Next Article in Journal
Innovative Multi-Site Photoplethysmography Analysis for Quantifying Pulse Amplitude and Timing Variability Characteristics in Peripheral Arterial Disease
Previous Article in Journal
“Holes” in the Jaw—A Report of Two Cases of Periapical Actinomycosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Therapeutic Benefits of Herbs and Supplements in Patients with NAFLD

1
Drexel University College of Medicine, Philadelphia, PA 19129, USA
2
Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
3
Department of Medicine, Mary Imogene Bassett Hospital, Cooperstown, NY 13326, USA
4
Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA 94305, USA
*
Author to whom correspondence should be addressed.
Diseases 2018, 6(3), 80; https://doi.org/10.3390/diseases6030080
Submission received: 9 August 2018 / Revised: 4 September 2018 / Accepted: 7 September 2018 / Published: 10 September 2018

Abstract

:
Our aim is to review the efficacy of various herbs and supplements as a possible therapeutic option in the treatment and/or prevention of nonalcoholic fatty liver disease (NAFLD). We performed a systematic review of medical literature using the PubMed Database by searching the chemical names of many common herbs and supplements with “AND (NAFLD or NASH)”. Studies and medical literature that discussed the roles and usage of herbs and supplements in NAFLD and nonalcoholic steatohepatitis (NASH) from inception until 20 June 2018 were reviewed. Many studies have claimed that the use of various herbs and supplements may improve disease endpoints and outcomes related to NAFLD and/or NASH. Improvement in liver function tests were noted. Amelioration or reduction of lobular inflammation, hepatic steatosis, and fibrosis were also noted. However, well-designed studies demonstrating improved clinical outcomes are lacking. Furthermore, experts remain concerned about the lack of regulation of herbs/supplements and the need for further research on potential adverse effects and herb–drug interactions. In conclusion, preliminary data on several herbs have demonstrated promising antioxidant, anti-inflammatory, anti-apoptotic, and anti-adipogenic properties that may help curtail the progression of NAFLD/NASH. Clinical trials testing the safety and efficacy must be completed before widespread use can be recommended.

1. Introduction

Nonalcoholic fatty liver disease (NAFLD) and its progressive subset, nonalcoholic steatohepatitis (NASH), affect a significant proportion of the population, with no approved drug treatment. The current treatment option is lifestyle modification through diet and exercise [1,2,3]. In this special issue, the discussion on potential drug targets, oral hypoglycemics, lipid-lowering agents, vitamins/minerals, and surgical options will be reviewed in other sections. The focus of this article is to review the role of herbs and supplements in patients with NASH, the less common but aggressive subset of NAFLD.
Alternative medicine has recently gained interest as a possible approach to disease therapy [4,5]. Herb usage is popular in eastern medicine, especially in countries such as India and China, and it is becoming increasingly popular in the Western hemisphere [4,5]. Many people are looking to herbs and supplements as a natural alternative to drugs. Herbs have many great antioxidant, anti-inflammatory, anti-apoptotic, and anti-adipogenic effects (decreased adipose tissue formation and weight loss) that allow them to be possible therapeutic agents in NAFLD treatment [4,5].

2. Herbs Studied with NAFLD

2.1. Milk Thistle

Silymarin is the extract from the milk thistle plant (Silybum marianum); both Silybum and silymarin have been consistently studied with respect to NAFLD and have been shown to have therapeutic effects [6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24]. Silymarin has been used for centuries as a natural remedy for liver disease [10]. Silymarin is a powerful antioxidant in the cell that can increase catalase and glutathione levels and also scavenge free lipid peroxyl radicals in hepatocytes [6,10,12,17,19,22,25]. Through its antioxidative capabilities, silymarin can restore NAD+ homeostasis, Sirtuin 1 (SIRT1) activity, and the AMP-activated protein kinase α (AMPKα) pathway to improve poly-(ADP-ribose)-polymerase (PARP) function, which protects the cell from the oxidative damage observed in NAFLD [22]. This effect also allows for improved hepatic lipid homeostasis [17,22]. This herb can also reduce hepatic de novo lipogenesis through the downregulation of peroxisome proliferator-activated receptor γ (PPARγ), acetyl-CoA carboxylase (ACC), and fatty acid synthase (FAS) [18]. Furthermore, silymarin can reduce steatosis and insulin resistance seen in NAFLD through restoration of the insulin receptor substrate-1 (IRS-1)/PI3K/Akt pathway [12]. Silymarin is also able to reduce hepatic inflammation through activation of the farnesyl X receptor (FXR) which correlates with the inhibition of NF-κB transactivity [12,16]. Due to the vast data supporting milk thistle as a therapeutic option from experimental mouse studies, there are multiple clinical trials testing its effects [7,9,11,12,16,17,18,20,21,22]. In a randomized double-blinded, placebo-controlled trial, Wah Kheong et al. tested the effects of silymarin supplementation on adult patients with biopsy-proven NASH and a NAFLD activity score (NAS) of greater than 4 [23]. The 99 patients were divided into a treatment group that received 700 mg three times daily and a placebo group [23] for 48 weeks [23]. The treatment group showed reduced aspartate aminotransferase with reduced fibrosis and liver stiffness without any issues in tolerability in patients [23]. Another study by Solhi et al. treated NASH patients with 210 mg of silymarin daily and after 8 weeks noted decreased alanine aminotransferase (ALT) and aspartate amino transferase (AST) levels in the treated group [13]. Zhong et al. completed a meta-analysis of eight randomized controlled trials (RCTs) studying NAFLD, and concluded that silymarin had statistically significant effect in reducing AST and ALT levels in patients [24].

2.2. Resveratrol

Resveratrol is another common alternative treatment studied with NAFLD [26,27,28,29,30,31,32,33,34,35,36]. Resveratrol has been shown to have similar effects to milk thistle in that is has antioxidative and anti-inflammatory properties [26,27,28,29,30,31,32,33,34,35,36]. Resveratrol supplementation is able to decrease inflammation and decrease hepatic steatosis through the inhibition of overactivated inflammatory agent NF-κB through the activation of the AMPKα/SIRT1 pathway which also regulates hepatic lipid metabolism and allows for lipid accumulation clearing through autophagy [31]. Resveratrol has also been shown to help decrease endoplasmic reticulum (ER) stress markers such as C/EBP homologous protein (CHOP) [33]. ER stress has been linked with NAFLD, lipotoxicity, hepatocyte apoptosis, and steatosis [33]. Sterol regulatory element-binding proteins (SREBPs) are key for de novo lipogenesis, and over-activation of this process is exhibited in NAFLD [36]. Resveratrol can block the activation of SREBP-1 and SREBP-2 by inhibiting their proteolytic cleavage [36]. Also, by inhibiting the carnitine palmitoyltransferase (CPT-1) and uncoupling protein (UCP-2), resveratrol can decrease free fatty acid β-oxidation and the production of reactive oxygen species (ROS) [36]. Although the experimental data on resveratrol supports its use with NAFLD, an early study found that there was no benefit to resveratrol supplementation [26,31,33,35,36]. This early study by Chachay et al. gave 10 obese NAFLD patients 3000 mg of resveratrol for 8 weeks, noting no improvement and even increased liver enzymes [26]. However, more recent studies have indicated that resveratrol could be a therapeutic agent for NAFLD patients [27,28,29,30,32,34]. In a double-blinded RCT by Faghihzadeh et al., 50 NAFLD patients were divided to receive lifestyle modification and placebo or 500 mg of resveratrol for 12 weeks and after the study period the treatment group had improved inflammatory cytokines, ALT, and hepatic steatosis compared to the placebo group [27,29]. A similar RCT by Chen et al. with 60 NAFLD patients randomized to either placebo or 600 mg of resveratrol for three months noted improved levels of ALT, AST, insulin resistance, and inflammatory factors [28]. Furthermore, Heeboll et al. observed that dosing affected the efficacy of resveratrol as a treatment [30]. Their study noted that high-dose resveratrol does not improve NAFLD histologically but has minimal ameliorating effects on liver enzymes [30]. Despite the beneficial effects of resveratrol in the clinical studies mentioned, meta-analysis by Elgebaly et al. concluded that current research is insufficient to claim that resveratrol improves NAFLD fibrosis and did not significantly improve liver enzymes [34].

3. Caffeine and Tea

3.1. Coffee

Consumed daily by many individuals, coffee has been noted as having an inverse relationship with NAFLD disease progression in many studies [37,38,39,40,41,42,43,44,45,46,47,48]. A cross-sectional study by Bambha et al. analyzed coffee drinking habits in 782 patients in the NASH Clinical Research Network and correlated coffee intake with decreased odds of advanced fibrosis in patients with lower insulin resistance [38]. In a similar study, Zelber-Sagi et al. examined cross-sectional as well as prospective patient data for 7 years in a sub cohort of patients [41]. In the cross-sectional portion of the study, increased coffee intake was inversely associated with clinically significant fibrosis [41]. Furthermore, the prospective analysis noted no association between new development of NAFLD and coffee intake but observed a protective effect of coffee on fibrosis progression [41]. When comparing patients with NAFLD to healthy individuals, Gutierrez-Grobe et al. noted that coffee intake was significantly higher in the healthy patients compared to those with NAFLD [49]. Coffee can upregulate ER and mitochondrial chaperone proteins such as glucose-related protein 78 (GRP78) which modulates ER homeostasis and prevents the activation of SREBP-1 [39]. Experimental studies with GRP78 knockout mice noted increased inflammation, steatosis, and apoptosis in hepatocytes due to its role in the unfolded protein response [50]. Coffee, contains polyphenols similar in structure to silymarin, which can also increase the production of antioxidant proteins [39]. Coffee notably increases periredoxin-1 (PRDX-1) which helps reduce ROS and decrease the oxidative stress in hepatocytes [39]. The synergistic effect of the polyphenols and caffeine in hepatocytes allow it to decrease insulin resistance and steatohepatitis [46]. Moreover, the polyphenols seem to be the cause of the antifibrotic effects of coffee over caffeine [46]. Finally, meta-analysis conducted by Wijarnpreecha et al. noted that coffee drinkers had a decreased risk of NAFLD and that regular daily consumption of coffee correlated with decreased risk of fibrosis [47].

3.2. Green Tea

Green tea, made from the leaves of the Camillia sinensis plant, like coffee, is commonly consumed and has been well documented to have beneficial effects with NAFLD [51,52,53,54,55,56,57,58,59,60,61,62]. The main therapeutic agent for NAFLD in green tea is epigallocatechin-3-gallate (EGCG) [54,55]. EGCG decreases hepatic inflammation through the reduction of hepatic cyclooxygenase-2 (COX2), prostaglandin E2, NF-κB, and toll-like receptor 4 (TLR4) [53,56,59]. EGCG also regulates hepatic lipid homeostasis through modulation of mitochondrial complex chain proteins and the previously mentioned AMPK, IRS-1, SREBP, and PPARγ pathways [54,55,60,61]. Furthermore, the polyphenols and EGCG in green tea allow for an antioxidative effect through NADPH oxidase and cytochrome P450 2E1 (CYP2E1) [62,63]. In the clinical setting, many trials have seen improvements in NAFLD patients with green tea therapy [51,52,57,58]. A double-blinded RCT with 80 NAFLD patients by Pezeshki et al. noted improved AST and ALT in patients supplemented with 500 mg of green tea extract daily compared to placebo [57]. Another study 12-week study by Hussein et al. with 80 patients randomized to receive 500 mg of green tea extract or placebo daily noted improvement in all parameters of NAFLD measured in the treatment group [58]. The green tea group observed improved inflammatory markers, aminotransferases, insulin resistance, adiponectin, and regression of fatty liver on ultrasound examination [58].

4. Household Herbs

4.1. Turmeric

Of all the common household spices and herbs, turmeric and garlic have the most research in the setting of treatment. Turmeric (active ingredient curcumin), also known as the Siam tulip, is a plant most notably cultivated in India as a spice recognized by its yellow color. Turmeric has been studied to be a therapeutic agent for NAFLD patients [64,65,66,67,68,69,70,71,72,73,74,75,76,77,78]. Curcumin is acknowledged as powerful antioxidant and anti-inflammatory agent, in addition to its many other beneficial features [74]. It acts as an antioxidant through the ability to neutralize free radicals and ROS that lead to oxidative stress in cells via the nuclear factor E2-related factor (Nrf2) and NADPH oxidase [68,74,77,78]. Curcumin can decrease major NAFLD risk factors such as hyperuricemia, dyslipidemia, and insulin resistance through the reduction of serum lipids and uric acid concentration [66]. It can also decrease hepatic steatosis through lipid homeostasis by increasing the inhibition of CD36 and PPARγ by the cAMP response element-binding (CREB) protein and the adiponectin precursor, ADIPOQ [64,72,77]. Curcumin can enhance the expression of PPARα and liver X receptor alpha (LXRα) and reduce SREBP activation and improve lipid balance [76,77]. Hepatic inflammation is controlled by the ability of curcumin to regulate the levels of hepatic proteins NF-κB, IP-10, IL-1β, IFN-γ, TLR4, and CD68 [68]. Also, the impairment intestinal mucosal mechanical barrier typical of NASH is reduced through curcumin’s upregulation of occludin [70]. Clinical trials have been used to confirm the experimental benefits of curcumin use in NAFLD patients [66,67,71,73,75]. Panahi et al. studied 102 NAFLD patients randomized to receive placebo or 1000 mg/day of curcumin for 8 weeks and concluded that curcumin improved hepatic steatosis determined through imaging and liver enzymes in NAFLD patients without any issues of tolerance [73]. A prospective cohort study by Selmanovic et al. with 400 mg curcumin supplementation also noted statistically significantly improved liver ultrasound results [74].

4.2. Garlic

Garlic is one of the most common herbs used around the world in cooking and medicine [79,80,81,82,83,84,85,86]. The main active components that give rise to the therapeutic effects of garlic are S-allyl cysteine (SAC), S-allymercaptocysteine (SAMC), diallyl disulfide (DADS), and cinnamoyloctopamines [79,80,81,82,83,84,85,86]. SAC is very similar to resveratrol is its ability to act as a SIRT1 activator that induces the activation of the AMPKα pathway and reduces hepatic lipogenesis and lipotoxicity [79]. It can also prevent lipid induced cell death by reducing free fatty acid ROS production and caspase activation [79]. SAMC and DADS act as antioxidants by inhibiting CYP2E1 and increasing catalase and glutathione peroxidase (GPx) [80,82]. Furthermore, it can decrease fibrosis factors, Transforming Growth Factor-β1 (TGF-β1) and α-Smooth Muscle Actin (α-SMA), and the inflammatory cytokines that would lead to the activation hepatic Kupffer cells and hepatic stellate cells (HSCs) that would cause collagen deposition [80]. Finally, SAMC can reduce inflammation through NF-κB and improve lipid homeostasis and insulin resistance through the AMPK and IRS-1/PI3K/Akt pathway [80,81]. DADS is also able to provide therapeutic effects through the stimulation of PPARα and CPT-1 [82]. The cinnamoyloctopamines are antioxidants that increase superoxide dismutase (SOD) and anti-inflammatory factors by decreasing COX-2 [84]. Clinically, Soleimani et al. noted that 800-mg garlic supplementation in NAFLD patients was able to significantly decrease body weight and body fat mass [85]. Furthermore, a double blinded RCT by Kim et al. observed improved ALT in patients with mild hepatic dysfunction who were given fermented garlic extract [86]

4.3. Rosemary, Peppermint, Basil, Lavender, Oregano, and Sage

Ursolic acid and carnosic acid are the active components of many herbs used in the common household [87,88]. Ursolic acid is found in rosemary, peppermint, basil, lavender, and oregano while carnosic acid is in rosemary and sage [87,88]. Ursolic acid’s ability to improve lipotoxicity and lipid homeostasis comes through regulation of PPARα, the AMPK pathway, and reduction of ER stress in hepatic cells [87,89,90]. Carnosic acid acts through modulation of hepatic SOD, SIRT1, NF-κB, PI3K/Akt, and SREBP-1c to exert antioxidant, anti-inflammatory, anti-adipogenic, and anti-apoptotic effects [88,91,92,93,94]. Further research on these herbs is necessary to determine their effects on NAFLD patients.

4.4. Ginger, Cocoa, and Cinnamon

Ginger, cocoa, and cinnamon have been explored as potential therapeutic agents, but not much is known about their use yet. Of the three, most of the research supports ginger supplementation [95,96,97]. Ginger has antioxidative capabilities through the inhibition of CYP2E1 and upregulation of SOD, catalase, and GPx to reduce ROS and oxidative stress [96] Furthermore, ginger has anti-inflammatory capabilities by reducing cytokines Tumor Necrosis Factor-α (TNF-α), IL-1β, and IL-6, as well as anti-lipotoxic/lipogenic capabilities by reducing SREBP-1c, ACC, and FAS [96]. A NAFLD 44-patient study by Rahimlou et al. observed improved ALT, insulin resistance, and hepatic steatosis in patients receiving two grams of ginger supplementation daily for 12 weeks versus placebo [97]. One particular type of ginger to be considered is Alpinia zerumbet [98,99]; which is a tropical plant commonly used as a Japanese herbal medicine and has been associated with the longevity of the people of Okinawa, Japan [98]. Its main component is dihydro-5,6-dehydrokavain (DDK), a powerful anti-inflammatory and antioxidative agent, which may be a promising therapy for patients with NAFLD patients; however, it requires further research [98,99].
Cocoa has gained interested based on its antioxidative potential and has been found to decrease NADPH oxidase levels in NASH patients and reduce oxidative stress [100,101]. Similarly, cinnamon has been studied for its antioxidative effects as well and a blinded trial in NAFLD patients by Askari et al. reported that 1500 mg of cinnamon supplementation for 12 weeks reduced AST, ALT, and insulin resistance compared to the placebo group [102].

5. Traditional Chinese Herbs

5.1. Ginkgo Biloba

Ginkgo biloba is one of the most common trees in China and its leaves have been studied for their therapeutic properties [103,104,105,106,107,108]. Through the upregulation of CPT-1A, ginkgo biloba can regulate fatty acid metabolism in the liver and reduce fat accumulation as well as decrease ROS production [103,104]. Furthermore, the ability of Gingko biloba to increase antioxidative enzymes SOD, GPx, and catalase allow it to reduce the oxidative stress in the liver as well [106]. Multiple experimental studies have been completed in NAFLD- and NASH-induced mice with Gingko biloba extract supplementation and noted improved AST, ALT, hepatic inflammation, and hepatic steatosis [103,104,105,106,107,108].

5.2. Ginseng

Ginseng, or Panax ginseng, has been often used in traditional Chinese medicine to treat multiple metabolic conditions, including hepatosteatosis [52,109,110,111,112,113]. The active agents in ginseng are ginsenosides and saponins [109,111,112,113]. Ginseng is able to reduce lipid accumulation through the induction of the SIRT1/AMPK pathway and activate auto-phagocytic pathways [111,112]. Moreover, ginseng is able to reduce hepatic fibrosis factors such as collagen-1 and α-SMA and prevent HSC activation [113]. Ginseng is able to also reduce inflammation and ER stress through GRP78 [113]. Multiple mouse studies observed improved steatosis, liver enzymes, hepatic inflammation, and fibrosis in NAFLD/NASH-induced mice supplemented with ginseng extract [109,110,111,112,113]. Current data on ginseng supplementation seems promising for future trials and use with NAFLD patients.

5.3. Lotus, Goji Berry, Astralagus, and Ciruwujia

These four herbs have been used in traditional Chinese medicine and have recently started being used in experimental studies which have indicated their potential for therapeutic use in NAFLD through improved liver function, steatosis, and inflammation in mice [114,115,116,117,118,119,120,121,122,123,124,125,126]. However, there are no clinical trials supporting their use. Lotus can be used to exert anti-inflammatory effects on the liver through its regulation of IL-6, NF-κB, TNF-α, and TGF-β1 [115,118,119,123]. Also, through its regulation of adiponectin, lotus can promote hepatic lipid homeostasis [115,118,119,123]. Goji berry, from the plant Lycium barbarum, exerts its hepatoprotective effects through regulation of lipid homeostasis (SIRT1/AMPK and SREBP-1c), and inflammation (NF-κB) [116,117,120,125]. Astragalus and ciruwujia have yet to be fully explored as therapeutic agents, but regression of fatty liver in NAFLD-induced mouse studies warrant more in-depth understanding [114,121,122,124,126]. Astragalus has hypothesized to function through CPT-1, FXR, PPARα, and SREBP-1c to give the its anti-inflammatory and lipid homeostatic effects to improve liver function and steatosis in mice [122,124,126]. Ciruwujia has been postulated to improve liver function through IL-6, CPT-1α, and TLR4 to confer anti-inflammatory effects [114,121].

6. Other Herbs

6.1. Cannibinoids

Cannabinoids have gained interest on their effects due to controversy of their usage across the United States and mixed understanding on their possible therapeutic effects on NAFLD [127,128,129,130,131,132,133,134]. A cross-sectional study by Adejumo et al. noted a statistically significant lower prevalence of NAFLD in cannabis users and another larger study by Kim et al. concluded an inverse relationship between cannabis use and suspected and ultrasonographically-diagnosed NAFLD [132,133]. Further research found that cannabinoids act through two receptors—cannabinoid receptors 1 and 2 (CB1 and CB2) [127,128,129,134]. In general, CB1 is pro-fibrogenic, while CB2 is anti-fibrogenic [127,128,129,134]. Tetrahtydrocannabinol (THC) acts via both receptors in a dose-dependent relationship, in which at low-levels it is a CB1 antagonist and has therapeutic effects, but at higher levels becomes a CB1 agonist and becomes a risk factor [127,128,129,134]. Further research on the exocannabinoid and endocannabinoid system is warranted to fully understand their possible therapeutic use.

6.2. Licorice, Red Clover, and Chamomile

Licorice, red clover, and chamomile are herbs currently being explored for their possible therapeutic use and are possible routes for future research. Licorice has been proposed to promote lipid homeostasis through SREBP-1c, FAS, ACC1, PPARα, and CPT-1α [135,136]. A double-blinded clinical trial by Hajiaghamohammadi et al. randomized 66 patients to receive either placebo or 2 g aqueous licorice root extract for 2 months and observed improved AST and ALT in the treated group [137]. Red clover and chamomile are hypothesized to have hepatoprotective effects due to their possible role in PPARα regulation, but currently not enough is known about how they function in terms of NAFLD and more studies are necessary [138,139] Table 1 summarizes the various herbs studied in NAFLD therapy.

7. Non-Herb Supplements

7.1. Fish Oils

Fish oils, mainly eicosapentaenoic acid (EPA) and docosahexanoic acid (DHA), represent one of the most highly marketed and widely used supplement types for many metabolic conditions [140,141,142,143,144,145,146,147,148,149,150,151,152,153]. They can improve lipid homeostasis and inflammation through the PPARα, AMPK, and adiponectin [140,141,142,143,144,145,146,147,148,149,150,151,152,153]. A 41-patient NASH study by Argo et al. randomized patients to receive 3000 mg/day of fish oil or placebo for 1 year and observed improved ALT in the treated patients [140]. Another study by Boyraz et al. in 52 obese NAFLD patients treated with 1000 mg fish oil reported improved AST, ALT, and steatosis [141] Nogueira et al. noted that fish oil supplementation improved steatosis and inflammation in patients [147]. Meta-analysis by Yu et al. calculated that fish oil supplementation improves ALT [152]. Another meta-analysis by Chen et al. noted that fish oil supplementation in NAFLD children improves steatosis, ALT, and AST [153].

7.2. Coenzyme Q-10

Like fish oil, coenzyme Q-10 (CoQ10) is commonly seen in stores and used widely. Many mouse studies have noted the hepatoprotective effects of CoQ10 [154,155,156]. A study by Farhangi et al. in NASH-induced mice noted improved liver biochemical enzymes after supplementation [155]. CoQ10 is able to modulate TNF-α and adiponectin levels to improve inflammation and lipid homeostasis in the liver [155,157]. A blinded RCT by Farsi et al. treated NAFLD patients with 100 mg CoQ10 and noted reduced AST, ALT, and hepatic inflammation compared to placebo [157].

8. Safety Concerns for Herb Supplementation

Despite the apparent therapeutic effects of the herbs discussed in this article, it is important to keep in mind that there are no guidelines or recommendations for herb usage in NAFLD management [2]. The lack of regulation in this field of treatment agents suggests caution with use of herbs in the context of NAFLD. Also, the lack of research indicates the possibility of drug interactions or adverse effects [158,159,160,161]. Herb–drug interactions have already been noted in warfarin, aspirin, alkylating agents, and cyclosporine, leading to toxicity in patients [158,161]. Furthermore, as natural herbs are converted to supplements, the final product may no longer be purely herbal and can contain trace metals that could become toxic if taken consistently at high levels [162]. Alhusban et al. discovered that some commonly used pharmaceutical herbal products contained toxic metals, such as Pb, Al, Ni, Cd, and As [162]. Of these, some products had recommended intakes that led to accumulation of the toxic metals greater than the tolerable daily intake set by the Food and Drug Administration (FDA) [162].
Furthermore, some studies have indicated that some herbs may have hepatotoxic characteristics [163,164,165,166]. Multiple studies have noted herb-induced liver injury (HILI), in particular with Chinese herbal medicine [163,164,165,166]. However, there have been obstacles in accurately assessing if liver injury is truly herb-induced versus drug-induced; more accurate diagnostic tools are currently being developed to make this distinction [163,164,165,166]. Nonetheless, the possible presence of hepatotoxins in some herbs and their unregulated, non-standardized use can lead to worsening of underlying liver disease, NASH, or other chronic hepatic diseases. Also, none of the studies reviewed noted obesity as a contraindication of herb usage. Extensive research is necessary to test the safety of herbs as a therapeutic agents before their use can be recommended in patients with NAFLD or other ailments.

9. Conclusions

All in all, there are many promising herbs and supplements that can be used as an alternative treatment for NAFLD. Some of them are basic nutrients that people use daily as spices and seasonings in their diet. However, to say that they should be part of a treatment for NAFLD and/or NASH is to overstep, as there is there is no clinical data that support this notion. Milk thistle, turmeric, resveratrol, coffee, and green tea have been extensively researched in the NAFLD population and have not been shown to have toxic effects. We can only recommend that they be used with caution and that further research is necessary to confirm the therapeutic role of herbs in patient with NAFLD.

Author Contributions

A.A.L., U.I., S.S., N.D.S., W.K., G.C., D.K. contributed significantly to data collection, compilation. B.J.P. prepared the manuscript. A.A. supervised the project.

Funding

No funding was needed to complete this project.

Conflicts of Interest

The authors declare no conflict of interest, including financial and/or material support for the preparation of this manuscript.

Abbreviations

NAFLDnonalcoholic fatty liver disease
NASHnonalcoholic steatohepatitis
PARPpoly-(ADP-ribose)-polymerase
PPARγperoxisome proliferator activated receptor γ
ACCacetyl-CoA carboxylase
FASfatty acid synthase
FXRfarnesyl X receptor
NASNAFLD activity score
ALTalanine amino transferase
ASTaspartate amino transferase
ERendoplasmic reticulum
CHOPC/EBP homologous protein
CPT-1carnitine palmitoyltransferase
UCP-2uncoupling protein
ROSreactive oxygen species
GRP78glucose-related protein 78
PRDX-1periredoxin-1
EGCGepigallocatechin-3-gallate
COX2cyclooxygenase-2
TLR4toll-like receptor 4
CYP2E1cytochrome P450 2E1
Nrf2nuclear factor E2 related factor
CREBcAMP response element-binding
SACS-allyl cysteine
SAMCS-allymercaptocysteine
DADSdiallyl disulfide
GPxglutathione peroxidase
HSCshepatic stellate cells
SODsuperoxide dismutase
DDKdihydro-5,6-dehydrokavain
CB1cannabinoid receptor 1
CB2cannabinoid receptors 2
EPAeicosapentaenoic acid
DHAdocosahexanoic acid
CoQ10coenzyme Q-10
FDAFood and Drug Administration
HILIherb-induced liver injury

References

  1. Blond, E.; Disse, E.; Cuerq, C.; Drai, J.; Valette, P.J.; Laville, M.; Thivolet, C.; Simon, C.; Caussy, C. Easl-EASD-EASO clinical practice guidelines for the management of non-alcoholic fatty liver disease in severely obese people: Do they lead to over-referral? Diabetologia 2017, 60, 1218–1222. [Google Scholar] [CrossRef] [PubMed]
  2. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Diehl, A.M.; Brunt, E.M.; Cusi, K.; Charlton, M.; Sanyal, A.J.; American Gastroenterological Association; American Association for the Study of Liver Diseases; et al. The diagnosis and management of non-alcoholic fatty liver disease: Practice guideline by the American gastroenterological association, American association for the study of liver diseases, and American college of gastroenterology. Gastroenterology 2012, 142, 1592–1609. [Google Scholar] [CrossRef] [PubMed]
  3. Oseini, A.M.; Sanyal, A.J. Therapies in non-alcoholic steatohepatitis (NASH). Liver Int. 2017, 37 (Suppl. 1), 97–103. [Google Scholar] [CrossRef] [PubMed]
  4. Liu, Z.L.; Xie, L.Z.; Zhu, J.; Li, G.Q.; Grant, S.J.; Liu, J.P. Herbal medicines for fatty liver diseases. Cochrane Database Syst. Rev. 2013, CD009059. [Google Scholar] [CrossRef] [PubMed]
  5. Xiao, J.; Fai So, K.; Liong, E.C.; Tipoe, G.L. Recent advances in the herbal treatment of non-alcoholic fatty liver disease. J. Tradit. Complement. Med. 2013, 3, 88–94. [Google Scholar] [CrossRef] [PubMed]
  6. Hajiaghamohammadi, A.A.; Ziaee, A.; Oveisi, S.; Masroor, H. Effects of metformin, pioglitazone, and silymarin treatment on non-alcoholic fatty liver disease: A randomized controlled pilot study. Hepat. Mon. 2012, 12, e6099. [Google Scholar] [CrossRef] [PubMed]
  7. Kim, M.; Yang, S.G.; Kim, J.M.; Lee, J.W.; Kim, Y.S.; Lee, J.I. Silymarin suppresses hepatic stellate cell activation in a dietary rat model of non-alcoholic steatohepatitis: Analysis of isolated hepatic stellate cells. Int. J. Mol. Med. 2012, 30, 473–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Loguercio, C.; Andreone, P.; Brisc, C.; Brisc, M.C.; Bugianesi, E.; Chiaramonte, M.; Cursaro, C.; Danila, M.; de Sio, I.; Floreani, A.; et al. Silybin combined with phosphatidylcholine and vitamin e in patients with nonalcoholic fatty liver disease: A randomized controlled trial. Free. Radic. Biol. Med. 2012, 52, 1658–1665. [Google Scholar] [CrossRef] [PubMed]
  9. Salamone, F.; Galvano, F.; Cappello, F.; Mangiameli, A.; Barbagallo, I.; Li Volti, G. Silibinin modulates lipid homeostasis and inhibits nuclear factor κB activation in experimental nonalcoholic steatohepatitis. Transl. Res. 2012, 159, 477–486. [Google Scholar] [CrossRef] [PubMed]
  10. Cacciapuoti, F.; Scognamiglio, A.; Palumbo, R.; Forte, R.; Cacciapuoti, F. Silymarin in non alcoholic fatty liver disease. World J. Hepatol. 2013, 5, 109–113. [Google Scholar] [CrossRef] [PubMed]
  11. Yao, J.; Zhi, M.; Gao, X.; Hu, P.; Li, C.; Yang, X. Effect and the probable mechanisms of silibinin in regulating insulin resistance in the liver of rats with non-alcoholic fatty liver. Braz. J. Med. Biol. Res. 2013, 46, 270–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Zhang, Y.; Hai, J.; Cao, M.; Zhang, Y.; Pei, S.; Wang, J.; Zhang, Q. Silibinin ameliorates steatosis and insulin resistance during non-alcoholic fatty liver disease development partly through targeting irs-1/pi3k/akt pathway. Int. Immunopharmacol. 2013, 17, 714–720. [Google Scholar] [CrossRef] [PubMed]
  13. Solhi, H.; Ghahremani, R.; Kazemifar, A.M.; Hoseini Yazdi, Z. Silymarin in treatment of non-alcoholic steatohepatitis: A randomized clinical trial. Casp. J. Intern. Med. 2014, 5, 9–12. [Google Scholar]
  14. Abenavoli, L.; Greco, M.; Nazionale, I.; Peta, V.; Milic, N.; Accattato, F.; Foti, D.; Gulletta, E.; Luzza, F. Effects of mediterranean diet supplemented with silybin-vitamin e-phospholipid complex in overweight patients with non-alcoholic fatty liver disease. Expert Rev. Gastroenterol. Hepatol. 2015, 9, 519–527. [Google Scholar] [CrossRef] [PubMed]
  15. Aller, R.; Izaola, O.; Gomez, S.; Tafur, C.; Gonzalez, G.; Berroa, E.; Mora, N.; Gonzalez, J.M.; de Luis, D.A. Effect of silymarin plus vitamin e in patients with non-alcoholic fatty liver disease. A randomized clinical pilot study. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 3118–3124. [Google Scholar] [PubMed]
  16. Gu, M.; Zhao, P.; Huang, J.; Zhao, Y.; Wang, Y.; Li, Y.; Li, Y.; Fan, S.; Ma, Y.M.; Tong, Q.; et al. Silymarin ameliorates metabolic dysfunction associated with diet-induced obesity via activation of farnesyl x receptor. Front. Pharmacol. 2016, 7, 345. [Google Scholar] [CrossRef] [PubMed]
  17. Ni, X.; Wang, H. Silymarin attenuated hepatic steatosis through regulation of lipid metabolism and oxidative stress in a mouse model of nonalcoholic fatty liver disease (NAFLD). Am. J. Transl. Res. 2016, 8, 1073–1081. [Google Scholar] [PubMed]
  18. Cui, C.X.; Deng, J.N.; Yan, L.; Liu, Y.Y.; Fan, J.Y.; Mu, H.N.; Sun, H.Y.; Wang, Y.H.; Han, J.Y. Silibinin capsules improves high fat diet-induced nonalcoholic fatty liver disease in hamsters through modifying hepatic de novo lipogenesis and fatty acid oxidation. J. Ethnopharmacol. 2017, 208, 24–35. [Google Scholar] [CrossRef] [PubMed]
  19. Federico, A.; Conti, V.; Russomanno, G.; Dallio, M.; Masarone, M.; Stiuso, P.; Tuccillo, C.; Caraglia, M.; Manzo, V.; Persico, M.; et al. A long-term treatment with silybin in patients with non-alcoholic steatohepatitis stimulates catalase activity in human endothelial cells. In Vivo 2017, 31, 609–618. [Google Scholar] [PubMed]
  20. Marcolino Assis-Junior, E.; Melo, A.T.; Pereira, V.B.M.; Wong, D.V.T.; Sousa, N.R.P.; Oliveira, C.M.G.; Malveira, L.R.C.; Moreira, L.S.; Souza, M.; Almeida, P.R.C.; et al. Dual effect of silymarin on experimental non-alcoholic steatohepatitis induced by irinotecan. Toxicol. Appl. Pharmacol. 2017, 327, 71–79. [Google Scholar] [CrossRef] [PubMed]
  21. Marin, V.; Gazzin, S.; Gambaro, S.E.; Dal Ben, M.; Calligaris, S.; Anese, M.; Raseni, A.; Avellini, C.; Giraudi, P.J.; Tiribelli, C.; et al. Effects of oral administration of silymarin in a juvenile murine model of non-alcoholic steatohepatitis. Nutrients 2017, 9. [Google Scholar] [CrossRef] [PubMed]
  22. Salomone, F.; Barbagallo, I.; Godos, J.; Lembo, V.; Currenti, W.; Cina, D.; Avola, R.; D’Orazio, N.; Morisco, F.; Galvano, F.; et al. Silibinin restores nad(+) levels and induces the sirt1/ampk pathway in non-alcoholic fatty liver. Nutrients 2017, 9. [Google Scholar] [CrossRef] [PubMed]
  23. Wah Kheong, C.; Nik Mustapha, N.R.; Mahadeva, S. A randomized trial of silymarin for the treatment of nonalcoholic steatohepatitis. Clin. Gastroenterol. Hepatol. 2017, 15, 1940–1949.e1948. [Google Scholar] [CrossRef] [PubMed]
  24. Zhong, S.; Fan, Y.; Yan, Q.; Fan, X.; Wu, B.; Han, Y.; Zhang, Y.; Chen, Y.; Zhang, H.; Niu, J. The therapeutic effect of silymarin in the treatment of nonalcoholic fatty disease: A meta-analysis (PRISMA) of randomized control trials. Medicine 2017, 96, e9061. [Google Scholar] [CrossRef] [PubMed]
  25. Shaker, E.; Mahmoud, H.; Mnaa, S. Silymarin, the antioxidant component and silybum marianum extracts prevent liver damage. Food Chem. Toxicol. 2010, 48, 803–806. [Google Scholar] [CrossRef] [PubMed]
  26. Chachay, V.S.; Macdonald, G.A.; Martin, J.H.; Whitehead, J.P.; O’Moore-Sullivan, T.M.; Lee, P.; Franklin, M.; Klein, K.; Taylor, P.J.; Ferguson, M.; et al. Resveratrol does not benefit patients with nonalcoholic fatty liver disease. Clin. Gastroenterol. Hepatol. 2014, 12, 2092–2103.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Faghihzadeh, F.; Adibi, P.; Rafiei, R.; Hekmatdoost, A. Resveratrol supplementation improves inflammatory biomarkers in patients with nonalcoholic fatty liver disease. Nutr. Res. 2014, 34, 837–843. [Google Scholar] [CrossRef] [PubMed]
  28. Chen, S.; Zhao, X.; Ran, L.; Wan, J.; Wang, X.; Qin, Y.; Shu, F.; Gao, Y.; Yuan, L.; Zhang, Q.; et al. Resveratrol improves insulin resistance, glucose and lipid metabolism in patients with non-alcoholic fatty liver disease: A randomized controlled trial. Dig. Liver Dis. 2015, 47, 226–232. [Google Scholar] [CrossRef] [PubMed]
  29. Faghihzadeh, F.; Adibi, P.; Hekmatdoost, A. The effects of resveratrol supplementation on cardiovascular risk factors in patients with non-alcoholic fatty liver disease: A randomised, double-blind, placebo-controlled study. Br. J. Nutr. 2015, 114, 796–803. [Google Scholar] [CrossRef] [PubMed]
  30. Heeboll, S.; Kreuzfeldt, M.; Hamilton-Dutoit, S.; Kjaer Poulsen, M.; Stodkilde-Jorgensen, H.; Moller, H.J.; Jessen, N.; Thorsen, K.; Kristina Hellberg, Y.; Bonlokke Pedersen, S.; et al. Placebo-controlled, randomised clinical trial: High-dose resveratrol treatment for non-alcoholic fatty liver disease. Scand. J. Gastroenterol. 2016, 51, 456–464. [Google Scholar] [CrossRef] [PubMed]
  31. Tian, Y.; Ma, J.; Wang, W.; Zhang, L.; Xu, J.; Wang, K.; Li, D. Resveratrol supplement inhibited the NF-κB inflammation pathway through activating ampkalpha-sirt1 pathway in mice with fatty liver. Mol. Cell. Biochem. 2016, 422, 75–84. [Google Scholar] [CrossRef] [PubMed]
  32. Charytoniuk, T.; Drygalski, K.; Konstantynowicz-Nowicka, K.; Berk, K.; Chabowski, A. Alternative treatment methods attenuate the development of NAFLD: A review of resveratrol molecular mechanisms and clinical trials. Nutrition 2017, 34, 108–117. [Google Scholar] [CrossRef] [PubMed]
  33. Ding, S.; Jiang, J.; Zhang, G.; Bu, Y.; Zhang, G.; Zhao, X. Resveratrol and caloric restriction prevent hepatic steatosis by regulating sirt1-autophagy pathway and alleviating endoplasmic reticulum stress in high-fat diet-fed rats. PLoS ONE 2017, 12, e0183541. [Google Scholar] [CrossRef] [PubMed]
  34. Elgebaly, A.; Radwan, I.A.; AboElnas, M.M.; Ibrahim, H.H.; Eltoomy, M.F.; Atta, A.A.; Mesalam, H.A.; Sayed, A.A.; Othman, A.A. Resveratrol supplementation in patients with non-alcoholic fatty liver disease: Systematic review and meta-analysis. J. Gastrointest. Liver Dis. 2017, 26, 59–67. [Google Scholar]
  35. Asghari, S.; Asghari-Jafarabadi, M.; Somi, M.H.; Ghavami, S.M.; Rafraf, M. Comparison of calorie-restricted diet and resveratrol supplementation on anthropometric indices, metabolic parameters, and serum sirtuin-1 levels in patients with nonalcoholic fatty liver disease: A randomized controlled clinical trial. J. Am. Coll. Nutr. 2018, 37, 223–233. [Google Scholar] [CrossRef] [PubMed]
  36. Khaleel, E.F.; Abdel-Aleem, G.A.; Mostafa, D.G. Resveratrol improves high-fat diet induced fatty liver and insulin resistance by concomitantly inhibiting proteolytic cleavage of sterol regulatory element-binding proteins, free fatty acid oxidation, and intestinal triglyceride absorption. Can. J. Physiol. Pharmacol. 2018, 96, 145–157. [Google Scholar] [CrossRef] [PubMed]
  37. Catalano, D.; Martines, G.F.; Tonzuso, A.; Pirri, C.; Trovato, F.M.; Trovato, G.M. Protective role of coffee in non-alcoholic fatty liver disease (NAFLD). Dig. Dis. Sci. 2010, 55, 3200–3206. [Google Scholar] [CrossRef] [PubMed]
  38. Bambha, K.; Wilson, L.A.; Unalp, A.; Loomba, R.; Neuschwander-Tetri, B.A.; Brunt, E.M.; Bass, N.M.; Nonalcoholic Steatohepatitis Clinical Research Network (NASH CRN). Coffee consumption in NAFLD patients with lower insulin resistance is associated with lower risk of severe fibrosis. Liver Int. 2014, 34, 1250–1258. [Google Scholar] [CrossRef] [PubMed]
  39. Salomone, F.; Li Volti, G.; Vitaglione, P.; Morisco, F.; Fogliano, V.; Zappala, A.; Palmigiano, A.; Garozzo, D.; Caporaso, N.; D’Argenio, G.; et al. Coffee enhances the expression of chaperones and antioxidant proteins in rats with nonalcoholic fatty liver disease. Transl. Res. 2014, 163, 593–602. [Google Scholar] [CrossRef] [PubMed]
  40. Graeter, T.; Niedermayer, P.C.; Mason, R.A.; Oeztuerk, S.; Haenle, M.M.; Koenig, W.; Boehm, B.O.; Kratzer, W.; EMIL-Study Group. Coffee consumption and NAFLD: A community based study on 1223 subjects. BMC Res. Notes 2015, 8, 640. [Google Scholar] [CrossRef] [PubMed]
  41. Zelber-Sagi, S.; Salomone, F.; Webb, M.; Lotan, R.; Yeshua, H.; Halpern, Z.; Santo, E.; Oren, R.; Shibolet, O. Coffee consumption and nonalcoholic fatty liver onset: A prospective study in the general population. Transl. Res. 2015, 165, 428–436. [Google Scholar] [CrossRef] [PubMed]
  42. Barros, R.K.; Cotrim, H.P.; Daltro, C.; Alves, E.; de Freitas, L.A.; Daltro, C.; Oliveira, Y. Nonalcoholic steatohepatitis in morbid obese patients: Coffee consumption vs. Disease severity. Ann. Hepatol. 2016, 15, 350–355. [Google Scholar] [PubMed]
  43. Hodge, A.; Lim, S.; Goh, E.; Wong, O.; Marsh, P.; Knight, V.; Sievert, W.; de Courten, B. Coffee intake is associated with a lower liver stiffness in patients with non-alcoholic fatty liver disease, hepatitis c, and hepatitis b. Nutrients 2017, 9. [Google Scholar] [CrossRef] [PubMed]
  44. Katsagoni, C.N.; Georgoulis, M.; Papatheodoridis, G.V.; Fragopoulou, E.; Ioannidou, P.; Papageorgiou, M.; Alexopoulou, A.; Papadopoulos, N.; Deutsch, M.; Kontogianni, M.D. Associations between lifestyle characteristics and the presence of nonalcoholic fatty liver disease: A case-control study. Metab. Syndr. Relat. Disord. 2017, 15, 72–79. [Google Scholar] [CrossRef] [PubMed]
  45. Shokouh, P.; Jeppesen, P.B.; Hermansen, K.; Norskov, N.P.; Laustsen, C.; Jacques Hamilton-Dutoit, S.; Qi, H.; Stodkilde-Jorgensen, H.; Gregersen, S. A combination of coffee compounds shows insulin-sensitizing and hepatoprotective effects in a rat model of diet-induced metabolic syndrome. Nutrients 2017, 10. [Google Scholar] [CrossRef] [PubMed]
  46. Watanabe, S.; Takahashi, T.; Ogawa, H.; Uehara, H.; Tsunematsu, T.; Baba, H.; Morimoto, Y.; Tsuneyama, K. Daily coffee intake inhibits pancreatic beta cell damage and nonalcoholic steatohepatitis in a mouse model of spontaneous metabolic syndrome, tsumura-suzuki obese diabetic mice. Metab. Syndr. Relat. Disord. 2017, 15, 170–177. [Google Scholar] [CrossRef] [PubMed]
  47. Wijarnpreecha, K.; Thongprayoon, C.; Ungprasert, P. Coffee consumption and risk of nonalcoholic fatty liver disease: A systematic review and meta-analysis. Eur. J. Gastroenterol. Hepatol. 2017, 29, e8–e12. [Google Scholar] [CrossRef] [PubMed]
  48. Veronese, N.; Notarnicola, M.; Cisternino, A.M.; Reddavide, R.; Inguaggiato, R.; Guerra, V.; Rotolo, O.; Zinzi, I.; Leandro, G.; Correale, M.; et al. Coffee intake and liver steatosis: A population study in a mediterranean area. Nutrients 2018, 10. [Google Scholar] [CrossRef] [PubMed]
  49. Gutierrez-Grobe, Y.; Chavez-Tapia, N.; Sanchez-Valle, V.; Gavilanes-Espinar, J.G.; Ponciano-Rodriguez, G.; Uribe, M.; Mendez-Sanchez, N. High coffee intake is associated with lower grade nonalcoholic fatty liver disease: The role of peripheral antioxidant activity. Ann. Hepatol. 2012, 11, 350–355. [Google Scholar] [PubMed]
  50. Ji, C.; Kaplowitz, N.; Lau, M.Y.; Kao, E.; Petrovic, L.M.; Lee, A.S. Liver-specific loss of glucose-regulated protein 78 perturbs the unfolded protein response and exacerbates a spectrum of liver diseases in mice. Hepatology 2011, 54, 229–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Sakata, R.; Nakamura, T.; Torimura, T.; Ueno, T.; Sata, M. Green tea with high-density catechins improves liver function and fat infiltration in non-alcoholic fatty liver disease (NAFLD) patients: A double-blind placebo-controlled study. Int. J. Mol. Med. 2013, 32, 989–994. [Google Scholar] [CrossRef] [PubMed]
  52. Miranda-Henriques, M.S.; Diniz Mde, F.; Araujo, M.S. Ginseng, green tea or fibrate: Valid options for nonalcoholic steatohepatitis prevention? Arq. Gastroenterol. 2014, 51, 255–260. [Google Scholar] [CrossRef] [PubMed]
  53. Chung, M.Y.; Mah, E.; Masterjohn, C.; Noh, S.K.; Park, H.J.; Clark, R.M.; Park, Y.K.; Lee, J.Y.; Bruno, R.S. Green tea lowers hepatic cox-2 and prostaglandin e2 in rats with dietary fat-induced nonalcoholic steatohepatitis. J. Med. Food 2015, 18, 648–655. [Google Scholar] [CrossRef] [PubMed]
  54. Santamarina, A.B.; Carvalho-Silva, M.; Gomes, L.M.; Okuda, M.H.; Santana, A.A.; Streck, E.L.; Seelaender, M.; do Nascimento, C.M.; Ribeiro, E.B.; Lira, F.S.; et al. Decaffeinated green tea extract rich in epigallocatechin-3-gallate prevents fatty liver disease by increased activities of mitochondrial respiratory chain complexes in diet-induced obesity mice. J. Nutr. Biochem. 2015, 26, 1348–1356. [Google Scholar] [CrossRef] [PubMed]
  55. Santamarina, A.B.; Oliveira, J.L.; Silva, F.P.; Carnier, J.; Mennitti, L.V.; Santana, A.A.; de Souza, G.H.; Ribeiro, E.B.; Oller do Nascimento, C.M.; Lira, F.S.; et al. Green tea extract rich in epigallocatechin-3-gallate prevents fatty liver by ampk activation via lkb1 in mice fed a high-fat diet. PLoS ONE 2015, 10, e0141227. [Google Scholar] [CrossRef] [PubMed]
  56. Li, J.; Sapper, T.N.; Mah, E.; Rudraiah, S.; Schill, K.E.; Chitchumroonchokchai, C.; Moller, M.V.; McDonald, J.D.; Rohrer, P.R.; Manautou, J.E.; et al. Green tea extract provides extensive Nrf2-independent protection against lipid accumulation and NF-κB pro- inflammatory responses during nonalcoholic steatohepatitis in mice fed a high-fat diet. Mol. Nutr. Food Res. 2016, 60, 858–870. [Google Scholar] [CrossRef] [PubMed]
  57. Pezeshki, A.; Safi, S.; Feizi, A.; Askari, G.; Karami, F. The effect of green tea extract supplementation on liver enzymes in patients with nonalcoholic fatty liver disease. Int. J. Prev. Med. 2016, 7, 28. [Google Scholar] [CrossRef] [PubMed]
  58. Hussain, M.; Habib Ur, R.; Akhtar, L. Therapeutic benefits of green tea extract on various parameters in non-alcoholic fatty liver disease patients. Pak. J. Med. Sci. 2017, 33, 931–936. [Google Scholar] [CrossRef] [PubMed]
  59. Li, J.; Sapper, T.N.; Mah, E.; Moller, M.V.; Kim, J.B.; Chitchumroonchokchai, C.; McDonald, J.D.; Bruno, R.S. Green tea extract treatment reduces NF-κB activation in mice with diet-induced nonalcoholic steatohepatitis by lowering tnfr1 and tlr4 expression and ligand availability. J. Nutr. Biochem. 2017, 41, 34–41. [Google Scholar] [CrossRef] [PubMed]
  60. Pan, M.H.; Yang, G.; Li, S.; Li, M.Y.; Tsai, M.L.; Wu, J.C.; Badmaev, V.; Ho, C.T.; Lai, C.S. Combination of citrus polymethoxyflavones, green tea polyphenols, and lychee extracts suppresses obesity and hepatic steatosis in high-fat diet induced obese mice. Mol. Nutr. Food. Res. 2017, 61. [Google Scholar] [CrossRef] [PubMed]
  61. Tan, Y.; Kim, J.; Cheng, J.; Ong, M.; Lao, W.G.; Jin, X.L.; Lin, Y.G.; Xiao, L.; Zhu, X.Q.; Qu, X.Q. Green tea polyphenols ameliorate non-alcoholic fatty liver disease through upregulating ampk activation in high fat fed zucker fatty rats. World J. Gastroenterol. 2017, 23, 3805–3814. [Google Scholar] [CrossRef] [PubMed]
  62. Zhu, W.; Chen, S.; Chen, R.; Peng, Z.; Wan, J.; Wu, B. Taurine and tea polyphenols combination ameliorate nonalcoholic steatohepatitis in rats. BMC Complement. Altern. Med. 2017, 17, 455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Chung, M.Y.; Park, H.J.; Manautou, J.E.; Koo, S.I.; Bruno, R.S. Green tea extract protects against nonalcoholic steatohepatitis in ob/ob mice by decreasing oxidative and nitrative stress responses induced by proinflammatory enzymes. J. Nutr. Biochem. 2012, 23, 361–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Chen, W.J.; Cai, B.; Chen, H.T.; Cao, C.Y.; Du, Y.L.; Li, Y.Y.; Nie, Y.Q.; Zhou, Y.J. The role of adipoq methylation in curcumin-administrated experimental nonalcoholic fatty liver disease. J. Dig. Dis. 2016, 17, 829–836. [Google Scholar] [CrossRef] [PubMed]
  65. Li, C.; Li, J.; Chen, Y.; Zhong, X.; Kang, M. Effect of curcumin on visfatin and zinc-alpha2-glycoprotein in a rat model of non-alcoholic fatty liver disease. Acta Cir. Bras. 2016, 31, 706–713. [Google Scholar] [CrossRef] [PubMed]
  66. Panahi, Y.; Kianpour, P.; Mohtashami, R.; Jafari, R.; Simental-Mendia, L.E.; Sahebkar, A. Curcumin lowers serum lipids and uric acid in subjects with nonalcoholic fatty liver disease: A randomized controlled trial. J. Cardiovasc. Pharmacol. 2016, 68, 223–229. [Google Scholar] [CrossRef] [PubMed]
  67. Rahmani, S.; Asgary, S.; Askari, G.; Keshvari, M.; Hatamipour, M.; Feizi, A.; Sahebkar, A. Treatment of non-alcoholic fatty liver disease with curcumin: A randomized placebo-controlled trial. Phytother. Res. 2016, 30, 1540–1548. [Google Scholar] [CrossRef] [PubMed]
  68. Afrin, R.; Arumugam, S.; Rahman, A.; Wahed, M.I.; Karuppagounder, V.; Harima, M.; Suzuki, H.; Miyashita, S.; Suzuki, K.; Yoneyama, H.; et al. Curcumin ameliorates liver damage and progression of nash in nash-hcc mouse model possibly by modulating hmgb1- NF-κB translocation. Int. Immunopharmacol. 2017, 44, 174–182. [Google Scholar] [CrossRef] [PubMed]
  69. Heritage, M.; Jaskowski, L.; Bridle, K.; Campbell, C.; Briskey, D.; Britton, L.; Fletcher, L.; Vitetta, L.; Subramaniam, V.N.; Crawford, D. Combination curcumin and vitamin e treatment attenuates diet-induced steatosis in hfe(-/-) mice. World J. Gastrointest. Pathophysiol. 2017, 8, 67–76. [Google Scholar] [CrossRef] [PubMed]
  70. Hou, H.T.; Qiu, Y.M.; Zhao, H.W.; Li, D.H.; Liu, Y.T.; Wang, Y.Z.; Su, S.H. [Effect of Curcumin on Intestinal Mucosal Mechanical Barrier in Rats with Non-Alcoholic Fatty Liver Disease]. Zhonghua Gan Zang Bing Za Zhi 2017, 25, 134–138. [Google Scholar] [PubMed]
  71. Inzaugarat, M.E.; De Matteo, E.; Baz, P.; Lucero, D.; Garcia, C.C.; Gonzalez Ballerga, E.; Daruich, J.; Sorda, J.A.; Wald, M.R.; Chernavsky, A.C. New evidence for the therapeutic potential of curcumin to treat nonalcoholic fatty liver disease in humans. PLoS ONE 2017, 12, e0172900. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, Y.; Cheng, F.; Luo, Y.; Zhan, Z.; Hu, P.; Ren, H.; Tang, H.; Peng, M. Pegylated curcumin derivative attenuates hepatic steatosis via creb/ppar-gamma/cd36 pathway. Biomed. Res. Int. 2017, 2017, 8234507. [Google Scholar] [CrossRef] [PubMed]
  73. Panahi, Y.; Kianpour, P.; Mohtashami, R.; Jafari, R.; Simental-Mendia, L.E.; Sahebkar, A. Efficacy and safety of phytosomal curcumin in non-alcoholic fatty liver disease: A randomized controlled trial. Drug Res. 2017, 67, 244–251. [Google Scholar] [CrossRef] [PubMed]
  74. Selmanovic, S.; Beganlic, A.; Salihefendic, N.; Ljuca, F.; Softic, A.; Smajic, E. Therapeutic effects of curcumin on ultrasonic morphological characteristics of liver in patients with metabolic syndrome. Acta Inf. Med. 2017, 25, 169–174. [Google Scholar] [CrossRef] [PubMed]
  75. Feng, W.W.; Kuang, S.Y.; Tu, C.; Ma, Z.J.; Pang, J.Y.; Wang, Y.H.; Zang, Q.C.; Liu, T.S.; Zhao, Y.L.; Xiao, X.H.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  76. Li, Y.Y.; Tang, D.; Du, Y.L.; Cao, C.Y.; Nie, Y.Q.; Cao, J.; Zhou, Y.J. Fatty liver mediated by ppar-alpha DNA methylation can be reversed by a methylation inhibitor and curcumin. J. Dig. Dis. 2018, 19, 421–430. [Google Scholar] [CrossRef] [PubMed]
  77. Yan, C.; Zhang, Y.; Zhang, X.; Aa, J.; Wang, G.; Xie, Y. Curcumin regulates endogenous and exogenous metabolism via Nrf2-FXR-LXR pathway in NAFLD mice. Biomed. Pharmacother. 2018, 105, 274–281. [Google Scholar] [CrossRef] [PubMed]
  78. 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. [Google Scholar] [CrossRef] [PubMed]
  79. Hwang, Y.P.; Kim, H.G.; Choi, J.H.; Do, M.T.; Chung, Y.C.; Jeong, T.C.; Jeong, H.G. S-allyl cysteine attenuates free fatty acid-induced lipogenesis in human hepg2 cells through activation of the amp-activated protein kinase-dependent pathway. J. Nutr. Biochem. 2013, 24, 1469–1478. [Google Scholar] [CrossRef] [PubMed]
  80. Xiao, J.; Ching, Y.P.; Liong, E.C.; Nanji, A.A.; Fung, M.L.; Tipoe, G.L. Garlic-derived s-allylmercaptocysteine is a hepato-protective agent in non-alcoholic fatty liver disease in vivo animal model. Eur. J. Nutr. 2013, 52, 179–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Xiao, J.; Guo, R.; Fung, M.L.; Liong, E.C.; Chang, R.C.; Ching, Y.P.; Tipoe, G.L. Garlic-derived s-allylmercaptocysteine ameliorates nonalcoholic fatty liver disease in a rat model through inhibition of apoptosis and enhancing autophagy. Evid. Based Complement. Altern. Med. 2013, 2013, 642920. [Google Scholar] [CrossRef] [PubMed]
  82. Lai, Y.S.; Chen, W.C.; Ho, C.T.; Lu, K.H.; Lin, S.H.; Tseng, H.C.; Lin, S.Y.; Sheen, L.Y. Garlic essential oil protects against obesity-triggered nonalcoholic fatty liver disease through modulation of lipid metabolism and oxidative stress. J. Agric. Food Chem. 2014, 62, 5897–5906. [Google Scholar] [CrossRef] [PubMed]
  83. Shin, J.H.; Lee, C.W.; Oh, S.J.; Yun, J.; Kang, M.R.; Han, S.B.; Park, H.; Jung, J.C.; Chung, Y.H.; Kang, J.S. Hepatoprotective effect of aged black garlic extract in rodents. Toxicol. Res. 2014, 30, 49–54. [Google Scholar] [CrossRef] [PubMed]
  84. Wu, Z.R.; Peng, C.; Yang, L.; Li, J.Y.; Xin, W.; Yong, W.; Guo, D.D.; Lei, C.; Guan, Q.G.; Li, H.Y. Two cinnamoyloctopamine antioxidants from garlic skin attenuates oxidative stress and liver pathology in rats with non-alcoholic steatohepatitis. Phytomedicine 2015, 22, 178–182. [Google Scholar] [CrossRef] [PubMed]
  85. Soleimani, D.; Paknahad, Z.; Askari, G.; Iraj, B.; Feizi, A. Effect of garlic powder consumption on body composition in patients with nonalcoholic fatty liver disease: A randomized, double-blind, placebo-controlled trial. Adv. Biomed. Res. 2016, 5, 2. [Google Scholar] [CrossRef] [PubMed]
  86. Kim, H.N.; Kang, S.G.; Roh, Y.K.; Choi, M.K.; Song, S.W. Efficacy and safety of fermented garlic extract on hepatic function in adults with elevated serum gamma-glutamyl transpeptidase levels: A double-blind, randomized, placebo-controlled trial. Eur. J. Nutr. 2017, 56, 1993–2002. [Google Scholar] [CrossRef] [PubMed]
  87. Li, S.; Liao, X.; Meng, F.; Wang, Y.; Sun, Z.; Guo, F.; Li, X.; Meng, M.; Li, Y.; Sun, C. 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. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, T.; Takikawa, Y.; Satoh, T.; Yoshioka, Y.; Kosaka, K.; Tatemichi, Y.; Suzuki, K. Carnosic acid prevents obesity and hepatic steatosis in ob/ob mice. Hepatol. Res. 2011, 41, 87–92. [Google Scholar] [CrossRef] [PubMed]
  89. Yin, J.; Luo, Y.; Deng, H.; Qin, S.; Tang, W.; Zeng, L.; Zhou, B. Hugan qingzhi medication ameliorates hepatic steatosis by activating ampk and pparalpha pathways in l02 cells and hepg2 cells. J. Ethnopharmacol. 2014, 154, 229–239. [Google Scholar] [CrossRef] [PubMed]
  90. 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. [Google Scholar] [CrossRef] [PubMed]
  91. Park, M.Y.; Sung, M.K. Carnosic acid attenuates obesity-induced glucose intolerance and hepatic fat accumulation by modulating genes of lipid metabolism in c57bl/6j-ob/ob mice. J. Sci. Food Agric. 2015, 95, 828–835. [Google Scholar] [CrossRef] [PubMed]
  92. Shan, W.; Gao, L.; Zeng, W.; Hu, Y.; Wang, G.; Li, M.; Zhou, J.; Ma, X.; Tian, X.; Yao, J. Activation of the sirt1/p66shc antiapoptosis pathway via carnosic acid-induced inhibition of mir-34a protects rats against nonalcoholic fatty liver disease. Cell Death Dis. 2015, 6, e1833. [Google Scholar] [CrossRef] [PubMed]
  93. Xu, T.; Zhou, J.; Zhu, J.; Zhang, S.; Zhang, N.; Zhao, Y.; Ding, C.; Shi, X.; Yao, J. Carnosic acid protects non-alcoholic fatty liver-induced dopaminergic neuron injury in rats. Metab. Brain Dis. 2017, 32, 483–491. [Google Scholar] [CrossRef] [PubMed]
  94. Song, H.M.; Li, X.; Liu, Y.Y.; Lu, W.P.; Cui, Z.H.; Zhou, L.; Yao, D.; Zhang, H.M. Carnosic acid protects mice from high-fat diet-induced NAFLD by regulating marcks. Int. J. Mol. Med. 2018, 42, 193–207. [Google Scholar] [CrossRef] [PubMed]
  95. Sahebkar, A. Potential efficacy of ginger as a natural supplement for nonalcoholic fatty liver disease. World J. Gastroenterol. 2011, 17, 271–272. [Google Scholar] [CrossRef] [PubMed]
  96. Lai, Y.S.; Lee, W.C.; Lin, Y.E.; Ho, C.T.; Lu, K.H.; Lin, S.H.; Panyod, S.; Chu, Y.L.; Sheen, L.Y. Ginger essential oil ameliorates hepatic injury and lipid accumulation in high fat diet-induced nonalcoholic fatty liver disease. J. Agric. Food Chem. 2016, 64, 2062–2071. [Google Scholar] [CrossRef] [PubMed]
  97. Rahimlou, M.; Yari, Z.; Hekmatdoost, A.; Alavian, S.M.; Keshavarz, S.A. Ginger supplementation in nonalcoholic fatty liver disease: A randomized, double-blind, placebo-controlled pilot study. Hepat. Mon. 2016, 16, e34897. [Google Scholar] [CrossRef] [PubMed]
  98. Teschke, R.; Xuan, T.D. Viewpoint: A Contributory Role of Shell Ginger (Alpinia Zerumbet (Pers.) B.L. Burtt & R.M. Sm) for Human Longevity in Okinawa, Japan? Nutrients 2018, 10. [Google Scholar] [CrossRef]
  99. Xuan, T.D.; Teschke, R. Dihydro-5,6-dehydrokavain (ddk) from alpinia zerumbet: Its isolation, synthesis, and characterization. Molecules 2015, 20, 16306–16319. [Google Scholar] [CrossRef] [PubMed]
  100. Janevski, M.; Antonas, K.N.; Sullivan-Gunn, M.J.; McGlynn, M.A.; Lewandowski, P.A. The effect of cocoa supplementation on hepatic steatosis, reactive oxygen species and LFABP in a rat model of NASH. Comp. Hepatol. 2011, 10, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Loffredo, L.; Del Ben, M.; Perri, L.; Carnevale, R.; Nocella, C.; Catasca, E.; Baratta, F.; Ceci, F.; Polimeni, L.; Gozzo, P.; et al. Effects of dark chocolate on nox-2-generated oxidative stress in patients with non-alcoholic steatohepatitis. Aliment. Pharmacol. Ther. 2016, 44, 279–286. [Google Scholar] [CrossRef] [PubMed]
  102. Askari, F.; Rashidkhani, B.; Hekmatdoost, A. Cinnamon may have therapeutic benefits on lipid profile, liver enzymes, insulin resistance, and high-sensitivity c-reactive protein in nonalcoholic fatty liver disease patients. Nutr. Res. 2014, 34, 143–148. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, S.D.; Xie, Z.Q.; Chen, J.; Wang, K.; Wei, T.; Zhao, A.H.; Zhang, Q.H. Inhibitory effect of ginkgo biloba extract on fatty liver: Regulation of carnitine palmitoyltransferase 1a and fatty acid metabolism. J. Dig. Dis. 2012, 13, 525–535. [Google Scholar] [CrossRef] [PubMed]
  104. Wei, T.; Xiong, F.F.; Wang, S.D.; Wang, K.; Zhang, Y.Y.; Zhang, Q.H. Flavonoid ingredients of ginkgo biloba leaf extract regulate lipid metabolism through sp1-mediated carnitine palmitoyltranferase 1a up-regulation. J. Biomed. Sci. 2014, 21, 87. [Google Scholar] [CrossRef] [PubMed]
  105. Li, H.Z.; Wang, J.H.; Niu, C.C.; Pan, S.H. [Intervention Effect and Mechanism of Compound Ginkgo Biloba Preparations on Nonalcoholic Fatty Liver]. Zhongguo Zhong Yao Za Zhi 2015, 40, 1580–1584. [Google Scholar] [PubMed]
  106. Yan, Z.; Fan, R.; Yin, S.; Zhao, X.; Liu, J.; Li, L.; Zhang, W.; Ge, L. Protective effects of ginkgo biloba leaf polysaccharide on nonalcoholic fatty liver disease and its mechanisms. Int. J. Biol. Macromol. 2015, 80, 573–580. [Google Scholar] [CrossRef] [PubMed]
  107. Yang, Q.; Zhao, H.; Zhou, A.Z.; Lou, Z.H. [Preventive and Therapeutic Effects of Compound Ginkgo Extract in Rats with Nonalcoholic Steatohepatitis Induced by High-Fat, High-Fructose Diet]. Zhonghua Gan Zang Bing Za Zhi 2016, 24, 852–858. [Google Scholar] [PubMed]
  108. Jeong, H.S.; Kim, K.H.; Lee, I.S.; Park, J.Y.; Kim, Y.; Kim, K.S.; Jang, H.J. Ginkgolide a ameliorates non-alcoholic fatty liver diseases on high fat diet mice. Biomed. Pharmacother. 2017, 88, 625–634. [Google Scholar] [CrossRef] [PubMed]
  109. Hong, S.H.; Suk, K.T.; Choi, S.H.; Lee, J.W.; Sung, H.T.; Kim, C.H.; Kim, E.J.; Kim, M.J.; Han, S.H.; Kim, M.Y.; et al. Anti-oxidant and natural killer cell activity of Korean red ginseng (panax ginseng) and urushiol (rhus vernicifera stokes) on non-alcoholic fatty liver disease of rat. Food Chem. Toxicol. 2013, 55, 586–591. [Google Scholar] [CrossRef] [PubMed]
  110. Hong, M.; Lee, Y.H.; Kim, S.; Suk, K.T.; Bang, C.S.; Yoon, J.H.; Baik, G.H.; Kim, D.J.; Kim, M.J. Anti-inflammatory and antifatigue effect of korean red ginseng in patients with nonalcoholic fatty liver disease. J. Ginseng. Res. 2016, 40, 203–210. [Google Scholar] [CrossRef] [PubMed]
  111. Chen, X.J.; Liu, W.J.; Wen, M.L.; Liang, H.; Wu, S.M.; Zhu, Y.Z.; Zhao, J.Y.; Dong, X.Q.; Li, M.G.; Bian, L.; et al. Ameliorative effects of compound k and ginsenoside rh1 on non-alcoholic fatty liver disease in rats. Sci. Rep. 2017, 7, 41144. [Google Scholar] [CrossRef] [PubMed]
  112. Huang, Q.; Wang, T.; Yang, L.; Wang, H.Y. Ginsenoside rb2 alleviates hepatic lipid accumulation by restoring autophagy via induction of Sirt1 and activation of AMPK. Int. J. Mol. Sci. 2017, 18. [Google Scholar] [CrossRef] [PubMed]
  113. Yuan, D.; Xiang, T.; Huo, Y.; Liu, C.; Wang, T.; Zhou, Z.; Dun, Y.; Zhao, H.; Zhang, C. Preventive effects of total saponins of Panax japonicus on fatty liver fibrosis in mice. Arch. Med. Sci. 2018, 14, 396–406. [Google Scholar] [CrossRef] [PubMed]
  114. Park, S.H.; Lee, S.G.; Kang, S.K.; Chung, S.H. Acanthopanax senticosus reverses fatty liver disease and hyperglycemia in ob/ob mice. Arch. Pharm. Res. 2006, 29, 768–776. [Google Scholar] [CrossRef] [PubMed]
  115. Tsuruta, Y.; Nagao, K.; Shirouchi, B.; Nomura, S.; Tsuge, K.; Koganemaru, K.; Yanagita, T. Effects of lotus root (the edible rhizome of Nelumbo nucifera) on the deveolopment of non-alcoholic fatty liver disease in obese diabetic db/db mice. Biosci. Biotechnol. Biochem. 2012, 76, 462–466. [Google Scholar] [CrossRef] [PubMed]
  116. Xiao, J.; Xing, F.; Huo, J.; Fung, M.L.; Liong, E.C.; Ching, Y.P.; Xu, A.; Chang, R.C.; So, K.F.; Tipoe, G.L. Lycium barbarum polysaccharides therapeutically improve hepatic functions in non-alcoholic steatohepatitis rats and cellular steatosis model. Sci. Rep. 2014, 4, 5587. [Google Scholar] [CrossRef] [PubMed]
  117. Lin, J.; Zhang, Y.; Wang, X.; Wang, W. Lycium ruthenicum extract alleviates high-fat diet-induced nonalcoholic fatty liver disease via enhancing the AMPK signaling pathway. Mol. Med. Rep. 2015, 12, 3835–3840. [Google Scholar] [CrossRef] [PubMed]
  118. Wu, Q.; Li, S.; Li, X.; Sui, Y.; Yang, Y.; Dong, L.; Xie, B.; Sun, Z. Inhibition of advanced glycation endproduct formation by lotus seedpod oligomeric procyanidins through rage-mapk signaling and NF-κB activation in high-fat-diet rats. J. Agric. Food Chem. 2015, 63, 6989–6998. [Google Scholar] [CrossRef] [PubMed]
  119. Zhang, D.D.; Zhang, J.G.; Wu, X.; Liu, Y.; Gu, S.Y.; Zhu, G.H.; Wang, Y.Z.; Liu, G.L.; Li, X.Y. Nuciferine downregulates per-arnt-sim kinase expression during its alleviation of lipogenesis and inflammation on oleic acid-induced hepatic steatosis in hepg2 cells. Front. Pharmacol. 2015, 6, 238. [Google Scholar] [CrossRef] [PubMed]
  120. Jia, L.; Li, W.; Li, J.; Li, Y.; Song, H.; Luan, Y.; Qi, H.; Ma, L.; Lu, X.; Yang, Y. Lycium barbarum polysaccharide attenuates high-fat diet-induced hepatic steatosis by up-regulating sirt1 expression and deacetylase activity. Sci. Rep. 2016, 6, 36209. [Google Scholar] [CrossRef] [PubMed]
  121. Kim, H.; Park, M.; Shin, J.H.; Kwon, O. Ethanolic extract of acanthopanax koreanum NAKAI alleviates alcoholic liver damage combined with a high-fat diet in c57bl/6j mice. Molecules 2016, 21. [Google Scholar] [CrossRef] [PubMed]
  122. Qian, W.; Hasegawa, J.; Cai, X.; Yang, J.; Ishihara, Y.; Ping, B.; Tsuno, S.; Endo, Y.; Matsuda, A.; Miura, N. Components of boiogito suppress the progression of hypercholesterolemia and fatty liver induced by high-cholesterol diet in rats. Yonago Acta Med. 2016, 59, 67–80. [Google Scholar] [PubMed]
  123. Yang, D.H.; Lou, Z.H.; Cheng, B.; Zhang, G.J.; Wang, Y.P.; Xu, H. [Effects of Lotus Leaf on Inflammatory Factors and Liver Adipor2 Expressions in Rats with NAFLD Induced by High Fat Diet and High Glucose]. Zhongguo Zhong Yao Za Zhi 2016, 41, 3406–3411. [Google Scholar] [PubMed]
  124. Duan, X.; Meng, Q.; Wang, C.; Liu, Z.; Liu, Q.; Sun, H.; Sun, P.; Yang, X.; Huo, X.; Peng, J.; et al. Calycosin attenuates triglyceride accumulation and hepatic fibrosis in murine model of non-alcoholic steatohepatitis via activating farnesoid x receptor. Phytomedicine 2017, 25, 83–92. [Google Scholar] [CrossRef] [PubMed]
  125. 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. [Google Scholar] [CrossRef] [PubMed]
  126. Duan, X.; Meng, Q.; Wang, C.; Liu, Z.; Sun, H.; Huo, X.; Sun, P.; Ma, X.; Peng, J.; Liu, K. Effects of calycosin against high-fat diet-induced nonalcoholic fatty liver disease in mice. J. Gastroenterol. Hepatol. 2018, 33, 533–542. [Google Scholar] [CrossRef] [PubMed]
  127. De Gottardi, A.; Spahr, L.; Ravier-Dall’Antonia, F.; Hadengue, A. Cannabinoid receptor 1 and 2 agonists increase lipid accumulation in hepatocytes. Liver Int. 2010, 30, 1482–1489. [Google Scholar] [CrossRef] [PubMed]
  128. Purohit, V.; Rapaka, R.; Shurtleff, D. Role of cannabinoids in the development of fatty liver (steatosis). AAPS J. 2010, 12, 233–237. [Google Scholar] [CrossRef] [PubMed]
  129. Yang, Y.Y.; Lin, H.C. Cross-regulation of cannabinoid cb(1) and cb(2) receptors governs hepatic steatosis. Liver Int. 2010, 30, 1397–1399. [Google Scholar] [CrossRef] [PubMed]
  130. Tarantino, G.; Citro, V.; Finelli, C. Recreational drugs: A new health hazard for patients with concomitant chronic liver diseases. J. Gastrointestin. Liver Dis. 2014, 23, 79–84. [Google Scholar] [PubMed]
  131. Silvestri, C.; Paris, D.; Martella, A.; Melck, D.; Guadagnino, I.; Cawthorne, M.; Motta, A.; Di Marzo, V. Two non-psychoactive cannabinoids reduce intracellular lipid levels and inhibit hepatosteatosis. J. Hepatol. 2015, 62, 1382–1390. [Google Scholar] [CrossRef] [PubMed]
  132. Adejumo, A.C.; Alliu, S.; Ajayi, T.O.; Adejumo, K.L.; Adegbala, O.M.; Onyeakusi, N.E.; Akinjero, A.M.; Durojaiye, M.; Bukong, T.N. Cannabis use is associated with reduced prevalence of non-alcoholic fatty liver disease: A cross-sectional study. PLoS ONE 2017, 12, e0176416. [Google Scholar] [CrossRef] [PubMed]
  133. Kim, D.; Kim, W.; Kwak, M.S.; Chung, G.E.; Yim, J.Y.; Ahmed, A. Inverse association of marijuana use with nonalcoholic fatty liver disease among adults in the united states. PLoS ONE 2017, 12, e0186702. [Google Scholar] [CrossRef] [PubMed]
  134. Dibba, P.; Li, A.A.; Cholankeril, G.; Iqbal, U.; Gadiparthi, C.; Khan, M.A.; Kim, D.; Ahmed, A. The role of cannabinoids in the setting of cirrhosis. Medicines 2018, 5. [Google Scholar] [CrossRef] [PubMed]
  135. Madak-Erdogan, Z.; Gong, P.; Zhao, Y.C.; Xu, L.; Wrobel, K.U.; Hartman, J.A.; Wang, M.; Cam, A.; Iwaniec, U.T.; Turner, R.T.; et al. Dietary licorice root supplementation reduces diet-induced weight gain, lipid deposition, and hepatic steatosis in ovariectomized mice without stimulating reproductive tissues and mammary gland. Mol. Nutr. Food Res. 2016, 60, 369–380. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, C.; Duan, X.; Sun, X.; Liu, Z.; Sun, P.; Yang, X.; Sun, H.; Liu, K.; Meng, Q. Protective effects of glycyrrhizic acid from edible botanical glycyrrhiza glabra against non-alcoholic steatohepatitis in mice. Food Funct. 2016, 7, 3716–3723. [Google Scholar] [CrossRef] [PubMed]
  137. Hajiaghamohammadi, A.A.; Ziaee, A.; Samimi, R. The efficacy of licorice root extract in decreasing transaminase activities in non-alcoholic fatty liver disease: A randomized controlled clinical trial. Phytother. Res. 2012, 26, 1381–1384. [Google Scholar] [CrossRef] [PubMed]
  138. Chen, T.; Zhong, F.J.; Hong, Y.M.; Su, W.J.; Zhuang, L.L.; Qiu, L.X. Effect of trifolium pratense extract on methionine-choline-deficient diet-induced steatohepatitis in c57bl/6 mice. Chin. J. Nat. Med. 2014, 12, 194–198. [Google Scholar] [CrossRef]
  139. Weidner, C.; Wowro, S.J.; Rousseau, M.; Freiwald, A.; Kodelja, V.; Abdel-Aziz, H.; Kelber, O.; Sauer, S. Antidiabetic effects of chamomile flowers extract in obese mice through transcriptional stimulation of nutrient sensors of the peroxisome proliferator-activated receptor (ppar) family. PLoS ONE 2013, 8, e80335. [Google Scholar] [CrossRef] [PubMed]
  140. Argo, C.K.; Patrie, J.T.; Lackner, C.; Henry, T.D.; de Lange, E.E.; Weltman, A.L.; Shah, N.L.; Al-Osaimi, A.M.; Pramoonjago, P.; Jayakumar, S.; et al. Effects of n-3 fish oil on metabolic and histological parameters in NASH: A double-blind, randomized, placebo-controlled trial. J. Hepatol. 2015, 62, 190–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Boyraz, M.; Pirgon, O.; Dundar, B.; Cekmez, F.; Hatipoglu, N. Long-term treatment with n-3 polyunsaturated fatty acids as a monotherapy in children with nonalcoholic fatty liver disease. J. Clin. Res. Pediatr. Endocrinol. 2015, 7, 121–127. [Google Scholar] [CrossRef] [PubMed]
  142. Gupta, V.; Mah, X.J.; Garcia, M.C.; Antonypillai, C.; van der Poorten, D. Oily fish, coffee and walnuts: Dietary treatment for nonalcoholic fatty liver disease. World J. Gastroenterol. 2015, 21, 10621–10635. [Google Scholar] [CrossRef] [PubMed]
  143. Qin, Y.; Zhou, Y.; Chen, S.H.; Zhao, X.L.; Ran, L.; Zeng, X.L.; Wu, Y.; Chen, J.L.; Kang, C.; Shu, F.R.; et al. Fish oil supplements lower serum lipids and glucose in correlation with a reduction in plasma fibroblast growth factor 21 and prostaglandin e2 in nonalcoholic fatty liver disease associated with hyperlipidemia: A randomized clinical trial. PLoS ONE 2015, 10, e0133496. [Google Scholar] [CrossRef] [PubMed]
  144. Adi, N.; Adi, J.; Lassance-Soares, R.M.; Kurlansky, P.; Yu, H.; Webster, K.A. High protein/fish oil diet prevents hepatic steatosis in noncnzo10 mice; association with diet/genetics-regulated micro-RNAs. J. Diabetes Metab. 2016, 7. [Google Scholar] [CrossRef]
  145. Della Corte, C.; Carpino, G.; De Vito, R.; De Stefanis, C.; Alisi, A.; Cianfarani, S.; Overi, D.; Mosca, A.; Stronati, L.; Cucchiara, S.; et al. Docosahexanoic acid plus vitamin d treatment improves features of NAFLD in children with serum vitamin d deficiency: Results from a single centre trial. PLoS ONE 2016, 11, e0168216. [Google Scholar] [CrossRef] [PubMed]
  146. He, X.X.; Wu, X.L.; Chen, R.P.; Chen, C.; Liu, X.G.; Wu, B.J.; Huang, Z.M. Effectiveness of omega-3 polyunsaturated fatty acids in non-alcoholic fatty liver disease: A meta-analysis of randomized controlled trials. PLoS ONE 2016, 11, e0162368. [Google Scholar] [CrossRef] [PubMed]
  147. Nogueira, M.A.; Oliveira, C.P.; Ferreira Alves, V.A.; Stefano, J.T.; Rodrigues, L.S.; Torrinhas, R.S.; Cogliati, B.; Barbeiro, H.; Carrilho, F.J.; Waitzberg, D.L. Omega-3 polyunsaturated fatty acids in treating non-alcoholic steatohepatitis: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2016, 35, 578–586. [Google Scholar] [CrossRef] [PubMed]
  148. Torrinhas, R.S.; Waitzberg, D.L. A double-blinded randomized placebo-controlled clinical trial of omega-3 fatty acids for the treatment of diabetic patients with nonalcoholic steatohepatitis. J. Clin. Gastroenterol. 2016, 50, 180. [Google Scholar] [CrossRef] [PubMed]
  149. Hodson, L.; Bhatia, L.; Scorletti, E.; Smith, D.E.; Jackson, N.C.; Shojaee-Moradie, F.; Umpleby, M.; Calder, P.C.; Byrne, C.D. Docosahexaenoic acid enrichment in NAFLD is associated with improvements in hepatic metabolism and hepatic insulin sensitivity: A pilot study. Eur. J. Clin. Nutr. 2017, 71, 973–979. [Google Scholar] [CrossRef] [PubMed]
  150. Liu, Y.; Shi, D.; Tian, Y.; Liu, Y.; Zhan, Q.; Xu, J.; Wang, J.; Xue, C. Eicosapentaenoic acid-enriched phosphatidylcholine attenuated hepatic steatosis through regulation of cholesterol metabolism in rats with nonalcoholic fatty liver disease. Lipids 2017, 52, 119–127. [Google Scholar] [CrossRef] [PubMed]
  151. Wang, H.; Shao, Y.; Yuan, F.; Feng, H.; Li, N.; Zhang, H.; Wu, C.; Liu, Z. Fish oil feeding modulates the expression of hepatic microRNAs in a western-style diet-induced nonalcoholic fatty liver disease rat model. Biomed. Res. Int. 2017, 2017, 2503847. [Google Scholar] [CrossRef] [PubMed]
  152. Yu, L.; Yuan, M.; Wang, L. The effect of omega-3 unsaturated fatty acids on non-alcoholic fatty liver disease: A systematic review and meta-analysis of rcts. Pak. J. Med. Sci. 2017, 33, 1022–1028. [Google Scholar] [CrossRef] [PubMed]
  153. Chen, L.H.; Wang, Y.F.; Xu, Q.H.; Chen, S.S. Omega-3 fatty acids as a treatment for non-alcoholic fatty liver disease in children: A systematic review and meta-analysis of randomized controlled trials. Clin. Nutr. 2018, 37, 516–521. [Google Scholar] [CrossRef] [PubMed]
  154. Bravo, E.; Palleschi, S.; Rossi, B.; Napolitano, M.; Tiano, L.; D’Amore, E.; Botham, K.M. Coenzyme q metabolism is disturbed in high fat diet-induced non-alcoholic fatty liver disease in rats. Int. J. Mol. Sci. 2012, 13, 1644–1657. [Google Scholar] [CrossRef] [PubMed]
  155. Farhangi, M.A.; Alipour, B.; Jafarvand, E.; Khoshbaten, M. Oral coenzyme q10 supplementation in patients with nonalcoholic fatty liver disease: Effects on serum vaspin, chemerin, pentraxin 3, insulin resistance and oxidative stress. Arch. Med. Res. 2014, 45, 589–595. [Google Scholar] [CrossRef] [PubMed]
  156. Saleh, D.O.; Ahmed, R.F.; Amin, M.M. Modulatory role of co-enzyme q10 on methionine and choline deficient diet-induced non-alcoholic steatohepatitis (NASH) in albino rats. Appl. Physiol. Nutr. Metab. 2017, 42, 243–249. [Google Scholar] [CrossRef] [PubMed]
  157. Farsi, F.; Mohammadshahi, M.; Alavinejad, P.; Rezazadeh, A.; Zarei, M.; Engali, K.A. Functions of coenzyme q10 supplementation on liver enzymes, markers of systemic inflammation, and adipokines in patients affected by nonalcoholic fatty liver disease: A double-blind, placebo-controlled, randomized clinical trial. J. Am. Coll. Nutr. 2016, 35, 346–353. [Google Scholar] [CrossRef] [PubMed]
  158. Awortwe, C.; Makiwane, M.; Reuter, H.; Muller, C.; Louw, J.; Rosenkranz, B. Critical evaluation of causality assessment of herb-drug interactions in patients. Br. J. Clin. Pharmacol. 2018, 84, 679–693. [Google Scholar] [CrossRef] [PubMed]
  159. Choi, S.; Oh, D.S.; Jerng, U.M. A systematic review of the pharmacokinetic and pharmacodynamic interactions of herbal medicine with warfarin. PLoS ONE 2017, 12, e0182794. [Google Scholar] [CrossRef] [PubMed]
  160. Leite, P.M.; de Freitas, A.A.; Mourao, A.O.M.; Martins, M.A.P.; Castilho, R.O. Warfarin safety: A cross-sectional study of the factors associated with the consumption of medicinal plants in a Brazilian anticoagulation clinic. Am. J. Cardiovasc. Drugs 2018, 18, 231–243. [Google Scholar] [CrossRef] [PubMed]
  161. Lim, J.W.; Chee, S.X.; Wong, W.J.; He, Q.L.; Lau, T.C. Traditional Chinese medicine: Herb-drug interactions with Aspirin. Singap. Med. J. 2018, 59, 230–239. [Google Scholar] [CrossRef] [PubMed]
  162. Alhusban, A.A.; Ata, S.A.; Shraim, S.A. The safety assessment of toxic metals in commonly used pharmaceutical herbal products and traditional herbs for infants in Jordanian market. Biol. Trace Elem. Res. 2018. [Google Scholar] [CrossRef] [PubMed]
  163. Shahbaz, O.; Mahajan, S.; Lewis, J.H. Highlights of drug- and herb- induced liver injury in the literature from 2016: How best to translate new information into clinical practice? Expert Opin. Drug Metab. Toxicol. 2017, 13, 935–951. [Google Scholar] [CrossRef] [PubMed]
  164. Teschke, R.; Andrade, R.J. Drug, herb, and dietary supplement hepatotoxicity. Int. J. Mol. Sci. 2016, 17. [Google Scholar] [CrossRef] [PubMed]
  165. Teschke, R.; Frenzel, C.; Schulze, J.; Eickhoff, A. Herbal hepatotoxicity: Challenges and pitfalls of causality assessment methods. World J. Gastroenterol. 2013, 19, 2864–2882. [Google Scholar] [CrossRef] [PubMed]
  166. Zhu, Y.; Niu, M.; Chen, J.; Zou, Z.S.; Ma, Z.J.; Liu, S.H.; Wang, R.L.; He, T.T.; Song, H.B.; Wang, Z.X.; et al. Hepatobiliary and pancreatic: Comparison between Chinese herbal medicine and western medicine-induced liver injury of 1985 patients. J. Gastroenterol. Hepatol. 2016, 31, 1476–1482. [Google Scholar] [CrossRef] [PubMed]
Table 1. The extent of research for each herb in the setting of nonalcoholic fatty liver disease (NAFLD) therapy.
Table 1. The extent of research for each herb in the setting of nonalcoholic fatty liver disease (NAFLD) therapy.
HerbLevel of Research with NAFLD ContextMechanism of Action
Milk thistle [6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25]Experimental and clinical studies (RCTs)Antioxidative
Anti-apoptotic
Anti-inflammatory
Anti-adipogenic
Resveratrol [26,27,28,29,30,31,32,33,34,35,36]Experimental and clinical studies (RCTs)Antioxidative
Anti-apoptotic
Anti-adipogenic
Coffee [37,38,39,40,41,42,43,44,45,46,47,48,49,50]Experimental and clinical studies (CSs, CCs)Antioxidative
Anti-apoptotic
Anti-inflammatory
Anti-fibrotic
Green tea [51,52,53,54,55,56,57,58,59,60,61,62,63]Experimental and clinical studies (RCTs)Antioxidative
Anti-inflammatory
Turmeric [64,65,66,67,68,69,70,71,72,73,74,75,76,77,78]Experimental and clinical studies (RCTs)Antioxidative
Anti-apoptotic
Anti-inflammatory
Anti-adipogenic
Garlic [79,80,81,82,83,84,85,86]Experimental and clinical studies (RCTs)Antioxidative
Anti-apoptotic
Anti-inflammatory
Ursolic acid (rosemary, peppermint, basil, lavender, and oregano) [87,89,90]Experimental studiesAnti-inflammatory
Anti-adipogenic
Carnosic acid (rosemary and sage) [88,91,92,93,94]Experimental studiesAntioxidative
Anti-apoptotic
Anti-inflammatory
Anti-adipogenic
Ginger [95,96,97]Experimental and clinical studies (RCTs)Antioxidative
Anti-apoptotic
Anti-inflammatory
Anti-adipogenic
Cocoa [100,101]Experimental studiesAntioxidative
Cinnamon [102]Clinical studies (RCTs)Antioxidative
Ginkgo biloba [103,104,105,106,107,108]Experimental studiesAntioxidative
Anti-apoptotic
Anti-inflammatory
Anti-adipogenic
Ginseng [52,109,110,111,112,113]Experimental and clinical studies (RCTs)Anti-inflammatory
Anti-adipogenic
Anti-fibrotic
Lotus [115,118,119,123]Experimental studiesAnti-inflammatory
Goji Berry [116,117,120,125]Experimental studiesAnti-inflammatory
Anti-adipogenic
Astragalus [122,124,126]Experimental studiesAnti-inflammatory
Anti-adipogenic
Ciruwuijia [114,121]Experimental studiesAnti-inflammatory
Cannabinoids [127,128,129,130,131,132,133,134]Experimental and clinical studies (CSs)Anti-fibrotic
Licorice [135,136,137]Experimental and clinical studies (RCTs)Anti-inflammatory
Red clover [138]Experimental studiesAnti-inflammatory
Chamomile [139]Experimental studiesAnti-inflammatory
Abbreviations: randomized control trial (RCT), cross-sectional study (CS), case–control study (CC).

Share and Cite

MDPI and ACS Style

Perumpail, B.J.; Li, A.A.; Iqbal, U.; Sallam, S.; Shah, N.D.; Kwong, W.; Cholankeril, G.; Kim, D.; Ahmed, A. Potential Therapeutic Benefits of Herbs and Supplements in Patients with NAFLD. Diseases 2018, 6, 80. https://doi.org/10.3390/diseases6030080

AMA Style

Perumpail BJ, Li AA, Iqbal U, Sallam S, Shah ND, Kwong W, Cholankeril G, Kim D, Ahmed A. Potential Therapeutic Benefits of Herbs and Supplements in Patients with NAFLD. Diseases. 2018; 6(3):80. https://doi.org/10.3390/diseases6030080

Chicago/Turabian Style

Perumpail, Brandon J., Andrew A. Li, Umair Iqbal, Sandy Sallam, Neha D. Shah, Waiyee Kwong, George Cholankeril, Donghee Kim, and Aijaz Ahmed. 2018. "Potential Therapeutic Benefits of Herbs and Supplements in Patients with NAFLD" Diseases 6, no. 3: 80. https://doi.org/10.3390/diseases6030080

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop