Planta Med
DOI: 10.1055/a-2277-4805
Reviews

Bioprotective Role of Phytocompounds Against the Pathogenesis of Non-alcoholic Fatty Liver Disease to Non-alcoholic Steatohepatitis: Unravelling Underlying Molecular Mechanisms

Tanmoy Banerjee
1   Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, India
,
Arnab Sarkar
1   Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, India
,
Sk Zeeshan Ali
1   Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, India
,
Rudranil Bhowmik
1   Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, India
,
Sanmoy Karmakar
1   Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, India
,
Amit Kumar Halder
2   Dr. B. C. Roy College of Pharmacy and Allied Health Sciences, Dr. Meghnad Saha Sarani, Bidhannagar, Durgapur, West Bengal, India
,
Nilanjan Ghosh
1   Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata, India
› Author Affiliations
 

Abstract

Non-alcoholic fatty liver disease (NAFLD), with a global prevalence of 25%, continues to escalate, creating noteworthy concerns towards the global health burden. NAFLD causes triglycerides and free fatty acids to build up in the liver. The excessive fat build-up causes inflammation and damages the healthy hepatocytes, leading to non-alcoholic steatohepatitis (NASH). Dietary habits, obesity, insulin resistance, type 2 diabetes, and dyslipidemia influence NAFLD progression. The disease burden is complicated due to the paucity of therapeutic interventions. Obeticholic acid is the only approved therapeutic agent for NAFLD. With more scientific enterprise being directed towards the understanding of the underlying mechanisms of NAFLD, novel targets like lipid synthase, farnesoid X receptor signalling, peroxisome proliferator-activated receptors associated with inflammatory signalling, and hepatocellular injury have played a crucial role in the progression of NAFLD to NASH. Phytocompounds have shown promising results in modulating hepatic lipid metabolism and de novo lipogenesis, suggesting their possible role in managing NAFLD. This review discusses the ameliorative role of different classes of phytochemicals with molecular mechanisms in different cell lines and established animal models. These compounds may lead to the development of novel therapeutic strategies for NAFLD progression to NASH. This review also deliberates on phytomolecules undergoing clinical trials for effective management of NAFLD.


#

Abbreviations

ABCC2: ATP-binding cassette subfamily C member 2
ACADS: acyl-CoA dehydrogenases
ACC1: acetyl-CoA carboxylase 1
ACSL1: acyl CoA synthetase long-chain 1
AdipoR1: adiponectin receptor 1
AdipoR2: adiponectin receptor 2
ALK-1: anaplastic lymphoma kinase-1
ALK-5: anaplastic lymphoma kinase-5
ALT: alanine aminotransferase
AST: aspartate aminotransferase
ATF6: activating transcription factor 6
ATG5: autophagy related 5
ATG7: autophagy related 7
ATGL: adipose triglyceride lipase
ATP: Adenosine triphosphate
C/EBPα : CCAAT/enhancer binding protein α
CAV-1: caveolin-1
Co-SMAD: common-mediator SMAD 4 proteins
COX-1: cyclooxygenase-1
COX-2: cyclooxygenase-2
CPT1A: carnitine palmitoyltransferase 1A
CPT2: carnitine palmitoyltransferase 2
CYP2E1: cytochrome p450 2E1
EIF2: eukaryotic initiation factor 2
ERR: estrogen-related receptor
ERRα : estrogen-related receptor α
FABP1: fatty acid-binding protein 1
FABP2: fatty acid-binding protein 2
FDFT1: farnesyl-diphosphate farnesyltransferase 1
FXR1: fragile X related 1
GLUT4: glucose transporter type 4
GPAT1: glycerol-3-phosphate acyltransferase 1
GRP78: glucose-regulated protein 78
HNF4-α : hepatocyte nuclear factor 4α
HSL: hormone-sensitive lipase
IFN-γ : interferon-γ
IL-10: interleukin-10
IL-18: interleukin-18
IL-1β : interleukin-1β
IL-6: interleukin-6
IRS-2: insulin receptor substrate 2
KEAP1: kelch-like ECH-associated protein 1
LC3-II: microtubule-associated protein light chain 3-II
LCAD: long-chain acyl-CoA dehydrogenase
LPL: lipoprotein lipase
MCAD: medium-chain acyl-CoA dehydrogenase
mRNA: messenger ribonucleic acid
mTORC1: mammalian target of rapamycin complex 1
NADPH: nicotinamide adenine dinucleotide phosphate
NR4A1: nuclear receptor 4A1
OA: oleic acid
PA: palmitic acid
PARP1: poly(ADP-ribose) polymerase 1
PCK1: phosphoenolpyruvate carboxykinase 1
PKB: protein kinase B
PPAR-α : peroxisome proliferator-activated receptor α
PPAR-δ: peroxisome proliferator-activated receptor δ
R-SMAD: receptor-activated SMAD
SCD1: stearoyl CoA desaturase 1
SMAD 2: suppressor of mothers against decapentaplegic homolog 2
SMAD 3: suppressor of mothers against decapentaplegic homolog 3
SREBF1: sterol regulatory element binding transcription factor 1
SREBP-2: sterol regulatory element-binding protein-2
SULT2A1: sulfotransferase 2A1
TAZ: transcriptional coactivator with PDZ-binding motif
TBXA2R: thromboxane A2 receptor
TIMP-1: tissue inhibitor of metalloproteinase-1
TIMP-2: tissue inhibitor of metalloproteinase-2
TNFR2: tumour necrosis factor receptor 2
UCP1: uncoupling protein 1
UCP2: uncoupling protein 2
VCAM-1: vascular cell adhesion molecule 1
VLDL: very-low-density lipoprotein
XBP-1: x-box binding protein 1
ZO-1: zonula occludens-1
 

Introduction

Non-alcoholic fatty liver disease (NAFLD), a primary cause of cirrhosis and HCC, is a global concern for clinicians [1]. The Middle East has the greatest prevalence of NAFLD at 31.8%, followed by Europe at 23% and Africa at 13.5% [2], [3]. NAFLD is expected to become the largest cause of liver-related mortality and morbidity and a leading cause of hepatic transplantation within 20 years [4]. In non-alcoholics, NAFLD causes fat accumulation in liver [5]. Steatosis with or without moderate inflammation of the non-alcoholic fatty liver (NAFL) progresses to non-alcoholic steatohepatitis (NASH), leading to enhanced fibrosis progression [6]. Unlike other hepatic disorders, NAFLD is linked to metabolic syndromes [7].

Diabetes has assumed global proportions and affected 8.7% of adults worldwide in 2014 [8]. Type 2 diabetes mellitus (T2DM), cardiovascular disease, obesity, insulin resistance (IR), and hypothyroidism enhance the risk of cirrhosis and accompanying consequences ([Fig. 1]). Reduced lipid oxidation and high FFAs cause IR and NAFLD. Dietary fat can cause hepatic steatosis (HS), which can worsen IR [9]. Another very interesting aspect of NAFLD, as pointed out by Tarantino et al., 2023, is the presence of sarcopenia in NAFLD patients, a pathological condition characterised by loss of skeletal muscle and strength [10]. Oxidative stress–a disparity between ROS generation and antioxidant responses–is another important factor in NAFLD pathophysiology associated with inflammation and FFA lipotoxicity, as depicted in [Fig. 1] [11]. There are many co-morbidities linked to NAFLD that share the main mechanism of low-grade chronic inflammation, with the involvement of the immune system (primarily overexpression of cytokines) partly mediated by the spleen [12].

Zoom Image
Fig. 1 Common risk factors associated with NAFLD.

Pioglitazone, a thiazolidinedione derivative and PPAR-γ agonist, improves glucose and lipid metabolism in T2DM by reducing IR but has not received USFDA approval for the management of NAFLD [13], [14]. Obeticholic acid (OCA), a selective agonist of farnesoid X receptors (FXR), has been approved by the FDA to manage NAFLD [15]. Natural compound research has increased over the past decade, providing new insights into mechanisms involved in controlling dyslipidemia, improving liver function in NAFLD. Products of natural origin have been found to retard the progression towards NASH by their modulatory actions on inflammatory and fibrotic pathways. This review deliberates on the mechanism of many potent lead molecules with potential in the management of NAFLD and NASH. Certain lead molecules with noteworthy activity in clinical trials have also been discussed.

The novelty of this review lies in the fact that, to date, there have been no correlation studies of phytoconstituents with clinical and preclinical outcomes explicitly pointing to phytomarkers [16]. The preclinical and clinical studies of phytoconstituents ameliorating NAFLD and its progression have been reported separately without any correlation [11], whereas this review aims to explore the mechanistic aspects of the lead phytotherapeutic molecules and map the mechanisms involved with the preclinical and clinical outcomes.


#

Pathophysiology of NAFLD Progressing to NASH

NAFLD is a medical disorder distinguished by the excessive buildup of adipose tissue inside the liver of individuals who exhibit little or no alcohol use. NAFLD is a prevalent hepatic illness on a global scale, exhibiting a range of severity levels from steatosis (simple fatty liver) to NASH, a more serious manifestation characterised by liver inflammation and damage. In the pathogenesis of NAFLD and the subsequent development of NASH, hepatic lipid build-up and inflammation play a pivotal role, as illustrated in [Fig. 2] [4]. The progression of NAFLD may be categorised into two distinct stages: non-alcoholic fatty liver (NAFL) and NASH. Nevertheless, NASH is characterised by not only the build-up of fat but also the presence of inflammation and damage to liver cells. Inflammation has a significant role in differentiating NASH from simple hepatic steatosis [1]. The immunological response is initiated in NASH due to the buildup of lipids inside hepatocytes. The precise processes behind this phenomenon remain incompletely elucidated; nevertheless, it is postulated that lipids originating from adipose tissue, together with cellular stress and injury, contribute to the attraction of immune cells, including macrophages, to the hepatic region. The immune cells secrete pro-inflammatory cytokines, including TNF-α and interleukins, which contribute to the amplification of the inflammatory response [17]. Hepatocellular death is a prominent indicator of NASH, as shown by the presence of enlarged hepatocytes seen during histological analysis of the liver. The condition is initiated by certain external and internal cellular stressors, such as the build-up of lipids in hepatocytes, leading to the occurrence of lipotoxicity. The phenomenon of lipotoxicity induces cellular apoptosis via the initiation of many intracellular processes, including endoplasmic reticulum stress and inflammation. Hepatocellular deaths are categorised into many routes, including apoptosis, necroptosis, and pyroptosis, based on their diverse underlying processes, which are characterised by specific morphological and molecular signs. Both the apoptosis and necroptosis signalling pathways are shown to be active in NASH, perhaps in a TNF-α-dependent manner. This is supported by the observed elevation in TNF-α levels, as well as the increased expression of its downstream effectors in the liver [18].

Zoom Image
Fig. 2 Pathophysiology of NAFLD progressing to NASH.

The occurrence of inflammation in NAFLD is initiated by the deposition of lipids inside hepatocytes, which then elicits an immune response. In response to hepatic inflammation, macrophages are attracted to the liver. Kupffer cells, which are specialised macrophages, reside in the liver. Macrophages serve as the primary barrier against invading microorganisms that gain access to the circulation through the portal vein, establishing a connection between the gastrointestinal tract and the liver. In the context of NAFLD and NASH, the activation of Kupffer cells occurs as a result of the presence of fatty acids and other inflammatory signals, hence playing a role in the promotion of hepatic inflammation [19]. The process of phagocytosis, facilitated by macrophages in the liver, aids in the removal of deceased or impaired liver cells. The liver cell death is heightened as a result of many variables, including oxidative stress and inflammation. Macrophages are recruited to eliminate these deceased cells; nevertheless, an excessive occurrence of cell death and subsequent macrophage activation might potentially exacerbate inflammation and fibrosis [20]. Fibrosis refers to the pathological process characterised by the accumulation of excessive scar tissue in the liver. This condition is primarily driven by the persistent activation of macrophages and the resulting inflammation. Notably, the activation of hepatic stellate cells assumes a pivotal role in the progression of liver fibrosis. Macrophages secrete several substances that induce hepatic stellate cells to synthesise collagen, so initiating the process of fibrosis, which is a characteristic feature of the course of NASH [21]. The role of oxidative stress in the pathogenesis and advancement of NAFLD is of considerable importance. Oxidative stress arises from a disparity between the generation of reactive oxygen species (ROS), often referred to as free radicals, and the organismʼs capacity to counteract their effects via the utilisation of antioxidants [22].

Lipid peroxidation is a process that may occur in NAFLD wherein an excessive build-up of lipids in hepatocytes can result in the peroxidation of these lipids. The aforementioned process encompasses the oxidative destruction of lipids, resulting in the production of very reactive compounds and free radicals. Reactive species can inflict harm against biological structures, including proteins, lipids, and DNA [23]. The impairment of mitochondrial activity, which is responsible for energy production inside hepatocytes, may be caused by oxidative stress. The impairment of mitochondrial activity might result in an increased generation of ROS, initiating a cascade of oxidative stress that detrimentally affects hepatocytes and further aggravates the underlying pathology [24]. The liver may initiate an inflammatory response when exposed to oxidative stress. ROS have the capability to initiate inflammatory signalling pathways and facilitate the attraction of immune cells, namely macrophages, to the liver. Consequently, the presence of inflammation may exacerbate the levels of oxidative stress, establishing a self-sustaining feedback loop [25]. Transforming growth factor-beta (TGF-β) is a cytokine that assumes a pivotal role in the processes of tissue healing and fibrosis. In the setting of NAFLD, specifically NASH, it is widely postulated that TGF-β plays a significant role in the pathogenesis of liver fibrosis. The activation of hepatic stellate cells, which are involved in collagen production and the promotion of liver fibrosis, is stimulated. In the context of liver inflammation, it is noteworthy that TGF-β has immunosuppressive characteristics and has the ability to regulate the inflammatory reaction. In the context of NAFLD, inflammation plays a pivotal role, particularly during the progression from simple steatosis to NASH. TGF-β potentially plays a crucial role in the modulation of the equilibrium between pro-inflammatory and anti-inflammatory mechanisms [26].

Satellite cells are a kind of stem cell found in skeletal muscle tissue. Satellite cells are a distinct population of stem cells that are specifically localised inside the skeletal muscle tissue. Skeletal muscle satellite cells are essential to the process of muscle repair and regeneration, particularly in response to instances of injury or physiological stress. Satellite cells are predominantly seen in muscle tissue, but their presence in the liver is often absent. The relationship between satellite cells and muscle tissue is of primary importance when examining the larger implications of physical exercise in the setting of NAFLD. Physical activity, including weight training and aerobic exercise, has been shown to have a beneficial influence on NAFLD. Engaging in regular physical exercise has been shown to enhance insulin sensitivity, diminish inflammation, and foster comprehensive metabolic well-being. These beneficial effects may contribute to the prevention and management of NAFLD [27]. Excessive caloric intake, especially from carbohydrates, is recognised as one of the main contributing factors to de novo lipogenesis (DNL) in NAFLD. When humans ingest excessive calories, particularly in the form of carbs such as sugars and starches, the surplus energy undergoes DNL in the liver, resulting in the conversion of the extra energy into triglycerides (TG) [28]. IR is a prevalent metabolic anomaly seen in individuals with NAFLD. IR is characterised by a diminished responsiveness of the bodyʼs cells, particularly hepatocytes, to the physiological actions of insulin. Insulin plays a pivotal role in the regulation of glucose metabolism and acts as an inhibitor of DNL. Nevertheless, in individuals with IR, the liver exhibits reduced sensitivity to the suppressive impact of insulin on DNL, resulting in heightened lipid synthesis [29]. Increased intake of dietary carbohydrates, i.e., fructose, may lead to DNL. The metabolism of fructose occurs mostly in the liver, where it undergoes conversion into glucose and fatty acids, hence facilitating the process of lipid production. The liver can produce additional fatty acids from acetyl-CoA thanks to DNL. First, acetyl-CoA carboxylase (ACC) converts acetyl-CoA to malonyl-CoA, which fatty acid synthase (FAS) subsequently converts to palmitate. Following a variety of desaturation, elongation, and esterification processes, the newly formed fatty acid may either be evacuated as VLDL particles or retained as TG. As a result, elevated DNL may result in hypertriglyceridemia, hepatic steatosis, and/or steatohepatitis because saturated fatty acids, such as palmitate, can trigger apoptosis and inflammation. Research indicated that unusually increased DNL, independent of fasting, is a key feature of NAFLD patients [30].

Certain individuals may possess genetic predispositions that render them more susceptible to DNL. The development of NASH requires the presence of patatin-like phospholipase 3 (PNPLA3), a protein that facilitates the production of lipids. There exists a strong correlation between the severity of NAFLD and the presence of polymorphisms in the PNPLA3 gene. The PNPLA3 gene exhibits interaction with SREBP-1c, hence facilitating the promotion of gene expression [31]. An increase in the activity of AMPK leads to a decrease in DNL and an enhancement of fatty acid oxidation (FAO), which hinders the pathogenesis of NAFLD via the phosphorylation of ACC. The activation of malonyl-CoA decarboxylase occurs subsequent to phosphorylation by AMPK, resulting in a decrease in malonyl-CoA levels and a concomitant reduction in the synthesis of FFAs. In the context of gene regulation, it has been shown that the genes FAS, ACC1, and SCD1 are known to be targeted by SREBP-1c. Notably, these genes have been found to be substantially inhibited by AMPK. This facilitates the promotion of the FAO and significantly adds to the regulation of lipogenesis balance [32]. In addition, it is essential to note that AMPK plays a crucial role in the maintenance of the mitochondrial matrix via the upregulation of carnitine palmitoyltransferase 1 (CPT1) in adipose tissue and hepatocytes. The AMPK functions by stimulating catabolic processes that generate ATP and suppressing ATP-utilising pathways to restore energy homeostasis. The efficiency of hepatic fat synthesis and storage may be influenced by genetic differences in enzymes associated with lipid metabolism [33]. Hormonal imbalances, characterised by elevated levels of hormones such as leptin and adiponectin, have the potential to disrupt lipid metabolism and have a role in DNL in NAFLD. The dietary choices made by individuals may have an impact on DNL since the specific kinds of lipids and carbs ingested play a significant role in this process. Diets that are rich in carbohydrates, particularly fructose, and saturated fats have a higher propensity to stimulate DNL and facilitate the storage of fat in the liver [34].

Hepatocyte ballooning is well recognised as a prominent and distinctive hallmark of NASH. Ballooning has been shown to be correlated with an increased risk for the development of NASH and fibrosis. Ballooned hepatocytes, while potentially progressing towards cell death and exhibiting increased susceptibility to apoptosis, are widely recognised as wounded but viable hepatocytes. Consequently, they have been referred to as “undead” hepatocytes. The hepatocytes that have undergone ballooning release sonic hedgehog (SHH), a protein that stimulates the activation of hepatic stellate cells (HSCs). However, it is probable that these hepatocytes also release other ligands that cause fibrosis. However, it is worth considering the possibility that ballooned “undead” hepatocytes may serve as an indicator of hepatocyte stress, suggesting that the progression of fibrosis in NASH can occur without hepatocytes reaching a state of ballooning or death [35]. The process of activating HSCs is crucial in the progression of fibrosis, since these cells are particular to the liver and have a pivotal role in this pathological condition. Upon receiving inflammatory signals and several other stimuli, dormant hematopoietic stem cells undergo activation and undergo a phenotypic transformation into cells resembling myofibroblasts. These activated cells then proceed to generate collagen and other constituents of the fibrous tissue. Cytokines and growth factors, including inflammatory cytokines (e.g., TNF-α and IL-6) and TGF-β, have been implicated in the promotion of fibrosis. These chemokines have a role in the activation of fibrogenic cells and promote the deposition of extracellular matrix components, leading to the formation of scar tissue [36]. The normal function of insulin involves its role as a hormone synthesised by the pancreas, which is crucial in the regulation of glucose levels in the bloodstream. The major role of this mechanism is to enhance the cellular absorption of glucose, with a particular emphasis on muscle and adipose cells, to support energy production. Additionally, it hinders the hepatic release of glucose. IR may be described as a physiological condition whereby the responsiveness of the bodyʼs cells, particularly those found in muscle, adipose tissue, and the liver, to the actions of insulin is diminished. Consequently, the impaired ability of cells to uptake glucose from the circulation and the ongoing hepatic glucose production contribute to the observed elevation in blood glucose levels [37].

Insulin plays a significant part in the metabolism of lipids, or fats. This process aids in the prevention of fat degradation inside adipocytes while also facilitating fat accumulation. IR gives rise to a compromised capacity of the body to control fat metabolism, resulting in heightened lipolysis (the breakdown of fat) and an elevation in circulating FFAs. These FFAs in the liver may be esterified to create TG as lipid droplets in hepatocytes, or they can be oxidised by β-oxidation in mitochondria to generate ATP or ketone bodies. Alternatively, they can be discharged into the bloodstream as very low-density lipoprotein (VLDL). Hepatic IR and lipid levels rise as a result of inadequate lipid oxidation. There is also a definite correlation between the quantity of visceral fat and hepatic IR. Different lipid intermediates, including ceramides and DAG, build up in cells of fatty livers and induce IR. Increased hepatic DAG concentration, which stimulates PKCε activity, might be a contributory factor of NAFLD-related hepatic IR. PKCε stimulation mediated by hepatic DAG is linked to the pathophysiology of hepatic IR linked to NAFLD. PKC activation caused by intracellular DAG closer to the cell membrane suppresses insulin receptor kinase, tyrosine phosphorylation of IRS-1 and − 2 downstream, and phosphorylation of inositol 3-kinase (PI3K), all of which contribute to decreased insulin signalling. In the end, this results in decreased hepatic glycogen synthesis because glycogen synthase is not as activated and in enhanced hepatic gluconeogenesis because forkhead box protein O1 (FOXO1) is less inactivated. This causes an excessive amount of glucose to be released via glucose transporter 2 (GLUT2). Elevated generation and release of hepatocellular ceramide may promote hepatic IR in NAFLD. Ceramide plays a significant role in IR. For example, the reduction in ceramide synthesis decreases hepatic steatosis and fibrosis in NAFLD [38]. The intricacy of NAFLD therapy is reflected in its pathophysiology since effective treatment of NAFLD patients necessitates an integrated, multi-systemic approach. Referrals to liver disease may be made at any point, although they are most common when fibrosis is diagnosed. Since there are not many already authorised medicines, the field of treatment possibilities is ready for innovation. Although weight reduction might cause NASH and fibrosis to decline. NAFLD patients may benefit from various pharmacological therapies available as discussed later.


#

Natural Compounds from the Phytochemical Class Impeding NAFLD

Secondary metabolites such alkaloids, flavonoids, glycosides, terpenoids, and tannins have previously shown anti-microbial [39], anti-hyperglycemic, and anti-cancer properties [40], [41]. There is an unmet, compelling need by physicians to use natural products to alleviate NAFLD, due to their large availability coupled with safety and low cost [42]. As shown in this section, several natural phytoconstituents that target lipid metabolism have demonstrated their effectiveness in treating fatty liver and can be used as therapies and natural remedies. This study describes the most current developments of isolated natural compounds and their chemotherapeutic characteristics in the prevention of NAFLD. The literature surveys for this review were performed using Scopus, PubMed, Web of Science, and Google Scholar. All the scientific data were collected from the years 2003 to 2023. The literature search was carried out using the following keywords: NAFLD, NASH, steatohepatitis, ethnomedicine for NAFLD, lipid metabolism, molecular targets for NAFLD & NASH, and novel phytocompound leads for NAFLD & NASH.


#

Alkaloids

Alkaloids are naturally occurring, alkaline, nitrogenous organic substances found in plants. Most alkaloids have complex cyclic structures and significant effects on lipid metabolism. The significant molecular target of alkaloids is AMPK, which increases the expression of PPAR-α and CPT1 and reduces the levels of SREBP-1c, ACC, PPAR-γ, and C/EBPα. Therefore, the availability of lipids like fatty acids might be restricted because natural substances in this class can boost lipid catabolic metabolism and hinder anabolic metabolism [43]. Some crucial alkaloids that have shown effect against NAFLD are briefly discussed below and depicted in [Fig. 3] and [Table 1].

Zoom Image
Fig. 3 Alkaloids involved in the management of NAFLD [berberine (I), betaine (II), nuciferine (III), matrine (IV), oxymatrine (V), piperine (VI), trigonelline (VII), and sophocarpine (VIII)].

Table 1 Structure of alkaloids targeting biomarkers involved in NAFLD.

Compound name with structure

Biological source

Research model

Biomarkers

Ref.

Liensinine

Nelumbo nucifera Gaertn

In vitro PA-induced L02 and AML12 cells; in vivo HFD-fed mice

Downregulated NF-κB, TAK1, ROS, ACC, PPAR-γ, FAS, and KEAP1; upregulated AMPK, Nrf2, PPAR-α, CPT1A, and UCP2

[210]

Theacrine

Camellia assamica var. kucha

In vitro OA-induced HepG2 and L02 cells; in vivo HFD-fed ApoE−/− and C57BL/6 J mice

Downregulated SREBF1, FAS, IL-1β, and IL-6; upregulated LCAD, and SIRT3

[211]

Kukoamine A

Lycium chinense (L. chinense) Miller

In vitro PA-induced AML-12 cells; in vivo HFD-fed mice

Downregulated SREBP-1c, FAS, ACC1, TNF-α, IL-1β, and IL-6

[212]

Conophylline

Tabernaemontana divaricate

In vivo HFD-fed BALB/c mice

Downregulated p62, TLR4, TGF-β, and TIMP-1; upregulated PPAR-α, CPT1, CPT2, LC3-II, and ACOX1

[213]

Tiliamosine

Tiliacora racemose

In vitro PO-BSA- induced HepG2 cells; in vivo HFD N-Nitrosodiethylamine-induced rats

Downregulated TNF-α; upregulated FXR expression

[214]

Koumine

Gelsemium elegans

In vivo HFD-fed rats

Downregulated IFN-γ, IL-6, IL-1β, IL-17A, and TNF-α; upregulated IL-10

[215]

Berberine

From Coptis chinensis and Phellodendron chinense, berberine (BRB), an isoquinoline alkaloid, is isolated [44], [45]. In HepG2 and 3 T3-L1 cells, BRB is said to activate AMPK, increasing FAO and decreasing lipid synthesis [46], [47]. Turning on the sirtuin1 (SIRT1), forkhead box protein O1 (FOXO1) and SREBP-2 signalling pathways, BRB could prevent HepG2 cells from developing steatosis by FFA [48]. Furthermore, BRB lowered serum TG in rats fed a high-fat diet (HFD) and improved HS in mice with diet-induced obesity. Moreover, it reduced the phosphorylation of protein kinase C (PKC) on Tyr311 and boosted the ATP-binding cassette transporter A1 (ABCA1), which regulated hepatic cholesterol and phospholipid transport [49]. Additionally, clinical studies supported the idea that BRB significantly reduced the amount of hepatic fat, specifically by inducing ceramide levels to decrease in NAFLD patients [50]. Overall, BRB therapy might be a natural alternative for treating NAFLD since it can increase FAO and decrease lipid build-up.


#

Betaine

Betaine (BET), a methyl derivative of glycine, is present in a wide range of foods, including spinach, mussels, wheat bread, and beets. It is also present in plants, animals, and microbes. According to reports, BET protects individuals against several metabolic diseases [51], [52]. For example, BET significantly reduced the severity of HS in C57BL/6 J mice by upregulating AMPK and downregulating SREBP-1c [53]. BET also lowered HS in HepG2 cells by preventing FOXO1/PPAR-α activation and reduced the effects of adenovirus-mediated FOXO1 overexpression, which noticeably elevated the mRNA expression levels of PPAR-γ and its target genes, including FAS and ACC [54]. Additionally, BET minimised methylation of the PPAR-γ promoter and increased PPAR-α expression, which brought down hepatic lipid build-up [55]. Similar outcomes were observed in rats with HFD, where BET increased hepatic lipid export and FAO by raising the expression of the PPAR-α and CPT1 genes [56]. To slow the course of metabolic illnesses, BET seems to influence various metabolic processes, including lipid absorption, transportation, and FAO.


#

Nuciferine

The leaves of Nelumbo nucifera Gaertn are the prime source of nuciferine (NUF), an active aporphine alkaloid [57], [58]. In 3 T3-L1 adipocytes, NUF has drastically lowered intracellular TG levels and improved lipid metabolism. AMPK phosphorylation and activation mediated by NUF may be the cause of the pathways [59]. In mice with diet-induced HS, NUF may aid in hepatic lipid metabolism via activating the PPAR-α/PGC1α pathway [60]. Similarly, NUF protects golden hamsters on HFD from developing HS. Supplementing with this substance dramatically reduced blood and hepatic lipid levels and boosted PPAR-α and CPT1 expression levels, leading to more FAO. Additionally, NUF therapy significantly reduced the HFD-induced rise in SREBP-1c and FAS protein levels in the liver, indicating that NUF might delay the onset of HFD-induced HS [61].


#

Matrine

Matrine (MAT) is a tetracyclic quinoline alkaloid identified in the dried roots of Sophora flavescens. It reduces blood lipid levels and has anti-inflammatory, anti-fibrosis, and anti-cancer properties [62], [63]. In China, MAT has been used as a liver-protective drug, primarily for the treatment of liver tumours and viral hepatitis [64]. By triggering the nuclear factor erythroid 2-related factor 2 (Nrf2) signalling pathway, the researchers demonstrated that MAT had a protective impact against NAFLD and reduced HS. In addition to facilitating Nrf2 translocation to the nucleus, MAT may enhance the synthesis of the antioxidant enzyme protein [65]. In mice fed an HFD, MAT has been shown to reduce HS, hepatic lipid synthesis, plasma insulin levels, and liver TG content. The activation of heat shock protein 72 (HSP72), which prevents cellular stress in the liver, was surprisingly credited with the protective effect [66]. More significantly, researchers verified that the anti-NASH activity of MAT was connected to the overexpression of HSP72 and the downregulation of the mammalian target of rapamycin (mTOR) [67].


#

Oxymatrine

A bioactive alkaloid derived from the Sophora (Kushen) plant called oxymatrine (OXM) has a variety of pharmacological properties, which include anti-pathogen, anti-inflammation, anti-hepatitis B virus infection, and immunological modulation [68]. Recently, it was shown that OXM and lipid metabolism are closely related. Rats consuming OXM have reduced liver and serum TG and total cholesterol (TC) levels. In addition, PPAR-α, CPT1A, and microsomal TG transfer protein (MTTP), which is in charge of VLDL assembly and hepatic outflow, were all significantly upregulated by OXM [69]. It was also discovered that OXM might lessen HS and liver lipid build-up in rats with NAFLD. Increased expression of genes including PPAR-α, CPT1A, and acyl-CoA oxidase 1 (ACOX1) involved in FAO is thought to be the causative factor, as well as decreased expression of genes involved in DNL, including SREBF1, ACC, and FAS [70]. Thus, OXM should successfully treat NAFLD by focusing on mechanisms involved in lipoprotein transportation and lipogenic synthesis.


#

Piperine

Piperine (PIP), a phytoconstituent present in black pepper, has anti-inflammatory, antioxidant, and anti-cancer activities [71]. The adipocyte differentiation and adipogenesis of 3 T3-L1 cells are significantly inhibited by PIP. The underlying molecular mechanism of PIP was revealed to be the inhibition of PPAR-γ expression and activity, as well as the downregulation of adipogenic transcription factors, such as SREBP-1c and C/EBP. Thus, the treatment of disorders linked to fat might benefit from using PIP [72]. Additionally, PIP might boost lipid catabolism by activating AMPK and PPAR-δ, and it can also prevent the onset of NAFLD by reducing hepatic TG build-up [73].


#

Trigonelline

Trigonella foenum-graecum is the principal source of the natural alkaloid known as trigonelline (TRG), which is also present in other food plants such as soybeans, onions, and maize. TRG demonstrates a range of therapeutic benefits, including anti-diabetic, anti-hypercholesterolemia, and anti-carcinogenic qualities [74], [75]. According to a study, TRG reduced adipocyte differentiation and fat accumulation in 3 T3-L1 cells by downregulating PPAR-γ and C/EBPα and could be effective in treating disorders connected with metabolism [76]. Western blot analysis showed that TRG markedly enhanced the expression of superoxide dismutase 1 (SOD1) in the hepatic tissues of rats receiving HFD. These proteins could protect the liver by hampering apoptosis and played a significant role in the treatment of NAFLD [77].


#

Sophocarpine

Sophocarpine (SPC), a member of the sophorae alkaloid (quinolizidines and pyridines) that is frequently used in traditional Chinese medicine, apparently successfully lowers NASH in rats and adipocytokine production, demonstrating that adipocytokines can influence hepatic lipid metabolism through the AMPK pathway [78]. In primary hepatocytes derived from particular pathogen-free rats and treated with OA, a recent study showed that SPC therapy resulted in noticeable relief from HS, reductions in leptin expression, and increases in adiponectin expression. This study also showed that SPC therapy significantly increased AMPK phosphorylation and decreased the expression of the protein hepatocyte nuclear factor-4. Additionally, SREBP-1c mRNA expression was markedly reduced by SPC. The results showed that SPC reduced HS by activating the AMPK signalling pathway [79].


#
#

Flavonoids

Flavonoids are a class of organic compounds with varying phenolic structures present in various plant components, including fruits, barks, and aerial parts. The antioxidative, anti-inflammatory, anti-mutagenic, and anti-carcinogenic qualities of flavonoids and their ability to influence important cellular enzyme activity are well known for positively impacting human health. Using FFA-induced HS in HepG2 and AML12 cells, the hepatoprotective effects of the flavonoids are discovered [80], and various flavonoids involved in retaliating against NAFLD are also enlisted below, shown in [Fig. 4]

Zoom Image
Fig. 4 Flavonoids involved in the management of NAFLD [nobiletin (I), dihydromyricetin (II), rutin (III), naringenin (IV), baicalin (V), quercetin (VI), luteolin (VII), scutellarin (VIII), genistein (IX), and fisetin (X)].

Nobiletin

Nobiletin (NOB), a polymethoxy flavonoid abundant in citrus fruit and isolated from Citrus depressa and Citrus reticulata [81], attenuated steatosis in high-glucose-treated HepG2 cells by downregulating SREBP-1c and FAS expression levels. Lipogenesis was restricted by inhibiting SREBP-1c activation and subsequent downregulation of FAS and ACC, which decreased malonyl-CoA generation upon AMPK activation. Therefore, the study concluded that NOB could effectively demolish hyperlipidemia and intracellular lipid accumulation in NAFLD [82]. Along with the upregulation of AdipoR1 expression, NOB substantially enhanced plasma levels of adiponectin. Lower amounts of malondialdehyde (MDA) in the plasma and liver tissue were associated with the downregulation of liver NADPH oxidase gp91phox subunit expression and thereby impaired ROS and MDA production in HFD-fed rats in NAFLD [83]. The activated NOD-like receptor family pyrin domain containing 3 (NLRP3) is likely to contribute to lipotoxicity and enhanced IL-1β and IL-18 expression in PA acid-induced AML-12 cells. NOB boosted the expression of SIRT1 while decreasing the expression of NLRP3, IL-1β, and IL-18. Additionally, it is proposed that SIRT1 might be a potential target to arrest the advancement of NAFLD [84].


#

Dihydromyricetin

Dihydromyricetin (DMY), a bioactive flavonoid isolated from Ampelopsis grossedentata and Cedrus deodara [85], was found to reduce lipid accumulation in OA-induced steatotic L02 and HepG2 cells. Additionally, OA-induced cell death was lessened by DMY, and peroxidation index generation was similarly reduced. By using qRT-PCR and Western blot analysis, DMY was shown to boost AMPK phosphorylation while lowering PPAR-γ expression and phosphorylating AKT. Therefore, DMY could efficiently treat NAFLD in the PPAR-γ, AMPK, and AKT signalling pathways [86]. DMY reduced NAFLD by enhancing redox homeostasis and mitochondrial respiratory capacity in PA-induced hepatocytes through a sirtuin 3 (SIRT3)-dependent mechanism by transfecting SIRT3 siRNA. Peroxisome proliferator-activated receptor gamma coactivator 1α (PGC1α) and SIRT3 expressions increased AMPK activation in steatosis-induced HepG2 cells. PGC1α activated ERRα to facilitate ERR binding to ERRE in the SIRT3 promoter, which encouraged SIRT3 mRNA production. The experiment demonstrated that DMY might boost SIRT3 expression by stimulating the AMPK-PGC1α signalling pathway [87]. A study revealed that DMY decreased oxidative stress, inflammation, and apoptosis, preventing the onset of the NAFLD rodent model. The DMY therapy increased PPAR-α expression while decreasing albumin and collagen I levels and reducing NF-κB and p53 expression levels. It can be said that DMY preferentially protected the liver against NAFLD via NF-κB/p53 signalling pathways [88].


#

Rutin

Rutin (RUT), a flavonol initially isolated from Ruta graveolens, is a plant pigment found in various fruits and vegetables [89]. The report suggests that RUT increased the activity of SOD, an antioxidant enzyme, and decreased cellular MDA, a lipid peroxidation marker, in hepatocytes. Administering RUT decreased the expression of SREBP-1c, diglyceride acyltransferase 1 and 2 (DGAT1 and DGAT2), and ACC while enhancing the expression of PPAR-α and its downstream substrates CPT1 and CPT2. By decreasing the levels of the vital autophagy biomarkers TNF-α and IL-1β, RUT has also been demonstrated to suppress the autophagic activity of liver tissues. The capacity of RUT to promote fatty acid metabolism while inhibiting lipogenesis might be the cause of its hypolipidemic effects. RUT might therefore be the possible treatment option for NAFLD [90]. In rats, HFD generated metabolic syndrome, which included obesity, dyslipidemia, hypertension, and NASH. Increased levels of glutathione peroxidase (GPx) and MDA, both biomarkers of oxidative stress, demonstrated that these clinical states are influenced by oxidative stress. These stress parameters were blocked and turned around by the RUT administration. Therefore, RUT successfully prevented these diet-related alterations by lowering liver and cardiac inflammation and oxidative stress [91].


#

Naringenin

Naringenin (NGN), a colourless flavanone exclusively available in citrus fruits [92], is beneficial against liver diseases by targeting several molecular pathways [93]. In order to encourage fatty acid oxidation and prevent lipogenesis in the liver, NGN enhanced AMPK activation. A reduction in the activity of ACC, HMG-CoA reductase (HMG-CoAR), and lipogenesis due to AMPK activation protected the fatty liver by reducing the production of TGs. In addition, the NF-κB and PPAR-α were upregulated, leading to a significant elevation of heme oxygenase 1 (HO-1) expression decrease in VLDL production in NAFLD models [94]. Furthermore, NGN reduced inflammation and lipid build-up in the liver by preventing NLRP3 activation in primary hepatocytes and HepG2 cells. However, it was also evident that NGN decreased the expression of CD68 and CD64, macrophage markers, in the livers of wild-type mice given the methionine-choline deficient (MCD) diet. Additionally, NGN lowered inflammation by inhibiting NF-κB, IL-1β, and IL-18, reducing hepatic inflammation and steatosis [95].


#

Baicalin

Baicalin (BAI) is a monomeric flavonoid in the Scutellaria baicalensis Georgi plant [96], which restricted the toll-like receptor 4 (TLR4) expressed on the immune system cell surface and parenchymal hepatocytes in the MCD diet NAFLD mice model. TNF-α, IL-1β, IL-6, and C – X-C motif chemokine ligand 2 were among the pro-inflammatory indicators that BAI also suppressed when mitogen-activated protein kinase (MAPK) and NF-κB were turned down. Therefore, hepatocyte damage, intracellular lipid accumulation, and liver fibrosis were abolished upon BAI treatment [97]. Hepatic cholestasis was also diminished by BAI via activating crucial receptors such as FXR and takeda G-protein-coupled receptor 5 (TGR5), which regulated the gene expression responsible for lipid and bile acid synthesis. Furthermore, by overexpressing AMPK and downregulating SREBP-1c, BAI helps reduce the toxicity of FFAs by preventing fat build-up in the liver. Scientists determined that BAI performed a therapeutic effect in NAFLD via the regulation of endoplasmic reticulum (ER) stress and the thioredoxin-interacting protein (TXNIP)/NLRP3 pathway in an investigation of PA-induced NAFLD HepG2 cells [98]. In addition to providing good protection against the harmful effects of an HFC-rich diet on the liver, BAI significantly reduced oxidative stress and liver inflammation in NASH patients. Furthermore, BAI might protect the liver from damage by inhibiting the activity of JNK and downregulating COX-2 and CYP2E1. Therefore, natural antioxidants like BAI might restrict NASH development [99].


#

Quercetin

Quercetin (QRC) is a bioflavonoid initially discovered in Quercus (oak genus) and reported to be available in a handful of plants, such as Ginkgo biloba, Sambucus canadensis, Oryza sativa L. [100], and Hypericum perforatum, with possible chemopreventive action and reverse lipid peroxidation [101]. In both in vitro and in vivo models of T2DM-induced NAFLD, the antioxidant QRC significantly reduced the production of IL-1β, IL-6, and TNF-α. Studies revealed that the lipid metabolism included the FXR1/TGR5 signalling pathways, which QRC enhanced to treat NAFLD [102]. Apart from possessing several beneficial effects in improving IR, QRC also ameliorated hyperlipidemia by downregulating SREBP-1c and FAS, thereby upregulating PPAR-α and LXRα expressions to accelerate cholesterol outflow from THP-1 macrophages and increasing the expression ATP-binding cassette transporter A1 (ABCA1) genes. Therefore, QRC can be a suitable molecule for NAFLD treatment [103]. In a study, QRC reduced excessive hepatic LDL-c accumulation and hepatic necrosis brought on by HFD in mice and enhanced autophagosome potential, which might have been involved in LDL-c degradation, in addition to blocking the expression of the receptors, such as macrophage scavenger receptor 1 (MSR1) and CD36, which are primarily involved in LDL-c uptake. An experiment revealed QRC has an impact on autophagy modification in the prevention of LDL-c and NAFLD [104].


#

Luteolin

Luteolin (LUT), a tetrahydroxyflavone with anti-inflammatory, anti-hypertensive, and anti-cancer properties [105], was observed to alleviate HS in the HFD-induced NAFLD mice model. The homeostatic model assessment of insulin resistance (HOMA-IR) was decreased upon LUT treatment, which was previously upregulated in the control group. Intestinal zonula occludens-1, occludin, and claudin-1 protein expressions were elevated following LUT treatment, as opposed to previously in the HFD group. Damage to the intestinal barrier allows lipopolysaccharides to reach the liver through the portal vein, activating TLR4 signalling to promote dyslipidemia and liver inflammation, exacerbating NAFLD. In addition to elevated IL-1β, IL-6, and TNF-α levels, the TLR-4 and NF-κB mRNA expressions were upregulated after LUT administration [106]. Additional in vitro research has shown that LUT can inhibit the stimulation of the LXR. This inhibited TG accumulation in HepG2 cells and primary hepatocytes. Results showed that LUT could prevent lipid build-up by overexpression of SREBP-1c both in vitro and in vivo, suggesting that it could help treat NAFLD. To summarise, the LXR-SREBP-1c signalling pathway, which was initially upregulated in the hepatocytes of the NAFLD mice model, was downregulated by LUT to decrease hepatic DNL [107].


#

Scutellarin

Scutellarin (SCU) is a flavonoid 7-O-glucuronide having cardioprotective attributes, mostly found in Salvia splendens and Scutellaria baicalensis [108]. In HFD rats, SCU prevented dyslipidemia via reducing oxidative damage. After one month of SCU treatment on the liver cells obtained from the established rat model, activated phosphoinositide 3-kinase (PI3K) and AKT expression with concomitant elevation of Nrf2 promoted the expression of HO-1 and NADPH quinone dehydrogenase 1 (NQO1). Therefore, the findings by Western blot suggested that SCU could be a necessary antioxidant in NAFLD treatment. Furthermore, these findings indicated that SCU prevented NAFLD by potentially reducing oxidative stress in the HFD rat model [109]. In the experiment conducted on in vitro HepG2 cells and in vivo HFD NAFLD mice models, SCU considerably lowered hyperlipidemia and boosted antioxidant capacity. Additionally, SCU administration markedly suppressed NF-κB and KEAP1 while activating PPAR-γ, Nrf2, PGC1α, HO-1, glutathione S-transferase (GST), and NQO1. The results indicated that SCU had potent antioxidative, hepatoprotective, and hypocholesterolemic properties that might effectively cure NAFLD via the PPAR-γ/PGC1α-Nrf2 signalling pathway [110].


#

Genistein

Genistein (GNT), an isoflavonoid isolated from Glycine max, commonly known as soybean, has been found effective in treating cancer and obesity, while currently showing a therapeutic role in retarding NAFLD [111]. An increase in plasma thromboxane A2 (TXA2) and the expression of TBXA2R in the HepG2 cells and liver of the HFD-induced NAFLD model were indicators that the TXA2 pathway was active. GNT, an isoflavonoid derived from soybean, targeted the COX-1 activity, inhibited TXA2 production, and affected NAFLD development by reducing insulin sensitivity. This research demonstrated the critical mechanism of GNT in the advancement of NAFLD in the TXA2 pathway [112]. Steatosis-induced primary human hepatocytes (PHHs) exhibited increased PPAR-α protein expression and elevated CPT1 and ACSL1. In addition, GNT therapy downregulated PPAR-α expression and reverted the activated SREBP-1c protein in steatotic PHHs [113].


#

Fisetin

Edible fruits and vegetables, including strawberries, grapes, and onions, contain the bioactive flavonol known as fisetin (FST), preventing LDL-c oxidation [114]. In the hepatocytes of HFD-induced obese mice, FST treatment also increased the production of SIRT-1 and CPT1 and decreased the levels of HS, FAS, and leptin. It also considerably boosted AMPK phosphorylation. Furthermore, FST accelerated lipolysis and β-oxidation in hepatocytes. According to this investigation, FST might improve NAFLD and hepatic lipid metabolism in mice via activating the SIRT1/AMPK and β-oxidation pathways [115]. Despite enhancing Nrf2 in the liver exposed to PA, FST substantially reduced cellular and mitochondrial ROS generation, inhibiting the inflammatory response and lipid deposition. Furthermore, FST strongly inhibited ER stress induced by PA. Surprisingly, ER stress inhibition governed by glucose-regulated protein 78 (GRP78) was substantially responsible for ROS production inhibited by FST [116]. In a rat model of NAFLD brought on by high fat/high sugar, FST decreased lipid accumulation in hepatocytes. The administration of FST resulted in an upregulation of HNF4-α gene expression and lipin-1 signalling. Additionally, it led to a decrease in oxidative stress, inhibition of ROS-induced synthesis of TXNIP, and activation of PARP1. Therefore, FST could offer protection from NAFLD and slow disease progression [117].

Apart from the discussed compounds, some alkaloids that contribute to reverting NAFLD are listed in [Table 2].

Table 2 Structure of flavonoids targeting biomarkers involved in NAFLD.

Compound name with structure

Biological source

Research model

Biomarkers

Ref

Quercetin-3-O- β -D-glucuronide

Polygonum perfoliatum and Polygonum aviculare

In vitro RAW264.7 cells; in vivo primary hepatocytes of HFD-fed rats

Downregulated SREBP-1c and FAS; upregulated PPAR-α, CPT1, and MCAD

[216], [217]

Quercitrin

Xylopia emarginata and Lotus ucrainicus

In vivo ovariectomised mice

Downregulated TNF-α, IL-1β, and IL-6

[218], [219]

Vitexin

Prosopis cineraria and Acer palmatum

In vivo liver tissue of HFD-fed mice

Downregulated PPAR-γ, SREBP-1c, ACC and FAS; upregulated PPAR-α, CPT1A, and AMPK

[220], [221], [222]

Chrysin

Passiflora caerulea and Passiflora incarnata

In vitro HepG2 cells; in vivo MCD mice, and HFD-fed rats

Downregulated PKC, HNF4-α, TNF-α, SREBP-1c and IL-6; upregulated PPAR-α

[223], [224], [225]

Galangin

Alpinia officinarum

In vitro OA : PA (2 : 1)-induced HepG2 cells; in vivo streptozotocin-induced hyperglycaemic rats and HFD-fed rats

Downregulated CD36, SREBP-1c and ChREBP; upregulated PPAR-α, CPT1A

[226], [227]

Eriocitrin

Citrus limon

In vitro PA-induced HepG2 cells; in vivo diet-induced obesity zebrafish and HFD-fed obese mice

Downregulated FAS, PPAR-γ, ACC, SCD1, and CD36; upregulated PPAR-α, CPT1A, PGC1α, and UCP1

[228], [229]

Silymarin

Silybum marianum L.

In vitro PA-induced HepG2 cells; in vivo HFD-fed mice and fructose-induced mice

Downregulated PARP, SREBP-1, FAS, CPT1A, GRP78 and XBP-1; upregulated PPAR-α, PPAR-δ, SIRT1, and AMPK

[230], [231], [232]

Hesperetin

Citrus genus

In vitro OA-induced HepG2 cells and in vivo HFD-fed and primary hepatocytes of rat

Downregulated NF-κB, TNF-α, IL-6; upregulated Nrf2, PI3K, AKT, SOD, GPx, HO-1, XBP-1, and GRP78

[233], [234], [235]

Hesperidin

Citrus sinensis

In vitro OA-induced HepG2 cells; in vivo HFD-fed mice

Downregulated SREBP-1c, ACC and FAS; upregulated AMPK

[236], [237]

Bavachinin

Psoralea corylifolia L. and Fructus Psoraleae

In vitro PA-induced HepaRG cells

Downregulated FDFT1, SREBP-2, AKT and mTOR

[238], [239]

Didymin

Citrus genus and Mentha spicata

In vitro insulin-resistant HepG2 cells; in vivo HFD-fed mice

Downregulated SREBP-1c, ACC1, FAS, TLR4, NF-κB and LXRα

[132], [240]

Ugonin J

Helminthostachys zeylanica

In vitro PA-induced HuS-E/2 cells; in vivo HFD-fed mice

Downregulated SREBP-1c; upregulated AMPK, ACC, AKT and CPT1

[241]

Hyperoside

Hypeticum perforatum and Artemisia capillaris

In vivo HFD-fed mice

Downregulated SREBP-1, SREBP-2 and ACC; upregulated CYP7A1, FXR, LXRα, and NR4A1

[242], [243]

Icariin

Epimedium genus

In vivo HFD-fed mice

Downregulated SREBP-1c and DGAT2; upregulated CPT1, ACC, PGC1α, and GLUT4

[244]

Xanthohumol

Humulus lupulus

In vivo HFD-fed mice and T2DM-induced steatosis rats

Downregulated PPAR-γ and NF-κB; upregulated Nrf2

[245], [246], [247]

Breviscapine

Erigeron breviscapus (Vant.) Hand. -Mazz.

In vivo HFD-fed, MCD, and HFHC diet-fed mice

Downregulated NF-κB, TAK1, FAS, ACC, SCD1, CD36, FABP1, and PPAR-γ; upregulated PPAR-α and CPT1A

[248], [249]

Myricetin

Euphorbia tirucalli L.

In vivo HFD-fed, and ob/ob mice

Downregulated ChREBP, SREBP-1c, TNF-α, IL-6, and PPAR-γ; upregulated Nrf2, SOD, GPx, HO-1, and NQO1

[250], [251], [252]


#
#

Glycosides

Glycosides are phytochemicals in which the sugar moiety has a glycosidic linkage with another functional group. They play various essential functions of glycosides in phytopharmacology. Inactive glycosides are the primary type of chemical storage in many plants. These can be made active by enzyme hydrolysis, which separates the aglycone part from the sugar component and makes it therapeutically active [118]. The crucial molecular targets of glycosides are sterol regulatory element-binding protein 1 (SREBP-1), ACC, FAS, NF-κB, PI3K-AKT pathway, PPAR-α, and several other pathways. Thus, this class of phytochemicals is a crucial player in controlling the progression of NAFLD. Some of the widely discovered glycosides are summarised below ([Fig. 5]) and portrayed in [Table 3].

Zoom Image
Fig. 5 Glycosides involved in the management of NAFLD [swertiamarin (I), glycyrrhizic acid (II), salidroside (III), and didymin (IV)].

Table 3 Structure of glycosides targeting biomarkers involved in NAFLD.

Compound name with structure

Biological source

Research model

Biomarkers

Ref.

Geniposide

Gardenia jasminoides

In vitro OA and PA-induced HepG2 cells; in vivo tyloxapol

(Ty)-induced mice

Downregulated SREBP-1c, MPO, mTORC, and PI3K; upregulated Nrf2, HO-1, PPAR-α, and AMPK

[253], [254]

Clitorin

Carica papaya L.

In vitro OA-induced HepG2 cells; in vivo male Western diet-fed mice

Downregulated SREBP-1, PPAR-γ, C/EBPα, LXR and HMGCR, ACC, and FAS; upregulated PPAR-α, CPT1, and AMPK

[255]

Gentiopicroside

Gentianaceae family

In vitro OA and PA at (2 : 1)-induced HepG2 cells; in vivo Ty-induced hyperlipidemia mice

Downregulated SREBP-1c and MDA; upregulated PI3K, AKT, Nrf2, NQO1, HO-1, PPAR-α, and SOD

[256]

Diosgenin

Dioscorea villosa and Rhizoma Dioscoreae Nipponicae

In vitro high glucose treated HepG2 cells; in vivo HFD-fed rats

Downregulated SREBP-1c, LXR; upregulated AMPK, and ACC

[257]

Schaftoside

Herba Desmodii Styracifolii

In vitro OA-induced Huh-7 cells and FXR knockout mice primary hepatocytes; in vivo HFD-fed mice

Downregulated SREBP-1 and CYP7A1; upregulated FXR

[258]

Swertiamarin

Swertiamarin (SWT), a bitter secoiridoid glycoside, improves IR in T2DM and has lipid-lowering properties. It is the main constituent in Enicostemma littorale Blume and other medicinal plants from the Gentianaceae family [119]. By maintaining membrane integrity and preventing apoptosis, as seen by decreased cleavage of caspase 3 and PARP1, SWT effectively lowered TG levels and drastically reduced lactate dehydrogenase release activity. With a concurrent decrease in p307 insulin receptor substrate 1 (IRS1), SWT also markedly boosted the expressions of essential insulin signalling proteins like IR, PI3K, and AKT. Noticeably, SWT controls hepatic glycemic load, fat deposition, IR, and ROS in HS by collectively targeting putative metabolic regulators AMPK and PPAR-α [120]. In a recent study, SWT was found to reduce weight gain, glucose intolerance, oxidative stress, and IR caused by HFD in mice and improve insulin signalling. The SWT-treated animals showed enhanced lipolysis and decreased adipocyte hypertrophy and macrophage infiltration in epididymal white adipose tissue (eWAT) compared to HFD-fed mice. In the liver of obese mice, SWT reduced HS and inflammation brought on by HFD by inhibiting p38 MAPK and NF-κB pathway activation in the eWAT [121]. In HFD, SWT administration to fructose-fed mice also decreased levels of ALT, AST, uric acid, blood glucose, and TGs. Histological analyses revealed that SWT therapy could reduce hepatic ballooning degeneration and steatosis. Additionally, SWT treatment reduced hepatic oxidative stress and hepatic pro-inflammatory cytokines, which was connected to a decline in hepatic xanthine oxidase activity and an increase in antioxidant defence system enzymes as activation of Nrf2 in mice fed fructose. Furthermore, SWT also reduced the expression of ACC1, FAS, and SREBP-1 in the liver of mice administered with fructose [122].


#

Glycyrrhizic acid

Glycyrrhiza glabra, an edible and medicinal plant with a long history of usage in Ayurveda and traditional Chinese medicine, yields a triterpene glycoside known as glycyrrhizic acid (GA) [123]. The relative liver weight, serum ALT and AST activity, lipid levels, blood glucose, and insulin levels were all dramatically decreased by GA therapy. In addition, GA reduced the build-up of lipids in the liver. Further research into the mechanism of action of GA revealed that it decreased the expression of SREBP-1c, FAS, and SCD1, raised the expression of PPAR-α, CPT1, and ACADS, and enhanced lipid metabolism via activating LPL. Additionally, GA boosted glycogen synthesis by inducing PDase and GSK3β gene expressions while decreasing gluconeogenesis by suppressing PEPCK and G6Pase. Furthermore, GA improved insulin sensitivity by upregulating IRS1 and IRS-2 phosphorylation [124]. Other research suggested that by lowering the levels of SREBP-1c, FAS, ACC1, and SCD1 in the liver, GA treatment decreased hepatic lipogenesis. It also increased lipid metabolism by inducing PPAR-α, CPT1A, ACADS, and LPL. Through a reduction in the expression of monocyte chemoattractant protein 1 (MCP1) and VCAM-1 in the liver, GA also decreased hepatic inflammation. By limiting HSC activation and collagen deposition, GA also decreased liver fibrosis. In conclusion, GA significantly reduced the risk of NASH in mice brought on by the MCD diet [125].


#

Salidroside

Salidroside (SAD), a phenylpropanoid glycoside [2-(4-hydroxyphenyl)-ethyl-β-d-glucopyranoside], is the main bioactive phytoconstituent isolated from Rhodiola rosea L. SAD has been studied for its potential biological effects, including its anti-inflammatory, antioxidant, antifibrotic, and neuro- and cardioprotective activities [126]. Interestingly, SAD improved oxidative stress and mitochondrial damage while reversing the damage caused by PA in a dose-dependent manner in AML-12 cells. It also protected against hepatic lipotoxicity by inhibiting TLR4/MAPK and p53 activation in AMPK and AKT pathways. Furthermore, SAD-reduced lipid accumulation was likewise influenced by TLR4 suppression [127]. According to another study, SAD treatment significantly improved their insulin sensitivity by reducing obesity, hepatic fat deposition, and blood glucose fluctuation caused by HFD in mice. Moreover, SAD also reduced the production of TXNIP and the activation of the NLRP3 inflammasome in the liver by increasing AMPK activity and insulin sensitivity in cultured hepatocytes by regulating lipid accumulation, ROS formation, and NLRP3 inflammasome activation. The positive effects of SAD on hepatocytes were suppressed due to inhibiting AMPK activation by an inhibitor or short interfering RNA [128]. While reducing hepatic tissue vacuolation and steatosis, SAD also markedly decreased liver TG and FFA levels. Treatment with SAD improved serum dysregulation of fasting blood sugar (FBS), HOMA-IR, ALT, and AST. In addition, SAD significantly increased the levels of important insulin signalling pathway molecules such as phosphorylated IRS1, PI3K, and PKB in the liver while considerably lowering the levels of SREBP-1c, curing the NASH condition [129].


#

Didymin

Didymin (DYM) is an orally bioactive dietary flavonoid glycoside in citrus fruits, including oranges, lemons, etc. [130], [131], which was found to decrease lipid imbalance and hepatocyte damage dramatically. Furthermore, DYM significantly reduced hepatocyte apoptosis by controlling the expressions of the caspase family and the B-cell lymphoma-2 (BCL-2) family. It also reduced the expression of TLR4, as well as the phosphorylation of NF-κB, showing that DYM inhibits the TLR4/NF-κB pathway. DYM similarly suppressed the PI3K/AKT pathway, as evidenced by the reduction in PI3K and AKT phosphorylation levels [132]. Recent research suggested that DYM dramatically reduced hepatic damage and fibrogenesis by CCl4, low transaminase activity, and reduced collagen build-up. In addition, it markedly reduced hepatocyte apoptosis by restoring the expression of the BCL-2 and caspase families and the mitochondrial dysfunction. It also significantly reduced the production of IL-1β and IL-6 while inhibiting the glycerophospholipid metabolism pathway by reducing the production of phosphatidylethanolamines and phosphatidylcholines [133].


#
#

Phenols and Polyphenols

Phenolic compounds are the primary antioxidants and are essential for human health, and they primarily act as free radical scavengers by hindering lipid oxidation. This reduces the formation of rancidity-causing decomposition products like aldehydes and ketones [134]. Dietary polyphenols are secondary plant metabolites with antioxidant capabilities that work as antioxidants to prevent the development of chronic illnesses. The phytochemicals in the polyphenolic subclass might have anti-cancer, anti-diabetic, and anti-neurodegenerative properties [135]. This section extensively investigates phenolic and polyphenolic compounds proven beneficial in treating NAFLD, as illustrated in [Fig. 6] and [Table 4].

Zoom Image
Fig. 6 Phenols and polyphenols involved in the management of NAFLD [mangiferin (I), resveratrol (II), curcumin (III), epigallocatechin-3-gallate (IV), hydroxytyrosol (V), rosmarinic acid (VI), and ellagic acid (VII)].

Table 4 Structure of phenols and polyphenols targeting biomarkers involved in NAFLD.

Compound name with structure

Biological source

Research model

Biomarkers

Ref

Dihydrocurcumin

Curcuma longa

In vitro OA-induced L02 and HepG2 cells

Downregulated PNPLA3, SREBP-1c; upregulated PPAR-α, CYP4A, PI3K, AKT, and Nrf2

[259]

Cichoric acid

Echinacea purpurea

In vitro PA-induced L02, AML-12 and HepG2 cells; in vivo larval zebrafish and MCD mice

Downregulated MDA, NF-κB, KEAP1, SREBP-1c, DGAT1, FAS, and SCD1; upregulated SOD, AMPK, Nrf2, and HO-1

[260], [261], [262]

Gallic acid

Jatropha podagrica

In vivo HFD rats

Downregulated TNF-α, NF-κβ, IL-6, IL-17, IL-1β, FAS, ACC-α, and HMGCR; upregulated Nrf2, HO-1, GST, SOD, and GPx

[263], [264], [265]

Diphlorethohydroxycarmalol

Ishige okamurae

In vitro PA-induced HepG2; in vivo transgenic zebrafish

Downregulated SREBP-1c, ChREBP, FAS, IL-1β, and TNF-α; upregulated AMPK and SIRT1

[266]

Catechin

Camellia sinensis

In vitro 3 T3-L1 preadipocytes and in vivo high fructose-fed rat

Downregulated TNF-α, MCP1, JNK, p38, AP-1, TLR4, TNFR1, and NF-κB; upregulated Nrf2

[267], [268]

Chlorogenic acid

Coffea arabica

In vivo HFD and db/db mice

Downregulated SREBP-1c, LPL, PPAR-γ, FAS, C/EBPα, and TGF-β1; upregulated PPAR-α, AMPK, AdipoR1 and AdipoR2

[269], [270], [271]

Salvianolic acid A

Radix Salvia miltiorrhiza

In vitro PA-induced HepG2 cells; in vivo HFD and high-carbohydrate diet mice

Downregulated ChREBP, FAS, MDA, TNF-α, TXNIP, NLRP3, IL-1β and IL-6; upregulated LC3-II AMPK, SIRT1, SOD

[272], [273]

6-Gingerol

Zingiber officinale

In vivo HFD and high-cholesterol diet NMRI mice

Downregulated ChREBP, DGAT2, and FOXO1; upregulated AKT, PPAR-α, CPT1α, and SIRT3

[274], [275]

Methyl brevifolincarboxylate

Canarium album and Geranium carolinianum

In vitro OA-induced SK-HEP-1 cells

Downregulated SREBP-1c, FAS, ACC1, TNF-α, NF-κβ, IL-6, IL-8, and IL-1β; upregulated PPAR-α, CPT1, ACOX1, and AMPK

[276], [277]

Mangiferin

Mangiferin (MGN) is a glucosylxanthone polyphenol primarily isolated from Mangifera indica [136], possessing antioxidant as well as anti-diabetic, hypolipidemic, and cardioprotective properties [137]. This phytocompound promoted glucose metabolism and diminished hepatic TG accumulation by reducing NLRP3 and IL-1β transcription levels while upregulating phosphorylated AMPK in both in vitro HepG2 cells and in vivo HFD-induced mice models. The authors have also mentioned that by activating AMPK, MGN elevated IR, which could be a promising agent to cure NAFLD [138]. The effective concentration of MGN for inhibiting inflammatory cytokines NF-κB and activating PI3K was 30 mg per kg. The protein level of hepatic phosphorylated JNK was elevated in the HFD-induced NAFLD model, which was downregulated after MGN treatment to suppress HS. Deprivation of mTOR and AMPK stimulation led to the masking of SREBP-1, which prevented DNL. The findings demonstrate that MGN improved lipid accumulation in NAFLD by promoting autophagy and inhibiting lipogenesis via AMPK/mTOR pathway activation [139]. In the liver of seven-week-old male HFD hamsters, MGN elevated the mRNA expression of PPAR-α, CD36, and CPT1 while downregulating SREBP-1c, ACC, DGAT2, and microsomal TG transfer protein genes responsible for lipogenesis [140].


#

Resveratrol

Resveratrol (RVT) is a polyphenol that is available in dietary plant sources such as grapes, apples, blueberries, and peanuts and is particularly abundant in Polygonum cuspidatum. Hyperlipidemia and inflammation have been cured by RVT [141]. Dephosphorylation of phosphatidic acid is catalysed by lipin-1 to produce DAG, which is necessary to produce lipids. RVT reduced HS and lipid deposition in HepG2 cells primarily by upregulating AMPK and ACC while suppressing SREBP-1c and lipin-1 expression [142]. Genes responsible for FAO, such as CPT1 and ACOX1, were elevated in the HFD mice group, reversely expressed upon RVT treatment. In addition, lipogenic genes, viz., GPAT1 and DGAT2, along with cytosolic FABP1 and FABP2 proteins, were suppressed by RVT. TLR4 and myeloid differentiation primary response 88 (MyD88), as well as inflammatory mediators IL-1β and TNF-α, were elevated in the liver of the NAFLD model, but these alterations were abolished in the RVT group [143]. Overfed zebrafish model exhibited higher TG in plasma by lowering AMPKα, SIRT1, and PGC1α expression, which were upregulated by RVT, suggesting that it could lower plasma TG primarily activating the AMPKα-SIRT1-PGC1α pathway. Finally, RVT also suppressed CAV-1 and PPAR-γ while elevating LC3-II protein levels [144].


#

Curcumin

Curcumin (CRM) is derived from the Curcuma longa rhizomes and has an extensive range of therapeutic benefits, including anti-inflammatory and anti-mutagenic effects [145], [146]. O-GlcNAc transferase (OGT) and O-GlcNAcase are the only two enzymes responsible for the crucial post-translational modification known as O-glcNAcylation. In MCD diet mice, O-linked β-N-acetylglucosamine (O-GlcNAc) modification is linked to developing HS into hepatitis. Administration with CRM reduces the degree of HS via reducing O-GlcNAcylation on NF-κB in inflammatory signalling. Conversely, CRM administration dramatically increased the expression of SOD1 and SIRT1 while downregulating ChREBP [147]. Besides lowering LDL-c and FFA levels in the serum of the NAFLD mice model, CRM suppressed SREBP-1c, extracellular-signal-regulated kinase (ERK), TNF-α, and JNK expressions, as confirmed by Western blot [148]. Eventually, CRM might enhance FAO via the elevation of AMPK and PPAR-α, decreasing SREBP-1 expression and FAS transcription [149]. A clinical study of subjects with evidence of HS had low AST and ALT values with reduced pro-inflammatory high-sensitivity C-reactive protein and TNF-α levels in the serum [150].


#

Epigallocatechin-3-gallate

Epigallocatechin-3-gallate (EGCG) is an abundant polyphenol in green tea (Camellia sinensis) and possesses a potent antioxidant property that significantly blocked the expressions of TGF-β1, α-smooth muscle actin (α-SMA), SMAD 2, and SMAD 4, which were elevated in female HFD Sprague–Dawley rats. In the NAFLD model, EGCG reduced the expression of matrix metalloproteinase-2 (MMP-2) and the level of TIMP-2 protein. Moreover, EGCG therapy reversed NAFLD-induced changes in the expression of FOXO1 and p27kip and enhanced the phosphorylated PI3K and AKT [151]. A rate-limiting enzyme called glycogen synthase kinase 3β (GSK3β) controls many signalling pathways, including hepatic glycogen production and lipogenesis. In addition to triggering the enzymes involved in the insulin-signalling pathway, phosphorylation of GSK3β also controls glycogen production in the liver by raising glycogen synthase (GS) levels. This GS expression in HepG2 cells treated with EGCG was substantially increased; AMPKα and ACC expressions were significantly higher than in glucose-induced lipogenic cell lines [152]. The NAFLD progression could be restricted as the bile acid synthesis was hampered by the elevation of the cholesterol 7 alpha-hydroxylase (CYP7A1) enzyme by EGCG [153].


#

Hydroxytyrosol

Hydroxytyrosol (HT), the predominant phenolic compound in the fruit and leaves of Olea europaea L., showed a significant decrease in TC and TG and higher HDL-c levels in hyperlipidemic rabbits [154]. mRNA expression of PPAR-α, CPT1A, and fibroblast growth factor (FGF21) was elevated along with ACC in HT-treated rats. Interestingly, HT reduced hepatic inflammation and HIR on downregulation of TNF-α, IL-6, MDA, and COX-2 by HT to combat steatosis [155]. When mice on the HFD were given HT, the physiological changes that the diet caused in the liver were lessened by counteracting the pro-lipogenic, pro-inflammatory, and oxidative stress states. These impacts could be related to the PPAR-α and Nrf2 stimulation and the silencing of NF-κB [156]. Stress-induced mitochondrial dysfunction is prevented by PTEN-induced kinase 1 (PINK1). Ultimately, HT boosted PINK1 and AMPK expressions in the HFD zebrafish liver model, which suggested a reduction in hepatic lipid accumulation and mitochondrial dysfunction, activating the AMPK/PINK1 pathway [157].


#

Rosmarinic acid

Rosmarinic acid (RA) is named after rosemary, a polyphenol primarily isolated from Rosmarinus officinalis belonging to the Lamiaceae family [158]. Hepatic ROS generation was reduced as RA efficiently increases the activities of both enzymatic antioxidants, viz., SOD, CAT, and GPx, and non-enzymatic antioxidants, viz., GSH in HepG2 cells. RA significantly diminished ER stress to revert NAFLD by suppressing the expressions of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) and ATF6. With RA treatment in the steatotic HepG2 cells, overexpression of the vital autophagic markers, including LC3-II, Beclin-1, ATG5, and ATG7, was noticed [159]. Findings from both in vivo and in vitro experiments showed that the PGC1α level improved noticeably following RA treatment compared to levels in both NAFLD model groups. In contrast, the yes-associated protein 1 (YAP1) levels and TAZ considerably dropped. However, through silencing YAP1, the activity of RA was severely thwarted [160].


#

Ellagic acid

Ellagic acid (EA) is a bioactive polyphenol in various fruits and vegetables, including strawberries, almonds, and pomegranates. In streptozotocin-induced diabetic rats, EA reduced HS by triggering AMPK. This AMPK could cure NAFLD by reducing lipid synthesis by preventing SREBP-1/2, FAS, and HMG-CoAR from overexpressing and activating PPAR-α and CPT1A. EA also decreased NF-κB translocation into the nucleus, ROS, MDA, TNF-α, and IL-6 in the liver while increasing Nrf2, GSH, and SOD [161]. EA treatment lowered the level of AKT and reduced the activity of ribosomal protein S6 (RPS6) and SREBP-1, the two downstream effectors of the AKT/mTORC1 pathway. The results suggested that the EA-mediated reduction in SREBP-1, FAS, and ACC was suppressed both in vitro and in vivo. The research revealed a unique approach by which EA reduced AKT-triggered DNL [162]. By overexpressing Nrf2 and downregulating p47phox, EA demonstrated antioxidant advantages, which ultimately led to a decrease in HIF-α expression. Additionally, EA enhanced HIR by elevated AKT phosphorylation, therefore pointing to the possibility that EA could be used to treat HS in NAFLD [163].


#
#

Terpenoids

Terpenoids, which range in size from minor lipophilic cannabinoids to lipophilic pentacyclic triterpenes and polar saponins, are the largest class of chemicals that can modulate PPARs [164]. In addition, terpenes, which are chemically hydrocarbons, have been found to have antioxidant, anti-inflammatory, anti-hyperglycemic, and immunomodulatory properties in various plant secondary metabolites [165]. Some essential terpenoids with remarkable attributes to combat NAFLD are enlisted below and represented in [Fig. 7].

Zoom Image
Fig. 7 Terpenoids involved in the management of NAFLD [ursolic acid (I), oleanolic acid (II), tanshinone IIA (III), lycopene (IV), fucoxanthin (V), and betulinic acid (VI)].

Ursolic acid

Ursolic acid (UA), a pentacyclic triterpenoid, is primarily present in Fructus Ligustri Lucidi and isolated from Lantana camara Linn. [166], [167]. Impairing hepatic ER stress and boosting β-oxidation of lipids are presumably the mechanisms by which UA supplementation greatly reduced the occurrence of NAFLD and liver damage in db/db mice with upregulation of PPAR-α. The UA treatment reduced inositol-requiring protein 1α (IRE1α) and phosphorylation of JNK. An important marker of ER stress, CHOP (C/EBP homologous protein), was also inhibited by UA. In PA-induced L02 cells showing elevated ER stress, proteins such as IRE1α, PERK, and JNK were significantly restricted with UA administration [168]. By decreasing DGAT-mediated processes in mRNA and protein levels, UA significantly decreased HS caused by HFD via the increase in PPAR-α, CPT1, and TGR5. Lipogenic enzymes like ACC and FAS, along with SREBP-1c, were downregulated by UA treatment. Pro-inflammatory cytokines such as TNF-α, MCP1, and interleukins were markedly reduced on UA administration. The oxidative stress-combating ability of UA was also analysed as serum SOD, and MDA levels were reversed [169]. T0901317 is a potent, high-affinity LXR agonist. In a T0901317-induced animal model, UA severely reduced the LXRα and SREBP-1c, lowering the hepatocellular fat level. The mRNA expression of SREBP-1c, FAS, SCD1, and ACC was markedly downregulated upon inducing UA. The small heterodimer partner-interacting leucine zipper protein (SMILE) and AMPK phosphorylation were influenced by UA in hepatocytes. However, steroid receptor coactivator-1 (SRC-1) binding to the SREBP-1c promoter area was suppressed [170].


#

Oleanolic acid

Oleanolic acid (OLA) is a pentacyclic triterpenoid abundant in Olea europaea [171], [172]. By lowering lipid accumulation and providing hepatoprotection in NAFLD, dietary supplementation with OLA delays the onset of high-fructose-induced NAFLD [173], which reduced the risk of developing pre-diabetes-related NAFLD by reducing HS, which lowered hepatic inflammation. Moreover, OLA controlled hepatic oxidative stress by reducing MDA levels and elevating GPx and SOD antioxidants. Plasma SREBP amount was also significantly lowered by OLA [174]. In hepatic cells, OLA confined SRC-1 to the SREBP-1c promoter region while increasing AMPK phosphorylation and SMILE. This phytonutrient OLA could be an antagonist of LXRα as it suppressed the expression of lipogenic genes such as LXRE- and SREBP-1c [175]. For lipid metabolism, OLA may overexpress genes including AMPK and GLUT-4. Noticeably, OLA highly expresses β-oxidation of fatty acids regulated by CPT1. Adiponectin was increased by onefold, and so was the AdipoR1 gene expression upon OLA treatment. Inflammatory biomarkers, viz., TNF-α, IL-6, and MCP1, were enhanced to establish that OLA might be a powerful anti-NAFLD agent [176].


#

Tanshinone IIA

Tanshinone IIA (TSO) is a terpenoid primarily obtained from the roots of Salvia miltiorrhiza Bunge [177], [178], which markedly lessened lipid accumulation in the liver of HFD-fed Sprague–Dawley rats by silencing TNF-α and IL-6 proteins. An in vivo study showed that TSO suppressed TLR4, as well as the NF-κB signalling pathways. The mRNA and protein expression of TLR4, MyD88, NF-κB, and IKKβ were suppressed by TSO. Deregulated PGC1α was enhanced upon TSO induction comparison to the HFD group [179]. By preventing the development of neutrophil extracellular traps linked to the production of MPO, TSO might diminish inflammatory TNF-α and IL-6 markers to control hepatocyte death in MCD diet-fed NASH mice. Overexpression of TGF-β was also reverted by TSO via SMAD signalling [180]. Furthermore, TSO minimised oxidative stress by reducing the formation of ROS and MDA while increasing the activities of SOD and GPx, which could also help to prevent cytotoxicity and improve HS [181].


#

Lycopene

Lycopene (LP) is a tetraterpenoid found in a wide range of vegetables in the kitchen and has been found to lower health hazards like CVD, prostate cancer, and other malignancies [182]. HDL-c levels were raised by LP, which also reduced MDA levels with the capability to treat NAFLD. In addition, serum amyloid A and TNF-α promoting ROS generation contributing to liver damage were inhibited. Supplementing Wistar rats with a hypercaloric diet increased their levels of SOD and CAT enzymes [183]. The hepatoprotective action of LP against HFD-induced NAFLD was observed via inhibition of TNF-α, MDA, and CYP2E1 levels. In addition, LP also escalated the SOD and GSH [184]. By promoting PPAR-α activity, LP dramatically reduced the build-up of lipids in HepG2 cells exposed to PA. Additionally, it had been demonstrated that LP inhibited mitochondrial dysfunction by increasing the expressions of the AMPK, SIRT1, and PGC1α pathways for mitochondrial biogenesis. In mice given a Western diet, LP treatment significantly decreased fat accumulation by increasing PPAR-α activity. In order to demonstrate its antioxidant properties, LP also boosted the production of mitochondrial complexes, HO-1, and NQO1 [185].


#

Fucoxanthin

Fucoxanthin (FX), a carotenoid abundant in the brown marine algal family Phaeophyceae, has exceptional biological features [186]. Dietary FX treatment dramatically reduced the build-up of hepatic lipids, alleviating the symptoms of hepatic damage by suppressing mRNA expression of inflammatory cytokines, such as TNF-α, IL-6, IL-1β, and MCP1, as well as the expression of infiltration markers, such as F4/80, C–C chemokine receptor type 2 (CCR2), and Ly6c. Fibrogenic genes, viz., TGF-β1, collagen type I alpha 1 chain (COL1α1), and TIMP-1, were negatively expressed by FX. At both mRNA and protein levels, FX reduced α-SMA, thereby decreasing steatosis [187]. Interestingly, FX downregulated the essential lipid metabolism-operated transcription factors such as SREBP-1c and FAS while upregulating the downstream proteins, viz., PPAR-α, AMPK, ACC, and CPT1. After FX induction, antioxidant proteins like HO-1 and NQO1, along with Nrf2, were restored, while KEAP1 was elevated. The TLR4/MyD88 signalling pathway was significantly activated by FX [188].


#

Betulinic acid

Betulinic acid (BETA) is a pentacyclic triterpenoid isolated from the bark of Betula alba [189], which reversed the mRNA overexpression of SREBP-1c, FAS, SCD1, and ChREBP. Compared to the HFD-fed mice group, TNF-α, IL-6, and IL-1β gene expression significantly decreased in the BETA-treated group. As determined by a Western blot study, BETA therapy lowered the levels of PERK, EIF2, XBP-1, and GRP78 proteins [190]. The BETA administration stimulated both CAMKK and AMPK. Hepatocytes exposed to BETA showed decreased SREBP-1, mTOR, and S6 kinase (S6K) protein contents, demonstrating that the CAMKK-AMPK-SREBP-1 signalling cascade was responsible for the BETA-induced decrease in HS [191]. The triterpenoid BETA behaved as an FXR agonist by directly binding to it in HepG2 cells to elevate SHP, ABCC2, SULT2A1, and CYP7A1 gene expressions. Protein levels of PPAR-α and CPT1A were significantly increased upon BETA treatment. An in vivo study revealed that SHP and PGC1α mRNA levels dramatically rose after BETA exposure, whereas the expression quantities of the genes for HNF4-α, PCK1, and SCD1 dropped significantly [192].

Some bioactive terpenoids suppressing NAFLD are also enrolled in [Table 5].

Table 5 Structure of terpenoids targeting biomarkers involved in NAFLD.

Compound name with structure

Biological source

Research model

Biomarkers

Ref.

Lutein

Chlorella vulgaris

In vivo HFD-fed rats and hypercholesterolemic diet-fed guinea pigs

Downregulated MDA, TNF-α, NF-κB, and IL-1β; upregulated PPAR-α, SIRT1, IRS2, PI3K, and GLUT2

[278], [279], [280]

Astaxanthin

Haematococcus pluvialis

In vitro OA and PA-treated L02 and HepG2 cells; in vivo HFD-fed mice

Downregulated SREBP-1c, ACC, FAS, CD36, CPT1α, FOXA, PGC1α, TNF-α, and IL-1β; upregulated AMPK and Nrf2

[281], [282], [283]

β-cryptoxanthin

Capsicum annuum L.

In vivo high carbohydrate diet-fed mice

Downregulated ACC, FAS, and SCD1; upregulated PPAR-α, ACOX1, CYP7A1, SHP, SIRT1, and AMPK

[284], [285]

Andrographolide

Andrographis paniculata

In vitro PA-induced HepG2 and OA-treated L02 cells; in vivo CDAA diet-fed and HFD-fed mice

Downregulated α-SMA, MMP-2, TIMP-1, TGF-β, MCP1, TNF-α, IL-1β, IFN-γ, NF-κB, and DGAT1

[286], [287], [288]

Actein

Cimicifuga foetida

In vitro fructose-treated L02 cells; in vivo HFD-fed mice

Downregulated TNF-α, SREBP-1c, IL-1β, FAS, MCP1, SCD1, α-SMA, CD36, IRS1, FOXO1, AKT, and GSK3β

[289]


#
#

Tannins

Tannins are a diverse range of large-molecular-weight, water-soluble chemicals found naturally in various fruits, grains, and vegetables [193]. Tannins, potent antioxidant substances, serve as scavengers preventing harmful ROS effects [194]. Therefore, the compounds belonging to the tannin class with anti-NAFLD characteristics are enlisted in this section and outlined in [Fig. 8]

Zoom Image
Fig. 8 Tannins involved in the management of NAFLD [tannic acid (I), dieckol (II), punicalagin (III), and corilagin (IV).

Tannic acid

Many nutritional plant products contain tannic acid (TA), a hydrolysable tannin primarily obtained from the Quercus genus that has appeared to resist inflammation as an antioxidant [195]. Histone H3 lysine 9 (H3K9) acetylation is a well-known epigenetic marker for NAFLD. Histone acetylation caused by histone acetyltransferase (HAT) inhibitors was inhibited by the TA, as demonstrated by in vitro and in vivo HAT experiments. Lipid-accumulation-induced increases in the acetylation levels of H3K9 were decreased by TA. The rise in both mRNA and protein expressions of SREBP-1c, ATP-citrate lyase (ACLY), PPAR-γ, and FAS were inhibited by TA in HepG2 cells and Western-diet-fed mice. Moreover, TA decreased the genetic expression of DGAT2 [196]. ATO-treated liver histopathological alterations were lessened by TA therapy, as primary observations revealed that increased serum levels of ALT and AST were reduced. As a result of TA therapy, MDA and ROS quantities were minimised. TA therapy reduced ATO-induced hepatic inflammatory reactions by lowering IL-1β, IL-6, and TNF-α. Additionally, TA treatment enhanced the concentrations of Nrf2, KEAP1, HO-1, and NQO1 proteins. Battling against oxidative stress and inflammation confirmed that TA had a preventive impact on hepatotoxicity and hepatic lipid accumulation, which seriously impacted NAFLD [197].


#

Dieckol

Dieckol (DK) is an eckol-type marine phlorotannin found in the marine brown algae Ecklonia cava possessing hypnotic behaviour [198], which was found to decrease IL-6 and TNF-α, which were elevated by HFD in mice. More importantly, DK restored vascular endothelial growth factor receptor-C and its receptor VEGFR-3. The PI3K, AKT, and ERK levels were also upregulated by DK treatment [199]. AMPK phosphorylation was significantly increased by DK, and this ultimately resulted in the suppression of FAS and SREBP-1c proteins. By inhibiting ACC, DK may have a major impact on hepatic fatty acid β-oxidation by activating CPT1A, SIRT1, PGC1α, and PPAR-α [200]. With the administration of DK, the TLR4 and NF-κB levels were suppressed, which lowered the NLRP3 inflammasome components, including ASC and NLRP3, in the liver. The expressions of lipogenic genes such as FAS, SREBP-2, PPAR-γ, and FFA-binding protein 4 (FABP4) were decreased, while ATGL and HSL lipolytic genes were enhanced with DK induction in the NAFLD mice model [201].


#

Punicalagin

Punicalagin (PCG) is one of the major ellagitannins found in pomegranate (Punica granatum L.) [202], [203]; it could preserve HepG2 cells against lipotoxicity induced by PA via stimulating Nrf2. By increasing MMP and lowering ROS generation caused by PA, PCG increased mitochondrial activity. The researchers observed that the JNK activation and p38 were suppressed by PCG. Furthermore, PCG significantly impacted enhanced mitochondrial performance by inducing ERK and AKT phosphorylation. Therefore, PCG could influence cellular antioxidant mechanisms through the ERK/Nrf2 pathway, defending mitochondrial function [204]. Treatment with PCG significantly reduced the expression of DNL-related genes such as CD36 and PPAR-γ. The PPAR-α expression was markedly increased in the PCG-treated animals, two genes involved in lipid clearance. AMPK phosphorylation, Nrf2, ZO-1, and HO-1 were enhanced by PCG. The mitochondrial biogenesis genes, viz., PGC1 and TFAM expressions, were higher in the PCG-treated mice model [205]. Hepatic lipid production and the dyslipidemia caused by the HFD were significantly decreased by PCG. Downregulation of TNF-α and interleukins while Nrf2 stimulation was brought back to baseline with PCG administration to revert NAFLD. Lastly, PCG therapy decreased the expression of UCP2 and enhanced ATP levels in the hepatocytes. However, mitochondrial density remained unaffected at a high PGC1α level after PCG treatment [206].


#

Corilagin

Among the main active ingredients in many traditional medicinal plants, corilagin (CORI), a gallotannin isolated from Caesalpinia coriaria (Jacq.) Willd., was reported to possess hepatoprotective, anti-inflammatory, and anti-tumour properties [207]. An experiment showed that CORI therapy markedly decreased the gene expressions involved in fatty acid synthesis, such as SREBP-1c, ACC1, and FAS, and markedly increased the FAO-related genes, such as PPAR-α, CPT1, and ACOX1. Enhanced MMP suggested that CORI therapy improved mitochondrial dysfunction. Furthermore, MCP1, TNF-α, and IL-6 genes were downregulated upon CORI therapy in HFD-induced animals. Among the antioxidant enzymes that CORI increased were SOD, GPx, and CAT [208]. Finally, FABP4 was lowered, while CD36 and CYP7A1 genes were upregulated upon CORI administration [209].


#
#

Clinical Trials on Potent Phytocompounds in NAFLD

Several natural compounds have shown specific termination of FFA synthesis and lipid accumulation on different in vitro cell lines and preclinical animal models targeting molecular levels. At the same time, Phase 1/2/3 clinical trials of numerous phytochemicals have shown encouraging results in diminishing NAFLD ([Table 6]).

Table 6 Clinical trials of phytocompounds from different classes engaged in the treatment of NAFLD.

Phytocompounds

Class

Phase

Clinical Trial ID

Primary outcome

Time frame

Ref.

Berberine

Alkaloid

Phase 4

NCT03198572

Improvement in histologic features of NASH by NAFLD activity score (NAS)

48 weeks

[290]

Silymarin

Flavonoid

Phase 2

NCT02006498

To determine the efficacy of silymarin on NAS

12 months

[291]

Phase 1

NCT00389376

Determining the adverse effects

10 days

[292]

Phase 2

NCT00680407

Improvement in NAS score

48 – 50 weeks

[293]

Phase 4

NCT02973295

Lowering of liver steatosis and liver fibrosis

24 – 25 weeks

[294]

Resveratrol

Polyphenol

Phase 2/3

NCT02216552

Adverse effect profile

8 weeks

[295]

N/A

NCT01446276

Hepatic VLDL-TG secretion and its clearance

6 months

[296]

Phase 2/3

NCT02030977

ALT levels

12 weeks

[297]

N/A

NCT01464801

Alteration in HS and inflammatory markers

6 months

[298]

N/A

NCT01150955

Parameters related to glucose, protein and fat metabolism

5 weeks

[299]

Curcumin

Polyphenol

N/A

NCT03864783

Percentage of steatosis in the liver tissue

42 days ± 3 days

[300]

Phase 2/3

NCT02908152

HS

12 weeks

[301]

Hydroxytyrosol

Phenol

Phase 3

NCT02842567

Improvement of markers for inflammation and oxidative stress

4 months

[302]

Anthocyanin

Flavonoid

Early Phase 1

NCT01940263

Oxidative stress and inflammatory biomarkers

12 weeks

[303]

In 2019, a randomised controlled trial with an open label was carried out for seven weeks to investigate the effect of BRB on the metabolic profiles and liver function of 24 patients with NAFLD. Employees eligible for the study were randomised into one of two groups using a computer-generated random-allocation sequence. The treatment group was administered BRB at 6.25 g per day, while the second group received no intervention. The primary outcome measures considered changes from baseline in serum levels of ALT, AST, and ALP. In contrast, secondary outcome measures included changes from baseline in FBS, TC, LDL-c, HDL-c, and TG levels (https://clinicaltrials.gov/ct2/show/study/NCT04 049 396).

Patients suffering from NAFLD received two capsules regularly for three months. Each capsule comprised 700 mg of silymarin and the additives in a randomised, controlled trial conducted at the ambulatory care setting in a Brazilian hospital. The patients were split into two groups, including control and intervention. Primary outcome measures would scrutinise variations in NAFLD severity, while glycated haemoglobin, FBS, lipid profile, serum ferritin, AST, and ALT would be surveyed by secondary outcome measures (https://clinicaltrials.gov/ct2/show/study/NCT03 749 070).


#

Conclusion

The development of NAFLD and its progression involves various molecular signalling pathways, as well as genomic alterations, in the liver. Biological markers involved in different signalling pathways can portray the underlying mechanisms associated with NAFLD. Therefore, modulating the signalling pathways involved with NAFLD could be a viable option for arresting its progression. Although dietary and exercise-related lifestyle modifications are now the frontline of therapy for NAFLD, there is an impending need to mitigate this disorder through medication. The disease progression in NAFLD is multi-factorial in nature and involves a myriad of events like insulin resistance, oxidative stress, inflammation, and disrupted cellular energy metabolism, which contribute to the pathogenesis of NAFLD. A key aspect of management of NAFLD treatment could be preventing fibrosis, as well as ameliorating oxidative stress and inflammation. Natural products may offer new perspectives for novel therapeutic strategies aiming to reduce the development or progression of complications associated with diabetes mellitus, obesity, and NAFLD. Major lipogenic enzymes, viz., ACC, FAS, and DGAT2, are stimulated by SREBP-1c and ChREBP transcriptional factors to enhance hepatic lipogenesis. In addition, pro-inflammatory cytokines such as IL-6, IL-1β, and TNF-α play pivotal roles in various phases of liver ailments, influencing key features such as dyslipidemia, cholestasis, and fibrosis. The FAS, a crucial enzyme in DNL, has been observed at elevated levels in HS. Therefore, downregulation of FAS could control the biosynthesis of the FFAs.

OCA, a synthetic derivative of natural bile acid called chenodeoxycholic acid, has been exemplified to decrease HS, glucogenesis, and lipogenesis in the liver by stimulating the FXR. Despite having an influential role in combating NAFLD, unfortunately, OCA was reported to cause numerous adverse effects including dyslipidemia, oropharyngeal pain, thyroid hormone disorder, and hypersensitivity reactions. The case studies of the mentioned side effects have created severe challenges for OCA in achieving FDA approval. So, to avoid such undesired adverse events, larger numbers of bioactive phytocompounds are now being investigated for the management of NAFLD.

Naturally occurring substances derived from a variety of biological sources seemed to be extremely important in mitigating oxidative stress by reducing insulin resistance and fat accumulation. Altering the levels of hepatic enzymes and biochemical parameters such as serum TG, TC, HDL-c, and LDL-c by the bioactive plant molecules provides a primary perception in alleviating liver disorders. Compounds from different chemical classes, viz., alkaloid, flavonoid, glycoside, polyphenol, tannin, and terpenoid, which have been proven to mitigate the onset of illness by modulating hepatic lipid metabolism through various molecular targets such as AMPK, TGF-β1, PPARs, etc., are discussed vividly in this present work. The preclinical studies (Table 1, 2, 3, 4, 5), including in vitro assays against human cell lines like HepG2 and in vivo diet-fed animal NAFLD models of a significant number of compounds from each natural chemical class, have been discussed. Due to the promising efficacy exhibited by certain phytomolecules in preclinical studies, some of them have progressed to clinical trials, which has been depicted in [Table 6]. Due to the rising popularity of plant-based remedies globally, it is imperative that medical professionals be aware of both natural products and conventional therapies. Evidently, this review will serve as a springboard for additional research on the bioactive phytocompounds to scrutinise and create novel treatments that might serve as alternatives or supplements to existing ones in the battle against NAFLD.


#

Contributorsʼ Statement

T. Banerjee, A. Sarkar, S. Z. Ali, R. Bhowmik; Design of the study: T. Banerjee, N. Ghosh; Analysis and interpretation: A. K. Halder, S. Karmakar; Drafting the manuscript: T. Banerjee, A. Sarkar, S. Z. Ali, R. Bhowmik, A. K. Halder, S. Karmakar, N. Ghosh; Critical revision of the manuscript: A. K. Halder, S. Karmakar, N. Ghosh”.


#
#

Conflict of Interest

The authors declare no conflicts of interest.

Acknowledgements

We acknowledge the support of the Department of Science and Technology and Biotechnology (WBDSTBT), Govt. of West Bengal, India Vide Memo. 2027 (Sanc.)/STBT-11 012 (19)/6/2023-ST SEC, dated 24 – 01 – 2024. We also thank the Central Council for Research in Unani Medicine, F.No. 3 – 72/2020-CCRUM/TECH, CCRUM (Ministry of AYUSH) India, AICTE, UGC-DSA, DST, UGC-UPE, RUSA 2.0, and the Swami Vivekananda Merit Cum Means Scholarship Scheme (SVMCM), Government of West Bengal, India for providing fellowship. We also acknowledge the State Government Research Fellowship Scheme (Ref. no.: P-1/RS/08/23, dated 19. 01. 2023). The authors gratefully acknowledge the scientific illustrations provided by the Smart Medical Art server (https://smart.servier.com/) and appreciate the support obtained from the Department of Pharmaceutical Technology, Jadavpur University, Kolkata.

  • References

  • 1 Mcpherson S, Hardy T, Henderson E, Burt A, Day C, Anstee Q. Evidence of NAFLD Progression from Steatosis to Fibrosing-Steatohepatitis Using Paired Biopsies: Implications for Prognosis & Clinical Management. J Hepatol 2014; 62
  • 2 Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018; 67: 328-357
  • 3 Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease–Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016; 64: 73-84
  • 4 Benedict M, Zhang X. Non-alcoholic fatty liver disease: An expanded review. World J Hepatol 2017; 9: 715
  • 5 Bikbov MM, Gilmanshin TR, Zainullin RM, Kazakbaeva GM, Iakupova EM, Fakhretdinova AA. Prevalence of non-alcoholic fatty liver disease in the Russian Ural Eye and Medical Study and the Ural Very Old Study. Sci Rep 2022; 12: 7842
  • 6 Zhong F, Zhou X, Xu J, Gao L. Rodent Models of Nonalcoholic Fatty Liver Disease. Digestion 2020; 101: 522-535
  • 7 Armstrong MJ, Adams LA, Canbay A, Syn WK. Extrahepatic complications of nonalcoholic fatty liver disease. Hepatology 2014; 59: 1174-1197
  • 8 Sarkar A, Saha S, Paul A, Maji A, Roy P, Maity TK. Understanding stem cells and its pivotal role in regenerative medicine. Life Sci 2021; 273: 119270
  • 9 Utzschneider KM, Kahn SE. The Role of Insulin Resistance in Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 2006; 91: 4753-4761
  • 10 Tarantino G, Sinatti G, Citro V, Santini jr S, Balsano C. Sarcopenia, a condition shared by various diseases: can we alleviate or delay the progression?. Intern Emerg Med 2023; 18: 1887-1895
  • 11 Bagherniya M, Nobili V, Blesso CN, Sahebkar A. Medicinal plants and bioactive natural compounds in the treatment of non-alcoholic fatty liver disease: A clinical review. Pharmacol Res 2018; 130: 213-240
  • 12 Balato N, Napolitano M, Ayala F, Patruno C, Megna M, Tarantino G. Nonalcoholic fatty liver disease, spleen and psoriasis: New aspects of low-grade chronic inflammation. World J Gastroenterol 2015; 21: 6892-6897
  • 13 Lian J, Fu J. Pioglitazone for NAFLD Patients With Prediabetes or Type 2 Diabetes Mellitus: A Meta-Analysis. Front Endocrinol 2021; 12
  • 14 Bril F, Biernacki D, Kalavalapalli S, Lomonaco R, Subbarayan S, Lai J. Role of Vitamin E for Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Care 2019; 42: dc190167
  • 15 Lin C, Yu B, Chen L, Zhang Z, Ye W, Zhong H. Obeticholic Acid Induces Hepatoxicity Via FXR in the NAFLD Mice. Front Pharmacol 2022; 13
  • 16 Cheng C, Li Z, Zhao X, Liao C, Quan J, Bode AM. Natural alkaloid and polyphenol compounds targeting lipid metabolism: Treatment implications in metabolic diseases. Eur J Pharmacol 2020; 870: 172922
  • 17 Parthasarathy G, Revelo X, Malhi H. Pathogenesis of Nonalcoholic Steatohepatitis: An Overview. Hepatol Commun 2020; 4
  • 18 Neuman MG, Cohen LB, Nanau RM. Biomarkers in Nonalcoholic Fatty Liver Disease. Can J Gastroenterol Hepatol 2014; 28: 607-618
  • 19 Thibaut R, Gage M, Torra I, Chabrier G, Venteclef N, Alzaid F. Liver macrophages and inflammation in physiology and physiopathology of non‐alcoholic fatty liver disease. FEBS J 2021; 289
  • 20 Shan Z, Ju C. Hepatic Macrophages in Liver Injury. Front Immunol 2020; 11
  • 21 Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 2012; 18: 1028-1040
  • 22 Arroyave-Ospina JC, Wu Z, Geng Y, Moshage H. Role of Oxidative Stress in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: Implications for Prevention and Therapy. Antioxidants 2021; 10
  • 23 Martín-Fernández M, Arroyo V, Carnicero C, Sigüenza R, Busta R, Mora N. Role of Oxidative Stress and Lipid Peroxidation in the Pathophysiology of NAFLD. Antioxidants (Basel) 2022; 11: 2217
  • 24 Guo C, Sun L, Chen X, Zhang D. Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regen Res 2013; 8: 2003-2014
  • 25 Mittal M, Siddiqui M, Tran KA, Pothireddy S, Malik A. Reactive Oxygen Species in Inflammation and Tissue Injury. Antioxid Redox Signal 2013; 20
  • 26 Peverill W, Powell L, Skoien R. Evolving Concepts in the Pathogenesis of NASH: Beyond Steatosis and Inflammation. Int J Mol Sci 2014; 15: 8591-8638
  • 27 Yin H, Price F, Rudnicki M. Satellite Cells and the Muscle Stem Cell Niche. Physiol Rev 2013; 93: 23-67
  • 28 Basaranoglu M, Basaranoglu G, Bugianesi E. Carbohydrate intake and nonalcoholic fatty liver disease: Fructose as a weapon of mass destruction. Hepatobiliary Surg Nutr 2015; 4: 109-116
  • 29 Valdes P, Cuevas-Ramos D, Aguilar-Salinas C. Metabolic syndrome and non-alcoholic fatty liver disease. Annals Hepatol 2009; 8. Suppl1:S18-24
  • 30 Listenberger LL, Han X, Lewis SE, Cases S, Farese RV, Ory DS. Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proc Natl Acad Sci U S A 2003; 100: 3077-3082
  • 31 Bianco C, Casirati E, Malvestiti F, Valenti L. Genetic predisposition similarities between NASH and ASH: Identification of new therapeutic targets. JHEP Rep 2021; 3: 100284
  • 32 Dabravolski S, Bezsonov E, Baig M, Popkova T, Orekhov A. Mitochondrial Lipid Homeostasis at the Crossroads of Liver and Heart Diseases. Int J Mol Sci 2021; 22: 6949
  • 33 Herzig S, Shaw R. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol 2017; 19
  • 34 Fabbrini E, Sullivan S, Klein S. Obesity and Nonalcoholic Fatty Liver Disease: Biochemical, Metabolic, and Clinical Implications. Hepatology 2010; 51: 679-689
  • 35 Ibrahim S, Hirsova P, Gores G. Non-alcoholic steatohepatitis pathogenesis: Sublethal hepatocyte injury as a driver of liver inflammation. Gut 2018; 67: gutjnl-2017
  • 36 Zhang C-Y, Yuan W-G, He P, Lei J-H, Wang C-X. Liver fibrosis and hepatic stellate cells: Etiology, pathological hallmarks and therapeutic targets. World J Gastroenterol 2016; 22: 10512
  • 37 Chitturi S, Abeygunasekera S, Farrell GC, Holmes-Walker J, Hui JM, Fung C. NASH and insulin resistance: Insulin hypersecretion and specific association with the insulin resistance syndrome. Hepatology 2002; 35: 373-379
  • 38 Zhang C, Zhou B, Sheng J, Chen Y, Cao Y, Chen C. Molecular mechanisms of hepatic insulin resistance in nonalcoholic fatty liver disease and potential treatment strategies. Pharmacol Res 2020; 159: 104984
  • 39 Sarkar A, Saha R, Saha S, Bhowmik R, Chatterjee A, Paul A. Transesterification, GC-MS profiling, and in vitro antimicrobial potential of oil obtained from seeds of Citrus maxima (Burm.) Merr. Ind Crops Prod 2022; 189: 115764
  • 40 Sarkar A, Banerjee T, Maji A, Paul A, Guria T. Mikania Species: Revealing Phytochemicals from the Pandoraʼs Box. In: Chakraborty R, Sen S, editors New Avenues in Drug Discovery and Bioactive Natural Products. BENTHAM SCIENCE PUBLISHERS; 2023: 149-167
  • 41 Sarkar S, Kar A, Shaw P, DasGupta B, Keithellakpam OS, Mukherjee PK, Bhardwaj PK, Sharma N, Haldar PK, Sinha S. Hydroalcoholic root extracts of Houttuynia cordata (Thunb.) standardized by UPLC-Q-TOF-MS/MS promotes apoptosis in human hepatocarcinoma cell HepG2 via GSK-3β/β-catenin/PDL-1 axis. Fitoterapia 2023; 171: 105684
  • 42 Tarantino G, Balsano C, Santini SJ, Brienza G, Clemente I, Cosimini B, Sinatti G. It Is High Time Physicians Thought of Natural Products for Alleviating NAFLD. Is There Sufficient Evidence to Use Them?. Int J Mol Sci 2021; 22: 13424
  • 43 Zheng X, Wu F, Lin X, Shen L, Feng Y. Developments in drug delivery of bioactive alkaloids derived from traditional Chinese medicine. Drug Deliv 2018; 25: 398-416
  • 44 Yang Y, Vong CT, Zeng S, Gao C, Chen Z, Fu C. Tracking evidences of Coptis chinensis for the treatment of inflammatory bowel disease from pharmacological, pharmacokinetic to clinical studies. J Ethnopharmacol 2021; 268: 113573
  • 45 Zhang Y, Ma J, Zhang W. Berberine for bone regeneration: Therapeutic potential and molecular mechanisms. J Ethnopharmacol 2021; 277: 114249
  • 46 Guo T, Woo S-L, Guo X, Li H, Zheng J, Botchlett R. Berberine Ameliorates Hepatic Steatosis and Suppresses Liver and Adipose Tissue Inflammation in Mice with Diet-induced Obesity. Sci Rep 2016; 6: 22612
  • 47 Li Y, Zhao X, Feng X, Liu X, Deng C, Hu C-H. Berberine Alleviates Olanzapine-Induced Adipogenesis via the AMPKα–SREBP Pathway in 3 T3-L1 Cells. Int J Mol Sci 2016; 17
  • 48 Shan M, Dai Y, Ren X, Zheng J, Zhang K, Chen B. Berberine mitigates nonalcoholic hepatic steatosis by downregulating SIRT1-FoxO1-SREBP2 pathway for cholesterol synthesis. J Integr Med 2021; 19: 545-554
  • 49 Liang H, Wang Y. Berberine alleviates hepatic lipid accumulation by increasing ABCA1 through the protein kinase C δ pathway. Biochem Biophys Res Commun 2018; 498: 473-480
  • 50 Chang X, Wang Z, Zhang J, Yan H, Bian H, Xia M. Lipid profiling of the therapeutic effects of berberine in patients with nonalcoholic fatty liver disease. J Transl Med 2016; 14: 266
  • 51 Day CR, Kempson SA. Betaine chemistry, roles, and potential use in liver disease. Biochim Biophys Acta 2016; 1860: 1098-1106
  • 52 Deminice R, da Silva RP, Lamarre SG, Kelly KB, Jacobs RL, Brosnan ME. Betaine supplementation prevents fatty liver induced by a high-fat diet: effects on one-carbon metabolism. Amino Acids 2015; 47: 839-846
  • 53 Xu G, Huang K, Zhou J. Hepatic AMP Kinase as a Potential Target for Treating Nonalcoholic Fatty Liver Disease: Evidence from Studies of Natural Products. Curr Med Chem 2018; 25: 889-907
  • 54 Kim DH, Lee B, Kim MJ, Park MH, An HJ, Lee EK. Molecular Mechanism of Betaine on Hepatic Lipid Metabolism: Inhibition of Forkhead Box O1 (FoxO1) Binding to Peroxisome Proliferator-Activated Receptor Gamma (PPARγ). J Agric Food Chem 2016; 64: 6819-6825
  • 55 Wang L, Chen L, Tan Y, Wei J, Chang Y, Jin T. Betaine supplement alleviates hepatic triglyceride accumulation of apolipoprotein E deficient mice via reducing methylation of peroxisomal proliferator-activated receptor alpha promoter. Lipids Health Dis 2013; 12: 34
  • 56 Xu L, Huang D, Hu Q, Wu J, Wang Y, Feng J. Betaine alleviates hepatic lipid accumulation via enhancing hepatic lipid export and fatty acid oxidation in rats fed with a high-fat diet. Br J Nutr 2015; 113: 1835-1843
  • 57 Liu W, Yi D-D, Guo J-L, Xiang Z-X, Deng L-F, He L. Nuciferine, extracted from Nelumbo nucifera Gaertn, inhibits tumor-promoting effect of nicotine involving Wnt/β-catenin signaling in non-small cell lung cancer. J Ethnopharmacol 2015; 165: 83-93
  • 58 Lu Y, He Y, Wang M, Zhang L, Yang L, Wang Z. Characterization of nuciferine metabolism by P450 enzymes and uridine diphosphate glucuronosyltransferases in liver microsomes from humans and animals. Acta Pharmacol Sin 2010; 31: 1635-1642
  • 59 Ma C, Li G, He Y, Xu B, Mi X, Wang H. Pronuciferine and nuciferine inhibit lipogenesis in 3 T3-L1 adipocytes by activating the AMPK signaling pathway. Life Sci 2015; 136: 120-125
  • 60 Zhang C, Deng J, Liu D, Tuo X, Xiao L, Lai B. Nuciferine ameliorates hepatic steatosis in high-fat diet/streptozocin-induced diabetic mice through PPARα/PGC1α pathway. Br J Pharmacol 2018; 175
  • 61 Guo F, Yang X, Li X, Feng R, Guan C, Wang Y. Nuciferine Prevents Hepatic Steatosis and Injury Induced by a High-Fat Diet in Hamsters. PloS one 2013; 8: e63770
  • 62 Gong X, Gao Y, Guo G, Vondran FWR, Schwartlander R, Efimova E. Effect of matrine on primary human hepatocytes in vitro. Cytotechnology 2015; 67: 255-265
  • 63 Liu Z, Zhang Y, Tang Z, Xu J, Ma M, Pan S. Matrine attenuates cardiac fibrosis by affecting ATF6 signaling pathway in diabetic cardiomyopathy. Eur J Pharmacol 2017; 804: 21-30
  • 64 Liu J, Zhao Y, Xia J, Qiu M. Matrine induces toxicity in mouse liver cells through an ROS-dependent mechanism. Res Vet Sci 2020; 132: 308-311
  • 65 Zhang H-F, Shi L-J, Song G-Y, Cai Z-G, Wang C, An R-J. Protective effects of matrine against progression of high-fructose diet-induced steatohepatitis by enhancing antioxidant and anti-inflammatory defences involving Nrf2 translocation. Food Chem Toxicol 2013; 55: 70-77
  • 66 Zeng X-Y, Wang H, Bai F, Zhou X, Li S-P, Ren L-P. Identification of matrine as a promising novel drug for hepatic steatosis and glucose intolerance with HSP72 as an upstream target. Br J Pharmacol 2015; 172: 4303-4318
  • 67 Mahzari A, Li S, Zhou X, Li D, Fouda S, Alhomrani M. Matrine Protects Against MCD-Induced Development of NASH via Upregulating HSP72 and Downregulating mTOR in a Manner Distinctive From Metformin. Front Pharmacol 2019; 10
  • 68 Fei Z-W, Qiu M-K, Qi X-Q, Dai Y-X, Wang S-Q, Quan Z-W. Oxymatrine suppresses proliferation and induces apoptosis of hemangioma cells through inhibition of HIF-1a signaling. Int J Immunopathol Pharmacol 2015; 28: 201-208
  • 69 Shi L, Shi L, Zhang H, Hu Z, Wang C, Zhang D. Oxymatrine ameliorates non-alcoholic fatty liver disease in rats through peroxisome proliferator-activated receptor-α activation. Mol Med Rep 2013; 8: 439-445
  • 70 Shi L, Shi L, Zhang H, Hu Z, Wang C, Zhang D. Oxymatrine ameliorates non-alcoholic fatty liver disease in rats through peroxisome proliferator-activated receptor-α activation. Mol Med Rep 2013; 8
  • 71 Derosa G, Maffioli P, Sahebkar A. Piperine and Its Role in Chronic Diseases. In: Gupta SC, Prasad S, Aggarwal BB, editors Anti-inflammatory Nutraceuticals and Chronic Diseases. Cham: Springer International Publishing; 2016: 173-184
  • 72 Park U-H, Jeong H-S, Jo E-Y, Park T, Yoon SK, Kim E-J. Piperine, a Component of Black Pepper, Inhibits Adipogenesis by Antagonizing PPARγ Activity in 3 T3-L1 Cells. J Agric Food Chem 2012; 60: 3853-3860
  • 73 Kim KJ, Lee M-S, Jo K, Hwang J-K. Piperidine alkaloids from Piper retrofractum Vahl. protect against high-fat diet-induced obesity by regulating lipid metabolism and activating AMP-activated protein kinase. Biochem Biophys Res Commun 2011; 411: 219-225
  • 74 Zhou J, Chan L, Zhou S. Trigonelline: A Plant Alkaloid with Therapeutic Potential for Diabetes and Central Nervous System Disease. Curr Med Chem 2012; 19: 3523-3531
  • 75 Zia T, Hasnain SN, Hasan SK. Evaluation of the oral hypoglycaemic effect of Trigonella foenum-graecum L. (methi) in normal mice. J Ethnopharmacol 2001; 75: 191-195
  • 76 Ilavenil S, Arasu MV, Lee J-C, Kim DH, Roh SG, Park HS. Trigonelline attenuates the adipocyte differentiation and lipid accumulation in 3 T3-L1 cells. Phytomedicine 2014; 21: 758-765
  • 77 Zhang D-F, Zhang F, Zhang J, Zhang R-M, Li R. Protection effect of trigonelline on liver of rats with non-alcoholic fatty liver diseases. Asian Pac J Trop Med 2015; 8: 651-654
  • 78 Song C-Y, Zeng X, Chen S-W, Hu P-F, Zheng Z-W, Ning B-F. Sophocarpine alleviates non-alcoholic steatohepatitis in rats. J Gastroenterol Hepatol 2011; 26: 765-774
  • 79 Song C-Y, Shi J, Zeng X, Zhang Y, Xie W-F, Chen Y-X. Sophocarpine alleviates hepatocyte steatosis through activating AMPK signaling pathway. Toxicol In Vitro 2013; 27: 1065-1071
  • 80 Lee W-Y, Lee C-Y, Lee J-S, Kim C-E. Identifying Candidate Flavonoids for Non-Alcoholic Fatty Liver Disease by Network-Based Strategy. Front Pharmacol 2022; 13
  • 81 Wei D, Zhang G, Zhu Z, Zheng Y, Yan F, Pan C. Nobiletin Inhibits Cell Viability via the SRC/AKT/STAT3/YY1AP1 Pathway in Human Renal Carcinoma Cells. Front Pharmacol 2019; 10
  • 82 Yuk T, Kim Y, Yang J, Sung J, Jeong HS, Lee J. Nobiletin Inhibits Hepatic Lipogenesis via Activation of AMP-Activated Protein Kinase. Evid Based Complement Alternat Med 2018; 2018: 7420265
  • 83 Bunbupha S, Pakdeechote P, Maneesai P, Prasarttong P. Nobiletin alleviates high-fat diet-induced nonalcoholic fatty liver disease by modulating AdipoR1 and gp91phox expression in rats. J Nutr Biochem 2021; 87: 108526
  • 84 Peng Z, Li X, Xing D, Du X, Wang Z, Liu G. Nobiletin alleviates palmitic acid-induced NLRP3 inflammasome activation in a sirtuin 1-dependent manner in AML-12 cells. Mol Med Rep 2018; 18: 5815-5822
  • 85 Liu D, Mao Y, Ding L, Zeng X-A. Dihydromyricetin: A review on identification and quantification methods, biological activities, chemical stability, metabolism and approaches to enhance its bioavailability. Trends Food Sci Technol 2019; 91: 586-597
  • 86 Xie C, Chen Z, Zhang C, Xu X, Jin J, Zhan W. Dihydromyricetin ameliorates oleic acid-induced lipid accumulation in L02 and HepG2 cells by inhibiting lipogenesis and oxidative stress. Life Sci 2016; 157: 131-139
  • 87 Zeng X, Yang J, Hu O, Huang J, Ran L, Chen M. Dihydromyricetin Ameliorates Nonalcoholic Fatty Liver Disease by Improving Mitochondrial Respiratory Capacity and Redox Homeostasis Through Modulation of SIRT3 Signaling. Antioxid Redox Signal 2019; 30: 163-183
  • 88 Guo L, Zhang H, Yan X. Protective effect of dihydromyricetin revents fatty liver through nuclear factor-κB/p 53/B-cell lymphoma 2-associated X protein signaling pathways in a rat model. Mol Med Rep 2019; 19: 1638-1644
  • 89 Ganeshpurkar A, Saluja AK. The Pharmacological Potential of Rutin. Saudi Pharm J 2017; 25: 149-164
  • 90 Liu Q, Pan R, Ding L, Zhang F, Hu L, Ding B. Rutin exhibits hepatoprotective effects in a mouse model of non-alcoholic fatty liver disease by reducing hepatic lipid levels and mitigating lipid-induced oxidative injuries. Int Immunopharmacol 2017; 49: 132-141
  • 91 Panchal SK, Poudyal H, Arumugam TV, Brown L. Rutin Attenuates Metabolic Changes, Nonalcoholic Steatohepatitis, and Cardiovascular Remodeling in High-Carbohydrate, High-Fat Diet-Fed Rats. J Nutr 2011; 141: 1062-1069
  • 92 Sarkar A, Paul A, Banerjee T, Maji A, Saha S, Bishayee A. Therapeutic advancements in targeting BCL-2 family proteins by epigenetic regulators, natural, and synthetic agents in cancer. Eur J Pharmacol 2023; 944: 175588
  • 93 Hernández-Aquino E, Muriel P. Beneficial effects of naringenin in liver diseases: Molecular mechanisms. World J Gastroenterol 2018; 24: 1679-1707
  • 94 Zobeiri M, Belwal T, Parvizi F, Naseri R, Farzaei HM, Nabavi FS. Naringenin and its Nano-formulations for Fatty Liver: Cellular Modes of Action and Clinical Perspective. Curr Pharm Biotechnol 2018; 19: 196-205
  • 95 Wang Q, Ou Y, Hu G, Wen C, Yue S, Chen C. Naringenin attenuates non-alcoholic fatty liver disease by down-regulating the NLRP3/NF-κB pathway in mice. Br J Pharmacol 2020; 177: 1806-1821
  • 96 Chang H-H, Yi P-L, Cheng C-H, Lu C-Y, Hsiao Y-T, Tsai Y-F. Biphasic effects of baicalin, an active constituent of Scutellaria baicalensis Georgi, in the spontaneous sleep–wake regulation. J Ethnopharmacol 2011; 135: 359-368
  • 97 Liu J, Yuan Y, Gong X, Zhang L, Zhou Q, Wu S. Baicalin and its nanoliposomes ameliorates nonalcoholic fatty liver disease via suppression of TLR4 signaling cascade in mice. Int Immunopharmacol 2020; 80: 106208
  • 98 Hu Q, Zhang W, Wu Z, Tian X, Xiang J, Li L. Baicalin and the liver-gut system: Pharmacological bases explaining its therapeutic effects. Pharmacol Res 2021; 165: 105444
  • 99 Zhong X, Liu H. Baicalin attenuates diet induced nonalcoholic steatohepatitis by inhibiting inflammation and oxidative stress via suppressing JNK signaling pathways. Biomed Pharmacother 2018; 98: 111-117
  • 100 Banerjee R, Dasgupta B, Kar A, Bhardwaj PK, Sharma N, Haldar PK. Quality evaluation of different black rice varieties of northeastern region of India. Phytochem Anal 2023; 34: 507-517
  • 101 Li Y, Yao J, Han C, Yang J, Chaudhry MT, Wang S. Quercetin, Inflammation and Immunity. Nutrients 2016; 8
  • 102 Yang H, Yang T, Heng C, Zhou Y, Jiang Z, Qian X. Quercetin improves nonalcoholic fatty liver by ameliorating inflammation, oxidative stress, and lipid metabolism in db/db mice. Phytother Res 2019; 33: 3140-3152
  • 103 Yi H, Peng H, Wu X, Xu X, Kuang T, Zhang J. The Therapeutic Effects and Mechanisms of Quercetin on Metabolic Diseases: Pharmacological Data and Clinical Evidence. Oxid Med Cell Longev 2021; 2021: 6678662
  • 104 Liu L, Gao C, Yao P, Gong Z. Quercetin Alleviates High-Fat Diet-Induced Oxidized Low-Density Lipoprotein Accumulation in the Liver: Implication for Autophagy Regulation. Biomed Res Int 2015; 2015: 607531
  • 105 Lin Y, Shi R, Wang X, Shen H-M. Luteolin, a Flavonoid with Potential for Cancer Prevention and Therapy. Curr Cancer Drug Targets 2008; 8: 634-646
  • 106 Liu X, Sun R, Li Z, Xiao R, Lv P, Sun X. Luteolin alleviates non-alcoholic fatty liver disease in rats via restoration of intestinal mucosal barrier damage and microbiota imbalance involving in gut-liver axis. Arch Biochem Biophys 2021; 711: 109019
  • 107 Yin Y, Gao L, Lin H, Wu Y, Han X, Zhu Y. Luteolin improves non-alcoholic fatty liver disease in db/db mice by inhibition of liver X receptor activation to down-regulate expression of sterol regulatory element binding protein 1c. Biochem Biophys Res Commun 2017; 482: 720-726
  • 108 Chen R, Chen X, Zhu T, Liu J, Xiang X, Yu J. Integrated Transcript and Metabolite Profiles Reveal That EbCHI Plays an Important Role in Scutellarin Accumulation in Erigeron breviscapus Hairy Roots. Front Plant Sci 2018; 9
  • 109 Hua Fan, Xiande Ma, Peng Lin, Qiang Kang, Zhilong Zhao, Lina Wang. Scutellarin Prevents Nonalcoholic Fatty Liver Disease (NAFLD) and Hyperlipidemia via PI3K/AKT-Dependent Activation of Nuclear Factor (Erythroid-Derived 2)-Like 2 (Nrf2) in Rats. Med Sci Monit 2017; 23: 5599-5612
  • 110 Zhang X, Ji R, Sun H, Peng J, Ma X, Wang C. Scutellarin ameliorates nonalcoholic fatty liver disease through the PPARγ/PGC-1α-Nrf2 pathway. Free Radic Res 2018; 52: 198-211
  • 111 Xin X, Chen C, Hu Y-Y, Feng Q. Protective effect of genistein on nonalcoholic fatty liver disease (NAFLD). Biomed Pharmacother 2019; 117: 109047
  • 112 Wang W, Chen J, Mao J, Li H, Wang M, Zhang H. Genistein Ameliorates Non-alcoholic Fatty Liver Disease by Targeting the Thromboxane A2 Pathway. J Agric Food Chem 2018; 66: 5853-5859
  • 113 Seidemann L, Krüger A, Kegel-Hübner V, Seehofer D, Damm G. Influence of Genistein on Hepatic Lipid Metabolism in an In Vitro Model of Hepatic Steatosis. Molecules 2021; 26
  • 114 Khan N, Syed DN, Ahmad N, Mukhtar H. Fisetin: A Dietary Antioxidant for Health Promotion. Antioxid Redox Signal 2013; 19: 151-162
  • 115 Liou C-J, Wei C-H, Chen Y-L, Cheng C-Y, Wang C-L, Huang W-C. Fisetin Protects Against Hepatic Steatosis Through Regulation of the Sirt1/AMPK and Fatty Acid β-Oxidation Signaling Pathway in High-Fat Diet-Induced Obese Mice. Cell Physiol Biochem 2018; 49: 1870-1884
  • 116 Dai X, Kuang Q, Sun Y, Xu M, Zhu L, Ge C. Fisetin represses oxidative stress and mitochondrial dysfunction in NAFLD through suppressing GRP78-mediated endoplasmic reticulum (ER) stress. J Funct Foods 2022; 90: 104954
  • 117 Gaballah HH, El-Horany HE, Helal DS. Mitigative effects of the bioactive flavonol fisetin on high-fat/high-sucrose induced nonalcoholic fatty liver disease in rats. J Cell Biochem 2019; 120: 12762-12774
  • 118 Brito-Arias M. Synthesis and Characterization of Glycosides. 2007
  • 119 Leong XY, Thanikachalam PV, Pandey M, Ramamurthy S. A systematic review of the protective role of swertiamarin in cardiac and metabolic diseases. Biomed Pharmacother 2016; 84: 1051-1060
  • 120 Patel TP, Rawal K, Soni S, Gupta S. Swertiamarin ameliorates oleic acid induced lipid accumulation and oxidative stress by attenuating gluconeogenesis and lipogenesis in hepatic steatosis. Biomed Pharmacother 2016; 83: 785-791
  • 121 Xu L, Li D, Zhu Y, Cai S, Liang X, Tang Y. Swertiamarin supplementation prevents obesity-related chronic inflammation and insulin resistance in mice fed a high-fat diet. Adipocyte 2021; 10: 160-173
  • 122 Yang Y, Li J, Wei C, He Y, Cao Y, Zhang Y. Amelioration of nonalcoholic fatty liver disease by swertiamarin in fructose-fed mice. Phytomedicine 2019; 59: 152782
  • 123 Wang J, Chen X, Wang W, Zhang Y, Yang Z, Jin Y. Glycyrrhizic acid as the antiviral component of Glycyrrhiza uralensis Fisch. against coxsackievirus A16 and enterovirus 71 of hand foot and mouth disease. J Ethnopharmacol 2013; 147: 114-121
  • 124 Sun X, Duan X, Wang C, Liu Z, Sun P, Huo X. Protective effects of glycyrrhizic acid against non-alcoholic fatty liver disease in mice. Eur J Pharmacol 2017; 806: 75-82
  • 125 Wang C, Duan X, Sun X, Liu Z, Sun P, Yang X. Protective effects of glycyrrhizic acid from edible botanical glycyrrhiza glabra against non-alcoholic steatohepatitis in mice. Food Funct 2016; 7: 3716-3723
  • 126 Nabavi SF, Braidy N, Orhan IE, Badiee A, Daglia M, Nabavi SM. Rhodiola rosea L. and Alzheimerʼs Disease: From Farm to Pharmacy. Phytother Res 2016; 30: 532-539
  • 127 Dou X, Ding Q, Lai S, Jiang F, Song Q, Zhao X. Salidroside alleviates lipotoxicity-induced cell death through inhibition of TLR4/MAPKs pathway, and independently of AMPK and autophagy in AML-12 mouse hepatocytes. J Funct Foods 2020; 65: 103691
  • 128 Zheng T, Yang X, Li W, Wang Q, Chen L, Wu D. Salidroside Attenuates High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease via AMPK-Dependent TXNIP/NLRP3 Pathway. Oxid Med Cell Longev 2018; 2018: 8597897
  • 129 Li H, Ying H, Hu A, Li D, Hu Y. Salidroside Modulates Insulin Signaling in a Rat Model of Nonalcoholic Steatohepatitis. Evid Based Complement Alternat Med 2017; 2017: 9651371
  • 130 Hung J-Y, Hsu Y-L, Ko Y-C, Tsai Y-M, Yang C-J, Huang M-S. Didymin, a dietary flavonoid glycoside from citrus fruits, induces Fas-mediated apoptotic pathway in human non-small-cell lung cancer cells in vitro and in vivo. Lung Cancer 2010; 68: 366-374
  • 131 Yao Q, Lin M-T, Zhu Y-D, Xu H-L, Zhao Y-Z. Recent Trends in Potential Therapeutic Applications of the Dietary Flavonoid Didymin. Molecules 2018; 23
  • 132 Feng Z, Pang L, Chen S, Pang X, Huang Y, Qiao Q. Didymin ameliorates dexamethasone-induced non-alcoholic fatty liver disease by inhibiting TLR4/NF-κB and PI3K/Akt pathways in C57BL/6 J mice. Int Immunopharmacol 2020; 88: 107003
  • 133 Li Y, Li C, Xiong Y, Fang B, Lin X, Huang Q. Didymin Ameliorates Liver Fibrosis by Alleviating Endoplasmic Reticulum Stress and Glycerophospholipid Metabolism: Based on Transcriptomics and Metabolomics. Drug Des Devel Ther 2022; 16: 1713-1729
  • 134 Tanase C, Coşarcă S, Muntean DL. A Critical Review of Phenolic Compounds Extracted from the Bark of Woody Vascular Plants and Their Potential Biological Activity. Molecules 2019; 24: 1182
  • 135 Ganesan K, Xu B. A Critical Review on Polyphenols and Health Benefits of Black Soybeans. Nutrients 2017; 9: 455
  • 136 Muruganandan S, Srinivasan K, Gupta S, Gupta PK, Lal J. Effect of mangiferin on hyperglycemia and atherogenicity in streptozotocin diabetic rats. J Ethnopharmacol 2005; 97: 497-501
  • 137 Du S, Liu H, Lei T, Xie X, Wang H, He X. Mangiferin: An effective therapeutic agent against several disorders (Review). Mol Med Rep 2018; 18: 4775-4786
  • 138 Yong Z, Ruiqi W, Hongji Y, Ning M, Chenzuo J, Yu Z. Mangiferin Ameliorates HFD-Induced NAFLD through Regulation of the AMPK and NLRP3 Inflammasome Signal Pathways. J Immunol Res 2021; 2021: 4084566
  • 139 Wang H, Zhu Y-Y, Wang L, Teng T, Zhou M, Wang S-G. Mangiferin ameliorates fatty liver via modulation of autophagy and inflammation in high-fat-diet induced mice. Biomed Pharmacother 2017; 96: 328-335
  • 140 Guo F, Huang C, Liao X, Wang Y, He Y, Feng R. Beneficial effects of mangiferin on hyperlipidemia in high-fat-fed hamsters. Mol Nutr Food Res 2011; 55: 1809-1818
  • 141 Koushki M, Amiri-Dashatan N, Ahmadi N, Abbaszadeh HA, Rezaei-Tavirani M. Resveratrol: A miraculous natural compound for diseases treatment. Food Sci Nutr 2018; 6: 2473-2490
  • 142 Tang LY, Chen Y, Rui BB, Hu CM. Resveratrol ameliorates lipid accumulation in HepG2 cells, associated with down-regulation of lipin1 expression. Can J Physiol Pharmacol 2016; 94: 185-189
  • 143 Du F, Huang R, Lin D, Wang Y, Yang X, Huang X. Resveratrol Improves Liver Steatosis and Insulin Resistance in Non-alcoholic Fatty Liver Disease in Association With the Gut Microbiota. Front Microbiol 2021; 12
  • 144 Ran G, Ying L, Li L, Yan Q, Yi W, Ying C. Resveratrol ameliorates diet-induced dysregulation of lipid metabolism in zebrafish (Danio rerio). PLOS ONE 2017; 12: e0180865
  • 145 Booker A, Frommenwiler D, Johnston D, Umealajekwu C, Reich E, Heinrich M. Chemical variability along the value chains of turmeric (Curcuma longa): A comparison of nuclear magnetic resonance spectroscopy and high performance thin layer chromatography. J Ethnopharmacol 2014; 152: 292-301
  • 146 Ghosh S, Banerjee S, Sil PC. The beneficial role of curcumin on inflammation, diabetes and neurodegenerative disease: A recent update. Food Chem Toxicol 2015; 83: 111-124
  • 147 Lee DE, Lee SJ, Kim SJ, Lee HS, Kwon OS. Curcumin Ameliorates Nonalcoholic Fatty Liver Disease through Inhibition of O-GlcNAcylation. Nutrients 2019; 11
  • 148 Feng W, Kuang S, Tu C, Ma Z, Pang J, Wang Y. Natural products berberine and curcumin exhibited better ameliorative effects on rats with non-alcohol fatty liver disease than lovastatin. Biomed Pharmacother 2018; 99: 325-333
  • 149 Kang O, Kim S, Seo Y, Joung D, Mun S, Choi J. Curcumin decreases oleic acid-induced lipid accumulation via AMPK phosphorylation in hepatocarcinoma cells. Eur Rev Med Pharmacol Sci 2013; 17: 2578-2586
  • 150 Saadati S, Sadeghi A, Mansour A, Yari Z, Poustchi H, Hedayati M. Curcumin and inflammation in non-alcoholic fatty liver disease: a randomized, placebo controlled clinical trial. BMC Gastroenterol 2019; 19: 133
  • 151 Xiao J, Ho CT, Liong EC, Nanji AA, Leung TM, Lau TYH. Epigallocatechin gallate attenuates fibrosis, oxidative stress, and inflammation in non-alcoholic fatty liver disease rat model through TGF/SMAD, PI3 K/Akt/FoxO1, and NF-kappa B pathways. Eur J Nutr 2014; 53: 187-199
  • 152 Kim JJY, Tan Y, Xiao L, Sun YL, Qu X. Green Tea Polyphenol Epigallocatechin-3-Gallate Enhance Glycogen Synthesis and Inhibit Lipogenesis in Hepatocytes. Biomed Res Int 2013; 2013: 920128
  • 153 Huang J, Feng S, Liu A, Dai Z, Wang H, Reuhl K. Green Tea Polyphenol EGCG Alleviates Metabolic Abnormality and Fatty Liver by Decreasing Bile Acid and Lipid Absorption in Mice. Mol Nutr Food Res 2018; 62: 1700696
  • 154 Vilaplana-Pérez C, Auñón D, García-Flores LA, Gil-Izquierdo A. Hydroxytyrosol and Potential Uses in Cardiovascular Diseases, Cancer, and AIDS. Front Nutr 2014; 1
  • 155 Pirozzi C, Lama A, Simeoli R, Paciello O, Pagano TB, Mollica MP. Hydroxytyrosol prevents metabolic impairment reducing hepatic inflammation and restoring duodenal integrity in a rat model of NAFLD. J Nutr Biochem 2016; 30: 108-115
  • 156 Valenzuela R, Illesca P, Echeverría F, Espinosa A, Rincón-Cervera MÁ, Ortiz M. Molecular adaptations underlying the beneficial effects of hydroxytyrosol in the pathogenic alterations induced by a high-fat diet in mouse liver: PPAR-α and Nrf2 activation, and NF-κB down-regulation. Food Funct 2017; 8: 1526-1537
  • 157 Dong Y, Yu M, Wu Y, Xia T, Wang L, Song K. Hydroxytyrosol Promotes the Mitochondrial Function through Activating Mitophagy. Antioxidants 2022; 11
  • 158 Hitl M, Kladar N, Gavarić N, Bozin B. Rosmarinic Acid–Human Pharmacokinetics and Health Benefits. Planta Med 2020; 87: 273-282
  • 159 Balachander GJ, Subramanian S, Ilango K. Rosmarinic acid attenuates hepatic steatosis by modulating ER stress and autophagy in oleic acid-induced HepG2 cells. RSC Adv 2018; 8: 26656-26663
  • 160 Luo C, Sun H, Peng J, Gao C, Bao L, Ji R. Rosmarinic acid exerts an antagonistic effect on nonalcoholic fatty liver disease by regulating the YAP1/TAZ-PPARγ/PGC-1α signaling pathway. Phytother Res 2021; 35: 1010-1022
  • 161 AlTamimi JZ, Alshammari GM, AlFaris NA, Alagal RI, Aljabryn DH, Albekairi NA, Alkhateeb MA, Yahya MA. Ellagic acid protects against non-alcoholic fatty liver disease in streptozotocin-diabetic rats by activating AMPK. Pharm Biol 2022; 60: 25-37
  • 162 Zhang C, Hu J, Sheng L, Yuan M, Wu Y, Chen L. Ellagic acid ameliorates AKT-driven hepatic steatosis in mice by suppressing de novo lipogenesis via the AKT/SREBP-1/FASN pathway. Food Funct 2019; 10: 3410-3420
  • 163 Polce SA, Burke C, França LM, Kramer B, de Andrade Paes AM, Carrillo-Sepulveda MA. Ellagic Acid Alleviates Hepatic Oxidative Stress and Insulin Resistance in Diabetic Female Rats. Nutrients 2018; 10
  • 164 Goto T, Takahashi N, Hirai S, Kawada T. Various Terpenoids Derived from Herbal and Dietary Plants Function as PPAR Modulators and Regulate Carbohydrate and Lipid Metabolism. PPAR Res 2010; 2010: 483958
  • 165 Cox-Georgian D, Ramadoss N, Dona C, Basu C. Therapeutic and Medicinal Uses of Terpenes. In: Joshee N, Dhekney SA, Parajuli P. editors Medicinal Plants: From Farm to Pharmacy. Cham: Springer International Publishing; 2019: 333-359
  • 166 Kazmi I, Rahman M, Afzal M, Gupta G, Saleem S, Afzal O. Anti-diabetic potential of ursolic acid stearoyl glucoside: A new triterpenic gycosidic ester from Lantana camara. Fitoterapia 2012; 83: 142-146
  • 167 Peng F, Zhang H, He X, Song Z. Effects of Ursolic Acid on Intestinal Health and Gut Bacteria Antibiotic Resistance in Mice. Front Physiol 2021; 12
  • 168 Li J-S, Wang W-J, Sun Y, Zhang Y-H, Zheng L. Ursolic acid inhibits the development of nonalcoholic fatty liver disease by attenuating endoplasmic reticulum stress. Food Funct 2015; 6: 1643-1651
  • 169 Li S, Meng F, Liao X, Wang Y, Sun Z, Guo F. Therapeutic Role of Ursolic Acid on Ameliorating Hepatic Steatosis and Improving Metabolic Disorders in High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease Rats. PLOS ONE 2014; 9: e86724
  • 170 Lin Y-N, Wang CCN, Chang H-Y, Chu F-Y, Hsu Y-A, Cheng W-K. Ursolic Acid, a Novel Liver X Receptor α (LXRα) Antagonist Inhibiting Ligand-Induced Nonalcoholic Fatty Liver and Drug-Induced Lipogenesis. J Agric Food Chem 2018; 66: 11647-11662
  • 171 Ayeleso TB, Matumba MG, Mukwevho E. Oleanolic Acid and Its Derivatives: Biological Activities and Therapeutic Potential in Chronic Diseases. Molecules 2017; 22
  • 172 Somova LI, Shode FO, Ramnanan P, Nadar A. Antihypertensive, antiatherosclerotic and antioxidant activity of triterpenoids isolated from Olea europaea, subspecies africana leaves. J Ethnopharmacol 2003; 84: 299-305
  • 173 Nyakudya TT, Mukwevho E, Nkomozepi P, Erlwanger KH. Neonatal intake of oleanolic acid attenuates the subsequent development of high fructose diet-induced non-alcoholic fatty liver disease in rats. J Dev Orig Health Dis 2018; 9: 500-510
  • 174 Gamede M, Mabuza-Mashaba L, Ngubane P, Khathi A. Plant-derived oleanolic acid ameliorates markers associated with non-alcoholic fatty liver disease in a diet-induced pre-diabetes rat model. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy. 2019;Volume 12; 1953.  –  1962
  • 175 Lin Y-N, Chang H-Y, Wang CCN, Chu F-Y, Shen H-Y, Chen C-J. Oleanolic Acid Inhibits Liver X Receptor Alpha and Pregnane X Receptor to Attenuate Ligand-Induced Lipogenesis. J Agric Food Chem 2018; 66: 10964-10976
  • 176 Matumba MG, Ayeleso AO, Nyakudya T, Erlwanger K, Chegou NN, Mukwevho E. Long-Term Impact of Neonatal Intake of Oleanolic Acid on the Expression of AMP-Activated Protein Kinase, Adiponectin and Inflammatory Cytokines in Rats Fed with a High Fructose Diet. Nutrients 2019; 11
  • 177 Fang Z, Zhang M, Liu J, Zhao X, Zhang Y, Fang L. Tanshinone IIA: A Review of its Anticancer Effects. Front Pharmacol 2021; 11
  • 178 Maione F, De Feo V, Caiazzo E, De Martino L, Cicala C, Mascolo N. Tanshinone IIA, a major component of Salvia milthorriza Bunge, inhibits platelet activation via Erk-2 signaling pathway. J Ethnopharmacol 2014; 155: 1236-1242
  • 179 Huang L, Ding W, Wang M-Q, Wang Z-G, Chen H-H, Chen W. Tanshinone IIA ameliorates non-alcoholic fatty liver disease through targeting peroxisome proliferator-activated receptor gamma and toll-like receptor 4. J Int Med Res 2019; 47: 5239-5255
  • 180 Xu L, Liu X, Jia T, Sun Y, Du Y, Wei S. Tanshinone IIA Ameliorates Nonalcoholic Steatohepatitis in Mice by Modulating Neutrophil Extracellular Traps and Hepatocyte Apoptosis. Evid Based Complement Alternat Med 2022; 2022: 5769350
  • 181 Yang G, Jia L, Wu J, Ma Y, Cao H, Song N. Effect of tanshinone IIA on oxidative stress and apoptosis in a rat model of fatty liver. Exp Ther Med 2017; 14: 4639-4646
  • 182 Li L, Liu Z, Jiang H, Mao X. Biotechnological production of lycopene by microorganisms. Appl Microbiol Biotechnol 2020; 104: 10307-10324
  • 183 Róvero Costa M, Leite Garcia J, Cristina Vágula de Almeida Silva C. Junio Togneri Ferron A, Valentini Francisqueti-Ferron F, Kurokawa Hasimoto F. Lycopene Modulates Pathophysiological Processes of Non-Alcoholic Fatty Liver Disease in Obese Rats. Antioxidants 2019; 8
  • 184 Jiang W, Guo M-H, Hai X. Hepatoprotective and antioxidant effects of lycopene on non-alcoholic fatty liver disease in rat. World J Gastroenterol 2016; 22: 10180
  • 185 Wang J, Geng T, Zou Q, Yang N, Zhao W, Li Y. Lycopene prevents lipid accumulation in hepatocytes by stimulating PPARα and improving mitochondrial function. J Funct Foods 2020; 67: 103857
  • 186 Peng J, Yuan J-P, Wu C-F, Wang J-H. Fucoxanthin, a Marine Carotenoid Present in Brown Seaweeds and Diatoms: Metabolism and Bioactivities Relevant to Human Health. Mar Drugs 2011; 9: 1806-1828
  • 187 Takatani N, Kono Y, Beppu F, Okamatsu-Ogura Y, Yamano Y, Miyashita K. Fucoxanthin inhibits hepatic oxidative stress, inflammation, and fibrosis in diet-induced nonalcoholic steatohepatitis model mice. Biochem Biophys Res Commun 2020; 528: 305-310
  • 188 Ye J, Zheng J, Tian X, Xu B, Yuan F, Wang B. Fucoxanthin Attenuates Free Fatty Acid-Induced Nonalcoholic Fatty Liver Disease by Regulating Lipid Metabolism/Oxidative Stress/Inflammation via the AMPK/Nrf2/TLR4 Signaling Pathway. Mar Drugs 2022; 20
  • 189 Shrikanth CB, Nandini CD. AMPK in microvascular complications of diabetes and the beneficial effects of AMPK activators from plants. Phytomedicine 2020; 73: 152808
  • 190 Mu Q, Wang H, Tong L, Fang Q, Xiang M, Han L. Betulinic acid improves nonalcoholic fatty liver disease through YY1/FAS signaling pathway. FASEB J 2020; 34: 13033-13048
  • 191 Quan HY, Kim DY, Kim SJ, Jo HK, Kim GW, Chung SH. Betulinic acid alleviates non-alcoholic fatty liver by inhibiting SREBP1 activity via the AMPK–mTOR–SREBP signaling pathway. Biochem Pharmacol 2013; 85: 1330-1340
  • 192 Gu M, Zhao P, Zhang S, Fan S, Yang L, Tong Q. Betulinic acid alleviates endoplasmic reticulum stress-mediated nonalcoholic fatty liver disease through activation of farnesoid X receptors in mice. Br J Pharmacol 2019; 176: 847-863
  • 193 Smeriglio A, Barreca D, Bellocco E, Trombetta D. Proanthocyanidins and hydrolysable tannins: occurrence, dietary intake and pharmacological effects. Br J Pharmacol 2017; 174: 1244-1262
  • 194 Liman MS, Hassen A, McGaw LJ, Sutovsky P, Holm DE. Potential Use of Tannin Extracts as Additives in Semen Destined for Cryopreservation: A Review. Animals 2022; 12
  • 195 Baldwin A, Booth BW. Biomedical applications of tannic acid. J Biomater Appl 2022; 36: 1503-1523
  • 196 Chung M-Y, Song J-H, Lee J, Shin EJ, Park JH, Lee S-H. Tannic acid, a novel histone acetyltransferase inhibitor, prevents non-alcoholic fatty liver disease both in vivo and in vitro model. Mol Metab 2019; 19: 34-48
  • 197 Li M, Liu P, Xue Y, Liang Y, Shi J, Han X. Tannic acid attenuates hepatic oxidative stress, apoptosis and inflammation by activating the Keap1-Nrf2/ARE signaling pathway in arsenic trioxide-toxicated rats. Oncol Rep 2020; 44: 2306-2316
  • 198 Yoon M, Kim J-S, Seo S, Lee K, Um MY, Lee J. Dieckol, a Major Marine Polyphenol, Enhances Non-Rapid Eye Movement Sleep in Mice via the GABAA-Benzodiazepine Receptor. Front Pharmacol 2020; 11
  • 199 Byun K-A, Oh S, Son M, Park C-H, Son K, Byun K. Dieckol Decreases Caloric Intake and Attenuates Nonalcoholic Fatty Liver Disease and Hepatic Lymphatic Vessel Dysfunction in High-Fat-Diet-Fed Mice. Mar Drugs 2021; 19: 495
  • 200 Eo H, Jeon Y, Lee M, Lim Y. Brown Alga Ecklonia cava Polyphenol Extract Ameliorates Hepatic Lipogenesis, Oxidative Stress, and Inflammation by Activation of AMPK and SIRT1 in High-Fat Diet-Induced Obese Mice. J Agric Food Chem 2015; 63: 349-359
  • 201 Oh S, Son M, Byun K-A, Jang JT, Choi CH, Son KH. Attenuating Effects of Dieckol on High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease by Decreasing the NLRP3 Inflammasome and Pyroptosis. Mar Drugs 2021; 19
  • 202 DellʼAgli M, Galli GV, Corbett Y, Taramelli D, Lucantoni L, Habluetzel A. Antiplasmodial activity of Punica granatum L. fruit rind. J Ethnopharmacol 2009; 125: 279-285
  • 203 Feng L, Yin Y, Yang X, Tang H, Jiao Q. Dynamic Variations in Punicalagin and Related Metabolic Substances in Pomegranate Fruit and Leaves During Development Periods. Horticult J 2019; 88: 444-454
  • 204 Yan C, Sun W, Wang X, Long J, Liu X, Feng Z. Punicalagin attenuates palmitate-induced lipotoxicity in HepG2 cells by activating the Keap1-Nrf2 antioxidant defense system. Mol Nutr Food Res 2016; 60: 1139-1149
  • 205 Liu H, Zhan Q, Miao X, Xia X, Yang G, Peng X. Punicalagin Prevents Hepatic Steatosis through Improving Lipid Homeostasis and Inflammation in Liver and Adipose Tissue and Modulating Gut Microbiota in Western Diet-Fed Mice. Mol Nutr Food Res 2021; 65: 2001031
  • 206 Zou X, Yan C, Shi Y, Cao K, Xu J, Wang X. Mitochondrial Dysfunction in Obesity-Associated Nonalcoholic Fatty Liver Disease: The Protective Effects of Pomegranate with Its Active Component Punicalagin. Antioxid Redox Signal 2014; 21: 1557-1570
  • 207 Li X, Deng Y, Zheng Z, Huang W, Chen L, Tong Q. Corilagin, a promising medicinal herbal agent. Biomed Pharmacother 2018; 99: 43-50
  • 208 Zhang R, Chu K, Zhao N, Wu J, Ma L, Zhu C. Corilagin Alleviates Nonalcoholic Fatty Liver Disease in High-Fat Diet-Induced C57BL/6 Mice by Ameliorating Oxidative Stress and Restoring Autophagic Flux. Front Pharmacol 2020; 10
  • 209 Liao M, Zhang R, Wang Y, Mao Z, Wu J, Guo H. Corilagin prevents non-alcoholic fatty liver disease via improving lipid metabolism and glucose homeostasis in high fat diet-fed mice. Front Nutr 2022; 9
  • 210 Liang L, Ye S, Jiang R, Zhou X, Zhou J, Meng S. Liensinine alleviates high fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) through suppressing oxidative stress and inflammation via regulating TAK1/AMPK signaling. Int Immunopharmacol 2022; 104: 108306
  • 211 Wang G-E, Li Y-F, Zhai Y-J, Gong L, Tian J-Y, Hong M. Theacrine protects against nonalcoholic fatty liver disease by regulating acylcarnitine metabolism. Metabolism 2018; 85: 227-239
  • 212 Li G, Zhou F, Chen Y, Zhang W, Wang N. Kukoamine A attenuates insulin resistance and fatty liver through downregulation of Srebp-1c. Biomed Pharmacother 2017; 89: 536-543
  • 213 Ohashi T, Nakade Y, Ibusuki M, Kitano R, Yamauchi T, Kimoto S. Conophylline inhibits high fat diet-induced non-alcoholic fatty liver disease in mice. PLOS ONE 2019; 14: e0210068
  • 214 Sylvester Darvin S, Toppo E, Esakkimuthu S, Ajeesh Krishna TP, Ceasar SA, Stalin A. Hepatoprotective effect of bisbenzylisoquinoline alkaloid tiliamosine from Tiliacora racemosa in high-fat diet/diethylnitrosamine-induced non-alcoholic steatohepatitis. Biomed Pharmacother 2018; 108: 963-973
  • 215 Yue R, Jin G, Wei S, Huang H, Su L, Zhang C. Immunoregulatory Effect of Koumine on Nonalcoholic Fatty Liver Disease Rats. J Immunol Res 2019; 2019: 8325102
  • 216 Wang LL, Zhang ZC, Hassan W, Li Y, Liu J, Shang J. Amelioration of free fatty acid-induced fatty liver by quercetin-3-O-β-D-glucuronide through modulation of peroxisome proliferator-activated receptor-alpha/sterol regulatory element-binding protein-1c signaling. Hepatol Res 2016; 46: 225-238
  • 217 Park J-Y, Lim M-S, Kim S-I, Lee H, Kim S-S, Kwon Y-S. Quercetin-3-O-β-D-Glucuronide Suppresses Lipopolysaccharide-Induced JNK and ERK Phosphorylation in LPS-Challenged RAW264.7 Cells. Biomol Ther 2016; 24
  • 218 Hur HJ, Jeong Y-H, Lee SH, Sung MJ. Quercitrin Ameliorates Hyperlipidemia and Hepatic Steatosis in Ovariectomized Mice. Life 2020; 10
  • 219 PubChem. Quercitrin. https://pubchem.ncbi.nlm.nih.gov/compound/5280459 Accessed 29 Oct 2022
  • 220 Kim JH, Lee BC, Kim JH, Sim GS, Lee DH, Lee KE. The isolation and antioxidative effects of vitexin fromAcer palmatum. Arch Pharm Res 2005; 28: 195-202
  • 221 Inamdar S, Joshi A, Malik S, Boppana R, Ghaskadbi S. Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice. Biochem Biophys Res Commun 2019; 519: 106-112
  • 222 Sarkar MK, Kar A, Jayaraman A, Kar Mahapatra S, Vadivel V. Vitexin isolated from Prosopis cineraria leaves induce apoptosis in K-562 leukemia cells via inhibition of the BCR-ABL-Ras-Raf pathway. J Pharm Pharmacol 2022; 74: 103-111
  • 223 Morissette M, Litim N, Paolo T. Natural Phytoestrogens: A Class of Promising Neuroprotective Agents for Parkinson Disease. In: Discovery and Development of Neuroprotective Agents from Natural Products: Natural Product Drug Discovery 2017: 9-61
  • 224 Pai SA, Munshi RP, Panchal FH, Gaur I-S, Juvekar AR. Chrysin ameliorates nonalcoholic fatty liver disease in rats. Naunyn Schmiedebergs Arch Pharmacol 2019; 392: 1617-1628
  • 225 Song Y, Wu W, Sheng L, Jiang B, Li X, Cai K. Chrysin ameliorates hepatic steatosis induced by a diet deficient in methionine and choline by inducing the secretion of hepatocyte nuclear factor 4α-dependent very low-density lipoprotein. J Biochem Mol Toxicol 2020; 34: e22497
  • 226 Aloud AA, Chinnadurai V, Govindasamy C, Alsaif MA, Al-Numair KS. Galangin, a dietary flavonoid, ameliorates hyperglycaemia and lipid abnormalities in rats with streptozotocin-induced hyperglycaemia. Pharm Biol 2018; 56: 302-308
  • 227 Zhang X, Deng Y, Xiang J, Liu H, Zhang J, Liao J. Galangin Improved Non-Alcoholic Fatty Liver Disease in Mice by Promoting Autophagy. Drug Des Devel Ther 2020; 14: 3393-3405
  • 228 Hiramitsu M, Shimada Y, Kuroyanagi J, Inoue T, Katagiri T, Zang L. Eriocitrin ameliorates diet-induced hepatic steatosis with activation of mitochondrial biogenesis. Sci Rep 2014; 4: 3708
  • 229 Kwon E-Y, Choi M-S. Eriocitrin Improves Adiposity and Related Metabolic Disorders in High-Fat Diet-Induced Obese Mice. J Med Food 2020; 23: 233-241
  • 230 Karimi G, Vahabzadeh M, Lari P, Rashedinia M, Moshiri M. “Silymarin”, a Promising Pharmacological Agent for Treatment of Diseases. Iran J Basic Med Sci 2011; 14: 308-317
  • 231 Salomone F, Barbagallo I, Godos J, Lembo V, Currenti W, Cinà D. Silibinin Restores NAD+ Levels and Induces the SIRT1/AMPK Pathway in Non-Alcoholic Fatty Liver. Nutrients 2017; 9
  • 232 Sahin E, Bagci R, Bektur Aykanat NE, Kacar S, Sahinturk V. Silymarin attenuated nonalcoholic fatty liver disease through the regulation of endoplasmic reticulum stress proteins GRP78 and XBP-1 in mice. J Food Biochem 2020; 44: e13194
  • 233 Geng Y, Wu Z, Buist-Homan M, Blokzijl H, Moshage H. Hesperetin protects against palmitate-induced cellular toxicity via induction of GRP78 in hepatocytes. Toxicol Appl Pharmacol 2020; 404: 115183
  • 234 Li J, Wang T, Liu P, Yang F, Wang X, Zheng W. Hesperetin ameliorates hepatic oxidative stress and inflammation via the PI3K/AKT-Nrf2-ARE pathway in oleic acid-induced HepG2 cells and a rat model of high-fat diet-induced NAFLD. Food Funct 2021; 12: 3898-3918
  • 235 Wdowiak K, Walkowiak J, Pietrzak R, Bazan-Woźniak A, Cielecka-Piontek J. Bioavailability of Hesperidin and Its Aglycone Hesperetin–Compounds Found in Citrus Fruits as a Parameter Conditioning the Pro-Health Potential (Neuroprotective and Antidiabetic Activity)–Mini-Review. Nutrients 2022; 14
  • 236 Stanisic D, Liu LHB, dos Santos RV, Costa AF, Durán N, Tasic L. New Sustainable Process for Hesperidin Isolation and Anti-Ageing Effects of Hesperidin Nanocrystals. Molecules 2020; 25
  • 237 Chen H, Nie T, Zhang P, Ma J, Shan A. Hesperidin attenuates hepatic lipid accumulation in mice fed high-fat diet and oleic acid induced HepG2 via AMPK activation. Life Sci 2022; 296: 120428
  • 238 Zarmouh NO, Mazzio EA, Elshami FM, Messeha SS, Eyunni SVK, Soliman KFA. Evaluation of the Inhibitory Effects of Bavachinin and Bavachin on Human Monoamine Oxidases A and B. Evid Based Complement Alternat Med 2015; 2015: 852194
  • 239 Dong X, Zhu Y, Wang S, Luo Y, Lu S, Nan F. Bavachinin inhibits cholesterol synthesis enzyme FDFT1 expression via AKT/mTOR/SREBP-2 pathway. Int Immunopharmacol 2020; 88: 106865
  • 240 Ali MY, Zaib S, Rahman MM, Jannat S, Iqbal J, Park SK. Didymin, a dietary citrus flavonoid exhibits anti-diabetic complications and promotes glucose uptake through the activation of PI3K/Akt signaling pathway in insulin-resistant HepG2 cells. Chem Biol Interact 2019; 305: 180-194
  • 241 Chang T-C, Chiou W-C, Lai W-H, Huang H-C, Huang Y-L, Liu H-K. Ugonin J improves metabolic disorder and ameliorates nonalcoholic fatty liver disease by regulating the AMPK/AKT signaling pathway. Pharmacol Res 2021; 163: 105298
  • 242 Sun B, Zhang R, Liang Z, Fan A, Kang D. Hyperoside attenuates non-alcoholic fatty liver disease through targeting Nr 4A1 in macrophages. Int Immunopharmacol 2021; 94: 107438
  • 243 Wang S, Sheng F, Zou L, Xiao J, Li P. Hyperoside attenuates non-alcoholic fatty liver disease in rats via cholesterol metabolism and bile acid metabolism. J Adv Res 2021; 34: 109-122
  • 244 Lin W, Jin Y, Hu X, Huang E, Zhu Q. AMPK/PGC-1α/GLUT4-Mediated Effect of Icariin on Hyperlipidemia-Induced Non-Alcoholic Fatty Liver Disease and Lipid Metabolism Disorder in Mice. Biochemistry (Mosc) 2021; 86: 1407-1417
  • 245 Dietz BM, Kang Y-H, Liu G, Eggler AL, Yao P, Chadwick LR. Xanthohumol Isolated from Humulus lupulus Inhibits Menadione-Induced DNA Damage through Induction of Quinone Reductase. Chem Res Toxicol 2005; 18: 1296-1305
  • 246 Zhang Y, Bobe G, Miranda CL, Lowry MB, Hsu VL, Lohr CV. Tetrahydroxanthohumol, a xanthohumol derivative, attenuates high-fat diet-induced hepatic steatosis by antagonizing PPARγ . eLife 2021; 10: e66398
  • 247 Wang W, Chen Z, Zheng T, Zhang M. Xanthohumol alleviates T2DM-induced liver steatosis and fibrosis by mediating the NRF2/RAGE/NF-κB signaling pathway. Future Med Chem 2021; 13: 2069-2081
  • 248 Gao J, Chen G, He H, Liu C, Xiong X, Li J. Therapeutic Effects of Breviscapine in Cardiovascular Diseases: A Review. Front Pharmacol 2017; 8
  • 249 Lan T, Jiang S, Zhang J, Weng Q, Yu Y, Li H. Breviscapine alleviates NASH by inhibiting TGF-β-activated kinase 1-dependent signaling. Hepatology 2022; 76: 155-171
  • 250 Xia S-F, Le G-W, Wang P, Qiu Y-Y, Jiang Y-Y, Tang X. Regressive Effect of Myricetin on Hepatic Steatosis in Mice Fed a High-Fat Diet. Nutrients 2016; 8
  • 251 de Lima M de FR, Cavalcante LA, Costa ECT de A, Veras B, da Silva MV, Cavalcanti LN, Araújo RM. Bioactivity flavonoids from roots of Euphorbia tirucalli L. Phytochem Lett 2021; 41: 186-192
  • 252 Choi H-N, Shin J-Y, Kim J-I. Ameliorative Effect of Myricetin on Nonalcoholic Fatty Liver Disease in ob/ob Mice. J Med Food 2021; 24: 1092-1099
  • 253 Fu Y, Liu B, Liu J, Liu Z, Liang D, Li F. Geniposide, from Gardenia jasminoides Ellis, inhibits the inflammatory response in the primary mouse macrophages and mouse models. Int Immunopharmacol 2012; 14: 792-798
  • 254 Shen B, Feng H, Cheng J, Li Z, Jin M, Zhao L. Geniposide alleviates non-alcohol fatty liver disease via regulating Nrf2/AMPK/mTOR signalling pathways. J Cell Mol Med 2020; 24: 5097-5108
  • 255 Cominguez DC, Park Y-J, Kang Y-M, Nugroho A, Kim S, An H-J. Clitorin ameliorates western diet-induced hepatic steatosis by regulating lipogenesis and fatty acid oxidation in vivo and in vitro. Sci Rep 2022; 12: 4154
  • 256 Jin M, Feng H, Wang Y, Yan S, Shen B, Li Z. Gentiopicroside Ameliorates Oxidative Stress and Lipid Accumulation through Nuclear Factor Erythroid 2-Related Factor 2 Activation. Oxid Med Cell Longev 2020; 2020: 2940746
  • 257 Cheng S, Liang S, Liu Q, Deng Z, Zhang Y, Du J. Diosgenin prevents high-fat diet-induced rat non-alcoholic fatty liver disease through the AMPK and LXR signaling pathways. Int J Mol Med 2018; 41: 1089-1095
  • 258 Liu M, Zhang G, Wu S, Song M, Wang J, Cai W. Schaftoside alleviates HFD-induced hepatic lipid accumulation in mice via upregulating farnesoid X receptor. J Ethnopharmacol 2020; 255: 112776
  • 259 Yu Q, Liu Y, Wu Y, Chen Y. Dihydrocurcumin ameliorates the lipid accumulation, oxidative stress and insulin resistance in oleic acid-induced L02 and HepG2 cells. Biomed Pharmacother 2018; 103: 1327-1336
  • 260 Kim M, Yoo G, Randy A, Kim HS, Nho CW. Chicoric acid attenuate a nonalcoholic steatohepatitis by inhibiting key regulators of lipid metabolism, fibrosis, oxidation, and inflammation in mice with methionine and choline deficiency. Mol Nutr Food Res 2017; 61: 1600632
  • 261 Ma J, Li M, Kalavagunta PK, Li J, He Q, Zhang Y. Protective effects of cichoric acid on H2O2-induced oxidative injury in hepatocytes and larval zebrafish models. Biomed Pharmacother 2018; 104: 679-685
  • 262 Ding X, Jian T, Li J, Lv H, Tong B, Li J. Chicoric Acid Ameliorates Nonalcoholic Fatty Liver Disease via the AMPK/Nrf2/NFκB Signaling Pathway and Restores Gut Microbiota in High-Fat-Diet-Fed Mice. Oxid Med Cell Longev 2020; 2020: 9734560
  • 263 Minh TN, Xuan TD, Tran H-D, Van TM, Andriana Y, Khanh TD. Isolation and Purification of Bioactive Compounds from the Stem Bark of Jatropha podagrica. Molecules 2019; 24
  • 264 Fanaei H, Mard SA, Sarkaki A, Goudarzi G, Khorsandi L. Gallic acid protects the liver against NAFLD induced by dust exposure and high-fat diet through inhibiting oxidative stress and repressing the inflammatory signaling pathways NF-κβ/TNF-α/IL-6 in Wistar rats. Avicenna J Phytomed 2021; 11: 527-540
  • 265 Ismail BS, Abdel-Reheim ES, Soliman HA, Mahmoud B. Protective Effect of Gallic Acid against Nonalcoholic Fatty Liver Disease Induced by High Fat Diet. J Pharm Res Int 2021; 378-397
  • 266 Cha S-H, Hwang Y, Heo S-J, Jun H-S. Diphlorethohydroxycarmalol Attenuates Palmitate-Induced Hepatic Lipogenesis and Inflammation. Mar Drugs 2020; 18
  • 267 Vazquez Prieto MA, Bettaieb A, Rodriguez Lanzi C, Soto VC, Perdicaro DJ, Galmarini CR. Catechin and quercetin attenuate adipose inflammation in fructose-fed rats and 3 T3-L1 adipocytes. Mol Nutr Food Res 2015; 59: 622-633
  • 268 Hodges JK, Sasaki GY, Bruno RS. Anti-inflammatory activities of green tea catechins along the gut–liver axis in nonalcoholic fatty liver disease: lessons learned from preclinical and human studies. J Nutr Biochem 2020; 85: 108478
  • 269 Suárez-Quiroz ML, Campos AA, Alfaro GV, González-Ríos O, Villeneuve P, Figueroa-Espinoza MC. Isolation of green coffee chlorogenic acids using activated carbon. J Food Compost Anal 2014; 33: 55-58
  • 270 Jin S, Chang C, Zhang L, Liu Y, Huang X, Chen Z. Chlorogenic Acid Improves Late Diabetes through Adiponectin Receptor Signaling Pathways in db/db Mice. PLOS ONE 2015; 10: e0120842
  • 271 Wang Z, Lam K-L, Hu J, Ge S, Zhou A, Zheng B. Chlorogenic acid alleviates obesity and modulates gut microbiota in high-fat-fed mice. Food Sci Nutr 2019; 7: 579-588
  • 272 Ding C, Zhao Y, Shi X, Zhang N, Zu G, Li Z. New insights into salvianolic acid A action: Regulation of the TXNIP/NLRP3 and TXNIP/ChREBP pathways ameliorates HFD-induced NAFLD in rats. Sci Rep 2016; 6: 28734
  • 273 Li S, Qian Q, Ying N, Lai J, Feng L, Zheng S. Activation of the AMPK-SIRT1 pathway contributes to protective effects of Salvianolic acid A against lipotoxicity in hepatocytes and NAFLD in mice. Front Pharmacol 2020; 11
  • 274 Li J, Wang S, Yao L, Ma P, Chen Z, Han T-L. 6-gingerol ameliorates age-related hepatic steatosis: Association with regulating lipogenesis, fatty acid oxidation, oxidative stress and mitochondrial dysfunction. Toxicol Appl Pharmacol 2019; 362: 125-135
  • 275 Sarrafan A, Ghobeh M, Yaghmaei P. The effect of 6-gingerol on biochemical and histological parameters in cholesterol-induced nonalcoholic fatty liver disease in NMRI mice. Braz J Pharm Sci 2021; 57: e18020
  • 276 Chen F, Yang L, Zhai L, Huang Y, Chen F, Duan W. Methyl brevifolincarboxylate, a novel influenza virus PB2 inhibitor from Canarium Album (Lour.) Raeusch. Chem Biol Drug Des 2020; 96: 1280-1291
  • 277 Geethangili M, Lin C-W, Mersmann HJ, Ding S-T. Methyl Brevifolincarboxylate Attenuates Free Fatty Acid-Induced Lipid Metabolism and Inflammation in Hepatocytes through AMPK/NF-κB Signaling Pathway. Int J Mol Sci 2021; 22
  • 278 Li H-B, Jiang Y, Chen F. Isolation and Purification of Lutein from the Microalga Chlorella vulgaris by Extraction after Saponification. J Agric Food Chem 2002; 50: 1070-1072
  • 279 Kim JE, Clark RM, Park Y, Lee J, Fernandez ML. Lutein decreases oxidative stress and inflammation in liver and eyes of guinea pigs fed a hypercholesterolemic diet. Nutr Res Pract 2012; 6: 113-119
  • 280 Qiu X, Gao D-H, Xiang X, Xiong Y-F, Zhu T-S, Liu L-G. Ameliorative effects of lutein on non-alcoholic fatty liver disease in rats. World J Gastroenterol 2015; 21: 8061-8072
  • 281 Shah MdMR, Liang Y, Cheng JJ, Daroch M. Astaxanthin-Producing Green Microalga Haematococcus pluvialis: From Single Cell to High Value Commercial Products. Front Plant Sci 2016; 7
  • 282 Wu L, Mo W, Feng J, Li J, Yu Q, Li S. et al. Astaxanthin attenuates hepatic damage and mitochondrial dysfunction in non-alcoholic fatty liver disease by up-regulating the FGF21/PGC-1α pathway. Br J Pharmacol 2020; 177: 3760-3777
  • 283 Li Y, Liu J, Ye B, Cui Y, Geng R, Liu S. Astaxanthin Alleviates Nonalcoholic Fatty Liver Disease by Regulating the Intestinal Flora and Targeting the AMPK/Nrf2 Signal Axis. J Agric Food Chem 2022; 70: 10620-10634
  • 284 Kobori M, Ni Y, Takahashi Y, Watanabe N, Sugiura M, Ogawa K. β-Cryptoxanthin Alleviates Diet-Induced Nonalcoholic Steatohepatitis by Suppressing Inflammatory Gene Expression in Mice. PLOS ONE 2014; 9: e98294
  • 285 Lim JY, Liu C, Hu K-Q, Smith DE, Wu D, Lamon-Fava S. Dietary β-Cryptoxanthin Inhibits High-Refined Carbohydrate Diet–Induced Fatty Liver via Differential Protective Mechanisms Depending on Carotenoid Cleavage Enzymes in Male Mice. J Nutr 2019; 149: 1553-1564
  • 286 Islam MT. Andrographolide, a New Hope in the Prevention and Treatment of Metabolic Syndrome. Front Pharmacol 2017; 8: 571
  • 287 Cabrera D, Wree A, Povero D, Solís N, Hernandez A, Pizarro M. Andrographolide Ameliorates Inflammation and Fibrogenesis and Attenuates Inflammasome Activation in Experimental Non-Alcoholic Steatohepatitis. Sci Rep 2017; 7: 3491
  • 288 Ran L-S, Wu Y-Z, Gan Y-W, Wang H-L, Wu L-J, Zheng C-M. Andrographolide ameliorates hepatic steatosis by suppressing FATP2-mediated fatty acid uptake in mice with nonalcoholic fatty liver disease. J Nat Med. 2022
  • 289 Chen H-J, Liu J. Actein ameliorates hepatic steatosis and fibrosis in high fat diet-induced NAFLD by regulation of insulin and leptin resistant. Biomed Pharmacother 2018; 97: 1386-1396
  • 290 Gao X. Efficacy and Safety of Berberine in Non-alcoholic Steatohepatitis: a Multicentre, Randomised, Placebo-controlled Trial. 2020. https://clinicaltrials.gov/ct2/show/NCT03198572 Accessed 21 Aug 2022
  • 291 Mahadeva S. A Randomised, Double-blind, Placebo-controlled, Phase II, Single-centre Study to Assess the Safety and Efficacy of Silymarin 700 mg Capsules TID for the Treatment of Non-Alcoholic Fatty Liver Disease (NAFLD). 2016. https://clinicaltrials.gov/ct2/show/NCT02006498 Accessed 25 Sep 2022
  • 292 Reddy KR, Navarro V, Afdhal N, Fried M. Single and Multiple Dose Escalation Phase I Study to Evaluate the Safety, Tolerability, and Pharmacokinetics of Orally Administered Silymarin (Legalon) in Non-Cirrhotic Subjects With Chronic Hepatitis C or Non-Alcoholic Fatty Liver Disease. 2008. https://clinicaltrials.gov/ct2/show/NCT00389376 Accessed 25 Aug 2022
  • 293 Fried M, Navarro V, Afdhal N, Reddy KR, Belle SH, Harrison SA. A Multicenter, Randomized, Placebo Controlled Study to Assess the Safety and Efficacy of Orally Administered Silymarin Preparation (Legalon) for the Treatment of Non-Cirrhotic Patients With Non-Alcoholic Steatohepatitis. 2019. https://clinicaltrials.gov/ct2/show/NCT00680407 Accessed 11 Sep 2022
  • 294 Hauser G. Silymarin® – Efficacy in Treatment of Non-alcoholic Fatty Liver Disease (NAFLD) Controlled by Laboratory and and Elastographic Parameters. 2020. https://clinicaltrials.gov/ct2/show/NCT02973295 Accessed 18 Sep 2022
  • 295 Wicklow B. Safety and Efficacy of Resveratrol for the Treatment of Non-Alcoholic Fatty Liver Disease and Associated Insulin Resistance in Overweight and Obese Adolescents. 2017. https://clinicaltrials.gov/ct2/show/NCT02216552 Accessed 12 Oct 2022
  • 296 Nielsen S. Long-term Investigation of Resveratrol on Lipid Turnover in Obese Men With Nonalcoholic Fatty Liver Disease. Effects on Liver Fat Content and Basal and Insulin Stimulated FFA and VLDL-triglyceride Metabolism. 2014. https://clinicaltrials.gov/ct2/show/NCT01446276 Accessed 22 Sep 2022
  • 297 Hekmatdoost A. The Effects of Resveratrol Supplement on Lipid Profile, Liver Enzymes, Inflammatory Factors and Hepatic Fibrosis in Patients With Nonalcoholic Steatohepatitis. 2014. https://clinicaltrials.gov/ct2/show/NCT02030977 Accessed 20 Oct 2022
  • 298 Grønbæk H. Long-term Investigation of Resveratrol on Management of Metabolic Syndrome, Osteoporosis and Inflammation, and Identification of Plant Derived Anti-inflammatory Compounds, Study 3. 2015. https://clinicaltrials.gov/ct2/show/NCT01464801 Accessed 14 Oct 2022
  • 299 Jørgensen JOL. Potential Beneficial Effects of Resveratrol on Obesity, Metabolic Syndrome and Inflammation – Emphasis on Description of the Molecular Biology Underpinning the Interplay Between Calorie Restriction, SIRT1, STAT5 and the GH/IGF-I Axis. 2012. https://clinicaltrials.gov/ct2/show/NCT01150955 Accessed 11 Sep 2022
  • 300 Hellmann PH. The Effect of Curcumin on Liver Fat Content in Obese Subjects. 2021. https://clinicaltrials.gov/ct2/show/NCT03864783 Accessed 14 Oct 2022
  • 301 Hekmatdoost A. The Effect of Curcumin Supplement on Metabolic Factors and Hepatic Fibrosis in Nonalcoholic Fatty Liver Patients. 2020. https://clinicaltrials.gov/ct2/show/NCT02908152 Accessed 30 Aug 2022
  • 302 Nobili V. Hydroxytyrosol and Vitamin E in the Treatment of Children With Biopsy-proven NASH. 2018. https://clinicaltrials.gov/ct2/show/NCT02842567 Accessed 27 Sep 2022
  • 303 Ling W. Effects of Purified Anthocyanin on Oxidative and Inflammatory Markers in Subjects With Nonalcoholic Fatty Liver Disease: A Randomized, Double-Blinded, Placebo-Controlled Trial. 2014. https://clinicaltrials.gov/ct2/show/NCT01940263 Accessed 11 Oct 2022

Correspondence

Dr. Nilanjan Ghosh
Department of Pharmaceutical Technology
Jadavpur University
188, Raja Subodh Chandra Mallick Rd, Jadavpur
Kolkata – 700 032
West Bengal, India   
Phone: 95 64 67 54 20   
Fax: 0 33 24 57 24 55   

Publication History

Received: 22 December 2023

Accepted after revision: 07 March 2024

Accepted Manuscript online:
08 March 2024

Article published online:
15 April 2024

© 2024. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Mcpherson S, Hardy T, Henderson E, Burt A, Day C, Anstee Q. Evidence of NAFLD Progression from Steatosis to Fibrosing-Steatohepatitis Using Paired Biopsies: Implications for Prognosis & Clinical Management. J Hepatol 2014; 62
  • 2 Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018; 67: 328-357
  • 3 Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease–Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016; 64: 73-84
  • 4 Benedict M, Zhang X. Non-alcoholic fatty liver disease: An expanded review. World J Hepatol 2017; 9: 715
  • 5 Bikbov MM, Gilmanshin TR, Zainullin RM, Kazakbaeva GM, Iakupova EM, Fakhretdinova AA. Prevalence of non-alcoholic fatty liver disease in the Russian Ural Eye and Medical Study and the Ural Very Old Study. Sci Rep 2022; 12: 7842
  • 6 Zhong F, Zhou X, Xu J, Gao L. Rodent Models of Nonalcoholic Fatty Liver Disease. Digestion 2020; 101: 522-535
  • 7 Armstrong MJ, Adams LA, Canbay A, Syn WK. Extrahepatic complications of nonalcoholic fatty liver disease. Hepatology 2014; 59: 1174-1197
  • 8 Sarkar A, Saha S, Paul A, Maji A, Roy P, Maity TK. Understanding stem cells and its pivotal role in regenerative medicine. Life Sci 2021; 273: 119270
  • 9 Utzschneider KM, Kahn SE. The Role of Insulin Resistance in Nonalcoholic Fatty Liver Disease. J Clin Endocrinol Metab 2006; 91: 4753-4761
  • 10 Tarantino G, Sinatti G, Citro V, Santini jr S, Balsano C. Sarcopenia, a condition shared by various diseases: can we alleviate or delay the progression?. Intern Emerg Med 2023; 18: 1887-1895
  • 11 Bagherniya M, Nobili V, Blesso CN, Sahebkar A. Medicinal plants and bioactive natural compounds in the treatment of non-alcoholic fatty liver disease: A clinical review. Pharmacol Res 2018; 130: 213-240
  • 12 Balato N, Napolitano M, Ayala F, Patruno C, Megna M, Tarantino G. Nonalcoholic fatty liver disease, spleen and psoriasis: New aspects of low-grade chronic inflammation. World J Gastroenterol 2015; 21: 6892-6897
  • 13 Lian J, Fu J. Pioglitazone for NAFLD Patients With Prediabetes or Type 2 Diabetes Mellitus: A Meta-Analysis. Front Endocrinol 2021; 12
  • 14 Bril F, Biernacki D, Kalavalapalli S, Lomonaco R, Subbarayan S, Lai J. Role of Vitamin E for Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Care 2019; 42: dc190167
  • 15 Lin C, Yu B, Chen L, Zhang Z, Ye W, Zhong H. Obeticholic Acid Induces Hepatoxicity Via FXR in the NAFLD Mice. Front Pharmacol 2022; 13
  • 16 Cheng C, Li Z, Zhao X, Liao C, Quan J, Bode AM. Natural alkaloid and polyphenol compounds targeting lipid metabolism: Treatment implications in metabolic diseases. Eur J Pharmacol 2020; 870: 172922
  • 17 Parthasarathy G, Revelo X, Malhi H. Pathogenesis of Nonalcoholic Steatohepatitis: An Overview. Hepatol Commun 2020; 4
  • 18 Neuman MG, Cohen LB, Nanau RM. Biomarkers in Nonalcoholic Fatty Liver Disease. Can J Gastroenterol Hepatol 2014; 28: 607-618
  • 19 Thibaut R, Gage M, Torra I, Chabrier G, Venteclef N, Alzaid F. Liver macrophages and inflammation in physiology and physiopathology of non‐alcoholic fatty liver disease. FEBS J 2021; 289
  • 20 Shan Z, Ju C. Hepatic Macrophages in Liver Injury. Front Immunol 2020; 11
  • 21 Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 2012; 18: 1028-1040
  • 22 Arroyave-Ospina JC, Wu Z, Geng Y, Moshage H. Role of Oxidative Stress in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: Implications for Prevention and Therapy. Antioxidants 2021; 10
  • 23 Martín-Fernández M, Arroyo V, Carnicero C, Sigüenza R, Busta R, Mora N. Role of Oxidative Stress and Lipid Peroxidation in the Pathophysiology of NAFLD. Antioxidants (Basel) 2022; 11: 2217
  • 24 Guo C, Sun L, Chen X, Zhang D. Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regen Res 2013; 8: 2003-2014
  • 25 Mittal M, Siddiqui M, Tran KA, Pothireddy S, Malik A. Reactive Oxygen Species in Inflammation and Tissue Injury. Antioxid Redox Signal 2013; 20
  • 26 Peverill W, Powell L, Skoien R. Evolving Concepts in the Pathogenesis of NASH: Beyond Steatosis and Inflammation. Int J Mol Sci 2014; 15: 8591-8638
  • 27 Yin H, Price F, Rudnicki M. Satellite Cells and the Muscle Stem Cell Niche. Physiol Rev 2013; 93: 23-67
  • 28 Basaranoglu M, Basaranoglu G, Bugianesi E. Carbohydrate intake and nonalcoholic fatty liver disease: Fructose as a weapon of mass destruction. Hepatobiliary Surg Nutr 2015; 4: 109-116
  • 29 Valdes P, Cuevas-Ramos D, Aguilar-Salinas C. Metabolic syndrome and non-alcoholic fatty liver disease. Annals Hepatol 2009; 8. Suppl1:S18-24
  • 30 Listenberger LL, Han X, Lewis SE, Cases S, Farese RV, Ory DS. Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proc Natl Acad Sci U S A 2003; 100: 3077-3082
  • 31 Bianco C, Casirati E, Malvestiti F, Valenti L. Genetic predisposition similarities between NASH and ASH: Identification of new therapeutic targets. JHEP Rep 2021; 3: 100284
  • 32 Dabravolski S, Bezsonov E, Baig M, Popkova T, Orekhov A. Mitochondrial Lipid Homeostasis at the Crossroads of Liver and Heart Diseases. Int J Mol Sci 2021; 22: 6949
  • 33 Herzig S, Shaw R. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol 2017; 19
  • 34 Fabbrini E, Sullivan S, Klein S. Obesity and Nonalcoholic Fatty Liver Disease: Biochemical, Metabolic, and Clinical Implications. Hepatology 2010; 51: 679-689
  • 35 Ibrahim S, Hirsova P, Gores G. Non-alcoholic steatohepatitis pathogenesis: Sublethal hepatocyte injury as a driver of liver inflammation. Gut 2018; 67: gutjnl-2017
  • 36 Zhang C-Y, Yuan W-G, He P, Lei J-H, Wang C-X. Liver fibrosis and hepatic stellate cells: Etiology, pathological hallmarks and therapeutic targets. World J Gastroenterol 2016; 22: 10512
  • 37 Chitturi S, Abeygunasekera S, Farrell GC, Holmes-Walker J, Hui JM, Fung C. NASH and insulin resistance: Insulin hypersecretion and specific association with the insulin resistance syndrome. Hepatology 2002; 35: 373-379
  • 38 Zhang C, Zhou B, Sheng J, Chen Y, Cao Y, Chen C. Molecular mechanisms of hepatic insulin resistance in nonalcoholic fatty liver disease and potential treatment strategies. Pharmacol Res 2020; 159: 104984
  • 39 Sarkar A, Saha R, Saha S, Bhowmik R, Chatterjee A, Paul A. Transesterification, GC-MS profiling, and in vitro antimicrobial potential of oil obtained from seeds of Citrus maxima (Burm.) Merr. Ind Crops Prod 2022; 189: 115764
  • 40 Sarkar A, Banerjee T, Maji A, Paul A, Guria T. Mikania Species: Revealing Phytochemicals from the Pandoraʼs Box. In: Chakraborty R, Sen S, editors New Avenues in Drug Discovery and Bioactive Natural Products. BENTHAM SCIENCE PUBLISHERS; 2023: 149-167
  • 41 Sarkar S, Kar A, Shaw P, DasGupta B, Keithellakpam OS, Mukherjee PK, Bhardwaj PK, Sharma N, Haldar PK, Sinha S. Hydroalcoholic root extracts of Houttuynia cordata (Thunb.) standardized by UPLC-Q-TOF-MS/MS promotes apoptosis in human hepatocarcinoma cell HepG2 via GSK-3β/β-catenin/PDL-1 axis. Fitoterapia 2023; 171: 105684
  • 42 Tarantino G, Balsano C, Santini SJ, Brienza G, Clemente I, Cosimini B, Sinatti G. It Is High Time Physicians Thought of Natural Products for Alleviating NAFLD. Is There Sufficient Evidence to Use Them?. Int J Mol Sci 2021; 22: 13424
  • 43 Zheng X, Wu F, Lin X, Shen L, Feng Y. Developments in drug delivery of bioactive alkaloids derived from traditional Chinese medicine. Drug Deliv 2018; 25: 398-416
  • 44 Yang Y, Vong CT, Zeng S, Gao C, Chen Z, Fu C. Tracking evidences of Coptis chinensis for the treatment of inflammatory bowel disease from pharmacological, pharmacokinetic to clinical studies. J Ethnopharmacol 2021; 268: 113573
  • 45 Zhang Y, Ma J, Zhang W. Berberine for bone regeneration: Therapeutic potential and molecular mechanisms. J Ethnopharmacol 2021; 277: 114249
  • 46 Guo T, Woo S-L, Guo X, Li H, Zheng J, Botchlett R. Berberine Ameliorates Hepatic Steatosis and Suppresses Liver and Adipose Tissue Inflammation in Mice with Diet-induced Obesity. Sci Rep 2016; 6: 22612
  • 47 Li Y, Zhao X, Feng X, Liu X, Deng C, Hu C-H. Berberine Alleviates Olanzapine-Induced Adipogenesis via the AMPKα–SREBP Pathway in 3 T3-L1 Cells. Int J Mol Sci 2016; 17
  • 48 Shan M, Dai Y, Ren X, Zheng J, Zhang K, Chen B. Berberine mitigates nonalcoholic hepatic steatosis by downregulating SIRT1-FoxO1-SREBP2 pathway for cholesterol synthesis. J Integr Med 2021; 19: 545-554
  • 49 Liang H, Wang Y. Berberine alleviates hepatic lipid accumulation by increasing ABCA1 through the protein kinase C δ pathway. Biochem Biophys Res Commun 2018; 498: 473-480
  • 50 Chang X, Wang Z, Zhang J, Yan H, Bian H, Xia M. Lipid profiling of the therapeutic effects of berberine in patients with nonalcoholic fatty liver disease. J Transl Med 2016; 14: 266
  • 51 Day CR, Kempson SA. Betaine chemistry, roles, and potential use in liver disease. Biochim Biophys Acta 2016; 1860: 1098-1106
  • 52 Deminice R, da Silva RP, Lamarre SG, Kelly KB, Jacobs RL, Brosnan ME. Betaine supplementation prevents fatty liver induced by a high-fat diet: effects on one-carbon metabolism. Amino Acids 2015; 47: 839-846
  • 53 Xu G, Huang K, Zhou J. Hepatic AMP Kinase as a Potential Target for Treating Nonalcoholic Fatty Liver Disease: Evidence from Studies of Natural Products. Curr Med Chem 2018; 25: 889-907
  • 54 Kim DH, Lee B, Kim MJ, Park MH, An HJ, Lee EK. Molecular Mechanism of Betaine on Hepatic Lipid Metabolism: Inhibition of Forkhead Box O1 (FoxO1) Binding to Peroxisome Proliferator-Activated Receptor Gamma (PPARγ). J Agric Food Chem 2016; 64: 6819-6825
  • 55 Wang L, Chen L, Tan Y, Wei J, Chang Y, Jin T. Betaine supplement alleviates hepatic triglyceride accumulation of apolipoprotein E deficient mice via reducing methylation of peroxisomal proliferator-activated receptor alpha promoter. Lipids Health Dis 2013; 12: 34
  • 56 Xu L, Huang D, Hu Q, Wu J, Wang Y, Feng J. Betaine alleviates hepatic lipid accumulation via enhancing hepatic lipid export and fatty acid oxidation in rats fed with a high-fat diet. Br J Nutr 2015; 113: 1835-1843
  • 57 Liu W, Yi D-D, Guo J-L, Xiang Z-X, Deng L-F, He L. Nuciferine, extracted from Nelumbo nucifera Gaertn, inhibits tumor-promoting effect of nicotine involving Wnt/β-catenin signaling in non-small cell lung cancer. J Ethnopharmacol 2015; 165: 83-93
  • 58 Lu Y, He Y, Wang M, Zhang L, Yang L, Wang Z. Characterization of nuciferine metabolism by P450 enzymes and uridine diphosphate glucuronosyltransferases in liver microsomes from humans and animals. Acta Pharmacol Sin 2010; 31: 1635-1642
  • 59 Ma C, Li G, He Y, Xu B, Mi X, Wang H. Pronuciferine and nuciferine inhibit lipogenesis in 3 T3-L1 adipocytes by activating the AMPK signaling pathway. Life Sci 2015; 136: 120-125
  • 60 Zhang C, Deng J, Liu D, Tuo X, Xiao L, Lai B. Nuciferine ameliorates hepatic steatosis in high-fat diet/streptozocin-induced diabetic mice through PPARα/PGC1α pathway. Br J Pharmacol 2018; 175
  • 61 Guo F, Yang X, Li X, Feng R, Guan C, Wang Y. Nuciferine Prevents Hepatic Steatosis and Injury Induced by a High-Fat Diet in Hamsters. PloS one 2013; 8: e63770
  • 62 Gong X, Gao Y, Guo G, Vondran FWR, Schwartlander R, Efimova E. Effect of matrine on primary human hepatocytes in vitro. Cytotechnology 2015; 67: 255-265
  • 63 Liu Z, Zhang Y, Tang Z, Xu J, Ma M, Pan S. Matrine attenuates cardiac fibrosis by affecting ATF6 signaling pathway in diabetic cardiomyopathy. Eur J Pharmacol 2017; 804: 21-30
  • 64 Liu J, Zhao Y, Xia J, Qiu M. Matrine induces toxicity in mouse liver cells through an ROS-dependent mechanism. Res Vet Sci 2020; 132: 308-311
  • 65 Zhang H-F, Shi L-J, Song G-Y, Cai Z-G, Wang C, An R-J. Protective effects of matrine against progression of high-fructose diet-induced steatohepatitis by enhancing antioxidant and anti-inflammatory defences involving Nrf2 translocation. Food Chem Toxicol 2013; 55: 70-77
  • 66 Zeng X-Y, Wang H, Bai F, Zhou X, Li S-P, Ren L-P. Identification of matrine as a promising novel drug for hepatic steatosis and glucose intolerance with HSP72 as an upstream target. Br J Pharmacol 2015; 172: 4303-4318
  • 67 Mahzari A, Li S, Zhou X, Li D, Fouda S, Alhomrani M. Matrine Protects Against MCD-Induced Development of NASH via Upregulating HSP72 and Downregulating mTOR in a Manner Distinctive From Metformin. Front Pharmacol 2019; 10
  • 68 Fei Z-W, Qiu M-K, Qi X-Q, Dai Y-X, Wang S-Q, Quan Z-W. Oxymatrine suppresses proliferation and induces apoptosis of hemangioma cells through inhibition of HIF-1a signaling. Int J Immunopathol Pharmacol 2015; 28: 201-208
  • 69 Shi L, Shi L, Zhang H, Hu Z, Wang C, Zhang D. Oxymatrine ameliorates non-alcoholic fatty liver disease in rats through peroxisome proliferator-activated receptor-α activation. Mol Med Rep 2013; 8: 439-445
  • 70 Shi L, Shi L, Zhang H, Hu Z, Wang C, Zhang D. Oxymatrine ameliorates non-alcoholic fatty liver disease in rats through peroxisome proliferator-activated receptor-α activation. Mol Med Rep 2013; 8
  • 71 Derosa G, Maffioli P, Sahebkar A. Piperine and Its Role in Chronic Diseases. In: Gupta SC, Prasad S, Aggarwal BB, editors Anti-inflammatory Nutraceuticals and Chronic Diseases. Cham: Springer International Publishing; 2016: 173-184
  • 72 Park U-H, Jeong H-S, Jo E-Y, Park T, Yoon SK, Kim E-J. Piperine, a Component of Black Pepper, Inhibits Adipogenesis by Antagonizing PPARγ Activity in 3 T3-L1 Cells. J Agric Food Chem 2012; 60: 3853-3860
  • 73 Kim KJ, Lee M-S, Jo K, Hwang J-K. Piperidine alkaloids from Piper retrofractum Vahl. protect against high-fat diet-induced obesity by regulating lipid metabolism and activating AMP-activated protein kinase. Biochem Biophys Res Commun 2011; 411: 219-225
  • 74 Zhou J, Chan L, Zhou S. Trigonelline: A Plant Alkaloid with Therapeutic Potential for Diabetes and Central Nervous System Disease. Curr Med Chem 2012; 19: 3523-3531
  • 75 Zia T, Hasnain SN, Hasan SK. Evaluation of the oral hypoglycaemic effect of Trigonella foenum-graecum L. (methi) in normal mice. J Ethnopharmacol 2001; 75: 191-195
  • 76 Ilavenil S, Arasu MV, Lee J-C, Kim DH, Roh SG, Park HS. Trigonelline attenuates the adipocyte differentiation and lipid accumulation in 3 T3-L1 cells. Phytomedicine 2014; 21: 758-765
  • 77 Zhang D-F, Zhang F, Zhang J, Zhang R-M, Li R. Protection effect of trigonelline on liver of rats with non-alcoholic fatty liver diseases. Asian Pac J Trop Med 2015; 8: 651-654
  • 78 Song C-Y, Zeng X, Chen S-W, Hu P-F, Zheng Z-W, Ning B-F. Sophocarpine alleviates non-alcoholic steatohepatitis in rats. J Gastroenterol Hepatol 2011; 26: 765-774
  • 79 Song C-Y, Shi J, Zeng X, Zhang Y, Xie W-F, Chen Y-X. Sophocarpine alleviates hepatocyte steatosis through activating AMPK signaling pathway. Toxicol In Vitro 2013; 27: 1065-1071
  • 80 Lee W-Y, Lee C-Y, Lee J-S, Kim C-E. Identifying Candidate Flavonoids for Non-Alcoholic Fatty Liver Disease by Network-Based Strategy. Front Pharmacol 2022; 13
  • 81 Wei D, Zhang G, Zhu Z, Zheng Y, Yan F, Pan C. Nobiletin Inhibits Cell Viability via the SRC/AKT/STAT3/YY1AP1 Pathway in Human Renal Carcinoma Cells. Front Pharmacol 2019; 10
  • 82 Yuk T, Kim Y, Yang J, Sung J, Jeong HS, Lee J. Nobiletin Inhibits Hepatic Lipogenesis via Activation of AMP-Activated Protein Kinase. Evid Based Complement Alternat Med 2018; 2018: 7420265
  • 83 Bunbupha S, Pakdeechote P, Maneesai P, Prasarttong P. Nobiletin alleviates high-fat diet-induced nonalcoholic fatty liver disease by modulating AdipoR1 and gp91phox expression in rats. J Nutr Biochem 2021; 87: 108526
  • 84 Peng Z, Li X, Xing D, Du X, Wang Z, Liu G. Nobiletin alleviates palmitic acid-induced NLRP3 inflammasome activation in a sirtuin 1-dependent manner in AML-12 cells. Mol Med Rep 2018; 18: 5815-5822
  • 85 Liu D, Mao Y, Ding L, Zeng X-A. Dihydromyricetin: A review on identification and quantification methods, biological activities, chemical stability, metabolism and approaches to enhance its bioavailability. Trends Food Sci Technol 2019; 91: 586-597
  • 86 Xie C, Chen Z, Zhang C, Xu X, Jin J, Zhan W. Dihydromyricetin ameliorates oleic acid-induced lipid accumulation in L02 and HepG2 cells by inhibiting lipogenesis and oxidative stress. Life Sci 2016; 157: 131-139
  • 87 Zeng X, Yang J, Hu O, Huang J, Ran L, Chen M. Dihydromyricetin Ameliorates Nonalcoholic Fatty Liver Disease by Improving Mitochondrial Respiratory Capacity and Redox Homeostasis Through Modulation of SIRT3 Signaling. Antioxid Redox Signal 2019; 30: 163-183
  • 88 Guo L, Zhang H, Yan X. Protective effect of dihydromyricetin revents fatty liver through nuclear factor-κB/p 53/B-cell lymphoma 2-associated X protein signaling pathways in a rat model. Mol Med Rep 2019; 19: 1638-1644
  • 89 Ganeshpurkar A, Saluja AK. The Pharmacological Potential of Rutin. Saudi Pharm J 2017; 25: 149-164
  • 90 Liu Q, Pan R, Ding L, Zhang F, Hu L, Ding B. Rutin exhibits hepatoprotective effects in a mouse model of non-alcoholic fatty liver disease by reducing hepatic lipid levels and mitigating lipid-induced oxidative injuries. Int Immunopharmacol 2017; 49: 132-141
  • 91 Panchal SK, Poudyal H, Arumugam TV, Brown L. Rutin Attenuates Metabolic Changes, Nonalcoholic Steatohepatitis, and Cardiovascular Remodeling in High-Carbohydrate, High-Fat Diet-Fed Rats. J Nutr 2011; 141: 1062-1069
  • 92 Sarkar A, Paul A, Banerjee T, Maji A, Saha S, Bishayee A. Therapeutic advancements in targeting BCL-2 family proteins by epigenetic regulators, natural, and synthetic agents in cancer. Eur J Pharmacol 2023; 944: 175588
  • 93 Hernández-Aquino E, Muriel P. Beneficial effects of naringenin in liver diseases: Molecular mechanisms. World J Gastroenterol 2018; 24: 1679-1707
  • 94 Zobeiri M, Belwal T, Parvizi F, Naseri R, Farzaei HM, Nabavi FS. Naringenin and its Nano-formulations for Fatty Liver: Cellular Modes of Action and Clinical Perspective. Curr Pharm Biotechnol 2018; 19: 196-205
  • 95 Wang Q, Ou Y, Hu G, Wen C, Yue S, Chen C. Naringenin attenuates non-alcoholic fatty liver disease by down-regulating the NLRP3/NF-κB pathway in mice. Br J Pharmacol 2020; 177: 1806-1821
  • 96 Chang H-H, Yi P-L, Cheng C-H, Lu C-Y, Hsiao Y-T, Tsai Y-F. Biphasic effects of baicalin, an active constituent of Scutellaria baicalensis Georgi, in the spontaneous sleep–wake regulation. J Ethnopharmacol 2011; 135: 359-368
  • 97 Liu J, Yuan Y, Gong X, Zhang L, Zhou Q, Wu S. Baicalin and its nanoliposomes ameliorates nonalcoholic fatty liver disease via suppression of TLR4 signaling cascade in mice. Int Immunopharmacol 2020; 80: 106208
  • 98 Hu Q, Zhang W, Wu Z, Tian X, Xiang J, Li L. Baicalin and the liver-gut system: Pharmacological bases explaining its therapeutic effects. Pharmacol Res 2021; 165: 105444
  • 99 Zhong X, Liu H. Baicalin attenuates diet induced nonalcoholic steatohepatitis by inhibiting inflammation and oxidative stress via suppressing JNK signaling pathways. Biomed Pharmacother 2018; 98: 111-117
  • 100 Banerjee R, Dasgupta B, Kar A, Bhardwaj PK, Sharma N, Haldar PK. Quality evaluation of different black rice varieties of northeastern region of India. Phytochem Anal 2023; 34: 507-517
  • 101 Li Y, Yao J, Han C, Yang J, Chaudhry MT, Wang S. Quercetin, Inflammation and Immunity. Nutrients 2016; 8
  • 102 Yang H, Yang T, Heng C, Zhou Y, Jiang Z, Qian X. Quercetin improves nonalcoholic fatty liver by ameliorating inflammation, oxidative stress, and lipid metabolism in db/db mice. Phytother Res 2019; 33: 3140-3152
  • 103 Yi H, Peng H, Wu X, Xu X, Kuang T, Zhang J. The Therapeutic Effects and Mechanisms of Quercetin on Metabolic Diseases: Pharmacological Data and Clinical Evidence. Oxid Med Cell Longev 2021; 2021: 6678662
  • 104 Liu L, Gao C, Yao P, Gong Z. Quercetin Alleviates High-Fat Diet-Induced Oxidized Low-Density Lipoprotein Accumulation in the Liver: Implication for Autophagy Regulation. Biomed Res Int 2015; 2015: 607531
  • 105 Lin Y, Shi R, Wang X, Shen H-M. Luteolin, a Flavonoid with Potential for Cancer Prevention and Therapy. Curr Cancer Drug Targets 2008; 8: 634-646
  • 106 Liu X, Sun R, Li Z, Xiao R, Lv P, Sun X. Luteolin alleviates non-alcoholic fatty liver disease in rats via restoration of intestinal mucosal barrier damage and microbiota imbalance involving in gut-liver axis. Arch Biochem Biophys 2021; 711: 109019
  • 107 Yin Y, Gao L, Lin H, Wu Y, Han X, Zhu Y. Luteolin improves non-alcoholic fatty liver disease in db/db mice by inhibition of liver X receptor activation to down-regulate expression of sterol regulatory element binding protein 1c. Biochem Biophys Res Commun 2017; 482: 720-726
  • 108 Chen R, Chen X, Zhu T, Liu J, Xiang X, Yu J. Integrated Transcript and Metabolite Profiles Reveal That EbCHI Plays an Important Role in Scutellarin Accumulation in Erigeron breviscapus Hairy Roots. Front Plant Sci 2018; 9
  • 109 Hua Fan, Xiande Ma, Peng Lin, Qiang Kang, Zhilong Zhao, Lina Wang. Scutellarin Prevents Nonalcoholic Fatty Liver Disease (NAFLD) and Hyperlipidemia via PI3K/AKT-Dependent Activation of Nuclear Factor (Erythroid-Derived 2)-Like 2 (Nrf2) in Rats. Med Sci Monit 2017; 23: 5599-5612
  • 110 Zhang X, Ji R, Sun H, Peng J, Ma X, Wang C. Scutellarin ameliorates nonalcoholic fatty liver disease through the PPARγ/PGC-1α-Nrf2 pathway. Free Radic Res 2018; 52: 198-211
  • 111 Xin X, Chen C, Hu Y-Y, Feng Q. Protective effect of genistein on nonalcoholic fatty liver disease (NAFLD). Biomed Pharmacother 2019; 117: 109047
  • 112 Wang W, Chen J, Mao J, Li H, Wang M, Zhang H. Genistein Ameliorates Non-alcoholic Fatty Liver Disease by Targeting the Thromboxane A2 Pathway. J Agric Food Chem 2018; 66: 5853-5859
  • 113 Seidemann L, Krüger A, Kegel-Hübner V, Seehofer D, Damm G. Influence of Genistein on Hepatic Lipid Metabolism in an In Vitro Model of Hepatic Steatosis. Molecules 2021; 26
  • 114 Khan N, Syed DN, Ahmad N, Mukhtar H. Fisetin: A Dietary Antioxidant for Health Promotion. Antioxid Redox Signal 2013; 19: 151-162
  • 115 Liou C-J, Wei C-H, Chen Y-L, Cheng C-Y, Wang C-L, Huang W-C. Fisetin Protects Against Hepatic Steatosis Through Regulation of the Sirt1/AMPK and Fatty Acid β-Oxidation Signaling Pathway in High-Fat Diet-Induced Obese Mice. Cell Physiol Biochem 2018; 49: 1870-1884
  • 116 Dai X, Kuang Q, Sun Y, Xu M, Zhu L, Ge C. Fisetin represses oxidative stress and mitochondrial dysfunction in NAFLD through suppressing GRP78-mediated endoplasmic reticulum (ER) stress. J Funct Foods 2022; 90: 104954
  • 117 Gaballah HH, El-Horany HE, Helal DS. Mitigative effects of the bioactive flavonol fisetin on high-fat/high-sucrose induced nonalcoholic fatty liver disease in rats. J Cell Biochem 2019; 120: 12762-12774
  • 118 Brito-Arias M. Synthesis and Characterization of Glycosides. 2007
  • 119 Leong XY, Thanikachalam PV, Pandey M, Ramamurthy S. A systematic review of the protective role of swertiamarin in cardiac and metabolic diseases. Biomed Pharmacother 2016; 84: 1051-1060
  • 120 Patel TP, Rawal K, Soni S, Gupta S. Swertiamarin ameliorates oleic acid induced lipid accumulation and oxidative stress by attenuating gluconeogenesis and lipogenesis in hepatic steatosis. Biomed Pharmacother 2016; 83: 785-791
  • 121 Xu L, Li D, Zhu Y, Cai S, Liang X, Tang Y. Swertiamarin supplementation prevents obesity-related chronic inflammation and insulin resistance in mice fed a high-fat diet. Adipocyte 2021; 10: 160-173
  • 122 Yang Y, Li J, Wei C, He Y, Cao Y, Zhang Y. Amelioration of nonalcoholic fatty liver disease by swertiamarin in fructose-fed mice. Phytomedicine 2019; 59: 152782
  • 123 Wang J, Chen X, Wang W, Zhang Y, Yang Z, Jin Y. Glycyrrhizic acid as the antiviral component of Glycyrrhiza uralensis Fisch. against coxsackievirus A16 and enterovirus 71 of hand foot and mouth disease. J Ethnopharmacol 2013; 147: 114-121
  • 124 Sun X, Duan X, Wang C, Liu Z, Sun P, Huo X. Protective effects of glycyrrhizic acid against non-alcoholic fatty liver disease in mice. Eur J Pharmacol 2017; 806: 75-82
  • 125 Wang C, Duan X, Sun X, Liu Z, Sun P, Yang X. Protective effects of glycyrrhizic acid from edible botanical glycyrrhiza glabra against non-alcoholic steatohepatitis in mice. Food Funct 2016; 7: 3716-3723
  • 126 Nabavi SF, Braidy N, Orhan IE, Badiee A, Daglia M, Nabavi SM. Rhodiola rosea L. and Alzheimerʼs Disease: From Farm to Pharmacy. Phytother Res 2016; 30: 532-539
  • 127 Dou X, Ding Q, Lai S, Jiang F, Song Q, Zhao X. Salidroside alleviates lipotoxicity-induced cell death through inhibition of TLR4/MAPKs pathway, and independently of AMPK and autophagy in AML-12 mouse hepatocytes. J Funct Foods 2020; 65: 103691
  • 128 Zheng T, Yang X, Li W, Wang Q, Chen L, Wu D. Salidroside Attenuates High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease via AMPK-Dependent TXNIP/NLRP3 Pathway. Oxid Med Cell Longev 2018; 2018: 8597897
  • 129 Li H, Ying H, Hu A, Li D, Hu Y. Salidroside Modulates Insulin Signaling in a Rat Model of Nonalcoholic Steatohepatitis. Evid Based Complement Alternat Med 2017; 2017: 9651371
  • 130 Hung J-Y, Hsu Y-L, Ko Y-C, Tsai Y-M, Yang C-J, Huang M-S. Didymin, a dietary flavonoid glycoside from citrus fruits, induces Fas-mediated apoptotic pathway in human non-small-cell lung cancer cells in vitro and in vivo. Lung Cancer 2010; 68: 366-374
  • 131 Yao Q, Lin M-T, Zhu Y-D, Xu H-L, Zhao Y-Z. Recent Trends in Potential Therapeutic Applications of the Dietary Flavonoid Didymin. Molecules 2018; 23
  • 132 Feng Z, Pang L, Chen S, Pang X, Huang Y, Qiao Q. Didymin ameliorates dexamethasone-induced non-alcoholic fatty liver disease by inhibiting TLR4/NF-κB and PI3K/Akt pathways in C57BL/6 J mice. Int Immunopharmacol 2020; 88: 107003
  • 133 Li Y, Li C, Xiong Y, Fang B, Lin X, Huang Q. Didymin Ameliorates Liver Fibrosis by Alleviating Endoplasmic Reticulum Stress and Glycerophospholipid Metabolism: Based on Transcriptomics and Metabolomics. Drug Des Devel Ther 2022; 16: 1713-1729
  • 134 Tanase C, Coşarcă S, Muntean DL. A Critical Review of Phenolic Compounds Extracted from the Bark of Woody Vascular Plants and Their Potential Biological Activity. Molecules 2019; 24: 1182
  • 135 Ganesan K, Xu B. A Critical Review on Polyphenols and Health Benefits of Black Soybeans. Nutrients 2017; 9: 455
  • 136 Muruganandan S, Srinivasan K, Gupta S, Gupta PK, Lal J. Effect of mangiferin on hyperglycemia and atherogenicity in streptozotocin diabetic rats. J Ethnopharmacol 2005; 97: 497-501
  • 137 Du S, Liu H, Lei T, Xie X, Wang H, He X. Mangiferin: An effective therapeutic agent against several disorders (Review). Mol Med Rep 2018; 18: 4775-4786
  • 138 Yong Z, Ruiqi W, Hongji Y, Ning M, Chenzuo J, Yu Z. Mangiferin Ameliorates HFD-Induced NAFLD through Regulation of the AMPK and NLRP3 Inflammasome Signal Pathways. J Immunol Res 2021; 2021: 4084566
  • 139 Wang H, Zhu Y-Y, Wang L, Teng T, Zhou M, Wang S-G. Mangiferin ameliorates fatty liver via modulation of autophagy and inflammation in high-fat-diet induced mice. Biomed Pharmacother 2017; 96: 328-335
  • 140 Guo F, Huang C, Liao X, Wang Y, He Y, Feng R. Beneficial effects of mangiferin on hyperlipidemia in high-fat-fed hamsters. Mol Nutr Food Res 2011; 55: 1809-1818
  • 141 Koushki M, Amiri-Dashatan N, Ahmadi N, Abbaszadeh HA, Rezaei-Tavirani M. Resveratrol: A miraculous natural compound for diseases treatment. Food Sci Nutr 2018; 6: 2473-2490
  • 142 Tang LY, Chen Y, Rui BB, Hu CM. Resveratrol ameliorates lipid accumulation in HepG2 cells, associated with down-regulation of lipin1 expression. Can J Physiol Pharmacol 2016; 94: 185-189
  • 143 Du F, Huang R, Lin D, Wang Y, Yang X, Huang X. Resveratrol Improves Liver Steatosis and Insulin Resistance in Non-alcoholic Fatty Liver Disease in Association With the Gut Microbiota. Front Microbiol 2021; 12
  • 144 Ran G, Ying L, Li L, Yan Q, Yi W, Ying C. Resveratrol ameliorates diet-induced dysregulation of lipid metabolism in zebrafish (Danio rerio). PLOS ONE 2017; 12: e0180865
  • 145 Booker A, Frommenwiler D, Johnston D, Umealajekwu C, Reich E, Heinrich M. Chemical variability along the value chains of turmeric (Curcuma longa): A comparison of nuclear magnetic resonance spectroscopy and high performance thin layer chromatography. J Ethnopharmacol 2014; 152: 292-301
  • 146 Ghosh S, Banerjee S, Sil PC. The beneficial role of curcumin on inflammation, diabetes and neurodegenerative disease: A recent update. Food Chem Toxicol 2015; 83: 111-124
  • 147 Lee DE, Lee SJ, Kim SJ, Lee HS, Kwon OS. Curcumin Ameliorates Nonalcoholic Fatty Liver Disease through Inhibition of O-GlcNAcylation. Nutrients 2019; 11
  • 148 Feng W, Kuang S, Tu C, Ma Z, Pang J, Wang Y. Natural products berberine and curcumin exhibited better ameliorative effects on rats with non-alcohol fatty liver disease than lovastatin. Biomed Pharmacother 2018; 99: 325-333
  • 149 Kang O, Kim S, Seo Y, Joung D, Mun S, Choi J. Curcumin decreases oleic acid-induced lipid accumulation via AMPK phosphorylation in hepatocarcinoma cells. Eur Rev Med Pharmacol Sci 2013; 17: 2578-2586
  • 150 Saadati S, Sadeghi A, Mansour A, Yari Z, Poustchi H, Hedayati M. Curcumin and inflammation in non-alcoholic fatty liver disease: a randomized, placebo controlled clinical trial. BMC Gastroenterol 2019; 19: 133
  • 151 Xiao J, Ho CT, Liong EC, Nanji AA, Leung TM, Lau TYH. Epigallocatechin gallate attenuates fibrosis, oxidative stress, and inflammation in non-alcoholic fatty liver disease rat model through TGF/SMAD, PI3 K/Akt/FoxO1, and NF-kappa B pathways. Eur J Nutr 2014; 53: 187-199
  • 152 Kim JJY, Tan Y, Xiao L, Sun YL, Qu X. Green Tea Polyphenol Epigallocatechin-3-Gallate Enhance Glycogen Synthesis and Inhibit Lipogenesis in Hepatocytes. Biomed Res Int 2013; 2013: 920128
  • 153 Huang J, Feng S, Liu A, Dai Z, Wang H, Reuhl K. Green Tea Polyphenol EGCG Alleviates Metabolic Abnormality and Fatty Liver by Decreasing Bile Acid and Lipid Absorption in Mice. Mol Nutr Food Res 2018; 62: 1700696
  • 154 Vilaplana-Pérez C, Auñón D, García-Flores LA, Gil-Izquierdo A. Hydroxytyrosol and Potential Uses in Cardiovascular Diseases, Cancer, and AIDS. Front Nutr 2014; 1
  • 155 Pirozzi C, Lama A, Simeoli R, Paciello O, Pagano TB, Mollica MP. Hydroxytyrosol prevents metabolic impairment reducing hepatic inflammation and restoring duodenal integrity in a rat model of NAFLD. J Nutr Biochem 2016; 30: 108-115
  • 156 Valenzuela R, Illesca P, Echeverría F, Espinosa A, Rincón-Cervera MÁ, Ortiz M. Molecular adaptations underlying the beneficial effects of hydroxytyrosol in the pathogenic alterations induced by a high-fat diet in mouse liver: PPAR-α and Nrf2 activation, and NF-κB down-regulation. Food Funct 2017; 8: 1526-1537
  • 157 Dong Y, Yu M, Wu Y, Xia T, Wang L, Song K. Hydroxytyrosol Promotes the Mitochondrial Function through Activating Mitophagy. Antioxidants 2022; 11
  • 158 Hitl M, Kladar N, Gavarić N, Bozin B. Rosmarinic Acid–Human Pharmacokinetics and Health Benefits. Planta Med 2020; 87: 273-282
  • 159 Balachander GJ, Subramanian S, Ilango K. Rosmarinic acid attenuates hepatic steatosis by modulating ER stress and autophagy in oleic acid-induced HepG2 cells. RSC Adv 2018; 8: 26656-26663
  • 160 Luo C, Sun H, Peng J, Gao C, Bao L, Ji R. Rosmarinic acid exerts an antagonistic effect on nonalcoholic fatty liver disease by regulating the YAP1/TAZ-PPARγ/PGC-1α signaling pathway. Phytother Res 2021; 35: 1010-1022
  • 161 AlTamimi JZ, Alshammari GM, AlFaris NA, Alagal RI, Aljabryn DH, Albekairi NA, Alkhateeb MA, Yahya MA. Ellagic acid protects against non-alcoholic fatty liver disease in streptozotocin-diabetic rats by activating AMPK. Pharm Biol 2022; 60: 25-37
  • 162 Zhang C, Hu J, Sheng L, Yuan M, Wu Y, Chen L. Ellagic acid ameliorates AKT-driven hepatic steatosis in mice by suppressing de novo lipogenesis via the AKT/SREBP-1/FASN pathway. Food Funct 2019; 10: 3410-3420
  • 163 Polce SA, Burke C, França LM, Kramer B, de Andrade Paes AM, Carrillo-Sepulveda MA. Ellagic Acid Alleviates Hepatic Oxidative Stress and Insulin Resistance in Diabetic Female Rats. Nutrients 2018; 10
  • 164 Goto T, Takahashi N, Hirai S, Kawada T. Various Terpenoids Derived from Herbal and Dietary Plants Function as PPAR Modulators and Regulate Carbohydrate and Lipid Metabolism. PPAR Res 2010; 2010: 483958
  • 165 Cox-Georgian D, Ramadoss N, Dona C, Basu C. Therapeutic and Medicinal Uses of Terpenes. In: Joshee N, Dhekney SA, Parajuli P. editors Medicinal Plants: From Farm to Pharmacy. Cham: Springer International Publishing; 2019: 333-359
  • 166 Kazmi I, Rahman M, Afzal M, Gupta G, Saleem S, Afzal O. Anti-diabetic potential of ursolic acid stearoyl glucoside: A new triterpenic gycosidic ester from Lantana camara. Fitoterapia 2012; 83: 142-146
  • 167 Peng F, Zhang H, He X, Song Z. Effects of Ursolic Acid on Intestinal Health and Gut Bacteria Antibiotic Resistance in Mice. Front Physiol 2021; 12
  • 168 Li J-S, Wang W-J, Sun Y, Zhang Y-H, Zheng L. Ursolic acid inhibits the development of nonalcoholic fatty liver disease by attenuating endoplasmic reticulum stress. Food Funct 2015; 6: 1643-1651
  • 169 Li S, Meng F, Liao X, Wang Y, Sun Z, Guo F. Therapeutic Role of Ursolic Acid on Ameliorating Hepatic Steatosis and Improving Metabolic Disorders in High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease Rats. PLOS ONE 2014; 9: e86724
  • 170 Lin Y-N, Wang CCN, Chang H-Y, Chu F-Y, Hsu Y-A, Cheng W-K. Ursolic Acid, a Novel Liver X Receptor α (LXRα) Antagonist Inhibiting Ligand-Induced Nonalcoholic Fatty Liver and Drug-Induced Lipogenesis. J Agric Food Chem 2018; 66: 11647-11662
  • 171 Ayeleso TB, Matumba MG, Mukwevho E. Oleanolic Acid and Its Derivatives: Biological Activities and Therapeutic Potential in Chronic Diseases. Molecules 2017; 22
  • 172 Somova LI, Shode FO, Ramnanan P, Nadar A. Antihypertensive, antiatherosclerotic and antioxidant activity of triterpenoids isolated from Olea europaea, subspecies africana leaves. J Ethnopharmacol 2003; 84: 299-305
  • 173 Nyakudya TT, Mukwevho E, Nkomozepi P, Erlwanger KH. Neonatal intake of oleanolic acid attenuates the subsequent development of high fructose diet-induced non-alcoholic fatty liver disease in rats. J Dev Orig Health Dis 2018; 9: 500-510
  • 174 Gamede M, Mabuza-Mashaba L, Ngubane P, Khathi A. Plant-derived oleanolic acid ameliorates markers associated with non-alcoholic fatty liver disease in a diet-induced pre-diabetes rat model. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy. 2019;Volume 12; 1953.  –  1962
  • 175 Lin Y-N, Chang H-Y, Wang CCN, Chu F-Y, Shen H-Y, Chen C-J. Oleanolic Acid Inhibits Liver X Receptor Alpha and Pregnane X Receptor to Attenuate Ligand-Induced Lipogenesis. J Agric Food Chem 2018; 66: 10964-10976
  • 176 Matumba MG, Ayeleso AO, Nyakudya T, Erlwanger K, Chegou NN, Mukwevho E. Long-Term Impact of Neonatal Intake of Oleanolic Acid on the Expression of AMP-Activated Protein Kinase, Adiponectin and Inflammatory Cytokines in Rats Fed with a High Fructose Diet. Nutrients 2019; 11
  • 177 Fang Z, Zhang M, Liu J, Zhao X, Zhang Y, Fang L. Tanshinone IIA: A Review of its Anticancer Effects. Front Pharmacol 2021; 11
  • 178 Maione F, De Feo V, Caiazzo E, De Martino L, Cicala C, Mascolo N. Tanshinone IIA, a major component of Salvia milthorriza Bunge, inhibits platelet activation via Erk-2 signaling pathway. J Ethnopharmacol 2014; 155: 1236-1242
  • 179 Huang L, Ding W, Wang M-Q, Wang Z-G, Chen H-H, Chen W. Tanshinone IIA ameliorates non-alcoholic fatty liver disease through targeting peroxisome proliferator-activated receptor gamma and toll-like receptor 4. J Int Med Res 2019; 47: 5239-5255
  • 180 Xu L, Liu X, Jia T, Sun Y, Du Y, Wei S. Tanshinone IIA Ameliorates Nonalcoholic Steatohepatitis in Mice by Modulating Neutrophil Extracellular Traps and Hepatocyte Apoptosis. Evid Based Complement Alternat Med 2022; 2022: 5769350
  • 181 Yang G, Jia L, Wu J, Ma Y, Cao H, Song N. Effect of tanshinone IIA on oxidative stress and apoptosis in a rat model of fatty liver. Exp Ther Med 2017; 14: 4639-4646
  • 182 Li L, Liu Z, Jiang H, Mao X. Biotechnological production of lycopene by microorganisms. Appl Microbiol Biotechnol 2020; 104: 10307-10324
  • 183 Róvero Costa M, Leite Garcia J, Cristina Vágula de Almeida Silva C. Junio Togneri Ferron A, Valentini Francisqueti-Ferron F, Kurokawa Hasimoto F. Lycopene Modulates Pathophysiological Processes of Non-Alcoholic Fatty Liver Disease in Obese Rats. Antioxidants 2019; 8
  • 184 Jiang W, Guo M-H, Hai X. Hepatoprotective and antioxidant effects of lycopene on non-alcoholic fatty liver disease in rat. World J Gastroenterol 2016; 22: 10180
  • 185 Wang J, Geng T, Zou Q, Yang N, Zhao W, Li Y. Lycopene prevents lipid accumulation in hepatocytes by stimulating PPARα and improving mitochondrial function. J Funct Foods 2020; 67: 103857
  • 186 Peng J, Yuan J-P, Wu C-F, Wang J-H. Fucoxanthin, a Marine Carotenoid Present in Brown Seaweeds and Diatoms: Metabolism and Bioactivities Relevant to Human Health. Mar Drugs 2011; 9: 1806-1828
  • 187 Takatani N, Kono Y, Beppu F, Okamatsu-Ogura Y, Yamano Y, Miyashita K. Fucoxanthin inhibits hepatic oxidative stress, inflammation, and fibrosis in diet-induced nonalcoholic steatohepatitis model mice. Biochem Biophys Res Commun 2020; 528: 305-310
  • 188 Ye J, Zheng J, Tian X, Xu B, Yuan F, Wang B. Fucoxanthin Attenuates Free Fatty Acid-Induced Nonalcoholic Fatty Liver Disease by Regulating Lipid Metabolism/Oxidative Stress/Inflammation via the AMPK/Nrf2/TLR4 Signaling Pathway. Mar Drugs 2022; 20
  • 189 Shrikanth CB, Nandini CD. AMPK in microvascular complications of diabetes and the beneficial effects of AMPK activators from plants. Phytomedicine 2020; 73: 152808
  • 190 Mu Q, Wang H, Tong L, Fang Q, Xiang M, Han L. Betulinic acid improves nonalcoholic fatty liver disease through YY1/FAS signaling pathway. FASEB J 2020; 34: 13033-13048
  • 191 Quan HY, Kim DY, Kim SJ, Jo HK, Kim GW, Chung SH. Betulinic acid alleviates non-alcoholic fatty liver by inhibiting SREBP1 activity via the AMPK–mTOR–SREBP signaling pathway. Biochem Pharmacol 2013; 85: 1330-1340
  • 192 Gu M, Zhao P, Zhang S, Fan S, Yang L, Tong Q. Betulinic acid alleviates endoplasmic reticulum stress-mediated nonalcoholic fatty liver disease through activation of farnesoid X receptors in mice. Br J Pharmacol 2019; 176: 847-863
  • 193 Smeriglio A, Barreca D, Bellocco E, Trombetta D. Proanthocyanidins and hydrolysable tannins: occurrence, dietary intake and pharmacological effects. Br J Pharmacol 2017; 174: 1244-1262
  • 194 Liman MS, Hassen A, McGaw LJ, Sutovsky P, Holm DE. Potential Use of Tannin Extracts as Additives in Semen Destined for Cryopreservation: A Review. Animals 2022; 12
  • 195 Baldwin A, Booth BW. Biomedical applications of tannic acid. J Biomater Appl 2022; 36: 1503-1523
  • 196 Chung M-Y, Song J-H, Lee J, Shin EJ, Park JH, Lee S-H. Tannic acid, a novel histone acetyltransferase inhibitor, prevents non-alcoholic fatty liver disease both in vivo and in vitro model. Mol Metab 2019; 19: 34-48
  • 197 Li M, Liu P, Xue Y, Liang Y, Shi J, Han X. Tannic acid attenuates hepatic oxidative stress, apoptosis and inflammation by activating the Keap1-Nrf2/ARE signaling pathway in arsenic trioxide-toxicated rats. Oncol Rep 2020; 44: 2306-2316
  • 198 Yoon M, Kim J-S, Seo S, Lee K, Um MY, Lee J. Dieckol, a Major Marine Polyphenol, Enhances Non-Rapid Eye Movement Sleep in Mice via the GABAA-Benzodiazepine Receptor. Front Pharmacol 2020; 11
  • 199 Byun K-A, Oh S, Son M, Park C-H, Son K, Byun K. Dieckol Decreases Caloric Intake and Attenuates Nonalcoholic Fatty Liver Disease and Hepatic Lymphatic Vessel Dysfunction in High-Fat-Diet-Fed Mice. Mar Drugs 2021; 19: 495
  • 200 Eo H, Jeon Y, Lee M, Lim Y. Brown Alga Ecklonia cava Polyphenol Extract Ameliorates Hepatic Lipogenesis, Oxidative Stress, and Inflammation by Activation of AMPK and SIRT1 in High-Fat Diet-Induced Obese Mice. J Agric Food Chem 2015; 63: 349-359
  • 201 Oh S, Son M, Byun K-A, Jang JT, Choi CH, Son KH. Attenuating Effects of Dieckol on High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease by Decreasing the NLRP3 Inflammasome and Pyroptosis. Mar Drugs 2021; 19
  • 202 DellʼAgli M, Galli GV, Corbett Y, Taramelli D, Lucantoni L, Habluetzel A. Antiplasmodial activity of Punica granatum L. fruit rind. J Ethnopharmacol 2009; 125: 279-285
  • 203 Feng L, Yin Y, Yang X, Tang H, Jiao Q. Dynamic Variations in Punicalagin and Related Metabolic Substances in Pomegranate Fruit and Leaves During Development Periods. Horticult J 2019; 88: 444-454
  • 204 Yan C, Sun W, Wang X, Long J, Liu X, Feng Z. Punicalagin attenuates palmitate-induced lipotoxicity in HepG2 cells by activating the Keap1-Nrf2 antioxidant defense system. Mol Nutr Food Res 2016; 60: 1139-1149
  • 205 Liu H, Zhan Q, Miao X, Xia X, Yang G, Peng X. Punicalagin Prevents Hepatic Steatosis through Improving Lipid Homeostasis and Inflammation in Liver and Adipose Tissue and Modulating Gut Microbiota in Western Diet-Fed Mice. Mol Nutr Food Res 2021; 65: 2001031
  • 206 Zou X, Yan C, Shi Y, Cao K, Xu J, Wang X. Mitochondrial Dysfunction in Obesity-Associated Nonalcoholic Fatty Liver Disease: The Protective Effects of Pomegranate with Its Active Component Punicalagin. Antioxid Redox Signal 2014; 21: 1557-1570
  • 207 Li X, Deng Y, Zheng Z, Huang W, Chen L, Tong Q. Corilagin, a promising medicinal herbal agent. Biomed Pharmacother 2018; 99: 43-50
  • 208 Zhang R, Chu K, Zhao N, Wu J, Ma L, Zhu C. Corilagin Alleviates Nonalcoholic Fatty Liver Disease in High-Fat Diet-Induced C57BL/6 Mice by Ameliorating Oxidative Stress and Restoring Autophagic Flux. Front Pharmacol 2020; 10
  • 209 Liao M, Zhang R, Wang Y, Mao Z, Wu J, Guo H. Corilagin prevents non-alcoholic fatty liver disease via improving lipid metabolism and glucose homeostasis in high fat diet-fed mice. Front Nutr 2022; 9
  • 210 Liang L, Ye S, Jiang R, Zhou X, Zhou J, Meng S. Liensinine alleviates high fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) through suppressing oxidative stress and inflammation via regulating TAK1/AMPK signaling. Int Immunopharmacol 2022; 104: 108306
  • 211 Wang G-E, Li Y-F, Zhai Y-J, Gong L, Tian J-Y, Hong M. Theacrine protects against nonalcoholic fatty liver disease by regulating acylcarnitine metabolism. Metabolism 2018; 85: 227-239
  • 212 Li G, Zhou F, Chen Y, Zhang W, Wang N. Kukoamine A attenuates insulin resistance and fatty liver through downregulation of Srebp-1c. Biomed Pharmacother 2017; 89: 536-543
  • 213 Ohashi T, Nakade Y, Ibusuki M, Kitano R, Yamauchi T, Kimoto S. Conophylline inhibits high fat diet-induced non-alcoholic fatty liver disease in mice. PLOS ONE 2019; 14: e0210068
  • 214 Sylvester Darvin S, Toppo E, Esakkimuthu S, Ajeesh Krishna TP, Ceasar SA, Stalin A. Hepatoprotective effect of bisbenzylisoquinoline alkaloid tiliamosine from Tiliacora racemosa in high-fat diet/diethylnitrosamine-induced non-alcoholic steatohepatitis. Biomed Pharmacother 2018; 108: 963-973
  • 215 Yue R, Jin G, Wei S, Huang H, Su L, Zhang C. Immunoregulatory Effect of Koumine on Nonalcoholic Fatty Liver Disease Rats. J Immunol Res 2019; 2019: 8325102
  • 216 Wang LL, Zhang ZC, Hassan W, Li Y, Liu J, Shang J. Amelioration of free fatty acid-induced fatty liver by quercetin-3-O-β-D-glucuronide through modulation of peroxisome proliferator-activated receptor-alpha/sterol regulatory element-binding protein-1c signaling. Hepatol Res 2016; 46: 225-238
  • 217 Park J-Y, Lim M-S, Kim S-I, Lee H, Kim S-S, Kwon Y-S. Quercetin-3-O-β-D-Glucuronide Suppresses Lipopolysaccharide-Induced JNK and ERK Phosphorylation in LPS-Challenged RAW264.7 Cells. Biomol Ther 2016; 24
  • 218 Hur HJ, Jeong Y-H, Lee SH, Sung MJ. Quercitrin Ameliorates Hyperlipidemia and Hepatic Steatosis in Ovariectomized Mice. Life 2020; 10
  • 219 PubChem. Quercitrin. https://pubchem.ncbi.nlm.nih.gov/compound/5280459 Accessed 29 Oct 2022
  • 220 Kim JH, Lee BC, Kim JH, Sim GS, Lee DH, Lee KE. The isolation and antioxidative effects of vitexin fromAcer palmatum. Arch Pharm Res 2005; 28: 195-202
  • 221 Inamdar S, Joshi A, Malik S, Boppana R, Ghaskadbi S. Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice. Biochem Biophys Res Commun 2019; 519: 106-112
  • 222 Sarkar MK, Kar A, Jayaraman A, Kar Mahapatra S, Vadivel V. Vitexin isolated from Prosopis cineraria leaves induce apoptosis in K-562 leukemia cells via inhibition of the BCR-ABL-Ras-Raf pathway. J Pharm Pharmacol 2022; 74: 103-111
  • 223 Morissette M, Litim N, Paolo T. Natural Phytoestrogens: A Class of Promising Neuroprotective Agents for Parkinson Disease. In: Discovery and Development of Neuroprotective Agents from Natural Products: Natural Product Drug Discovery 2017: 9-61
  • 224 Pai SA, Munshi RP, Panchal FH, Gaur I-S, Juvekar AR. Chrysin ameliorates nonalcoholic fatty liver disease in rats. Naunyn Schmiedebergs Arch Pharmacol 2019; 392: 1617-1628
  • 225 Song Y, Wu W, Sheng L, Jiang B, Li X, Cai K. Chrysin ameliorates hepatic steatosis induced by a diet deficient in methionine and choline by inducing the secretion of hepatocyte nuclear factor 4α-dependent very low-density lipoprotein. J Biochem Mol Toxicol 2020; 34: e22497
  • 226 Aloud AA, Chinnadurai V, Govindasamy C, Alsaif MA, Al-Numair KS. Galangin, a dietary flavonoid, ameliorates hyperglycaemia and lipid abnormalities in rats with streptozotocin-induced hyperglycaemia. Pharm Biol 2018; 56: 302-308
  • 227 Zhang X, Deng Y, Xiang J, Liu H, Zhang J, Liao J. Galangin Improved Non-Alcoholic Fatty Liver Disease in Mice by Promoting Autophagy. Drug Des Devel Ther 2020; 14: 3393-3405
  • 228 Hiramitsu M, Shimada Y, Kuroyanagi J, Inoue T, Katagiri T, Zang L. Eriocitrin ameliorates diet-induced hepatic steatosis with activation of mitochondrial biogenesis. Sci Rep 2014; 4: 3708
  • 229 Kwon E-Y, Choi M-S. Eriocitrin Improves Adiposity and Related Metabolic Disorders in High-Fat Diet-Induced Obese Mice. J Med Food 2020; 23: 233-241
  • 230 Karimi G, Vahabzadeh M, Lari P, Rashedinia M, Moshiri M. “Silymarin”, a Promising Pharmacological Agent for Treatment of Diseases. Iran J Basic Med Sci 2011; 14: 308-317
  • 231 Salomone F, Barbagallo I, Godos J, Lembo V, Currenti W, Cinà D. Silibinin Restores NAD+ Levels and Induces the SIRT1/AMPK Pathway in Non-Alcoholic Fatty Liver. Nutrients 2017; 9
  • 232 Sahin E, Bagci R, Bektur Aykanat NE, Kacar S, Sahinturk V. Silymarin attenuated nonalcoholic fatty liver disease through the regulation of endoplasmic reticulum stress proteins GRP78 and XBP-1 in mice. J Food Biochem 2020; 44: e13194
  • 233 Geng Y, Wu Z, Buist-Homan M, Blokzijl H, Moshage H. Hesperetin protects against palmitate-induced cellular toxicity via induction of GRP78 in hepatocytes. Toxicol Appl Pharmacol 2020; 404: 115183
  • 234 Li J, Wang T, Liu P, Yang F, Wang X, Zheng W. Hesperetin ameliorates hepatic oxidative stress and inflammation via the PI3K/AKT-Nrf2-ARE pathway in oleic acid-induced HepG2 cells and a rat model of high-fat diet-induced NAFLD. Food Funct 2021; 12: 3898-3918
  • 235 Wdowiak K, Walkowiak J, Pietrzak R, Bazan-Woźniak A, Cielecka-Piontek J. Bioavailability of Hesperidin and Its Aglycone Hesperetin–Compounds Found in Citrus Fruits as a Parameter Conditioning the Pro-Health Potential (Neuroprotective and Antidiabetic Activity)–Mini-Review. Nutrients 2022; 14
  • 236 Stanisic D, Liu LHB, dos Santos RV, Costa AF, Durán N, Tasic L. New Sustainable Process for Hesperidin Isolation and Anti-Ageing Effects of Hesperidin Nanocrystals. Molecules 2020; 25
  • 237 Chen H, Nie T, Zhang P, Ma J, Shan A. Hesperidin attenuates hepatic lipid accumulation in mice fed high-fat diet and oleic acid induced HepG2 via AMPK activation. Life Sci 2022; 296: 120428
  • 238 Zarmouh NO, Mazzio EA, Elshami FM, Messeha SS, Eyunni SVK, Soliman KFA. Evaluation of the Inhibitory Effects of Bavachinin and Bavachin on Human Monoamine Oxidases A and B. Evid Based Complement Alternat Med 2015; 2015: 852194
  • 239 Dong X, Zhu Y, Wang S, Luo Y, Lu S, Nan F. Bavachinin inhibits cholesterol synthesis enzyme FDFT1 expression via AKT/mTOR/SREBP-2 pathway. Int Immunopharmacol 2020; 88: 106865
  • 240 Ali MY, Zaib S, Rahman MM, Jannat S, Iqbal J, Park SK. Didymin, a dietary citrus flavonoid exhibits anti-diabetic complications and promotes glucose uptake through the activation of PI3K/Akt signaling pathway in insulin-resistant HepG2 cells. Chem Biol Interact 2019; 305: 180-194
  • 241 Chang T-C, Chiou W-C, Lai W-H, Huang H-C, Huang Y-L, Liu H-K. Ugonin J improves metabolic disorder and ameliorates nonalcoholic fatty liver disease by regulating the AMPK/AKT signaling pathway. Pharmacol Res 2021; 163: 105298
  • 242 Sun B, Zhang R, Liang Z, Fan A, Kang D. Hyperoside attenuates non-alcoholic fatty liver disease through targeting Nr 4A1 in macrophages. Int Immunopharmacol 2021; 94: 107438
  • 243 Wang S, Sheng F, Zou L, Xiao J, Li P. Hyperoside attenuates non-alcoholic fatty liver disease in rats via cholesterol metabolism and bile acid metabolism. J Adv Res 2021; 34: 109-122
  • 244 Lin W, Jin Y, Hu X, Huang E, Zhu Q. AMPK/PGC-1α/GLUT4-Mediated Effect of Icariin on Hyperlipidemia-Induced Non-Alcoholic Fatty Liver Disease and Lipid Metabolism Disorder in Mice. Biochemistry (Mosc) 2021; 86: 1407-1417
  • 245 Dietz BM, Kang Y-H, Liu G, Eggler AL, Yao P, Chadwick LR. Xanthohumol Isolated from Humulus lupulus Inhibits Menadione-Induced DNA Damage through Induction of Quinone Reductase. Chem Res Toxicol 2005; 18: 1296-1305
  • 246 Zhang Y, Bobe G, Miranda CL, Lowry MB, Hsu VL, Lohr CV. Tetrahydroxanthohumol, a xanthohumol derivative, attenuates high-fat diet-induced hepatic steatosis by antagonizing PPARγ . eLife 2021; 10: e66398
  • 247 Wang W, Chen Z, Zheng T, Zhang M. Xanthohumol alleviates T2DM-induced liver steatosis and fibrosis by mediating the NRF2/RAGE/NF-κB signaling pathway. Future Med Chem 2021; 13: 2069-2081
  • 248 Gao J, Chen G, He H, Liu C, Xiong X, Li J. Therapeutic Effects of Breviscapine in Cardiovascular Diseases: A Review. Front Pharmacol 2017; 8
  • 249 Lan T, Jiang S, Zhang J, Weng Q, Yu Y, Li H. Breviscapine alleviates NASH by inhibiting TGF-β-activated kinase 1-dependent signaling. Hepatology 2022; 76: 155-171
  • 250 Xia S-F, Le G-W, Wang P, Qiu Y-Y, Jiang Y-Y, Tang X. Regressive Effect of Myricetin on Hepatic Steatosis in Mice Fed a High-Fat Diet. Nutrients 2016; 8
  • 251 de Lima M de FR, Cavalcante LA, Costa ECT de A, Veras B, da Silva MV, Cavalcanti LN, Araújo RM. Bioactivity flavonoids from roots of Euphorbia tirucalli L. Phytochem Lett 2021; 41: 186-192
  • 252 Choi H-N, Shin J-Y, Kim J-I. Ameliorative Effect of Myricetin on Nonalcoholic Fatty Liver Disease in ob/ob Mice. J Med Food 2021; 24: 1092-1099
  • 253 Fu Y, Liu B, Liu J, Liu Z, Liang D, Li F. Geniposide, from Gardenia jasminoides Ellis, inhibits the inflammatory response in the primary mouse macrophages and mouse models. Int Immunopharmacol 2012; 14: 792-798
  • 254 Shen B, Feng H, Cheng J, Li Z, Jin M, Zhao L. Geniposide alleviates non-alcohol fatty liver disease via regulating Nrf2/AMPK/mTOR signalling pathways. J Cell Mol Med 2020; 24: 5097-5108
  • 255 Cominguez DC, Park Y-J, Kang Y-M, Nugroho A, Kim S, An H-J. Clitorin ameliorates western diet-induced hepatic steatosis by regulating lipogenesis and fatty acid oxidation in vivo and in vitro. Sci Rep 2022; 12: 4154
  • 256 Jin M, Feng H, Wang Y, Yan S, Shen B, Li Z. Gentiopicroside Ameliorates Oxidative Stress and Lipid Accumulation through Nuclear Factor Erythroid 2-Related Factor 2 Activation. Oxid Med Cell Longev 2020; 2020: 2940746
  • 257 Cheng S, Liang S, Liu Q, Deng Z, Zhang Y, Du J. Diosgenin prevents high-fat diet-induced rat non-alcoholic fatty liver disease through the AMPK and LXR signaling pathways. Int J Mol Med 2018; 41: 1089-1095
  • 258 Liu M, Zhang G, Wu S, Song M, Wang J, Cai W. Schaftoside alleviates HFD-induced hepatic lipid accumulation in mice via upregulating farnesoid X receptor. J Ethnopharmacol 2020; 255: 112776
  • 259 Yu Q, Liu Y, Wu Y, Chen Y. Dihydrocurcumin ameliorates the lipid accumulation, oxidative stress and insulin resistance in oleic acid-induced L02 and HepG2 cells. Biomed Pharmacother 2018; 103: 1327-1336
  • 260 Kim M, Yoo G, Randy A, Kim HS, Nho CW. Chicoric acid attenuate a nonalcoholic steatohepatitis by inhibiting key regulators of lipid metabolism, fibrosis, oxidation, and inflammation in mice with methionine and choline deficiency. Mol Nutr Food Res 2017; 61: 1600632
  • 261 Ma J, Li M, Kalavagunta PK, Li J, He Q, Zhang Y. Protective effects of cichoric acid on H2O2-induced oxidative injury in hepatocytes and larval zebrafish models. Biomed Pharmacother 2018; 104: 679-685
  • 262 Ding X, Jian T, Li J, Lv H, Tong B, Li J. Chicoric Acid Ameliorates Nonalcoholic Fatty Liver Disease via the AMPK/Nrf2/NFκB Signaling Pathway and Restores Gut Microbiota in High-Fat-Diet-Fed Mice. Oxid Med Cell Longev 2020; 2020: 9734560
  • 263 Minh TN, Xuan TD, Tran H-D, Van TM, Andriana Y, Khanh TD. Isolation and Purification of Bioactive Compounds from the Stem Bark of Jatropha podagrica. Molecules 2019; 24
  • 264 Fanaei H, Mard SA, Sarkaki A, Goudarzi G, Khorsandi L. Gallic acid protects the liver against NAFLD induced by dust exposure and high-fat diet through inhibiting oxidative stress and repressing the inflammatory signaling pathways NF-κβ/TNF-α/IL-6 in Wistar rats. Avicenna J Phytomed 2021; 11: 527-540
  • 265 Ismail BS, Abdel-Reheim ES, Soliman HA, Mahmoud B. Protective Effect of Gallic Acid against Nonalcoholic Fatty Liver Disease Induced by High Fat Diet. J Pharm Res Int 2021; 378-397
  • 266 Cha S-H, Hwang Y, Heo S-J, Jun H-S. Diphlorethohydroxycarmalol Attenuates Palmitate-Induced Hepatic Lipogenesis and Inflammation. Mar Drugs 2020; 18
  • 267 Vazquez Prieto MA, Bettaieb A, Rodriguez Lanzi C, Soto VC, Perdicaro DJ, Galmarini CR. Catechin and quercetin attenuate adipose inflammation in fructose-fed rats and 3 T3-L1 adipocytes. Mol Nutr Food Res 2015; 59: 622-633
  • 268 Hodges JK, Sasaki GY, Bruno RS. Anti-inflammatory activities of green tea catechins along the gut–liver axis in nonalcoholic fatty liver disease: lessons learned from preclinical and human studies. J Nutr Biochem 2020; 85: 108478
  • 269 Suárez-Quiroz ML, Campos AA, Alfaro GV, González-Ríos O, Villeneuve P, Figueroa-Espinoza MC. Isolation of green coffee chlorogenic acids using activated carbon. J Food Compost Anal 2014; 33: 55-58
  • 270 Jin S, Chang C, Zhang L, Liu Y, Huang X, Chen Z. Chlorogenic Acid Improves Late Diabetes through Adiponectin Receptor Signaling Pathways in db/db Mice. PLOS ONE 2015; 10: e0120842
  • 271 Wang Z, Lam K-L, Hu J, Ge S, Zhou A, Zheng B. Chlorogenic acid alleviates obesity and modulates gut microbiota in high-fat-fed mice. Food Sci Nutr 2019; 7: 579-588
  • 272 Ding C, Zhao Y, Shi X, Zhang N, Zu G, Li Z. New insights into salvianolic acid A action: Regulation of the TXNIP/NLRP3 and TXNIP/ChREBP pathways ameliorates HFD-induced NAFLD in rats. Sci Rep 2016; 6: 28734
  • 273 Li S, Qian Q, Ying N, Lai J, Feng L, Zheng S. Activation of the AMPK-SIRT1 pathway contributes to protective effects of Salvianolic acid A against lipotoxicity in hepatocytes and NAFLD in mice. Front Pharmacol 2020; 11
  • 274 Li J, Wang S, Yao L, Ma P, Chen Z, Han T-L. 6-gingerol ameliorates age-related hepatic steatosis: Association with regulating lipogenesis, fatty acid oxidation, oxidative stress and mitochondrial dysfunction. Toxicol Appl Pharmacol 2019; 362: 125-135
  • 275 Sarrafan A, Ghobeh M, Yaghmaei P. The effect of 6-gingerol on biochemical and histological parameters in cholesterol-induced nonalcoholic fatty liver disease in NMRI mice. Braz J Pharm Sci 2021; 57: e18020
  • 276 Chen F, Yang L, Zhai L, Huang Y, Chen F, Duan W. Methyl brevifolincarboxylate, a novel influenza virus PB2 inhibitor from Canarium Album (Lour.) Raeusch. Chem Biol Drug Des 2020; 96: 1280-1291
  • 277 Geethangili M, Lin C-W, Mersmann HJ, Ding S-T. Methyl Brevifolincarboxylate Attenuates Free Fatty Acid-Induced Lipid Metabolism and Inflammation in Hepatocytes through AMPK/NF-κB Signaling Pathway. Int J Mol Sci 2021; 22
  • 278 Li H-B, Jiang Y, Chen F. Isolation and Purification of Lutein from the Microalga Chlorella vulgaris by Extraction after Saponification. J Agric Food Chem 2002; 50: 1070-1072
  • 279 Kim JE, Clark RM, Park Y, Lee J, Fernandez ML. Lutein decreases oxidative stress and inflammation in liver and eyes of guinea pigs fed a hypercholesterolemic diet. Nutr Res Pract 2012; 6: 113-119
  • 280 Qiu X, Gao D-H, Xiang X, Xiong Y-F, Zhu T-S, Liu L-G. Ameliorative effects of lutein on non-alcoholic fatty liver disease in rats. World J Gastroenterol 2015; 21: 8061-8072
  • 281 Shah MdMR, Liang Y, Cheng JJ, Daroch M. Astaxanthin-Producing Green Microalga Haematococcus pluvialis: From Single Cell to High Value Commercial Products. Front Plant Sci 2016; 7
  • 282 Wu L, Mo W, Feng J, Li J, Yu Q, Li S. et al. Astaxanthin attenuates hepatic damage and mitochondrial dysfunction in non-alcoholic fatty liver disease by up-regulating the FGF21/PGC-1α pathway. Br J Pharmacol 2020; 177: 3760-3777
  • 283 Li Y, Liu J, Ye B, Cui Y, Geng R, Liu S. Astaxanthin Alleviates Nonalcoholic Fatty Liver Disease by Regulating the Intestinal Flora and Targeting the AMPK/Nrf2 Signal Axis. J Agric Food Chem 2022; 70: 10620-10634
  • 284 Kobori M, Ni Y, Takahashi Y, Watanabe N, Sugiura M, Ogawa K. β-Cryptoxanthin Alleviates Diet-Induced Nonalcoholic Steatohepatitis by Suppressing Inflammatory Gene Expression in Mice. PLOS ONE 2014; 9: e98294
  • 285 Lim JY, Liu C, Hu K-Q, Smith DE, Wu D, Lamon-Fava S. Dietary β-Cryptoxanthin Inhibits High-Refined Carbohydrate Diet–Induced Fatty Liver via Differential Protective Mechanisms Depending on Carotenoid Cleavage Enzymes in Male Mice. J Nutr 2019; 149: 1553-1564
  • 286 Islam MT. Andrographolide, a New Hope in the Prevention and Treatment of Metabolic Syndrome. Front Pharmacol 2017; 8: 571
  • 287 Cabrera D, Wree A, Povero D, Solís N, Hernandez A, Pizarro M. Andrographolide Ameliorates Inflammation and Fibrogenesis and Attenuates Inflammasome Activation in Experimental Non-Alcoholic Steatohepatitis. Sci Rep 2017; 7: 3491
  • 288 Ran L-S, Wu Y-Z, Gan Y-W, Wang H-L, Wu L-J, Zheng C-M. Andrographolide ameliorates hepatic steatosis by suppressing FATP2-mediated fatty acid uptake in mice with nonalcoholic fatty liver disease. J Nat Med. 2022
  • 289 Chen H-J, Liu J. Actein ameliorates hepatic steatosis and fibrosis in high fat diet-induced NAFLD by regulation of insulin and leptin resistant. Biomed Pharmacother 2018; 97: 1386-1396
  • 290 Gao X. Efficacy and Safety of Berberine in Non-alcoholic Steatohepatitis: a Multicentre, Randomised, Placebo-controlled Trial. 2020. https://clinicaltrials.gov/ct2/show/NCT03198572 Accessed 21 Aug 2022
  • 291 Mahadeva S. A Randomised, Double-blind, Placebo-controlled, Phase II, Single-centre Study to Assess the Safety and Efficacy of Silymarin 700 mg Capsules TID for the Treatment of Non-Alcoholic Fatty Liver Disease (NAFLD). 2016. https://clinicaltrials.gov/ct2/show/NCT02006498 Accessed 25 Sep 2022
  • 292 Reddy KR, Navarro V, Afdhal N, Fried M. Single and Multiple Dose Escalation Phase I Study to Evaluate the Safety, Tolerability, and Pharmacokinetics of Orally Administered Silymarin (Legalon) in Non-Cirrhotic Subjects With Chronic Hepatitis C or Non-Alcoholic Fatty Liver Disease. 2008. https://clinicaltrials.gov/ct2/show/NCT00389376 Accessed 25 Aug 2022
  • 293 Fried M, Navarro V, Afdhal N, Reddy KR, Belle SH, Harrison SA. A Multicenter, Randomized, Placebo Controlled Study to Assess the Safety and Efficacy of Orally Administered Silymarin Preparation (Legalon) for the Treatment of Non-Cirrhotic Patients With Non-Alcoholic Steatohepatitis. 2019. https://clinicaltrials.gov/ct2/show/NCT00680407 Accessed 11 Sep 2022
  • 294 Hauser G. Silymarin® – Efficacy in Treatment of Non-alcoholic Fatty Liver Disease (NAFLD) Controlled by Laboratory and and Elastographic Parameters. 2020. https://clinicaltrials.gov/ct2/show/NCT02973295 Accessed 18 Sep 2022
  • 295 Wicklow B. Safety and Efficacy of Resveratrol for the Treatment of Non-Alcoholic Fatty Liver Disease and Associated Insulin Resistance in Overweight and Obese Adolescents. 2017. https://clinicaltrials.gov/ct2/show/NCT02216552 Accessed 12 Oct 2022
  • 296 Nielsen S. Long-term Investigation of Resveratrol on Lipid Turnover in Obese Men With Nonalcoholic Fatty Liver Disease. Effects on Liver Fat Content and Basal and Insulin Stimulated FFA and VLDL-triglyceride Metabolism. 2014. https://clinicaltrials.gov/ct2/show/NCT01446276 Accessed 22 Sep 2022
  • 297 Hekmatdoost A. The Effects of Resveratrol Supplement on Lipid Profile, Liver Enzymes, Inflammatory Factors and Hepatic Fibrosis in Patients With Nonalcoholic Steatohepatitis. 2014. https://clinicaltrials.gov/ct2/show/NCT02030977 Accessed 20 Oct 2022
  • 298 Grønbæk H. Long-term Investigation of Resveratrol on Management of Metabolic Syndrome, Osteoporosis and Inflammation, and Identification of Plant Derived Anti-inflammatory Compounds, Study 3. 2015. https://clinicaltrials.gov/ct2/show/NCT01464801 Accessed 14 Oct 2022
  • 299 Jørgensen JOL. Potential Beneficial Effects of Resveratrol on Obesity, Metabolic Syndrome and Inflammation – Emphasis on Description of the Molecular Biology Underpinning the Interplay Between Calorie Restriction, SIRT1, STAT5 and the GH/IGF-I Axis. 2012. https://clinicaltrials.gov/ct2/show/NCT01150955 Accessed 11 Sep 2022
  • 300 Hellmann PH. The Effect of Curcumin on Liver Fat Content in Obese Subjects. 2021. https://clinicaltrials.gov/ct2/show/NCT03864783 Accessed 14 Oct 2022
  • 301 Hekmatdoost A. The Effect of Curcumin Supplement on Metabolic Factors and Hepatic Fibrosis in Nonalcoholic Fatty Liver Patients. 2020. https://clinicaltrials.gov/ct2/show/NCT02908152 Accessed 30 Aug 2022
  • 302 Nobili V. Hydroxytyrosol and Vitamin E in the Treatment of Children With Biopsy-proven NASH. 2018. https://clinicaltrials.gov/ct2/show/NCT02842567 Accessed 27 Sep 2022
  • 303 Ling W. Effects of Purified Anthocyanin on Oxidative and Inflammatory Markers in Subjects With Nonalcoholic Fatty Liver Disease: A Randomized, Double-Blinded, Placebo-Controlled Trial. 2014. https://clinicaltrials.gov/ct2/show/NCT01940263 Accessed 11 Oct 2022

Zoom Image
Fig. 1 Common risk factors associated with NAFLD.
Zoom Image
Fig. 2 Pathophysiology of NAFLD progressing to NASH.
Zoom Image
Fig. 3 Alkaloids involved in the management of NAFLD [berberine (I), betaine (II), nuciferine (III), matrine (IV), oxymatrine (V), piperine (VI), trigonelline (VII), and sophocarpine (VIII)].
Zoom Image
Fig. 4 Flavonoids involved in the management of NAFLD [nobiletin (I), dihydromyricetin (II), rutin (III), naringenin (IV), baicalin (V), quercetin (VI), luteolin (VII), scutellarin (VIII), genistein (IX), and fisetin (X)].
Zoom Image
Fig. 5 Glycosides involved in the management of NAFLD [swertiamarin (I), glycyrrhizic acid (II), salidroside (III), and didymin (IV)].
Zoom Image
Fig. 6 Phenols and polyphenols involved in the management of NAFLD [mangiferin (I), resveratrol (II), curcumin (III), epigallocatechin-3-gallate (IV), hydroxytyrosol (V), rosmarinic acid (VI), and ellagic acid (VII)].
Zoom Image
Fig. 7 Terpenoids involved in the management of NAFLD [ursolic acid (I), oleanolic acid (II), tanshinone IIA (III), lycopene (IV), fucoxanthin (V), and betulinic acid (VI)].
Zoom Image
Fig. 8 Tannins involved in the management of NAFLD [tannic acid (I), dieckol (II), punicalagin (III), and corilagin (IV).