, Z.; Ai, M.; Sun, J.; Hou, B.; Cai, W.; et al. Protective Effects and Mechanisms of Vaccarin on Vascular Endothelial Dysfunction in Diabetic Angiopathy. Int. J. Mol. Sci. 2019, 20, 4587. [Google Scholar] [CrossRef]
  91. Azimova, K.; Rude, J.; Mallawaarachchi, I.; Dwivedi, A.; Sarosiek, J.; Mukherjee, D. Glucose levels and depression in Hispanic patients admitted to the cardiovascular intensive care unit: A cross-sectional study. Angiology 2015, 66, 57–64. [Google Scholar] [CrossRef] [PubMed]
  92. Stanisic, J.; Koricanac, G.; Culafic, T.; Romic, S.; Stojiljkovic, M.; Kostic, M.; Pantelic, M.; Tepavcevic, S. Low intensity exercise prevents disturbances in rat cardiac insulin signaling and endothelial nitric oxide synthase induced by high fructose diet. Mol. Cell. Endocrinol. 2016, 420, 97–104. [Google Scholar] [CrossRef] [PubMed]
  93. Goldstein, B.J. Regulation of insulin receptor signaling by protein-tyrosine dephosphorylation. Receptor 1993, 3, 1–15. [Google Scholar] [PubMed]
  94. Evans, J.L.; Goldfine, I.D.; Maddux, B.A.; Grodsky, G.M. Are oxidative stress-activated signaling pathways mediators of insulin resistance and beta-cell dysfunction? Diabetes 2003, 52, 1–8. [Google Scholar] [CrossRef] [PubMed]
  95. Mao, X.; Gu, C.; Chen, D.; Yu, B.; He, J. Oxidative stress-induced diseases and tea polyphenols. Oncotarget 2017, 8, 81649–81661. [Google Scholar] [CrossRef]
  96. Keane, K.N.; Cruzat, V.F.; Carlessi, R.; de Bittencourt, P.I.H., Jr.; Newsholme, P. Molecular Events Linking Oxidative Stress and Inflammation to Insulin Resistance and beta-Cell Dysfunction. Oxid. Med. Cell. Longev. 2015, 2015, 181643. [Google Scholar] [CrossRef]
  97. Guo, Q.; Xu, L.; Li, H.; Sun, H.; Liu, J.; Wu, S.; Zhou, B. Progranulin causes adipose insulin resistance via increased autophagy resulting from activated oxidative stress and endoplasmic reticulum stress. Lipids Health Dis. 2017, 16, 25. [Google Scholar] [CrossRef]
  98. Ma, L.; Li, Y. SIRT1: Role in cardiovascular biology. Clin. Chim. Acta 2015, 440, 8–15. [Google Scholar] [CrossRef]
  99. Jamal, J.; Mustafa, M.R.; Wong, P.F. Paeonol protects against premature senescence in endothelial cells by modulating Sirtuin 1 pathway. J. Ethnopharmacol. 2014, 154, 428–436. [Google Scholar] [CrossRef]
  100. Do, M.T.; Kim, H.G.; Choi, J.H.; Jeong, H.G. Metformin induces microRNA-34a to downregulate the Sirt1/Pgc-1alpha/Nrf2 pathway, leading to increased susceptibility of wild-type p53 cancer cells to oxidative stress and therapeutic agents. Free Radic. Biol. Med. 2014, 74, 21–34. [Google Scholar]
  101. Yin, L.H.; Xu, Y.W.; Qi, Y.; Han, X.; Xu, L.N.; Peng, J.Y.; Sun, C.K. A green and efficient protocol for industrial-scale preparation of dioscin from Dioscorea nipponica Makino by two-step macroporous resin column chromatography. Chem. Eng. J. 2010, 165, 281–289. [Google Scholar] [CrossRef]
  102. Chen, W.; Gao, R.; Liu, L.; Zhu, M.; Wang, W.; Wang, Y.; Wu, Z.; Li, H.; Zheng, Z.; Jiang, L.; et al. Outline of the report on cardiovascular diseases in China, 2014. Eur. Heart J. Suppl. 2016, 18, F2–F11. [Google Scholar]
  103. Le Bouc, Y.; Brioude, F. Is there a relationship between the growth hormone dose and tumoral or cardiovascular complications? Bull. Acad. Natl. Med. 2012, 196, 127–135. [Google Scholar] [PubMed]
  104. Sadowska, J.; Bruszkowska, M. Comparing the effects of sucrose and high-fructose corn syrup on lipid metabolism and the risk of cardiovascular disease in male rats. Acta Sci. Pol. Technol. Aliment. 2017, 16, 231–240. [Google Scholar] [PubMed]
  105. Traverse, J.H.; Henry, T.D. Myocardial Injury as a New Target for Cell Therapy in Patients with Chronic Heart Failure When Something Bad Is Actually Good? Circ. Res. 2017, 120, 1857–1859. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, H.; Jing, X.; Dong, A.; Bai, B.; Wang, H. Overexpression of TIMP3 Protects Against Cardiac Ischemia/Reperfusion Injury by Inhibiting Myocardial Apoptosis through ROS/Mapks Pathway. Cell. Physiol. Biochem. 2017, 44, 1011–1023. [Google Scholar] [CrossRef] [PubMed]
  107. Zhang, X.; Pan, L.; Yang, K.; Fu, Y.; Liu, Y.; Chi, J.; Zhang, X.; Hong, S.; Ma, X.; Yin, X. H3 Relaxin Protects Against Myocardial Injury in Experimental Diabetic Cardiomyopathy by Inhibiting Myocardial Apoptosis, Fibrosis and Inflammation. Cell. Physiol. Biochem. 2017, 43, 1311–1324. [Google Scholar] [CrossRef]
  108. Li, F.; Zong, J.; Zhang, H.; Zhang, P.; Xu, L.; Liang, K.; Yang, L.; Yong, H.; Qian, W. Orientin Reduces Myocardial Infarction Size via eNOS/NO Signaling and thus Mitigates Adverse Cardiac Remodeling. Front. Pharmacol. 2017, 8, 926. [Google Scholar] [CrossRef]
  109. Zhang, Y.; Zhang, L.; Chu, W.F.; Wang, B.; Zhang, J.L.; Zhao, M.; Li, X.L.; Li, B.X.; Lu, Y.J.; Yang, B.F.; et al. Tanshinone IIA Inhibits miR-1 Expression through p38 MAPK Signal Pathway in Post-infarction Rat Cardiomyocytes. Cell. Physiol. Biochem. 2010, 26, 991–998. [Google Scholar] [CrossRef]
  110. Zhang, L.; Wu, Y.; Li, Y.; Xu, C.; Li, X.; Zhu, D.; Zhang, Y.; Xing, S.; Wang, H.; Zhang, Z.; et al. Tanshinone IIA improves miR-133 expression through MAPK ERK1/2 pathway in hypoxic cardiac myocytes. Cell. Physiol. Biochem. 2012, 30, 843–852. [Google Scholar] [CrossRef]
  111. Shan, H.; Li, X.; Pan, Z.; Zhang, L.; Cai, B.; Zhang, Y.; Xu, C.; Chu, W.; Qiao, G.; Li, B.; et al. Tanshinone IIA protects against sudden cardiac death induced by lethal arrhythmias via repression of microRNA-1. Br. J. Pharmacol. 2009, 158, 1227–1235. [Google Scholar] [CrossRef]
  112. Miano, J.M.; Ramanan, N.; Georger, M.A.; de Mesy Bentley, K.L.; Emerson, R.L.; Balza, R.O., Jr.; Xiao, Q.; Weiler, H.; Ginty, D.D.; Misra, R.P. Restricted inactivation of serum response factor to the cardiovascular system. Proc. Natl. Acad. Sci. USA 2004, 101, 17132–17137. [Google Scholar] [CrossRef]
  113. Yang, B.; Ma, S.; Wang, Y.B.; Xu, B.; Zhao, H.; He, Y.Y.; Li, C.W.; Zhang, J.; Cao, Y.K.; Feng, Q.Z. Resveratrol exerts protective effects on anoxia/reoxygenation injury in cardiomyocytes via miR-34a/Sirt1 signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2734–2741. [Google Scholar]
  114. Chang, L.; Shi, R.; Wang, X.; Bao, Y. Gypenoside A protects ischemia/reperfusion injuries by suppressing miR-143-3p level via the activation of AMPK/Foxo1 pathway. Biofactors 2020, 46, 432–440. [Google Scholar] [CrossRef] [PubMed]
  115. Hussein, R.M.; Youssef, A.M.; Magharbeh, M.K.; Al-Dalaen, S.M.; Al-Jawabri, N.A.; Al-Nawaiseh, T.N.; Al-Jwanieh, A.; Al-Ani, F.S. Protective Effect of Portulaca oleracea Extract Against Lipopolysaccharide-Induced Neuroinflammation, Memory Decline, and Oxidative Stress in Mice: Potential Role of miR-146a and miR-let 7. J. Med. Food 2022, 25, 807–817. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, L.; Tao, X.; Qi, Y.; Xu, L.; Yin, L.; Peng, J. Protective effect of dioscin against doxorubicin-induced cardiotoxicity via adjusting microRNA-140-5p-mediated myocardial oxidative stress. Redox Biol. 2018, 16, 189–198. [Google Scholar] [CrossRef]
  117. Qin, F.; Siwik, D.A.; Pimentel, D.R.; Morgan, R.J.; Biolo, A.; Tu, V.H.; Kang, Y.J.; Cohen, R.A.; Colucci, W.S. Cytosolic H2O2 mediates hypertrophy, apoptosis, and decreased SERCA activity in mice with chronic hemodynamic overload. Am. J. Physiol. Heart Circ. Physiol. 2014, 306, H1453–H1463. [Google Scholar] [CrossRef] [PubMed]
  118. Yang, T.R.; Zhang, T.; Mu, N.H.; Ruan, L.B.; Duan, J.L.; Zhang, R.P.; Miao, Y.B. Resina draconis inhibits the endoplasmic-reticulum-induced apoptosis of myocardial cells via regulating miR-423-3p/ERK signaling pathway in a tree shrew myocardial ischemia- reperfusion model. J. Biosci. 2019, 44, 53. [Google Scholar] [CrossRef] [PubMed]
  119. Lin, J.; Zheng, X. Salvianolate Blocks Apoptosis During Myocardial Infarction by Down Regulating miR-122-5p. Curr. Neurovasc. Res. 2017, 14, 323–329. [Google Scholar] [CrossRef]
  120. Yin, K.; Zhao, L.; Feng, D.; Ma, W.; Liu, Y.; Wang, Y.; Liang, J.; Yang, F.; Bi, C.; Chen, H.; et al. Resveratrol Attenuated Low Ambient Temperature-Induced Myocardial Hypertrophy via Inhibiting Cardiomyocyte Apoptosis. Cell. Physiol. Biochem. 2015, 35, 2451–2462. [Google Scholar] [CrossRef]
  121. Lee, B.S.; Oh, J.; Kang, S.K.; Park, S.; Lee, S.H.; Choi, D.; Chung, J.H.; Chung, Y.W.; Kang, S.M. Insulin Protects Cardiac Myocytes from Doxorubicin Toxicity by Sp1-Mediated Transactivation of Survivin. PLoS ONE 2015, 10, e0135438. [Google Scholar] [CrossRef]
  122. Geng, H.H.; Li, R.; Su, Y.M.; Xiao, J.; Pan, M.; Cai, X.X.; Ji, X.P. Curcumin protects cardiac myocyte against hypoxia-induced apoptosis through upregulating miR-7a/b expression. Biomed. Pharmacother. 2016, 81, 258–264. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, L.; Cui, Y.; Tang, M.; Hu, X.; Luo, H.; Hescheler, J.; Xi, J. Puerarin facilitates T-tubule development of murine embryonic stem cell-derived cardiomyocytes. Cell. Physiol. Biochem. 2014, 34, 383–392. [Google Scholar] [CrossRef]
  124. Yan, X.; Liu, J.; Wu, H.; Liu, Y.; Zheng, S.; Zhang, C.; Yang, C. Impact of miR-208 and its Target Gene Nemo-Like Kinase on the Protective Effect of Ginsenoside Rb1 in Hypoxia/Ischemia Injuried Cardiomyocytes. Cell. Physiol. Biochem. 2016, 39, 1187–1195. [Google Scholar] [CrossRef] [PubMed]
  125. Zeng, J.; Deng, Z.; Zou, Y.; Liu, C.; Fu, H.; Gu, Y.; Chang, H. Theaflavin alleviates oxidative injury and atherosclerosis progress via activating microRNA-24-mediated Nrf2/HO-1 signal. Phytother. Res. 2021, 35, 3418–3427. [Google Scholar] [CrossRef]
  126. Wang, Y.; Li, J.; Xuan, L.; Liu, Y.; Shao, L.; Ge, H.; Gu, J.; Wei, C.; Zhao, M. Astragalus Root dry extract restores connexin43 expression by targeting miR-1 in viral myocarditis. Phytomedicine 2018, 46, 32–38. [Google Scholar] [CrossRef]
  127. Li, X.; Zhao, D.; Guo, Z.; Li, T.; Qili, M.; Xu, B.; Qian, M.; Liang, H.; E, X.; Chege Gitau, S.; et al. Overexpression of SerpinE2/protease nexin-1 Contribute to Pathological Cardiac Fibrosis via increasing Collagen Deposition. Sci. Rep. 2016, 6, 37635. [Google Scholar] [CrossRef]
  128. Kannaiyan, R.; Shanmugam, M.K.; Sethi, G. Molecular targets of celastrol derived from Thunder of God Vine: Potential role in the treatment of inflammatory disorders and cancer. Cancer Lett. 2011, 303, 9–20. [Google Scholar] [CrossRef]
  129. Cheng, M.; Wu, G.; Song, Y.; Wang, L.; Tu, L.; Zhang, L.; Zhang, C. Celastrol-Induced Suppression of the MiR-21/ERK Signalling Pathway Attenuates Cardiac Fibrosis and Dysfunction. Cell. Physiol. Biochem. 2016, 38, 1928–1938. [Google Scholar] [CrossRef] [PubMed]
  130. Ning, B.B.; Zhang, Y.; Wu, D.D.; Cui, J.G.; Liu, L.; Wang, P.W.; Wang, W.J.; Zhu, W.L.; Chen, Y.; Zhang, T. Luteolin-7-diglucuronide attenuates isoproterenol-induced myocardial injury and fibrosis in mice. Acta Pharmacol. Sin. 2017, 38, 331–341. [Google Scholar] [CrossRef]
  131. Wei, Y.; Wu, Y.; Feng, K.; Zhao, Y.; Tao, R.; Xu, H.; Tang, Y. Astragaloside IV inhibits cardiac fibrosis via miR-135a-TRPM7-TGF-β/Smads pathway. J. Ethnopharmacol. 2020, 249, 112404. [Google Scholar] [CrossRef]
  132. Noratto, G.D.; Angel-Morales, G.; Talcott, S.T.; Mertens-Talcott, S.U. Polyphenolics from acai (Euterpe oleracea Mart.) and red muscadine grape (Vitis rotundifolia) protect human umbilical vascular Endothelial cells (HUVEC) from glucose- and lipopolysaccharide (LPS)-induced inflammation and target microRNA-126. J. Agric. Food Chem. 2011, 59, 7999–8012. [Google Scholar] [CrossRef] [PubMed]
  133. Nie, W.; Zhang, X.; Yan, H.; Li, S.; Zhu, W.; Fan, F.; Zhu, J. Xiaoxianggou attenuates atherosclerotic plaque formation in endogenous high Ang II ApoE(−/−) mice via the inhibition of miR-203 on the expression of Ets-2 in endothelial cells. Biomed. Pharmacother. 2016, 82, 173–179. [Google Scholar] [CrossRef]
  134. Yin, S.; Bai, W.; Li, P.; Jian, X.; Shan, T.; Tang, Z.; Jing, X.; Ping, S.; Li, Q.; Miao, Z.; et al. Berberine suppresses the ectopic expression of miR-133a in endothelial cells to improve vascular dementia in diabetic rats. Clin. Exp. Hypertens. 2019, 41, 708–716. [Google Scholar] [CrossRef]
  135. Hua, J.Y.; He, Y.Z.; Xu, Y.; Jiang, X.H.; Ye, W.; Pan, Z.M. Emodin prevents intima thickness via Wnt4/Dvl-1/beta-catenin signaling pathway mediated by miR-126 in balloon-injured carotid artery rats. Exp. Mol. Med. 2015, 47, e170. [Google Scholar] [CrossRef]
  136. Kwok, H.H.; Chan, L.S.; Poon, P.Y.; Yue, P.Y.; Wong, R.N. Ginsenoside-Rg1 induces angiogenesis by the inverse regulation of MET tyrosine kinase receptor expression through miR-23a. Toxicol. Appl. Pharmacol. 2015, 287, 276–283. [Google Scholar] [CrossRef] [PubMed]
  137. Hibender, S.; Franken, R.; van Roomen, C.; Ter Braake, A.; van der Made, I.; Schermer, E.E.; Gunst, Q.; van den Hoff, M.J.; Lutgens, E.; Pinto, Y.M.; et al. Resveratrol Inhibits Aortic Root Dilatation in the Fbn1C1039G/+ Marfan Mouse Model. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 1618–1626. [Google Scholar] [CrossRef]
  138. Chen, W.; Li, X.; Guo, S.; Song, N.; Wang, J.; Jia, L.; Zhu, A. Tanshinone IIA harmonizes the crosstalk of autophagy and polarization in macrophages via miR-375/KLF4 pathway to attenuate atherosclerosis. Int. Immunopharmacol. 2019, 70, 486–497. [Google Scholar] [CrossRef] [PubMed]
  139. Wang, X.L.; Fu, A.; Spiro, C.; Lee, H.C. Proteomic Analysis of Vascular Endothelial Cells-Effects of Laminar Shear Stress and High Glucose. J. Proteom. Bioinform. 2009, 2, 445. [Google Scholar] [CrossRef]
  140. Incalza, M.A.; D’Oria, R.; Natalicchio, A.; Perrini, S.; Laviola, L.; Giorgino, F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vascul. Pharmacol. 2017, 100, 1–19. [Google Scholar] [CrossRef]
  141. Eren, E.; Yilmaz, N.; Aydin, O. Functionally defective high-density lipoprotein and paraoxonase: A couple for endothelial dysfunction in atherosclerosis. Cholesterol 2013, 2013, 792090. [Google Scholar] [CrossRef] [PubMed]
  142. Dong, Q.; Xing, W.; Su, F.; Liang, X.; Tian, F.; Gao, F.; Wang, S.; Zhang, H. Tetrahydroxystilbene Glycoside Improves Microvascular Endothelial Dysfunction and Ameliorates Obesity-Associated Hypertension in Obese ZDF Rats Via Inhibition of Endothelial Autophagy. Cell. Physiol. Biochem. 2017, 43, 293–307. [Google Scholar] [CrossRef]
  143. Wilkinson, E.L.; Sidaway, J.E.; Cross, M.J. Statin regulated ERK5 stimulates tight junction formation and reduces permeability in human cardiac endothelial cells. J. Cell. Physiol. 2018, 233, 186–200. [Google Scholar] [CrossRef] [PubMed]
  144. Cai, X.; Bao, L.; Ding, Y.; Dai, X.; Zhang, Z.; Li, Y. Quercetin alleviates cell apoptosis and inflammation via the ER stress pathway in vascular endothelial cells cultured in high concentrations of glucosamine. Mol. Med. Rep. 2017, 15, 825–832. [Google Scholar] [CrossRef] [PubMed]
  145. Cheng, C.; Tempel, D.; den Dekker, W.K.; Haasdijk, R.; Chrifi, I.; Bos, F.L.; Wagtmans, K.; van de Kamp, E.H.; Blonden, L.; Biessen, E.A.; et al. Ets2 determines the inflammatory state of endothelial cells in advanced atherosclerotic lesions. Circ. Res. 2011, 109, 382–395. [Google Scholar] [CrossRef] [PubMed]
  146. Tsaousi, A.; Williams, H.; Lyon, C.A.; Taylor, V.; Swain, A.; Johnson, J.L.; George, S.J. Wnt4/beta-catenin signaling induces VSMC proliferation and is associated with intimal thickening. Circ. Res. 2011, 108, 427–436. [Google Scholar] [CrossRef]
  147. Chen, Y.; Lüttmann, F.F.; Schoger, E.; Schöler, H.R.; Zelarayán, L.C.; Kim, K.P.; Haigh, J.J.; Kim, J.; Braun, T. Reversible reprogramming of cardiomyocytes to a fetal state drives heart regeneration in mice. Science 2021, 373, 1537–1540. [Google Scholar] [CrossRef]
  148. Cowan, C.E.; Kohler, E.E.; Dugan, T.A.; Mirza, M.K.; Malik, A.B.; Wary, K.K. Kruppel-like factor-4 transcriptionally regulates VE-cadherin expression and endothelial barrier function. Circ. Res. 2010, 107, 959–966. [Google Scholar] [CrossRef]
  149. Hale, A.T.; Tian, H.; Anih, E.; Recio, F.O., 3rd; Shatat, M.A.; Johnson, T.; Liao, X.; Ramirez-Bergeron, D.L.; Proweller, A.; Ishikawa, M.; et al. Endothelial Kruppel-like factor 4 regulates angiogenesis and the Notch signaling pathway. J. Biol. Chem. 2014, 289, 12016–12028. [Google Scholar] [CrossRef]
  150. Zhou, G.; Hamik, A.; Nayak, L.; Tian, H.; Shi, H.; Lu, Y.; Sharma, N.; Liao, X.; Hale, A.; Boerboom, L.; et al. Endothelial Kruppel-like factor 4 protects against atherothrombosis in mice. J. Clin. Investig. 2012, 122, 4727–4731. [Google Scholar] [CrossRef]
  151. Dambal, S.; Shah, M.; Mihelich, B.; Nonn, L. The microRNA-183 cluster: The family that plays together stays together. Nucleic Acids Res. 2015, 43, 7173–7188. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustration of the main mechanisms by which natural products improve lipid metabolism disorders mediated by miRNAs. The red arrow means an increase, and the green arrow means a decrease.
Figure 1. Schematic illustration of the main mechanisms by which natural products improve lipid metabolism disorders mediated by miRNAs. The red arrow means an increase, and the green arrow means a decrease.
Molecules 28 06202 g001
Figure 2. Schematic illustration of the main mechanisms by which natural products improve glucose metabolism disorders mediated by miRNAs. The red arrow means an increase, and the green arrow means a decrease.
Figure 2. Schematic illustration of the main mechanisms by which natural products improve glucose metabolism disorders mediated by miRNAs. The red arrow means an increase, and the green arrow means a decrease.
Molecules 28 06202 g002
Figure 3. Schematic illustration of the main mechanisms by which natural products improve cardiovascular diseases mediated by miRNAs. The red arrow means an increase, and the green arrow means a decrease.
Figure 3. Schematic illustration of the main mechanisms by which natural products improve cardiovascular diseases mediated by miRNAs. The red arrow means an increase, and the green arrow means a decrease.
Molecules 28 06202 g003
Table 1. The effects of natural products (extracts) on lipid metabolism disorders.
Table 1. The effects of natural products (extracts) on lipid metabolism disorders.
Natural Products (Extracts)Relevant miRNAsDoseAdministration
Methods
Experimental ModelsTargetsObserved EffectsReferences
Averrhoa carambola free phenolic extractmiR-33↓
miR-34a↓
10, 20, 30 g/kg/d for 8 weeksGavagedb/db mice/● Reduced liver TG;
● Inhibited the signal transduction of hepatic lipogenesis;
● Exhibited a potent hepatic steatosis-relieving effect.
[20]
Cerasus humilis polyphenol extractmiR-7a/b↓40 μg/mL for 48 h;
250 g/kg/day for 12 weeks
Cell culture;
gavage
3T3-L1 pre-adipocyte cells; obese miceSirt1, Prdm16● Reduced body weight;
● Improved abnormal serum lipid and glucose levels;
● Inhibited adipocyte differentiation;
● Reduced fat accumulation by mitigating fat deposition, inflammation, and oxidation.
[21]
Citrus peel flavonoidsmiR-33↓
miR-122↓
10 μg/mL for 0.5, 1, 3 and 6 hCell cultureOleic acid-treated HepG2 cellsFAS, CPT1a● Attenuated intracellular lipid accumulation.[22]
Coffee polyphenols miR-122↑2.5 × 10−4%;
diet containing 0.5% or 1.0% coffee polyphenols for 15 weeks
Cell culture;
diet
Hepa 1-6 cells;
HFD-fed mice
SREBP1c● Activated AMPK;
● Enhanced energy metabolism;
● Reduced lipogenesis;
● Reduced body weight gain, abdominal and liver fat accumulation.
[23]
Ginger extractmiR-21↓
miR-132↓
Diet containing 0.8% ginger extract for 10 weeksDietHFD-fed rats/● Lowered body weight and white adipose tissue mass;
● Reduced serum and hepatic lipid levels;
● Enhanced AMPK activity;
● Ameliorated obesity and inflammation.
[24]
Grape seed proanthocyanidins extractmiR-33a↓
miR-122↓
5, 25, 50 mg/kg for 3 weeksGavageHFD–induced obese rats ABCA1;
FAS, PPARβ/δ
● Hypolipidemic;
● Decreased total liver fat.
[16]
miR-33a↓
miR-122↓
5, 15, 25, 50 mg/kg for 3 weeksGavageHealthy Wistar ratsABCA1;
FAS
● Improved postprandial hyperlipemia;
● Increased liver cholesterol efflux to HDL formation;
● Reduced fatty acid synthesis.
[25]
miR-33↓
miR-122↓
10, 25, 50, or 100 mg/L for 0.5, 1, 3, or 5 h;
250 mg/kg for 1 or 3 h
Cell culture;
gavage
FAO cells;
Wistar rats
ABCA1;
FAS
● Hypolipidemic;
● Reduced lipogenesis;
● Increased liver cholesterol efflux to HDL formation.
[26]
miR-33a↓
miR-122↓
25 mg/kg for 3 weeksGavageDyslipidemic obese ratsABCA1, CPT1a;
FAS, PPARβ/δ
● Improved dyslipidemia;
● Decreased total liver fat.
[27]
miR-96↓200 mg/kg/day for 180 daysDietHFD-fed micemTOR, FOXO1● Decreased the weight gain, serum levels of triglycerides, total cholesterol, and low-density lipoprotein cholesterol but increased high-density lipoprotein cholesterol;
● Clearance of lipid accumulation.
[28]
miR-33↓
miR-122↓
250 mg/kg onceGavageHFD-fed grass carp/● Decreased TG accumulation by reducing de novo lipogenesis and enhancing lipolysis and β-oxidation.[29]
Green tea extractmiR-34a↓
miR-194↑
500 mg/kg for 12 weeks (5 days/week)GavageHFD-fed miceSirt1, PPARα, INSIG2; HMGCS, APOA5● Protected against NAFLD development by altering lipid metabolism, increasing gene expression involved in triglycerides and fatty acid catabolism, and decreasing uptake and lipid accumulation.[30]
miR-335↓500 mg/kg for 12 weeks (5 days/week)GavageHFD-fed miceFOXO1, GSK3β● Reduced weight gain, adiposity and inflammation;
● Increased energy expenditure;
● Improved insulin sensitivity.
[31]
Guarana extractmiR-27b↓
miR-34b↓
miR-760↓
150 µg/mL for 48 hCell culture3T3-L1 pre-adipocyte cellsWnt3a, Wnt1, Wnt10b● Anti-adipogenic effect.[32]
Lychee pulp phenolicsmiR-33↓
miR-122↓
500 mg/kg for 10 weeksGavageHFD-fed miceABCA1, ABCG1, NPC1;
FAS, ACC1, ACC2, SCD1, ACLY
● Hypolipidemic;
● Repressed fatty acid synthesis and promoting fatty acid β-oxidation and cholesterol efflux in the liver;
● Decreased body fat accumulation;
● Ameliorated lipid metabolism.
[33]
Mulberry fruit extractmiR-33↓Diet containing 0.4% mulberry fruit extract for 4 weeksDietHigh cholesterol/cholic acid diet-fed rats/● Promoted serum high-density lipoprotein cholesterol levels;
● Decreased serum and hepatic cholesterol, serum low-density lipoprotein cholesterol, and fecal bile acid levels.
[34]
Mulberry leaf extractmiR-34a↓3 mg/mL for 24 hCell cultureGlucolipotoxicity-induced HepG2 cells Sirt1● Reduced liver fat accumulation;
● Decreased inflammatory responses and steatohepatitis;
● Exerted anti-glucolipotoxicity effects.
[35]
Moringa oleifera leaf extractmiR-21a↓
miR-103↓
miR-122↓
miR-34a↓
9.375 mg/d for 8 weeksGavageHFD-fed mice/● Improved ITT and decreased SREBP1c hepatic protein, while Sirt1 increased;
● Reduced insulin resistance, de novo lipogenesis, hepatic inflammation, and ER stress;
● Prevented progression of liver damage in a model of NASH.
[36]
Portulaca oleracea extract miR-122↓25, 50, 100 mg/kg/d for 7 daysGavageAcute alcoholic liver injury rats/● Reduced the ethanol-elevated serum level of ALT, AST, ALP, and TG;
● Enhanced activities of SOD and GSH-Px;
● Decreased content of NO and MDA;
● Increased antioxidant capacity;
● Relieved the inflammatory injury;
● Improved the lipid metabolism disorder.
[37]
miR-33↓
miR-34a↓
Diet containing 0.8% portulaca oleracea L. extract for 4 weeksDietHigh-cholesterol diet-fed rats/● Improved serum, liver, and fecal lipid profiles;
● Promoted cholesterol efflux and bile acid synthesis;
● Enhanced hepatic AMPK activity.
[38]
Rosmarinus officinalis extract miR let-7f-1↑30 μg/mL for 35 daysCell cultureHuman primary omental pre-adipocytes and adipocytes/● Decreased triglyceride accumulation;
● Increased glycerol release;
● Stimulated lipolytic activity in differentiating pre-adipocytes and mature adipocytes;
● Modulated the adipocyte life cycle at different levels.
[39]
The up arrow means an increase, and the down arrow means a decrease.
Table 2. The effects of natural products (compounds) on lipid metabolism disorders.
Table 2. The effects of natural products (compounds) on lipid metabolism disorders.
Natural Products (Compounds)Relevant miRNAsDoseAdministration MethodsExperimental ModelsTargetsObserved EffectsReferences
A-type ECG and EGCG dimers
Molecules 28 06202 i001
miR-7a/b↑ECG dimer: 20 μg/mL for 1–8 days;
ECGG dimer: 60 μg/mL for 1–8 days
Cell culture3T3-L1 pre-adipocyte cellsPPARγ● Inhibited pre-adipocyte differentiation;
● Reduced intracellular lipid accumulation;
● Blocked MCE process;
● Decreased the fluidity and hydrophobicity and increased the permeability of membrane.
[40]
Curcumin
Molecules 28 06202 i002
miR-17↓2 μM or 10 μM for 6 h;Cell culture3T3-L1 pre-adipocyte cells;
HFD-fed mice
TCF7L2● Inhibited adipocyte differentiation and adipogenesis;
● Stimulated the Wnt signaling pathway.
[41]
Grape seed procyanidin B2
Molecules 28 06202 i003
miR-483↓150 μg/mL for 48 hCell culture3T3-L1 pre-adipocyte cells PPARγ● Inhibited pre-adipocyte differentiation;
● Reduced intracellular lipid accumulation.
[42]
EGCG
Molecules 28 06202 i004
miR-143↑ 50 μM for 24 h Cell culture3T3-L1 pre-adipocyte cellsMAPK7● Inhibited 3T3-L1 cell growth.[43]
Lycopene
Molecules 28 06202 i005
miR-21↑50 μM for 24 h;
diet containing 0.05% lycopene for 8 weeks
Cell culture;
gavage
Hepa 1–6 cells;
HFD-fed mice
FABP7● Lowered body weight;
● Inhibited intracellular lipid accumulation;
● Protected against HFD-induced hepatic steatosis.
[44]
Nonivamide
Molecules 28 06202 i006
miR let-7d↑1 μM for 12 daysCell culture3T3-L1 pre-adipocyte cellsPPARγ● Impaired adipogenesis;
● Reduced mean lipid accumulation;
● Activated TRPV1.
[45]
Oleanolic acid
Molecules 28 06202 i007
miR-98↑10 mM for 6, 12, 24 h;
20 mg/kg for 4 weeks
Cell cultureHFD-fed mice;
db/db mice
PGC1β● Hypolipidemic.[46]
Persimmon tannin
Molecules 28 06202 i008
miR-27↑20, 40, or 60 μg/mL for 1–8 daysCell culture3T3-L1 pre-adipocyte cellsPPARγ, C/EBPα● Inhibited pre-adipocyte differentiation;
● Reduced intracellular lipid accumulation;
● Delayed MCE process.
[47]
Pseudoprotodioscin
Molecules 28 06202 i009
miR-33a/b↓25 μM for 24 hCell cultureHuman HepG2 cells and THP-1 monocytic cells SREBP1c, SREBP2● Promoted the cholesterol effluxion.[48]
Resveratrol
Molecules 28 06202 i010
miR-103↓
miR-107↓
miR-122↓
30 mg/kg for 6 weeksDietObesogenic diet-fed ratsSREBP1;
SREBP1, CPT1a;
FAS
● Reduced obesogenic diet-induced hepatic steatosis;
● Activated AMPK.
[49]
miR-539↑30 mg/kg for 6 weeksDietObesogenic diet-fed ratsSP1● Inhibited de novo lipogenesis.[50]
miR-155↑25 μM for 1–8 daysCell culture3T3-L1 pre-adipocyte cellsCEBP/α● Inhibited adipogenesis.[51]
Zerumbone
Molecules 28 06202 i011
miR-46b↓25 μM for 48 h;
diet containing 0.025% zerumbone for 8 weeks
Cell culture;
diet
3T3-L1 fibroblasts;
HFD-fed mice
Sirt1● Induced AMPK activation and phosphorylation of acetyl-CoA carboxylase;
● Ameliorated diet-induced obesity and inhibited adipogenesis.
[52]
The up arrow means an increase, and the down arrow means a decrease.
Table 3. The effects of natural products (extracts) on glucose metabolism disorders.
Table 3. The effects of natural products (extracts) on glucose metabolism disorders.
Natural Products (Extracts)Relevant miRNAsDoseAdministration MethodsExperimental ModelsTargetsObserved EffectsReferences
Alpinia oxyphylla extractmiR-let-7k, miR-378d: ↑;
miR-129, miR-21a, miR-29c, miR-203, miR-7a: ↓
50 mg/kg/d for 8 weeks GavageDB/DB and db-/db- mice/● Lowered concentrations of blood glucose;
● Changed the expressions of specific miRNAs.
[76]
Blueberry anthocyanins extractmiR-182↓200 mg/kg/d for 6 days GavageSTZ-induced diabetic ratsOGG1● Restored the increase of apoptosis, ROS level, and ERS induced by high-concentration glucose.[77]
Coreopsis tinctoria nutt extractmiR-192↓
miR-200b↓
300 mg/kg/d for 10 weeks Gavagedb/db miceZEB2, PTEN● Decreased body weight, fasting blood glucose, and 24 h urinary albumin excretion;
alleviated kidney damage;
● Modulated the activity of the PTEN/PI3K/AKT pathway to reduce the degree of renal fibrosis.
[78]
Crataegus persica extractmiR-126↓300 mg/kg/d for 10 weeks GavageDiabetic ratsNrf2● Decreased elevated levels of renal oxidative stress, glomerular filtration rate, insulin sensitivity, and pathological score.[79]
Licorice flavonoidmiR-122↑30 mg/kg for 5 weeks, 5 times per weekGavageHFD-fed micePTP1B● Reduced blood glucose;
● Restored IR and IRS1/2 tyrosine phosphorylation and insulin signaling;
● Abrogated hepatic insulin resistance induced by HFD diet.
[80]
Nigella sativa oilmiR-34a↓
miR-26b↓
2.0 mL for 21 daysGavageDiabetic rats/● Suppressed oxidative stress;
● Improved insulin resistance and insulin signaling pathway.
[81]
The up arrow means an increase, and the down arrow means a decrease.
Table 4. The effects of natural products (compounds) on glucose metabolism disorders.
Table 4. The effects of natural products (compounds) on glucose metabolism disorders.
Natural Products (Compounds)Relevant miRNAsDoseAdministration MethodsExperimental ModelsTargetsObserved EffectsReferences
Astragaloside IV
Molecules 28 06202 i012
miR-138↓25 mM for 48 hCell cultureHigh glucose cultured retinal pigment epithelial cellsSirt1, Nrf2● Alleviated high glucose-induced RPE cell damage;
● Increased Sirt1/Nrf2 activity and cellular antioxidant capacity;
● Alleviated ferroptosis;
● Decreased cell death.
[82]
Curcumin
Molecules 28 06202 i013
miR-206↑15, 30, and 60 mg/kg/d for 6 weeks GavageFructose-fed ratsPTP1B● Improved insulin signaling;
● Improved glucose intolerance and insulin sensitivity.
[83]
Dioscin
Molecules 28 06202 i014
miR-34a↓50, 100, and 200 ng/mL for 12 h;
10, 20, and 40 mg/kg for 12 days;
15, 30, and 60 mg/kg/d for 10 days
Cell culture; gavageNRK-52E and HK-2 cells;
Wistar rats;
C57BL/6J mice
Sirt1● Decreased the ROS levels;
● Suppressed oxidative stress.
[84]
Genistein
Molecules 28 06202 i015
miR-34a↓1000 nM for 6 hCell cultureHUVECsSirt1● Restrained ROS and MDA production;
● Ameliorated the inhibitory effect on SOD, CAT, GSH, GPx activity;
● Suppressed oxidative stress.
[85]
Gypenoside A
Molecules 28 06202 i016
miR-150↓50 or 100 mg/kg/d for 12 weeks GavageHFD-fed micePDX1● Alleviated pancreatic impairments;
● Improved the dysfunction of β pancreatic cells.
[86]
Oridonin
Molecules 28 06202 i017
miR-1305, miR-152, miR-182, miR-29b, miR-4298, miR-939: ↑;
miR-1246, miR-1268, miR-1290, miR-135a, miR-181b, miR-1973, miR-210, miR-30c, miR-30e, miR-3162, miR-4299, miR-572, miR-575, miR-630, miR-642b: ↓
5 μM for 24 h Cell cultureH2O2-exposed HaCaT cells/● Protected against H2O2-induced oxidative stress.[87]
Polydatin
Molecules 28 06202 i018
miR-200a↑10, 20, and 40 μM for 24 h;
7.5, 15, and 30 mg/kg/d for 7 weeks
Cell culture;
gavage
High fructose-treated buffalo rat liver cells and HepG2 cells;
fructose-fed rats
Keap1● Alleviated hepatic oxidative stress, inflammation, and lipid deposition;
● Activated Keap1/Nrf2 pathway.
[88]
Sodium tanshinone IIA sulfonate
Molecules 28 06202 i019
miR-133a↓ 10 mg/kg/d for 3 weeks GavageSTZ-induced diabetic mice/● Improved perfusion recovery;
● Increased capillary densities;
● Decreased ROS level and increased GCH-1 protein level.
[89]
Vaccarin
Molecules 28 06202 i020
miR-34a↓1 mg/kg/d for 4 weeks Intraperitoneal injectionSTZ/HFD-induced T2DM mice/● Reduced blood glucose;
● Increased glucose and insulin tolerance;
● Relieved the disorder of lipid metabolism and oxidative stress;
● Improved endothelium-dependent vasorelaxation.
[90]
The up arrow means an increase, and the down arrow means a decrease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Liu, J.; Chen, H.; Li, X.; Song, C.; Wang, L.; Wang, D. Micro-Executor of Natural Products in Metabolic Diseases. Molecules 2023, 28, 6202. https://doi.org/10.3390/molecules28176202

AMA Style

Liu J, Chen H, Li X, Song C, Wang L, Wang D. Micro-Executor of Natural Products in Metabolic Diseases. Molecules. 2023; 28(17):6202. https://doi.org/10.3390/molecules28176202

Chicago/Turabian Style

Liu, Jinxin, Huanwen Chen, Xiaoli Li, Chunmei Song, Li Wang, and Deguo Wang. 2023. "Micro-Executor of Natural Products in Metabolic Diseases" Molecules 28, no. 17: 6202. https://doi.org/10.3390/molecules28176202

Article Metrics

Back to TopTop