Next Article in Journal
Design and Synthesis of Novel Antioxidant 2-Substituted-5,7,8-Trimethyl-1,4-Benzoxazine Hybrids: Effects on Young and Senescent Fibroblasts
Previous Article in Journal
Exploitation of Natural By-Products for the Promotion of Healthy Outcomes in Humans: Special Focus on Antioxidant and Anti-Inflammatory Mechanisms and Modulation of the Gut Microbiota
Previous Article in Special Issue
Role of Mitochondrial Reactive Oxygen Species-Mediated Chaperone-Mediated Autophagy and Lipophagy in Baicalin and N-Acetylcysteine Mitigation of Cadmium-Induced Lipid Accumulation in Liver
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

The Role of Antioxidants in the Treatment of Metabolic Dysfunction-Associated Fatty Liver Disease: A Systematic Review

by
Kiana Mohammadian
1,
Fatemeh Fakhar
1,
Shayan Keramat
2,3 and
Agata Stanek
2,4,*
1
Division of Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz 71348, Iran
2
VAS-European Independent Foundation in Angiology/Vascular Medicine, Via GB Grassi 74, 20157 Milan, Italy
3
Support Association of Patients of Buerger’s Disease, Buerger’s Disease NGO, Mashhad 9183785195, Iran
4
Department and Clinic of Internal Medicine, Angiology, and Physical Medicine, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-902 Bytom, Poland
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(7), 797; https://doi.org/10.3390/antiox13070797
Submission received: 18 May 2024 / Revised: 19 June 2024 / Accepted: 27 June 2024 / Published: 29 June 2024

Abstract

:
Non-alcoholic fatty liver disease (NAFLD) is a global public health problem that causes liver-related morbidity and mortality. It is also an independent risk factor for non-communicable diseases. In 2020, a proposal was made to refer to it as “metabolic dysfunction-associated fatty liver disease (MAFLD)”, with concise diagnostic criteria. Given its widespread occurrence, its treatment is crucial. Increased levels of oxidative stress cause this disease. This review aims to evaluate various studies on antioxidant therapies for patients with MAFLD. A comprehensive search for relevant research was conducted on the PubMed, SCOPUS, and ScienceDirect databases, resulting in the identification of 87 studies that met the inclusion criteria. In total, 31.1% of human studies used natural antioxidants, 53.3% used synthetic antioxidants, and 15.5% used both natural and synthetic antioxidants. In human-based studies, natural antioxidants showed 100% efficacy in the treatment of MAFLD, while synthetic antioxidants showed effective results in only 91% of the investigations. In animal-based research, natural antioxidants were fully effective in the treatment of MAFLD, while synthetic antioxidants demonstrated effectiveness in only 87.8% of the evaluations. In conclusion, antioxidants in their natural form are more helpful for patients with MAFLD, and preserving the correct balance of pro-oxidants and antioxidants is a useful way to monitor antioxidant treatment.

Graphical Abstract

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) encompasses a range of liver conditions, from harmless non-alcoholic fatty liver (NAFL) to more severe non-alcoholic steatohepatitis (NASH) with or without fibrosis, NASH cirrhosis, and hepatocellular carcinoma (HCC) [1]. NAFLD is a significant public health concern, as it is a leading cause of liver-related morbidity and mortality globally, and it is also an independent risk factor for non-communicable diseases [2]. A combination of invasive and noninvasive tests is essential to diagnose NAFLD. The most comprehensive test for diagnosing and scoring fatty liver disease is a liver biopsy. The lesion at the most clinically benign end of the spectrum is fatty liver (hepatic steatosis). Both large (macro-) and microscopic (micro-) fat vesicles, mostly consisting of triglycerides, build up inside hepatocytes without significantly inducing scarring, liver cell death, or hepatic inflammation. The lesion at the other extreme end of the spectrum is known as cirrhosis. Hepatic steatosis frequently disappears by the time this level of architectural distortion manifests. Steatohepatitis is a type of liver damage that is characterized by hepatic steatosis and the development of restricted hepatic inflammation and hepatocyte death. Inflammatory infiltration is frequently evident in association with enlarged hepatocytes, which sometimes include Mallory’s hyalin. It consists of both mononuclear and polymorphonuclear leukocytes. These damage foci are primarily found in acinar zone 3 and are sometimes associated with bridging, perivenular, or perisinusoidal fibrosis [3]. The term proposed in 2020 to denote fatty liver disease associated with systemic metabolic dysregulation is “metabolic dysfunction-associated fatty liver disease (MAFLD).” The terminological transition from NAFLD to MAFLD was accompanied by a concise set of diagnostic criteria, facilitating convenient identification at the patient’s bedside for the broader medical community, including primary care providers [4].
Numerous interrelated mechanisms are involved in the pathophysiology of MAFLD. These processes include the infiltration of proinflammatory cells, which results in hepatic injury and ultimately leads to hepatic stellate cell (HSC) activation and fibrogenesis; lipotoxicity, which results from the accumulation of toxic lipid species; and insulin resistance (IR), which determines the metabolic syndrome. Although the proximal processes, such as inflammation, lipid excess, and lipotoxicity, have been extensively characterized, the downstream molecular mechanisms, including fibrogenesis, hepatocyte lipoapoptosis, and inflammatory processes, are not completely understood [5].
Liver diseases have a substantial impact on global health, with NAFLD being the most common worldwide, affecting 20–30% of the general population [6]. It affects 20–35% of adults, 15% of children, and up to 80% of individuals with obesity [7]. Cases increase markedly in patients with a history of type 2 diabetes mellitus (T2DM) and hyperlipidemia due to their connection to insulin resistance and metabolic dysfunction. However, NAFLD can affect individuals who have a normal weight and do not have metabolic problems, making up approximately 16% of cases [8]. Furthermore, NAFLD has led to an increase in death rates and liver transplants, particularly in the United States. Due to the fact that NAFLD is asymptomatic in its early stages, the actual burden of the disease can exceed the reported numbers [9]. The prevalence rates differ by region, ranging from 13.5% in Africa to 46% in America and an estimated 20–30% in Europe [2].
There is an opinion that a combination of several supportive treatments may be suitable for treatment. Due to the direct effect of body mass on NAFLD, weight loss treatment methods are used, the most important of which are changes in lifestyle through changes in eating habits and physical activity, as they reduce liver fat, and they should be used as the first line of treatment [10]. Over the past few decades, researchers have investigated several therapeutic agents that target different aspects of metabolic disorders, such as lipotoxicity, oxidative stress, mitochondrial dysfunction, and fibrosis, but some of these agents have been associated with disadvantages. However, the treatment used today includes antioxidants, such as vitamin E, and antidiabetic drugs, such as pioglitazone, and one of the treatments that has received a lot of attention in recent years is the targeting of intestinal bacteria [11,12].
Oxidative stress (OS) is characterized by an inconsistency between the generation of reactive species (RS) and antioxidant (AO) defenses [13]. A comprehensive description characterizes it as “an imbalance between oxidants and AOs, with oxidants having a greater advantage, resulting in a disturbance of redox signaling and regulation and/or molecular damage” [14]. OS is a crucial mechanism that contributes significantly to liver damage in MAFLD, playing a critical role in the transition from simple steatosis to NASH. Previous studies have obtained evidence showing that an increased production of reactive oxygen species (ROS) can trigger lipid peroxidation, resulting in inflammation and fibrogenesis via the activation of stellate cells [15]. Furthermore, ROS hinders the production of very-low-density lipoprotein (VLDL) by hepatocytes, leading to a buildup of fat in the liver. Additionally, ROS have the potential to induce hepatic insulin resistance and necroinflammation, as well as activate many intracellular pathways, which might ultimately result in hepatocyte apoptosis [16].
Different results have been obtained regarding the effect of antioxidants, which requires discussion. This systematic review aims to collect information on the impact of antioxidants in the treatment of MAFLD.

2. Materials and Methods

2.1. Literature Search

In this packccsystematic review, the recommendations stated in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. The systematic review protocol was registered in the PROSPERO database under the registration ID CRD42024534095, and a detailed exploration of the PubMed, SCOPUS, and ScienceDirect databases was performed. The primary sources of antioxidants are natural and dietary sources, as well as synthetic and medicinal supplements. Additionally, a search was performed using relevant keywords or title headings. Full details of the search strategy can be found in Table 1.

2.2. Study Selection

The inclusion and exclusion criteria for this review are described in Table 2. As an essential requirement for inclusion in the review, the research had to satisfy all established inclusion criteria. Publications were individually screened by two authors, KM and FF, to ensure compliance with the established inclusion criteria. Consequently, comprehensive reports were obtained for studies that indicated inconsistency or seemed to satisfy the inclusion criteria. Furthermore, disagreements were effectively resolved through an intensive evaluation process, resulting in the achievement of a consensus. On occasions when consensus could not be reached, the opinion of a third reviewer (SK) was sought.

3. Data Extraction and Quality Assessment

The reviewers autonomously extracted and documented data, including the author and publication year, population attributes such as sample size, antioxidant intervention (supplementation/dietary and medicinal forms), the duration of follow-up, post-intervention status, and quality control score determined by the two independent reviewers. The studies were evaluated using a study design and sampling method suitable for research. The sample size was sufficient, taking into account the prevalence of MAFLD. The evaluation of the results was performed using approved criteria. The outcomes were analyzed impartially, and the response rate was adequate. The statistics were reported with confidence intervals, and detailed descriptions of the study subjects were provided. Ultimately, the quality assessment determined that the selected studies met all the specified criteria and could be considered acceptable.

4. Results

4.1. Search Results

Figure 1 provides a concise overview of the procedure used to select pertinent studies. The search approach identified an overall number of 1015 articles. Furthermore, an additional 83 articles were found through an extensive review of the reference lists of pertinent reviews. Following the exclusion of duplicates, a total of 653 articles were selected based on their title and abstracts to determine their eligibility. A total of 445 articles were evaluated using their full texts, while 358 articles were removed due to factors such as untrustworthy study designs (including case reports, ethnographies, and observational designs), patient populations, interventions, outcome measurements, sample sizes smaller than 10, or the inaccessibility of the full text. As a result, a total of 87 articles were included in this review.

4.2. Study Characteristics

Of the 45 human studies, 14 (31.1%) used natural antioxidants, 24 (53.3%) used synthetic forms of antioxidants in dietary supplements and medications, and 7 used natural and synthetic antioxidants. Of the 24 studies that studied synthetic forms of antioxidants, 14 (58.8%) used dietary supplements, 4 (16.6%) used medications, and 6 (25%) used both dietary supplements and medications.
Among the human studies, 43 (95.5%) revealed a considerable effectiveness of antioxidant therapy, while 2 (4.4%) did not find significant differences from the placebo group. Of the 43 studies that had a significant effect, 14 examined natural antioxidants, 22 examined synthetic antioxidants, and 7 examined both natural and synthetic antioxidants. Furthermore, the two studies where no significant difference was seen examined synthetic antioxidants. Also, 30 studies examined both genders (66.6%), 8 studies (17.7) and 1 study (2.2%) included only males and females, respectively, and 6 studies did not mention the gender of cases. In addition, most of the studies, 38 of 45, have been designed as experimental, while there were 4 human studies, and 3 other studies used both experimental and human study design. The findings are summarized in Table 3.
Of the 42 animal studies, 10 (23.8%) used natural antioxidants, and 32 (76.1%) studies used synthetic forms of antioxidants in dietary supplements and medications. Of the 32 studies that studied synthetic forms of antioxidants, 7 (21.8%) used dietary supplements, and 25 (78.1%) studies used medications.
Among the animal studies, a total of 38 investigations, representing 90.4% of the studies, indicated considerable effectiveness of antioxidant therapy. On the contrary, in four studies (9.5%), no significant changes were detected between the antioxidant therapy group and the placebo group. Of the 38 studies that indicated significant effectiveness, 10 examined natural antioxidants, and 28 studies examined synthetic antioxidants. All four studies where no significant difference was seen examined synthetic antioxidants. The findings are summarized in Table 4.

5. Discussion

The present study was conducted as a systematic review to investigate the impact of antioxidant therapy on patients with MAFLD. This study investigated two types of antioxidants: natural antioxidants, found in fruits and vegetables, and synthetic antioxidants, such as dietary supplements or medications.
The analyzed studies indicate that natural antioxidants have a high level of efficacy in the treatment of MAFLD (100%). However, there is a scarcity of research using this method. The positive impact of synthetic antioxidants is not consistently observed, and only 91% of the studies demonstrate successful outcomes. On the basis of the available data, it may be inferred that natural antioxidants show greater efficacy than synthetic antioxidants in the treatment of MAFLD. Furthermore, fruits and vegetables are abundant in essential elements, as well as antioxidants. In contrast, previous studies have shown that the correct combination of minerals, such as sodium, potassium, selenium, magnesium, zinc, copper, and calcium, in conjunction with antioxidants, can improve the efficacy of antioxidants [104].
The impact of genetics on liver steatosis, inflammatory modifications, and fibrosis has been established by several studies. In genome-wide research, two genes have been associated with an increased risk of MAFLD: patatin-like phospholipase domain-containing 3 (PNPLA3) and trans-membrane 6 superfamily member 2 (TM6SF2) [9]. Lean MAFLD is a condition where individuals demonstrate a fatty liver but a normal body mass index (BMI). The main risk factors for this disease are visceral obesity, insulin resistance, high cholesterol, fructose intake, and specific genes. The triacylglycerol lipase enzyme, which is produced by the PNPLA3 gene, regulates lipid hydrolysis and assists in preserving balance between energy and its utilization in adipose tissue. Steatosis, inflammation, fibrosis, and hepatocellular carcinoma (HCC) can all be caused by a particular SNP in this gene. However, patients with MAFLD have a significantly decreased hepatic gene expression of TM6SF2, a gene essential for the generation of very-low-density lipoprotein (VLDL) [105]. An important gene, designated G-protein-coupled receptor 120 (GPR120), is present in hepatocytes, Kupffer cells, and adipocytes. It functions as a receptor for polyunsaturated fatty acids (PUFAs). Hepatocyte damage is the basis of abnormal liver function tests (LFTs) in individuals with the GPR120 270H mutation. Targeted by glitazone diabetes medications, PPARγ is frequently expressed in adipose tissue and influences both the arrival and departure of fatty acids to and from the liver, as well as the development of adipocytes. Liver steatosis is caused by these gene mutations [9].
Both in vitro and in vivo studies have shown that silybin or silibinin restores nicotinamide adenine dinucleotide+ (NAD+), a coenzyme essential for redox processes, by blocking poly (ADP-ribose) polymerase and triggering the SIRT1/AMP-activated protein kinase (AMPK) pathway. Reduced AMPK activity has been linked to the de novo lipogenesis process in MAFLD. Silybin’s anti-inflammatory action is achieved through the activity of SIRT2. Supplementing NAD+ with silybin has been found to be beneficial in preserving SIRT2 activity. Silybin has been shown to inhibit endoplasmic reticulum stress and the activation of the NLRP3 inflammasome in mice with fatty liver disease associated with metabolic dysfunction fed a high-fat diet [106].
Vitamin C has been reported to reduce mitochondrial ROS production, elevate levels of antioxidant enzymes such as superoxide dismutase and glutathione peroxidase, and enhance electron transport chain function in the liver. Vitamin C affects the balance of lipids and glucose and reduces visceral obesity and MAFLD by activating PPARα [9].
Multiple in vitro investigations on mouse and human adipocytes have shown that vitamin D has an anti-inflammatory impact by reducing the expression of chemokines and cytokines through the activation of p38 mitogen-activated protein (MAP) kinase and the NF-κB classical inflammatory pathway [9].
Vitamin E can boost antioxidant enzymes such as superoxide dismutase, catalase, and glutathione peroxidase; conversely, it reduces pro-oxidant contributions such as cellular myelocytomatosis (c-myc) and transforming growth factor-alpha (TGF-α), nitric oxide synthase, and NADPH. The antisteatotic activity of this substance is due to its capacity to decrease fatty acid uptake by hepatocytes via the downregulation of the hepatic cluster of differentiation 36 (CD36) protein, thereby limiting the amount of lipids available for peroxidation. Vitamin E reduces hepatic inflammation and fibrosis by downregulating the expression of pro-apoptotic BCL2 associated X (BAX), TGF-β, cyclooxygenase-2 (COX-2), and matrix metalloproteinase-2 (MMP-2) genes [1].
Plant flavonoids contain the naturally occurring flavonoid molecule quercetin. Quercetin’s antibacterial, anticancer, and antioxidant activities protect against free radicals, in addition to providing pharmacological advantages. One of the most abundant and substantial sources of quercetin is acacia rice. Numerous studies have demonstrated the effectiveness of quercetin, a flavonoid with potent antioxidant properties, in reducing lipid accumulation and the expression of SREBP1c and XBP-1 in adipocytes. A direct antilipogenic impact is produced by inhibiting the DNL pathway through its effects on the AMPK pathways [107].
Turmeric contains a polyphenolic compound known as curcumin, which has a number of medicinal effects, including anti-inflammatory, antiproliferative, antioxidant, and antiangiogenic effects. According to research, curcumin may prevent mice from developing MAFLD caused by high-fat and high-fructose diets. By regulating the LXR pathway, curcumin inhibits the expression of cytochrome P4503A (CYP3A) and cytochrome P4507A (CYP7A). Additionally, by inhibiting the FAS and Nrf2 pathways, it decreases the expression of CD36, SREBP1c, and the small heterodimer partner (SHP), which decreases hepatic steatosis. In an in vitro study, curcumin was found to reduce the development of liver fat by preventing citrate transport in the AMPK pathway, regulating the aberrant expression of SLC13A5/ACL. This prevents citrate from being carried or broken down. Curcumin inhibits the expression of SREBP1c and the suppressor of cytokine signaling 3 (SOCS-3), and it increases the phosphorylation of the hepatic activator of transcription 3 (STAT3), preventing liver steatosis. This assists in decreasing hepatic fat accumulation and regulating lipid metabolism [107]. A previous study demonstrated that, by enhancing the expression of the tight junction protein occludin 1, curcumin improved intestinal barrier function in MAFLD mice. The results of this study showed that it inhibited p65 nuclear translocation and NF-θB DNA-binding activity and that it decreased the expression of myeloid differentiation factor 88 (MyD88) in the liver, which, in turn, decreased hepatic steatosis [108]. MAFLD development is influenced by PPAR gene methylation. Curcumin significantly reduced methylation levels, elevated PPAR protein expression, and significantly reduced fat storage in MAFLD rats in other studies [109]. Curcumin protects LO2 cells from oleic acid-induced MAFLD by increasing the activity of certain proteins such as pAKT and P13K. Through Nrf2 signaling, it increases the absorption of glucose by liver cells and reduces NO and ROS levels. The initial phases of clinical trials have indicated that an excessive amount of 12 g/day of curcumin is safe for human intake. Because of its high metabolism and insufficient absorption, it has a low bioavailability. Currently, research is being conducted to increase the drug’s bioavailability and make it an unprecedented medication [107].
Resveratrol is a phenolic molecule that belongs to the stilbene family of phenols. It has a structure of C6-C2-C6 and is commonly found in plants such as cassia seeds, grape skins, and white tea. The substance comes primarily from the rhizome extract of Polygonum multiflorum. Resveratrol exhibits potent antioxidant, anticancer, and anti-inflammatory properties. Research has shown that resveratrol stimulates the sirtuin pathway (STRT1) for the treatment of MAFLD. Resveratrol reduces fat storage by activating the STRT1-FOXO1 pathway, which prevents SREBPE-1c acetylation and decreases metabolic abnormalities. A reduction in STRT1 levels in hepatocytes can lead to liver inflammation [110]. The stimulation of 3T3-L1 cells with TNF-ɑ leads to an increase in cytokine mRNA expression through the siRNA-mediated activation of SIRT1 [107]. Meanwhile, elevated SIRT1 levels suppress the generation of pro-inflammatory cytokines such as NF-ĸB and TNF-ɑ, thus protecting the liver’s metabolism from the harm caused by a high-fat diet. Research has shown that resveratrol can decrease apoptosis, mitochondrial dysfunction, and reactive oxygen species (ROS) production in OA-induced L02 cells. It can treat metabolic dysfunction-associated fatty liver disease (MAFLD) by decreasing caspase-3 and p53 and increasing B-cell lymphoma 2 (Bcl-2) levels, which helps to reduce liver fat accumulation [111]. The molecular mechanisms of non-alcoholic fatty liver disease (NAFLD) mediated by antioxidants in humans is summarized in Figure 2.
Furthermore, in 4.4% of the studies analyzed, there was no statistically significant impact on patient improvement when antioxidants were used. Furthermore, the reviewed studies also indicated that the efficacy of antioxidant therapy was not significantly influenced by the dosage or duration of treatment.
In the present systematic review, a study of animal research was also conducted. The reviewed animal studies showed that natural antioxidants are thoroughly effective in treating MAFLD (100%), but few studies used this method. This beneficial effect was not consistently seen with synthetic antioxidants, and only 87.8% of the studies showed effective results. On the basis of these data, it can be assumed that antioxidants may be more effective in their natural form than in a synthetic form in treating MAFLD.
Feeding a high-fat diet (HFD) to mice leads to the accumulation and fibrosis of liver collagen. Ascorbic acid supplementation decreases collagen levels and the mRNA expression of TGF-β and collagen. It also inhibits hepatocyte apoptosis and liver injury by increasing Bcl-2 protein levels and decreasing caspase 8. It effectively reduces liver inflammation in obese mice by suppressing the expression of inflammatory genes, including TNFα and MCP-1, in hepatocytes [112].
By controlling the proteins involved in lipid homeostasis, in which CES1 activation plays a crucial role, vitamin E in mice reduces the effects of a high-fat diet on liver lipid accumulation and glucose homeostasis. The biochemical mechanism responsible for the vitamin E-mediated overexpression of CES1 is, at least partially, the activation of nuclear Nrf2 [113].
Quercetin may help HFD-fed mice overcome MAFLD. An HFD causes fat peroxidation, ferroptosis, and fat accumulation; therapy clearly reverses these effects. For example, quercetin inhibits ferroptosis by increasing the expression of anti-glutathione peroxidase four (GPX4) and decreasing the expression of anticyclooxygenase 2 (COX2) and the long-chain family member acyl-coenzyme A synthase four (ACSL4) [114].
The administration of resveratrol to obese mice improves liver steatosis by improving glycolipid metabolic parameters, liver histology, inflammation, and lipid content. It could potentially have further positive effects by increasing T-SOD and GPX activities, inhibiting TNF-α production, suppressing TLR4 and CD36 expression [115], and improving steatohepatitis through the inhibition of the NF-jB inflammation pathway by further activating the AMPKa-SIRT1 pathway [116]. The molecular mechanisms of non-alcoholic fatty liver disease (NAFLD) mediated by antioxidants in animals is summarized in Figure 3.
In general, research examining the impact of antioxidants on the improvement of the condition of patients with MAFLD reveals contradictory results. On the contrary, the observed contradiction related to antioxidant properties in MAFLD therapy could also be attributed to the oxidative impact of antioxidants. Research has shown that, when the concentration of antioxidants in the human body is above the necessary threshold, oxidative effects are increased. In addition, an excess of antioxidants can contribute to elevated levels of oxidative stress [117]. Therefore, despite the positive therapeutic perspective of antioxidant supplements, it is essential to exercise caution and avoid their indiscriminate and/or excessive administration. The cellular intrinsic antioxidant system provides protection against damage caused by oxidants. Therefore, an assessment of the pro-oxidant–antioxidant balance can serve as an effective strategy for the management of antioxidant therapy. Measurement of oxidant and antioxidant capacity has been shown to facilitate the understanding of their balance. As a result, the administration of antioxidants may provide greater efficacy in patients when considering the balance or ratio between pro-oxidants and antioxidants [110,118].
The efficacy of antioxidants as an alternative therapy for MAFLD may be limited due to its multifactorial nature. The use of supplementary treatments based on the patient’s underlying disease, in conjunction with a suitable dose of antioxidants, appears to result in greater efficacy. The concept of combination therapy is interesting because it allows for the simultaneous targeting of multiple pathologic contributors to the disease. In the context of hepatic fibrosis, combination drugs can also be used to address the same target. In this context, these medications may have additive or synergistic effects on the target. Furthermore, the inclusion of a drug can potentially lead to a reduction in the dosage of other medication, thereby improving safety. It should be noted that the inclusion of a drug may reduce the negative consequences associated with an otherwise efficacious drug [119,120]. Furthermore, many fruits and vegetables, such as oranges, contain potassium and calcium, which have the potential to effectively manage blood pressure. On the basis of the evidence mentioned above, it can be inferred that the natural antioxidants present in fruits and vegetables may demonstrate greater effectiveness [104]. It is possible that medicinal supplements contain a combination of ingredients consisting of natural antioxidants that are effective. This study also highlights the absence of other comparable studies investigating the efficacy of antioxidants in preventing MAFLD in at-risk groups.

6. Current Limitations of Knowledge on the Effectiveness of Antioxidants in Treating MAFLD and Their Prospects

Although some evidence suggests the possible advantages of antioxidants for MAFLD, there are still certain issues that need to be clarified. First, because the vast majority of studies had an average duration of less than six months or 1 year, there has been an absence of substantial and outstanding scientific evidence indicating the long-term advantages of MAFLD antioxidant treatment. Second, it is currently questionable whether the general population and specific populations, such as men and women, the elderly, those with diabetes mellitus, children, and adolescents, should be prescribed antioxidant treatment. Third, how does long-term use of this antioxidant treatment affect people’s health? The essential concern is to assess who will benefit the most from an antioxidant treatment for MAFLD based on their personal characteristics, especially those with overlapping health conditions. Fourth, it is challenging to determine consistent findings because the included studies varied according to the characteristics of their study structure, patient demographics, antioxidant types, dosages, and treatment durations. Fifth, confounding variables can affect the results and make it challenging to differentiate between the impact of antioxidants. These variables include differences in the initial characteristics of the study participants, lifestyle factors, and concurrent treatments. Sixth, data analysis may be limited by the variations in the methods used by studies to assess and present outcomes associated with antioxidant levels, metabolic parameters, and liver function.

7. Conclusions

Antioxidants have been indicated to have similar effects on humans and animals. These effects include regulating the SIRT1/AMPK, NFKB, and LXR pathways; decreasing the expression of proapoptotic genes such as BAX, inflammatory genes such as TNF-ɑ, as well as TGF-β and COX2 genes; inhibiting the FAS and NrF2; and activating PPARα. Although natural antioxidants may be beneficial in treating MAFLD patients, combination therapy has been demonstrated to be more effective. However, more research is required to completely comprehend the effect of natural antioxidants. To improve the efficacy of MAFLD treatment, it is essential to take into account two crucial criteria. An effective approach to controlling treatment with antioxidants involves evaluating the appropriate balance between pro-oxidants and antioxidants. In essence, the administration of antioxidants at a suitable dose improves their efficacy, indicating the need for the development of customized therapeutic approaches. Furthermore, it is highly recommended to use the ideal dose of antioxidants in conjunction with other medications, such as antihypertensives, hypoglycemic agents, antidiabetic agents, and other cholesterol-balancing therapies, to effectively treat the underlying condition in patients with MAFLD.

Author Contributions

Conceptualization, S.K. and A.S.; methodology, S.K. and A.S.; formal analysis, K.M., F.F. and A.S.; writing—original draft preparation, K.M. and F.F.; writing—review and editing, S.K. and A.S.; visualization, K.M., F.F. and S.K.; supervision A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

We used PubMed, SCOPUS, and ScienceDirect databases to screen articles for this review. We did not report any data.

Conflicts of Interest

The authors declare no conflicts of interests.

References

  1. Usman, M.; Bakhtawar, N. Vitamin E as an adjuvant treatment for non-alcoholic fatty liver disease in adults: A systematic review of randomized controlled trials. Cureus 2020, 12, e9018. [Google Scholar] [CrossRef]
  2. Abenavoli, L.; Milanović, M.; Milić, N.; Luzza, F.; Giuffrè, A.M. Olive oil antioxidants and non-alcoholic fatty liver disease. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 739–749. [Google Scholar] [CrossRef]
  3. Clark, J.M.; Brancati, F.L.; Diehl, A.M. Nonalcoholic fatty liver disease. Gastroenterology 2002, 122, 1649–1657. [Google Scholar] [CrossRef]
  4. Gofton, C.; Upendran, Y.; Zheng, M.-H.; George, J. MAFLD: How is it different from NAFLD? Clin. Mol. Hepatol. 2023, 29, S17. [Google Scholar] [CrossRef]
  5. Kuchay, M.S.; Choudhary, N.S.; Mishra, S.K. Pathophysiological mechanisms underlying MAFLD. Diabetes Metab. Syndr. Clin. Res. Rev. 2020, 14, 1875–1887. [Google Scholar] [CrossRef]
  6. Abdel-Maboud, M.; Menshawy, A.; Menshawy, E.; Emara, A.; Alshandidy, M.; Eid, M. The efficacy of vitamin E in reducing non-alcoholic fatty liver disease: A systematic review, meta-analysis, and meta-regression. Ther. Adv. Gastroenterol. 2020, 13, 1756284820974917. [Google Scholar] [CrossRef]
  7. Coelho, J.M.; Cansanção, K.; Perez, R.M.; Leite, N.C.; Padilha, P.; Ramalho, A.; Peres, W. Association between serum and dietary antioxidant micronutrients and advanced liver fibrosis in non-alcoholic fatty liver disease: An observational study. PeerJ 2020, 8, e9838. [Google Scholar] [CrossRef]
  8. Cimini, F.A.; Barchetta, I.; Carotti, S.; Bertoccini, L.; Baroni, M.G.; Vespasiani-Gentilucci, U.; Cavallo, M.G.; Morini, S. Relationship between adipose tissue dysfunction, vitamin D deficiency and the pathogenesis of non-alcoholic fatty liver disease. World J. Gastroenterol. 2017, 23, 3407. [Google Scholar] [CrossRef]
  9. Abe, R.A.M.; Masroor, A.; Khorochkov, A.; Prieto, J.; Singh, K.B.; Nnadozie, M.C.; Abdal, M.; Shrestha, N.; Mohammed, L. The role of vitamins in non-alcoholic fatty liver disease: A systematic review. Cureus 2021, 13, e16855. [Google Scholar] [CrossRef]
  10. Romero-Gómez, M.; Zelber-Sagi, S.; Trenell, M. Treatment of NAFLD with diet, physical activity and exercise. J. Hepatol. 2017, 67, 829–846. [Google Scholar] [CrossRef]
  11. Nassir, F. NAFLD: Mechanisms, treatments, and biomarkers. Biomolecules 2022, 12, 824. [Google Scholar] [CrossRef]
  12. Rong, L.; Zou, J.; Ran, W.; Qi, X.; Chen, Y.; Cui, H.; Guo, J. Advancements in the treatment of non-alcoholic fatty liver disease (NAFLD). Front. Endocrinol. 2023, 13, 1087260. [Google Scholar] [CrossRef]
  13. Sies, H.; Berndt, C.; Jones, D. Oxidative stress. Annu. Rev. Biochem. 2017, 86, 715–748. [Google Scholar] [CrossRef]
  14. Ore, A.; Akinloye, O.A. Oxidative stress and antioxidant biomarkers in clinical and experimental models of non-alcoholic fatty liver disease. Medicina 2019, 55, 26. [Google Scholar] [CrossRef]
  15. Day, C. Pathogenesis of steatohepatitis. Best Pract. Res. Clin. Gastroenterol. 2002, 16, 663–678. [Google Scholar] [CrossRef]
  16. Polimeni, L.; Del Ben, M.; Baratta, F.; Perri, L.; Albanese, F.; Pastori, D.; Violi, F.; Angelico, F. Oxidative stress: New insights on the association of non-alcoholic fatty liver disease and atherosclerosis. World J. Hepatol. 2015, 7, 1325. [Google Scholar] [CrossRef]
  17. Mansour-Ghanaei, F.; Pourmasoumi, M.; Hadi, A.; Joukar, F. Efficacy of curcumin/turmeric on liver enzymes in patients with non-alcoholic fatty liver disease: A systematic review of randomized controlled trials. Integr. Med. Res. 2019, 8, 57–61. [Google Scholar] [CrossRef]
  18. Mosca, A.; Crudele, A.; Smeriglio, A.; Braghini, M.R.; Panera, N.; Comparcola, D.; Alterio, A.; Sartorelli, M.R.; Tozzi, G.; Raponi, M.; et al. The anti-inflammatory effects of hydroxytyrosol and vitamin e on paediatric NAFLD. Dig. Liver Dis. 2020, 52, e42–e43. [Google Scholar] [CrossRef]
  19. Khachidze, T. Comparison of the efficacy of Ursodeoxycholic acid (UDCA) versus vitamin E plus vitamin C in non-diabetic patients with nonalcoholic Steatohepatitis (NASH). HPB 2019, 21, S398–S399. [Google Scholar] [CrossRef]
  20. Anushiravani, A.; Haddadi, N.; Pourfarmanbar, M.; Mohammadkarimi, V. Treatment options for nonalcoholic fatty liver disease: A double-blinded randomized placebo-controlled trial. Eur. J. Gastroenterol. Hepatol. 2019, 31, 613–617. [Google Scholar] [CrossRef] [PubMed]
  21. Barchetta, I.; Cimini, F.A.; Cavallo, M.G. Vitamin D and metabolic dysfunction-associated fatty liver disease (MAFLD): An update. Nutrients 2020, 12, 3302. [Google Scholar] [CrossRef] [PubMed]
  22. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685. [Google Scholar] [CrossRef] [PubMed]
  23. Foster, T.; Budoff, M.J.; Saab, S.; Ahmadi, N.; Gordon, C.; Guerci, A.D. Atorvastatin and antioxidants for the treatment of nonalcoholic fatty liver disease: The St Francis Heart Study randomized clinical trial. Off. J. Am. Coll. Gastroenterol. ACG 2011, 106, 71–77. [Google Scholar] [CrossRef] [PubMed]
  24. Hoofnagle, J.H.; Van Natta, M.L.; Kleiner, D.E.; Clark, J.M.; Kowdley, K.V.; Loomba, R.; Neuschwander-Tetri, B.A.; Sanyal, A.J.; Tonascia, J. Non-alcoholic Steatohepatitis Clinical Research Network (NASH CRN). Vitamin E and changes in serum alanine aminotransferase levels in patients with non-alcoholic steatohepatitis. Aliment. Pharmacol. Ther. 2013, 38, 134–143. [Google Scholar] [CrossRef] [PubMed]
  25. Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S.; Polymerou, V.; Katsinelos, P. Effects of combined low-dose spironolactone plus vitamin E vs vitamin E monotherapy on insulin resistance, non-invasive indices of steatosis and fibrosis, and adipokine levels in non-alcoholic fatty liver disease: A randomized controlled trial. Diabetes Obes. Metab. 2017, 19, 1805–1809. [Google Scholar] [CrossRef] [PubMed]
  26. Pervez, M.A.; Khan, D.A.; Ijaz, A.; Khan, S. Effects of delta-tocotrienol supplementation on liver enzymes, inflammation, oxidative stress and hepatic steatosis in patients with nonalcoholic fatty liver disease. Turk. J. Gastroenterol. 2018, 29, 170. [Google Scholar] [CrossRef] [PubMed]
  27. Bril, F.; Biernacki, D.M.; Kalavalapalli, S.; Lomonaco, R.; Subbarayan, S.K.; Lai, J.; Tio, F.; Suman, A.; Orsak, B.K.; Hecht, J.; et al. Role of vitamin E for nonalcoholic steatohepatitis in patients with type 2 diabetes: A randomized controlled trial. Diabetes Care 2019, 42, 1481–1488. [Google Scholar] [CrossRef] [PubMed]
  28. 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]
  29. 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]
  30. 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]
  31. Heebøll, S.; Kreuzfeldt, M.; Hamilton-Dutoit, S.; Kjær Poulsen, M.; Stødkilde-Jørgensen, H.; Møller, H.J.; Jessen, N.; Thorsen, K.; Kristina Hellberg, Y.; Bønløkke 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]
  32. 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]
  33. Hussain, M.; 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. [Google Scholar] [CrossRef] [PubMed]
  34. Tabatabaee, S.M.; Alavian, S.M.; Ghalichi, L.; Miryounesi, S.M.; Mousavizadeh, K.; Jazayeri, S.; Vafa, M.R. Green tea in non-alcoholic fatty liver disease: A double blind randomized clinical trial. Hepat. Mon. 2017, 17, 6. [Google Scholar] [CrossRef]
  35. 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]
  36. Fukuzawa, Y.; Kapoor, M.P.; Yamasaki, K.; Okubo, T.; Hotta, Y.; Juneja, L.R. Effects of green tea catechins on nonalcoholic steatohepatitis (NASH) patients. J. Funct. Foods 2014, 9, 48–59. [Google Scholar] [CrossRef]
  37. Rezaei, S.; Akhlaghi, M.; Sasani, M.R.; Boldaji, R.B. Olive oil lessened fatty liver severity independent of cardiometabolic correction in patients with non-alcoholic fatty liver disease: A randomized clinical trial. Nutrition 2019, 57, 154–161. [Google Scholar] [CrossRef]
  38. Sofi, F.; Giangrandi, I.; Cesari, F.; Corsani, I.; Abbate, R.; Gensini, G.F.; Casini, A. Effects of a 1-year dietary intervention with n-3 polyunsaturated fatty acid-enriched olive oil on non-alcoholic fatty liver disease patients: A preliminary study. Int. J. Food Sci. Nutr. 2010, 61, 792–802. [Google Scholar] [CrossRef]
  39. Shidfar, F.; Bahrololumi, S.S.; Doaei, S.; Mohammadzadeh, A.; Gholamalizadeh, M.; Mohammadimanesh, A. The effects of extra virgin olive oil on alanine aminotransferase, aspartate aminotransferase, and ultrasonographic indices of hepatic steatosis in nonalcoholic fatty liver disease patients undergoing low calorie diet. Can. J. Gastroenterol. Hepatol. 2018, 2018, 1053710. [Google Scholar] [CrossRef]
  40. Nigam, P.; Bhatt, S.; Misra, A.; Chadha, D.S.; Vaidya, M.; Dasgupta, J.; Pasha, Q.M. Effect of a 6-month intervention with cooking oils containing a high concentration of monounsaturated fatty acids (olive and canola oils) compared with control oil in male Asian Indians with nonalcoholic fatty liver disease. Diabetes Technol. Ther. 2014, 16, 255–261. [Google Scholar] [CrossRef]
  41. Cueto-Galán, R.; Barón, F.J.; Valdivielso, P.; Pintó, X.; Corbella, E.; Gómez-Gracia, E.; Wärnberg, J.; los investigadores del Estudio PREDIMED. Changes in fatty liver index after consuming a Mediterranean diet: 6-year follow-up of the PREDIMED-Malaga trial. Med. Clínica (Engl. Ed.) 2017, 148, 435–443. [Google Scholar] [CrossRef]
  42. Kontogianni, M.D.; Tileli, N.; Margariti, A.; Georgoulis, M.; Deutsch, M.; Tiniakos, D.; Fragopoulou, E.; Zafiropoulou, R.; Manios, Y.; Papatheodoridis, G. Adherence to the Mediterranean diet is associated with the severity of non-alcoholic fatty liver disease. Clin. Nutr. 2014, 33, 678–683. [Google Scholar] [CrossRef] [PubMed]
  43. Ryan, M.C.; Itsiopoulos, C.; Thodis, T.; Ward, G.; Trost, N.; Hofferberth, S.; O’Dea, K.; Desmond, P.V.; Johnson, N.A.; Wilson, A.M. The Mediterranean diet improves hepatic steatosis and insulin sensitivity in individuals with non-alcoholic fatty liver disease. J. Hepatol. 2013, 59, 138–143. [Google Scholar] [CrossRef] [PubMed]
  44. Trovato, F.M.; Catalano, D.; Martines, G.F.; Pace, P.; Trovato, G.M. Mediterranean diet and non-alcoholic fatty liver disease: The need of extended and comprehensive interventions. Clin. Nutr. 2015, 34, 86–88. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, Z.F.; Fan, S.H.; Zheng, Y.L.; Lu, J.; Wu, D.M.; Shan, Q.; Hu, B. Troxerutin improves hepatic lipid homeostasis by restoring NAD+-depletion-mediated dysfunction of lipin 1 signaling in high-fat diet-treated mice. Biochem. Pharmacol. 2014, 91, 74–86. [Google Scholar] [CrossRef] [PubMed]
  46. Suda, I.; Ishikawa, F.; Hatakeyama, M.; Miyawaki, M.; Kudo, T.; Hirano, K.; Ito, A.; Yamakawa, O.; Horiuchi, S. Intake of purple sweet potato beverage affects on serum hepatic biomarker levels of healthy adult men with borderline hepatitis. Eur. J. Clin. Nutr. 2008, 62, 60–67. [Google Scholar] [CrossRef]
  47. 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]
  48. Stiuso, P.; Scognamiglio, I.; Murolo, M.; Ferranti, P.; De Simone, C.; Rizzo, M.R.; Tuccillo, C.; Caraglia, M.; Loguercio, C.; Federico, A. Serum oxidative stress markers and lipidomic profile to detect NASH patients responsive to an antioxidant treatment: A pilot study. Oxidative Med. Cell. Longev. 2014, 2014, 169216. [Google Scholar] [CrossRef]
  49. 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]
  50. Solhi, H.; Ghahremani, R.; Kazemifar, A.M.; Yazdi, Z.H. Silymarin in treatment of non-alcoholic steatohepatitis: A randomized clinical trial. Casp. J. Intern. Med. 2014, 5, 9. [Google Scholar]
  51. Soleimani, D.; Paknahad, Z.; Rouhani, M.H. Therapeutic effects of garlic on hepatic steatosis in nonalcoholic fatty liver disease patients: A randomized clinical trial. Diabetes Metab. Syndr. Obes. 2020, 13, 2389–2397. [Google Scholar] [CrossRef] [PubMed]
  52. Sangouni, A.A.; Azar, M.R.M.H.; Alizadeh, M. Effect of garlic powder supplementation on hepatic steatosis, liver enzymes and lipid profile in patients with non-alcoholic fatty liver disease: A double-blind randomised controlled clinical trial. Br. J. Nutr. 2020, 124, 450–456. [Google Scholar] [CrossRef] [PubMed]
  53. 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]
  54. Zhang, S.; Gu, Y.; Wang, L.; Zhang, Q.; Liu, L.; Lu, M.; Meng, G.; Yao, Z.; Wu, H.; Xia, Y.; et al. Association between dietary raw garlic intake and newly diagnosed nonalcoholic fatty liver disease: A population-based study. Eur. J. Endocrinol. 2019, 181, 591–602. [Google Scholar] [CrossRef] [PubMed]
  55. Emamat, H.; Farhadnejad, H.; Tangestani, H.; Saneei Totmaj, A.; Poustchi, H.; Hekmatdoost, A. Association of allium vegetables intake and non-alcoholic fatty liver disease risk: A case-control study. Nutr. Food Sci. 2020, 50, 1075–1083. [Google Scholar] [CrossRef]
  56. Loffredo, L.; Baratta, F.; Ludovica, P.; Battaglia, S.; Carnevale, R.; Nocella, C.; Novo, M.; Pannitteri, G.; Ceci, F.; Angelico, F.; et al. Effects of dark chocolate on endothelial function in patients with non-alcoholic steatohepatitis. Nutr. Metab. Cardiovasc. Dis. 2018, 28, 143–149. [Google Scholar] [CrossRef]
  57. Rafie, R.; Hosseini, S.A.; Hajiani, E.; Saki Malehi, A.; Mard, S.A. Effect of ginger powder supplementation in patients with non-alcoholic fatty liver disease: A randomized clinical trial. Clin. Exp. Gastroenterol. 2020, 13, 35–45. [Google Scholar] [CrossRef]
  58. Daneshi-Maskooni, M.; Keshavarz, S.A.; Qorbani, M.; Mansouri, S.; Alavian, S.M.; Badri-Fariman, M.; Jazayeri-Tehrani, S.A.; Sotoudeh, G. Green cardamom supplementation improves serum irisin, glucose indices, and lipid profiles in overweight or obese non-alcoholic fatty liver disease patients: A double-blind randomized placebo-controlled clinical trial. BMC Complement. Altern. Med. 2019, 19, 59. [Google Scholar] [CrossRef]
  59. 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]
  60. Anty, R.; Marjoux, S.; Iannelli, A.; Patouraux, S.; Schneck, A.S.; Bonnafous, S.; Gire, C.; Amzolini, A.; Ben-Amor, I.; Saint-Paul, M.C.; et al. Regular coffee but not espresso drinking is protective against fibrosis in a cohort mainly composed of morbidly obese European women with NAFLD undergoing bariatric surgery. J. Hepatol. 2012, 57, 1090–1096. [Google Scholar] [CrossRef]
  61. 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]
  62. 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. [Google Scholar] [PubMed]
  63. Mengesha, T.; Gnanasekaran, N.; Mehare, T. Hepatoprotective effect of silymarin on fructose induced nonalcoholic fatty liver disease in male albino wistar rats. BMC Complement. Med. Ther. 2021, 21, 104. [Google Scholar]
  64. Sun, R.; Xu, D.; Wei, Q.; Zhang, B.; Aa, J.; Wang, G.; Xie, Y. Silybin ameliorates hepatic lipid accumulation and modulates global metabolism in an NAFLD mouse model. Biomed. Pharmacother. 2020, 123, 109721. [Google Scholar] [CrossRef] [PubMed]
  65. Zhu, S.Y.; Jiang, N.; Yang, J.; Tu, J.; Zhou, Y.; Xiao, X.; Dong, Y. Silybum marianum oil attenuates hepatic steatosis and oxidative stress in high fat diet-fed mice. Biomed. Pharmacother. 2018, 100, 191–197. [Google Scholar] [CrossRef]
  66. Ou, Q.; Weng, Y.; Wang, S.; Zhao, Y.; Zhang, F.; Zhou, J.; Wu, X. Silybin alleviates hepatic steatosis and fibrosis in NASH mice by inhibiting oxidative stress and involvement with the Nf-κB pathway. Dig. Dis. Sci. 2018, 63, 3398–3408. [Google Scholar] [CrossRef] [PubMed]
  67. Ye, J.H.; Chao, J.; Chang, M.L.; Peng, W.H.; Cheng, H.Y.; Liao, J.W.; Pao, L.H. Pentoxifylline ameliorates non-alcoholic fatty liver disease in hyperglycaemic and dyslipidaemic mice by upregulating fatty acid β-oxidation. Sci. Rep. 2016, 6, 33102. [Google Scholar] [CrossRef]
  68. Massart, J.; Robin, M.A.; Noury, F.; Fautrel, A.; Lettéron, P.; Bado, A.; Eliat, P.A.; Fromenty, B. Pentoxifylline aggravates fatty liver in obese and diabetic ob/ob mice by increasing intestinal glucose absorption and activating hepatic lipogenesis. Br. J. Pharmacol. 2012, 165, 1361–1374. [Google Scholar] [CrossRef]
  69. Saeed, A.; Bartuzi, P.; Heegsma, J.; Dekker, D.; Kloosterhuis, N.; de Bruin, A.; Jonker, J.W.; van de Sluis, B.; Faber, K.N. Impaired hepatic vitamin A metabolism in NAFLD mice leading to vitamin A accumulation in hepatocytes. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 309–325.e3. [Google Scholar] [CrossRef]
  70. Ipsen, D.H.; Skat-Rørdam, J.; Svenningsen, M.; Andersen, M.; Latta, M.; Buelund, L.E.; Lintrup, K.; Skaarup, R.; Lykkesfeldt, J.; Tveden-Nyborg, P. The effect of acetylsalicylic acid and pentoxifylline in guinea pigs with non-alcoholic steatohepatitis. Basic Clin. Pharmacol. Toxicol. 2021, 128, 583–593. [Google Scholar] [CrossRef]
  71. Oliveira, C.P.; Gayotto, L.C.; Tatai, C.; Della Nina, B.I.; Lima, E.S.; Abdalla, D.S.; Lopasso, F.P.; Laurindo, F.R.; Carrilho, F.J. Vitamin C and vitamin E in prevention of nonalcoholic fatty liver disease (NAFLD) in choline deficient diet fed rats. Nutr. J. 2003, 2, 9. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, C.W.; Huang, C.C.; Hsu, C.F.; Li, T.H.; Tsai, Y.L.; Lin, M.W.; Tsai, H.C.; Huang, S.F.; Yang, Y.Y.; Hsieh, Y.C.; et al. SIRT1-dependent mechanisms and effects of resveratrol for amelioration of muscle wasting in NASH mice. BMJ Open Gastroenterol. 2020, 7, e000381. [Google Scholar] [CrossRef] [PubMed]
  73. Karimian, G.; Kirschbaum, M.; Veldhuis, Z.J.; Bomfati, F.; Porte, R.J.; Lisman, T. Vitamin E attenuates the progression of non-alcoholic fatty liver disease caused by partial hepatectomy in mice. PLoS ONE 2015, 10, e0143121. [Google Scholar] [CrossRef]
  74. Presa, N.; Clugston, R.D.; Lingrell, S.; Kelly, S.E.; Merrill, A.H., Jr.; Jana, S.; Kassiri, Z.; Gómez-Muñoz, A.; Vance, D.E.; Jacobs, R.L.; et al. Vitamin E alleviates non-alcoholic fatty liver disease in phosphatidylethanolamine N-methyltransferase deficient mice. Biochim. Et Biophys. Acta (BBA)-Mol. Basis Dis. 2019, 1865, 14–25. [Google Scholar] [CrossRef] [PubMed]
  75. Lee, S.W.; Baek, S.M.; Kang, K.K.; Lee, A.R.; Kim, T.U.; Choi, S.K.; Roh, Y.S.; Hong, I.H.; Park, S.J.; Kim, T.H.; et al. Vitamin C deficiency inhibits nonalcoholic fatty liver disease progression through impaired de novo lipogenesis. Am. J. Pathol. 2021, 191, 1550–1563. [Google Scholar] [CrossRef] [PubMed]
  76. Zeng, Q.; Zhao, L.; Meng, C.; Zhao, X.; Liu, Y.; Shi, R.; Han, X.; Wang, T.; Li, J. Prophylactic and therapeutic effects of different doses of vitamin C on high-fat-diet-induced non-alcoholic fatty liver disease in mice. Biomed. Pharmacother. 2020, 131, 110792. [Google Scholar] [CrossRef] [PubMed]
  77. Milton-Laskibar, I.; Marcos-Zambrano, L.J.; Gómez-Zorita, S.; Fernández-Quintela, A.; Carrillo de Santa Pau, E.; Martínez, J.A.; Portillo, M.P. Gut microbiota induced by pterostilbene and resveratrol in high-fat-high-fructose fed rats: Putative role in steatohepatitis onset. Nutrients 2021, 13, 1738. [Google Scholar] [CrossRef]
  78. Sun, M.; Gu, Y.; Glisan, S.L.; Lambert, J.D. Dietary cocoa ameliorates non-alcoholic fatty liver disease and increases markers of antioxidant response and mitochondrial biogenesis in high fat-fed mice. J. Nutr. Biochem. 2021, 92, 108618. [Google Scholar] [CrossRef]
  79. Reda, D.; Elshopakey, G.E.; Risha, E.F.; El-Boshy, M.E.; Abdelhamid, F.M. Vitamin D3 alleviates nonalcoholic fatty liver disease in rats by inhibiting hepatic oxidative stress and inflammation via the SREBP-1-c/PPARα-NF-κB/IR-S2 signaling pathway. Front. Pharmacol. 2023, 14, 1164512. [Google Scholar] [CrossRef]
  80. SEIFEL-DINSH; Sabra, A.-N.A.; Hammam, O.A.; Ebeid, F.A.; El-Lakkany, N.M. Pharmacological and antioxidant actions of garlic and/or onion in non-alcoholic fatty liver disease (NAFLD) in rats. J. Egypt. Soc. Parasitol. 2014, 44, 295–308. [Google Scholar]
  81. Wang, J.-H.; Hwang, S.-J.; Lim, D.-W.; Son, C.-G. Cynanchum atratum alleviates non-alcoholic fatty liver by balancing lipogenesis and fatty acid oxidation in a high-fat, high-fructose diet mice model. Cells 2021, 11, 23. [Google Scholar] [CrossRef]
  82. 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]
  83. Wang, Q.; Ou, Y.; Hu, G.; Wen, C.; Yue, S.; Chen, C.; Xu, L.; Xie, J.; Dai, H.; Xiao, H.; et al. Naringenin attenuates non-alcoholic fatty liver disease by down-regulating the NLRP3/NF-κB pathway in mice. Br. J. Pharmacol. 2020, 177, 1806–1821. [Google Scholar] [CrossRef] [PubMed]
  84. Zhou, J.; Yu, Y.; Ding, L.; Xu, P.; Wang, Y. Matcha green tea alleviates non-alcoholic fatty liver disease in high-fat diet-induced obese mice by regulating lipid metabolism and inflammatory responses. Nutrients 2021, 13, 1950. [Google Scholar] [CrossRef] [PubMed]
  85. Chao, J.; Huo, T.I.; Cheng, H.Y.; Tsai, J.C.; Liao, J.W.; Lee, M.S.; Qin, X.M.; Hsieh, M.T.; Pao, L.H.; Peng, W.H. Gallic acid ameliorated impaired glucose and lipid homeostasis in high fat diet-induced NAFLD mice. PLoS ONE 2014, 9, e96969. [Google Scholar] [CrossRef] [PubMed]
  86. Gao, X.; Zhao, X.; Liu, M.; Zhao, H.; Sun, Y. Lycopene prevents non-alcoholic fatty liver disease through regulating hepatic NF-κB/NLRP3 inflammasome pathway and intestinal microbiota in mice fed with high-fat and high-fructose diet. Front. Nutr. 2023, 10, 1120254. [Google Scholar] [CrossRef] [PubMed]
  87. Gu, M.; Zhao, P.; Zhang, S.; Fan, S.; Yang, L.; Tong, Q.; Ji, G.; Huang, C. 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. [Google Scholar] [CrossRef] [PubMed]
  88. Luo, D.; Yang, L.; Pang, H.; Zhao, Y.; Li, K.; Rong, X.; Guo, J. Tianhuang formula reduces the oxidative stress response of NAFLD by regulating the gut microbiome in mice. Front. Microbiol. 2022, 13, 984019. [Google Scholar] [CrossRef]
  89. Acedo, S.C.; Gotardo, É.M.F.; Pereira, J.A.; Pedrazzoli, J.; Ribeiro, M.L.; Gambero, A. Role of pentoxifylline in non-alcoholic fatty liver disease in high-fat diet-induced obesity in mice. World J. Hepatol. 2015, 7, 2551. [Google Scholar] [CrossRef]
  90. Panchal, S.K.; Poudyal, H.; Brown, L. Quercetin ameliorates cardiovascular, hepatic, and metabolic changes in diet-induced metabolic syndrome in rats. J. Nutr. 2012, 142, 1026–1032. [Google Scholar] [CrossRef]
  91. Marcolin, E.; San-Miguel, B.; Vallejo, D.; Tieppo, J.; Marroni, N.; González-Gallego, J.; Tuñón, M.J. Quercetin Treatment Ameliorates Inflammation and Fibrosis in Mice with Nonalcoholic Steatohepatitis, 3. J. Nutr. 2012, 142, 1821–1828. [Google Scholar] [CrossRef] [PubMed]
  92. Panchal, S.K.; Poudyal, H.; Arumugam, T.V.; 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. [Google Scholar] [CrossRef] [PubMed]
  93. Suguro, R.; Pang, X.-C.; Yuan, Z.-W.; Chen, S.-Y.; Zhu, Y.-Z.; Xie, Y. Combinational applicaton of silybin and tangeretin attenuates the progression of non-alcoholic steatohepatitis (NASH) in mice via modulating lipid metabolism. Pharmacol. Res. 2020, 151, 104519. [Google Scholar] [CrossRef] [PubMed]
  94. Kuzu, N.; Bahcecioglu, I.H.; Dagli, A.F.; Ozercan, I.H.; Ustündag, B.; Sahin, K. Epigallocatechin gallate attenuates experimental non-alcoholic steatohepatitis induced by high fat diet. J. Gastroenterol. Hepatol. 2008, 23 Pt 2, e465–e470. [Google Scholar] [CrossRef] [PubMed]
  95. Mao, Q.Q.; Li, B.Y.; Meng, J.M.; Gan, R.Y.; Xu, X.Y.; Gu, Y.Y.; Wang, X.H.; Li, H.B. Effects of several tea extracts on nonalcoholic fatty liver disease in mice fed with a high-fat diet. Food Sci. Nutr. 2021, 9, 2954–2967. [Google Scholar] [CrossRef] [PubMed]
  96. Zhou, D.D.; Mao, Q.Q.; Li, B.Y.; Saimaiti, A.; Huang, S.Y.; Xiong, R.G.; Shang, A.; Luo, M.; Li, H.Y.; Gan, R.Y.; et al. Effects of different green teas on obesity and non-alcoholic fatty liver disease induced by a high-fat diet in mice. Front. Nutr. 2022, 9, 929210. [Google Scholar] [CrossRef]
  97. Liu, B.; Zhang, J.; Sun, P.; Yi, R.; Han, X.; Zhao, X. Raw bowl tea (Tuocha) polyphenol prevention of nonalcoholic fatty liver disease by regulating intestinal function in mice. Biomolecules 2019, 9, 435. [Google Scholar] [CrossRef] [PubMed]
  98. Li, H.Y.; Huang, S.Y.; Zhou, D.D.; Xiong, R.G.; Luo, M.; Saimaiti, A.; Han, M.K.; Gan, R.Y.; Zhu, H.L.; Li, H.B. Theabrownin inhibits obesity and non-alcoholic fatty liver disease in mice via serotonin-related signaling pathways and gut-liver axis. J. Adv. Res. 2023, 52, 59–72. [Google Scholar] [CrossRef]
  99. Li, B.; Mao, Q.; Xiong, R.; Zhou, D.; Huang, S.; Saimaiti, A.; Shang, A.; Luo, M.; Li, H.; Li, H.; et al. Preventive effects of different black and dark teas on obesity and non-alcoholic fatty liver disease and modulate gut microbiota in high-fat diet fed mice. Foods 2022, 11, 3457. [Google Scholar] [CrossRef]
  100. Torres, L.; Cogliati, B.; Otton, R. Green tea prevents NAFLD by modulation of miR-34a and miR-194 expression in a high-fat diet mouse model. Oxidative Med. Cell. Longev. 2019, 2019, 4168380. [Google Scholar] [CrossRef]
  101. Noori, M.; Jafari, B.; Hekmatdoost, A. Pomegranate juice prevents development of non-alcoholic fatty liver disease in rats by attenuating oxidative stress and inflammation. J. Sci. Food Agric. 2017, 97, 2327–2332. [Google Scholar] [CrossRef] [PubMed]
  102. El-Lakkany, N.M.; El-Din, S.H.S.; Sabra, A.-N.A.-A.; Hammam, O.A.; Ebeid, F.A.-L. Co-administration of metformin and N-acetylcysteine with dietary control improves the biochemical and histological manifestations in rats with non-alcoholic fatty liver. Res. Pharm. Sci. 2016, 11, 374–382. [Google Scholar] [CrossRef] [PubMed]
  103. Seif El-Din, S.H.; El-Lakkany, N.M.; El-Naggar, A.A.; Hammam, O.A.; Abd El-Latif, H.A.; Ain-Shoka, A.A.; Ebeid, F.A. Effects of rosuvastatin and/or β-carotene on non-alcoholic fatty liver in rats. Res. Pharm. Sci. 2015, 10, 275–287. [Google Scholar] [PubMed]
  104. Chiu, H.-F.; Venkatakrishnan, K.; Golovinskaia, O.; Wang, C.-K. Impact of micronutrients on hypertension: Evidence from clinical trials with a special focus on meta-analysis. Nutrients 2021, 13, 588. [Google Scholar] [CrossRef] [PubMed]
  105. Kong, L.; Lu, Y.; Zhang, S.; Nan, Y.; Qiao, L. Role of nutrition, gene polymorphism, and gut microbiota in non-alcoholic fatty liver disease. Discov. Med. 2017, 24, 95–106. [Google Scholar] [PubMed]
  106. Ezhilarasan, D.; Lakshmi, T. A molecular insight into the role of antioxidants in nonalcoholic fatty liver diseases. Oxidative Med. Cell. Longev. 2022, 2022, 9233650. [Google Scholar] [CrossRef] [PubMed]
  107. Fang, X.; Song, J.; Zhou, K.; Zi, X.; Sun, B.; Bao, H.; Li, L. Molecular mechanism pathways of natural compounds for the treatment of non-alcoholic fatty liver disease. Molecules 2023, 28, 5645. [Google Scholar] [CrossRef] [PubMed]
  108. Feng, D.; Zou, J.; Su, D.; Mai, H.; Zhang, S.; Li, P.; Zheng, X. Curcumin prevents high-fat diet-induced hepatic steatosis in ApoE−/− mice by improving intestinal barrier function and reducing endotoxin and liver TLR4/NF-κB inflammation. Nutr. Metab. 2019, 16, 79. [Google Scholar] [CrossRef]
  109. Li, Y.Y.; Tang, D.; Du, Y.L.; Cao, C.Y.; Nie, Y.Q.; Cao, J.; Zhou, Y.J. Fatty liver mediated by peroxisome proliferator-activated receptor-α DNA methylation can be reversed by a methylation inhibitor and curcumin. J. Dig. Dis. 2018, 19, 421–430. [Google Scholar] [CrossRef]
  110. Purushotham, A.; Schug, T.T.; Xu, Q.; Surapureddi, S.; Guo, X.; Li, X. Hepatocyte-specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation. Cell Metab. 2009, 9, 327–338. [Google Scholar] [CrossRef]
  111. Yuan, W.; Zhang, M.; Wang, C.; Li, B.; Li, L.; Ye, F.; Xu, C. Resveratrol Attenuates High-Fat Diet–Induced Hepatic Lipotoxicity by Upregulating Bmi-1 Expression. J. Pharmacol. Exp. Ther. 2022, 381, 96–105. [Google Scholar] [CrossRef] [PubMed]
  112. Geng, C.; Xu, H.; Zhang, Y.; Gao, Y.; Li, M.; Liu, X.; Gao, M.; Wang, X.; Liu, X.; Fang, F.; et al. Retinoic acid ameliorates high-fat diet-induced liver steatosis through sirt1. Sci. China Life Sci. 2017, 60, 1234–1241. [Google Scholar] [CrossRef] [PubMed]
  113. He, W.; Xu, Y.; Ren, X.; Xiang, D.; Lei, K.; Zhang, C.; Liu, D. Vitamin E ameliorates lipid metabolism in mice with nonalcoholic fatty liver disease via Nrf2/CES1 signaling pathway. Dig. Dis. Sci. 2019, 64, 3182–3191. [Google Scholar] [CrossRef] [PubMed]
  114. Jiang, J.-J.; Zhang, G.-F.; Zheng, J.-Y.; Sun, J.-H.; Ding, S.-B. Targeting mitochondrial ROS-mediated ferroptosis by quercetin alleviates high-fat diet-induced hepatic lipotoxicity. Front. Pharmacol. 2022, 13, 876550. [Google Scholar] [CrossRef] [PubMed]
  115. Cheng, K.; Song, Z.; Zhang, H.; Li, S.; Wang, C.; Zhang, L.; Wang, T. The therapeutic effects of resveratrol on hepatic steatosis in high-fat diet-induced obese mice by improving oxidative stress, inflammation and lipid-related gene transcriptional expression. Med. Mol. Morphol. 2019, 52, 187–197. [Google Scholar] [CrossRef] [PubMed]
  116. Tian, Y.; Ma, J.; Wang, W.; Zhang, L.; Xu, J.; Wang, K.; Li, D. Resveratrol supplement inhibited the NF-κB inflammation pathway through activating AMPKα-SIRT1 pathway in mice with fatty liver. Mol. Cell. Biochem. 2016, 422, 75–84. [Google Scholar] [CrossRef] [PubMed]
  117. Alamdari, D.H.; Paletas, K.; Pegiou, T.; Sarigianni, M.; Befani, C.; Koliakos, G. A novel assay for the evaluation of the prooxidant–antioxidant balance, before and after antioxidant vitamin administration in type II diabetes patients. Clin. Biochem. 2007, 40, 248–254. [Google Scholar] [CrossRef]
  118. Keramat, S.; Sharebiani, H.; Patel, M.; Fazeli, B.; Stanek, A. The Potential Role of Antioxidants in the Treatment of Peripheral Arterial Disease: A Systematic Review. Antioxidants 2022, 11, 2126. [Google Scholar] [CrossRef]
  119. Makri, E.S.; Makri, E.; Polyzos, S.A. Combination therapies for nonalcoholic fatty liver disease. J. Pers. Med. 2022, 12, 1166. [Google Scholar] [CrossRef]
  120. Sharebiani, H.; Keramat, S.; Chavoshan, A.; Fazeli, B.; Stanek, A. The influence of antioxidants on oxidative stress-induced vascular aging in obesity. Antioxidants 2023, 12, 1295. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram showing the study selection and identification. A total of 1015 publications were identified, with 535 articles in the human studies category and 480 articles in the animal studies category. By looking through the reference lists of pertinent reviews, an additional 83 publications were found, 38 of which were animal studies and 48 of which were human studies. After duplicates were eliminated, 653 articles were chosen for eligibility according to their title and abstract. Of these, 445 articles underwent full-text evaluations; 358 articles were omitted due to faulty data. As a result, in this review, 87 publications were included, with 45 being human studies and 42 being animal studies.
Figure 1. PRISMA flow diagram showing the study selection and identification. A total of 1015 publications were identified, with 535 articles in the human studies category and 480 articles in the animal studies category. By looking through the reference lists of pertinent reviews, an additional 83 publications were found, 38 of which were animal studies and 48 of which were human studies. After duplicates were eliminated, 653 articles were chosen for eligibility according to their title and abstract. Of these, 445 articles underwent full-text evaluations; 358 articles were omitted due to faulty data. As a result, in this review, 87 publications were included, with 45 being human studies and 42 being animal studies.
Antioxidants 13 00797 g001
Figure 2. The molecular mechanisms underlying the regression of non-alcoholic fatty liver disease (NAFLD) mediated by antioxidants in humans. Different antioxidants can control the expression of different genes through distinct molecular pathways, leading to the recovery and regression of this disease, according to several human studies. For example, the disease is improved through the AMPK, LXR, FAS, and NRF2 pathways by altering the expression of the SIRT1, P4507A, P4503A, and SREBP1C genes, respectively. MAFLD—metabolic dysfunction-associated fatty liver disease; AMPK—AMP-activated protein kinase; LXR—liver X receptors; FAS—FS-7-associated surface; NRF2—nuclear factor erythroid 2-related factor 2; SIRT1—sirtuin 1; P4503A—cytochrome P450 3A; GPR120-G-protein-coupled receptor 120; TM6SF2-trans-membrane 6 superfamily member 2; PNPLA3-patatin-like phospholipase domain-containing 3; PPARα-Peroxisome proliferator-activated receptor α; P4507A—cytochrome P450 7A; SREBP1C—sterol regulatory element binding protein 1c.
Figure 2. The molecular mechanisms underlying the regression of non-alcoholic fatty liver disease (NAFLD) mediated by antioxidants in humans. Different antioxidants can control the expression of different genes through distinct molecular pathways, leading to the recovery and regression of this disease, according to several human studies. For example, the disease is improved through the AMPK, LXR, FAS, and NRF2 pathways by altering the expression of the SIRT1, P4507A, P4503A, and SREBP1C genes, respectively. MAFLD—metabolic dysfunction-associated fatty liver disease; AMPK—AMP-activated protein kinase; LXR—liver X receptors; FAS—FS-7-associated surface; NRF2—nuclear factor erythroid 2-related factor 2; SIRT1—sirtuin 1; P4503A—cytochrome P450 3A; GPR120-G-protein-coupled receptor 120; TM6SF2-trans-membrane 6 superfamily member 2; PNPLA3-patatin-like phospholipase domain-containing 3; PPARα-Peroxisome proliferator-activated receptor α; P4507A—cytochrome P450 7A; SREBP1C—sterol regulatory element binding protein 1c.
Antioxidants 13 00797 g002
Figure 3. The molecular mechanisms underlying the regression of non-alcoholic fatty liver disease (NAFLD) mediated by antioxidants in animals. In animal studies, antioxidants have been shown to improve this disease by affecting the expression of various genes in hepatocytes through molecular pathways, for example, by reducing the expression of inflammatory genes such as TNFα and MCP-1, as well as reducing the expression of TGF-βmRNA or the overexpression of CES1. MAFLD—metabolic dysfunction-associated fatty liver disease; TNF-α—tumor necrosis factor alpha; MCP-1—monocyte chemoattractant protein-1; TGF-β—transforming growth factor beta; CES1—carboxylesterase 1.
Figure 3. The molecular mechanisms underlying the regression of non-alcoholic fatty liver disease (NAFLD) mediated by antioxidants in animals. In animal studies, antioxidants have been shown to improve this disease by affecting the expression of various genes in hepatocytes through molecular pathways, for example, by reducing the expression of inflammatory genes such as TNFα and MCP-1, as well as reducing the expression of TGF-βmRNA or the overexpression of CES1. MAFLD—metabolic dysfunction-associated fatty liver disease; TNF-α—tumor necrosis factor alpha; MCP-1—monocyte chemoattractant protein-1; TGF-β—transforming growth factor beta; CES1—carboxylesterase 1.
Antioxidants 13 00797 g003
Table 1. Full details of the search strategy terms.
Table 1. Full details of the search strategy terms.
Terms 1Search Strategy Terms
Term 1“Antioxidants” OR “Antioxidant treatments” “Antioxidant therapy”
Term 2“Antioxidant drugs” OR “vitamin C” OR “Ascorbic acid” OR “Vitamin A” OR “Vitamin D” OR “Vitamin E” OR “Resveratrol” OR “Tocopherol” OR “Pentoxifylline” OR “Silymarin” OR “Green Tea” OR “Chocolate” OR “Garlic” OR “Ginger” OR “Red Wine”
Term 3Consumption OR “Dietary intake” OR Supplement OR Supplementation OR “Nutritional supplement” OR “high-fat diet” OR “chow”
Term 4“Non-alcoholic fatty liver disease” OR “Metabolic dysfunction-associated fatty liver disease” OR “NAFLD” OR “MAFLD”
Term 5“Clinical Trials” OR “Animal Study” OR “Mice” OR “Rat” OR “Rabbit” OR “Guinea pig”
1 Terms 1, 2, 3, and 4 were joined with “AND”.
Table 2. Inclusion and exclusion criteria.
Table 2. Inclusion and exclusion criteria.
Inclusion CriteriaExclusion Criteria
Human studiesHuman studies
Studies that include men (males) and women (females)
Studies that are randomized controlled trialsStudies that include participants at risk of MAFLD, including otherwise healthy smokers and hypertensive or diabetic patients
Studies with a sample size of 10 participantsStudies that administer mixed nutrient supplementation where no group receives any specific antioxidant supplement alone
Studies that include participants who either are healthy or have
established MAFLD
Studies that incorporate dual treatments such as exercise and supplementation
Studies in which there are no data relating to the dose of natural antioxidants
Studies in which there is no control group
Studies that orally administer a single antioxidant intervention
through supplementation or dietary or drug interventions
Animal studiesAnimal studies
Studies that include males and females
Studies that include participants in whom the diet induced MAFLDStudies that apply other treatments along with antioxidants
Studies that provide an oral or injectable antioxidant interventionStudies that administer mixed nutrient supplementation where no group receives any specific antioxidant supplement alone
Studies that administer a single antioxidant or a combination of antioxidants through supplementation or dietary intervention
Table 3. The summarized results of human studies that fulfilled the inclusion criteria.
Table 3. The summarized results of human studies that fulfilled the inclusion criteria.
StudyYearType of StudySample SizeAgeGenderInterventionForm of Intervention
(Natural or Synthetic)
Dose/DayDurationControlStatus after Intervention
Mansour-Ghanaei et al. [17]2019Experimental228Mean age: 50Not
mentioned
CurcuminNatural: curcumin80 to 3000 mg/dayRanged between 8 and 12 weeksThere were
no controls
in this study
Increased dosages of curcumin/turmeric may be beneficial for NAFLD.
Mosca et al. [18]2020Experimental80 Adolescents (age
range: 4–16 years)
Not
mentioned
Group 1: vitamin E and hydroxytyrosol
Group 2: placebo
Synthetic: vitamin ENot
mentioned
4 months PlaceboHydroxytyrosol and vitamin E significantly decreased IL-6, which, in turn, decreased systemic inflammation. The combination increased the expression of IL-10; this can hinder the release of cytokines, which increase inflammation.
Khachidze
et al. [19]
2019Both human and experimental107Not
Mentioned
Not
mentioned
Group 1: vitamin E
Group 2: lifestyle
modification
Group 3: ursodeoxycholic acid (UDCA)
Synthetic: vitamin EGroup 1: 400 IU once daily
Group 2: __ Group 3: 15 mg/kg once daily
12 monthsPlaceboCombining vitamins E and C is a safe, affordable, and effective therapeutic option for patients with NASH. This may help minimize oxidative stress-related damage and prevent the progression of cirrhosis.
Anushiravani
et al. [20]
2019Experimental150Aged between
18 and 65 years
Both gendersGroup 1: vitamin E
Group 2: placebo
Group 3: metformin
Group 4: silymarin
Group 5: pioglitazone
Synthetic: vitamin E and metformin and silymarin and pioglitazoneGroup 1: vitamin E 400 IU once daily
Group 2: placebo
Group 3: metformin 500 mg once daily
Group 4: silymarin 140 mg once daily
Group 5: pioglitazone 15 mg once daily
3 monthsPlaceboIn NAFLD patients, vitamin E, silymarin, and pioglitazone were shown to improve liver aminotransferases.
Barchetta et al. [21]2011Experimental5557.4 ± 10.7Females = 20
Males = 35
Vitamin D (cholecalciferol)Synthetic: vitamin D2000 IU/day24 weeksNot
mentioned
Clinical studies did not demonstrate that vitamin D treatment has a beneficial effect on liver impairment markers in MAFLD patients.
Sanyal et al. [22]2010Experimentaln = 247 Group 1 (pioglitazone): 80 Group 2 (vitamin E): 84 Group 3
(placebo): 83
46.3Females: 60%
Males: 40%
Group 1: pioglitazone Group 2: vitamin E Synthetic: vitamin E and
pioglitazone
Group 1: 30 mg once daily Group 2: 800 IU once daily2.5 yearsGroup 3: placeboVitamin E was more effective than placebo in treating non-alcoholic steatohepatitis in persons without diabetes. Pioglitazone did not provide any advantages over placebo in the main outcome, although it did show substantial benefits for some secondary outcomes.
Foster et
et al. [23]
2011Experimentaln = 455 Group 1 (atorvastatin, vitamin C, and vitamin E): 229 Group 2
(placebo): 226
59Females: 29.1%
Males: 70.9%
Atorvastatin, vitamin C, and vitamin E Synthetic: vitamin C, vitamin E, and
atorvastatin
Atorvastatin 20 mg, vitamin C 1 g, and vitamin E 1000 IU once daily4 yearsPlaceboThe combination of atorvastatin 20 mg with vitamins C and E was found to effectively reduce the likelihood of hepatic steatosis by 71% in healthy adults with non-alcoholic fatty liver disease (NAFLD) at baseline.
Hoofnagle et
al. [24]
2013Experimental13945.5Females: 57.5%
Males: 42.5%
Group 1: pioglitazone 30 mg and vitamin E placebo
Group 2: vitamin E 80 once daily and pioglitazone placebo
Synthetic: vitamin E and
pioglitazone
Group 1: pioglitazone 30 mg and vitamin E placebo
Group 2: vitamin E once daily and pioglitazone placebo
2.5 yearsPlaceboPatients with NASH showed a reduction in ALT levels, as well as an improvement in histological activity.
Polyzos et al. [25]2017Experimentaln = 31 Group 1 (vitamin E alone): 17
Group 2 (spironolactone plus vitamin
E): 12
Study
included
individuals
≥18 years)
Females:
74.2% Males:
25.8%
Group 1: vitamin E
Group 2: spironolactone
plus vitamin E
Synthetic: vitamin E and
spironolactone
Group 1: vitamin E 400 IU twice a day Group 2: spironolactone 25 mg once
a day plus vitamin E 400 IU twice a day
52
months
The combination of low-dose spironolactone and vitamin E resulted in a considerable reduction in the NAFLD liver fat score.
Pervez et al. [26]2018Experimentaln = 64 Group 1 (δ-tocotrienol): 31 Group 2 (placebo): 3344.3Females:
54.7% Males:
45.3%
δ-tocotrienol Synthetic: δ-tocotrienolGroup 1: δ-tocotrienol 300 mg twice daily 3
months
PlaceboThe administration of δ-tocotrienol substantially increased AST activity and reduced inflammatory and oxidative stress markers in individuals diagnosed with NAFLD.
Bril et al. [27]2019 n = 105 Group 1 (vitamin E plus placebo): 36
Group 2 (vitamin E plus
pioglitazone): 37 Group 3 (both placebo): 32
59Female: 11.4%
Males: 88.6%
Group 1: vitamin E plus placebo
Group 2: vitamin E
plus pioglitazone
Synthetic: vitamin E and
pioglitazone
Group 1: vitamin E 400 IU twice a day plus placebo
Group 2: vitamin E 400
IU twice a day plus pioglitazone 30 mg/day increased after 2 months to 45
mg/day
18
months
PlaceboCombination therapy provided better outcomes than a placebo in enhancing liver histology in patients with NASH and T2DM. The administration of vitamin E alone did not have any significant effect on the primary histological outcome.
Asghari et al. [28]2018Both human and experimental6039.53Males: 58.33%Pure trans- resveratrolSynthetic: capsules,
pure
trans-resveratrol
600 mg\day84 daysPlaceboA calorie-restricted (CR) diet resulting in moderate weight loss had beneficial effects on NAFLD. Furthermore, weight loss induced by resveratrol supplementation also had positive effects. However, it should be noted that these interventions did not replicate all the benefits of a CR diet.
Chachay et al. [29]2014Experimental2048.15Males: 100%ResveratrolSynthetic: capsules3000 mg\day56 daysPlaceboThe administration of resveratrol did not result in significant improvements in any of the characteristics of NAFLD when compared to a placebo. However, it did result in an increase in hepatic stress, demonstrated by an observed increase in the levels of liver enzymes.
Chen et al. [30]2015Experimental6044.30Males: 70%ResveratrolSynthetic: capsules
(from
natural
products)
600 mg\day90 daysPlaceboResveratrol reduced levels of ASPAT, ALAT, LDL-cholesterol, total cholesterol, TNF-α, cytokeratin fragment 18, and fibroblast growth factor 21, as well as the IR index, in patients with NAFLD. This suggests that resveratrol supplementation may be beneficial for individuals with NAFLD.
Heebøll et al. [31]2016Experimental2843.2Males: 65.38Pure
trans-resveratrol
Synthetic: capsules,
pure
trans-resveratrol
1500 mg\day180 daysPlaceboThe administration of resveratrol did not consistently improve the clinical or histological symptoms of NAFLD. However, there may have been a slight improvement in liver function tests and a reduction in the accumulation of liver fat.
Pezeshki et al. [32]2016Experimental7135Both gendersGreen
tea
Synthetic: green tea extract (GTE) tablets500 mg/day84 daysPlacebo
(cellulose)
In NAFLD patients, GTE supplementation decreased liver enzyme levels. It is possible to suggest that administering GTE to NAFLD patients can help them achieve better blood liver enzyme levels.
Hussain et al. [33]2017Experimental8026.5Both gendersGreen
tea
Synthetic: capsule (GTE) 1000 mg/day84 daysPlacebo
(cellulose)
The metabolic, chemical, inflammatory, and radiological characteristics of patients with NAFLD and dyslipidemia but without diabetes significantly improved as a result of GTE therapy.
Tabatabaee et al. [34]2017Experimental4540Both gendersGreen
tea
Synthetic: green tea tablets550 mg/day90 daysPlaceboThe two groups mainly differed in anthropometrics, liver enzyme levels, and metabolic markers; however, the results of the routine therapies that both groups received may have concealed some of the differences.
Sakata et al. [35]2013Experimental1755.9Both gendersGreen
tea
Synthetic: green
Tea
Group 1: 1080 mg/
700 mL catechins
Group 2: mg/700 mL
catechins
84 daysPlacebo (green
tea-flavored
beverage)
The consumption of a high-catechin tea reduced blood biochemistry, liver inflammation, and liver fat in addition to an oxidative stress marker.
Fukuzawa et al. [36]2014Experimental3850Both gendersGreen
tea
Synthetic: green
tea
Dietary and exercise
therapy + 600 mg/
day catechins
180 daysDietary and
exercise
therapy
Green tea catechins have the capability to inhibit NASH by efficiently improving NASH-related parameters such as insulin resistance recovery and anti-inflammatory activities.
Rezaei et al. [37]2019Experimental66Age ≥ 18 yearsNot
mentioned
Olive oilNatural: olive oil20 g per day12 weeksSunflower oilIn NAFLD patients, olive oil decreased body fat percentage and attenuated fatty liver grade; it had no effect on liver enzymes or cardiometabolic risk factors.
Sofi et al. [38]2010Both human and experimental11Age > 18 yearsMales: four
Females: two
Olive oil with n-3 PUFASynthetic: olive oil with n-3 PUFA6.5 mL per day12 monthsDietary recommendations and a similar package of olive oil not enriched with n-3 PUFAPatients with NAFLD may significantly increase their adiponectin levels and reduce their circulating triglycerides and liver enzymes via the long-term use of olive oil enriched with n-3 PUFA.
Shidfar et al. [39]2018Experimental5045.91 ± 9.61 19 women and 31 menOlive oilNatural: olive oilA hypocaloric diet enriched
with olive oil (20% of the total energy intake)
12 weeksA hypocaloric diet with normal fatA normal fat percentage of 30% in a diet containing olive oil (with the consumption of 20% of total calories from virgin olive oil) combined with a slight decrease in weight (about 5%) supported the intended effects of weight loss in increasing the levels of the enzymes ALT and AST.
Nigam et al. [40]2014Experimental93Age from 20 to 50 yearsMales: 100%Olive oil, canola oil, and soybeanNatural: olive oil, canola oil, and soybeanCooking medium (not exceeding 20 g/day)6 monthsSafflower oil The severity of fatty liver and liver span in NAFLD significantly reduced when olive and canola oils—which are high in MUFAs and have a balanced n-6/n-3 PUFA ratio—were used as a cooking medium. Insulin resistance and dyslipidemia both improved along with the fatty liver.
Cueto-Galán
et al. [41]
2017Human study276 participants, 57% NAFLD67Females: 66%
Males: 34%
Mediterranean diet (MD) with extra virgin olive oil and MD with nutsNatural: MD with extra virgin olive oil and MD with nutsMD with extra virgin olive oil (1 l/week), MD with nuts (30 g/day)6-year follow-upLow-fat dietA Mediterranean diet could be used as a dietary intervention to prevent or treat NAFL by preventing or reducing the disease’s natural course.
Kontogianni et al. [42]2014Human study7318–65
years old
Both gendersMD via MedDietScoreNatural: MD via MedDietScore_12 monthsStable dietary and exercise habitsAlthough increased Mediterranean diet adherence was associated with less severe liver disease and a reduced level of insulin resistance in NAFLD patients, it was not associated with a decreased likelihood of developing the disease.
Ryan et al. [43]2013Human study12Not
Mentioned
6 females/6 malesMDNatural: MD_6-week wash-out period in betweenControl diet (low-fat–high-carbohydrate diet)Compared to current dietary recommendations, MD improved insulin sensitivity and decreased liver steatosis in an insulin-resistant NAFLD population, even in the absence of weight loss.
Trovato et al. [44]2015Human study9050.13 ±13.68Females: 46
Males: 44
MD diet interventionNatural: MD diet intervention_6 months_Maintaining the Mediterranean diet is a strong indicator of alterations in the liver’s fat composition in overweight NAFLD patients. The diet has a beneficial, progressive effect that occurs on its own without requiring other lifestyle modifications.
Zhang et al. [45]2015Experimental74Age from 25 to 65Both gendersAnthocyanin\red orange juice and raw red raspberry (equivalent dose from dietary
source (according to Phenol Explorer))
Synthetic: anthocyanin
Natural: red orange juice and raw red raspberry
Anthocyanin 320 mg, once
a day\3.17 mg/100 mL red orange juice,
72.47 mg/100 g of raw red raspberry
12 weeksPlaceboDietary supplements containing anthocyanins may reduce the symptoms of NAFLD. Furthermore, these supplements are an inexpensive, effective treatment that can improve general health by decreasing the risk of T2DM, cardiovascular disease, and chronic liver disease at the same time.
Suda et al. [46]2008Experimental4830–60 yearsMales: 100%Purple sweet
potato
beverage
(anthocyanins)\red orange juice and raw red raspberry (equivalent dose of dietary source (according to Phenol Explorer))
Synthetic: purple sweet
potato
beverage
(anthocyanins)
Natural: red orange juice and raw red raspberry
125 mL/bottle, 2 bottles per day\3.17 mg/100 mL red orange juice,
72.47 mg/100 g of raw red raspberry
8 weeks PlaceboHepatic biomarker serum levels were considerably reduced after consuming the purple sweet potato beverage.
Faghihzadeh et al. [47]2014Experimental5044.04Males: 18 (72%)Resveratrol\red wine and
black grapes (equivalent dose from the dietary source (according to Phenol Explorer))
Synthetic: resveratrol
Natural: red wine;
black grapes
Resveratrol 500 mg, once a day\0.15 mg/100 mL red wine; 0.18
mg/100 g black grapes
12 weeksPlaceboFor the treatment of NAFLD, resveratrol supplementation is favorable to lifestyle modifications alone. The body’s hepatocellular apoptosis and the attenuation of inflammatory indicators are at least substantially accountable for this.
Stiuso et al. [48]2014Experimental30_Not
mentioned
Silymarin (Realsil, silybin, phosphatidylcholine, ɑ-tocopherol\dried milk thistle
fruit (112) (equivalent dose from dietary source (according to Phenol Explorer))
Synthetic: silymarin
Natural: dried milk thistle
fruit
Realsil, 94 mg silybin, 194 mg
phosphatidylcholine, 30 mg ɑ-tocopherol, twice a day\2.68% w/w of dried milk thistle
fruit (112)
12 months_Realsil may be effective in improving a patient’s metabolic asset if they have moderate NASH. In conclusion, this research indicates that the lipidomic profile of these RA patients changed in a distinctive way as a result of their therapy. This is probably because every patient has a different metabolic response, and they should be further categorized depending on other metabolic factors such as age, sex, AST, ALT, and GGT levels.
Loguercio et al. [49]2012Experimental179Age 18–65Not
mentioned
Silymarin (Realsil, silybin, phosphatidylcholine, ɑ-tocopherol\dried milk thistle
fruit (equivalent dose from dietary source (according to Phenol Explorer))
Synthetic: silymarin
Natural: dried milk thistle
fruit
Realsil, 94 mg silybin, 194 mg
phosphatidylcholine, 30 mg ɑ-tocopherol, twice a day\2.68% w/w of dried milk thistle
fruit
12 monthsPlaceboRealsil-treated NAFLD patients showed improvements in insulin resistance, liver histology, γGT levels, and transaminase levels without increasing weight.
Solhi et al. [50]2014Experimental64Patients in case group: 43.6 ± 8.3 Patients in control groups: 39.4 ± 10.5Both gendersSilymarin\ dried milk thistle fruit (equivalent dose from dietary source (according to Phenol Explorer))Synthetic: silymarin
Natural: dried milk thistle
fruit
Silymarin 70 mg, three times a
day\2.68% w/w of dried milk thistle fruit
8 weeksPlaceboFor the treatment of NASH, silymarin is a beneficial herbal medication. Patients who consumed it showed a more noticeable decrease in their hepatic enzyme levels.
Soleimani et al. [51]2020Experimental110Treatment: 45.6 ± 11.3
Control: 42.9 ±1 2.21
Both gendersGarlic powder tabletsSynthetic: garlic powder tablets400 mg garlic powder tablets15 weeksPlaceboSupplementation with garlic could assist NAFLD patients in decreasing body fat mass. Garlic may consequently decrease liver fat levels and prevent or impede the development of NAFLD.
Sangouni et al. [52]2020Experimental90Treatment: 45.2 ± 12.4
Control: 44.2 ± 11.1
Both gendersGarlic tablets
daily
Synthetic: garlic tablets
daily
Four garlic tablets
daily (each tablet
contained 400 mg
garlic powder)
12 weeksPlaceboThis clinical experiment demonstrated that supplementing with garlic powder improves liver enzymes, lipid profile, and hepatic steatosis as a dyslipidemia indicator. Therefore, implementing garlic into a patient’s adjusted diet seems reasonable and is an essential aspect of the treatment strategy for NAFLD.
Kim et al. [53]2017Experimental75Treatment: 54.5 ± 12.2
Control: 54.2 ± 9.3
Both gendersGarlicNatural: garlic sachetsTwo sachets of garlic/per day12 weeksPlaceboFor individuals without underlying hepatic disease, fermented garlic extracts (FGEs) might serve as a reliable and beneficial treatment for fatigue and reduce hepatic dysfunction caused by oxidative stress and inflammation.
Zhang et al. [54]2019Experimental24,106Males: 41.0 ± 12.2
Females: 40.3 ± 11.6
Both gendersRaw garlicNatural: raw garlicGarlic (≥7 t/week vs.
<1 t/week)
Garlic (4–6 t/week vs.
<1 t/week)
Garlic (1–3 t/week
vs. <1 t/week)
Garlic ≥7 t/week vs.
<1 t/week)
Garlic (4–6 t/week vs.
<1 t/week)
Garlic (1–3 t/week vs.
<1 t/week)
Consuming raw garlic consistently is inversely associated with NAFLD.
Emamat et al. [55]2020Experimental999Cases: 42.3 ± 11.9
Control: 43.5 ± 14.5
Both gendersDietary allium vegetable intakesNatural: dietary allium vegetable intake7.17 (2.52–15.21) g/day Higher allium vegetable consumption was associated with a decreased incidence of NAFLD.
Loffredo et al. [56]2017Experimental57 (3 groups: NASH (n = 19)
FLD (n = 19)
Controls (n = 19)
NASH (46 ± 11)
FLD (46 ± 6)
Controls (47 ± 8)
Both genders
(males/females: 11/8 per group)
NASH: dark chocolate, cocoa
>85% and milk chocolate, cocoa <35%
Natural: dark
chocolate and milk chocolate
40 g/day dark
chocolate and 40 g milk chocolate
2 weeks By comparing patients with NASH with individuals with fatty liver disease (FLD) and healthy controls, it was found that the former showed decreased flow-mediated dilation (FMD) and higher NOX2 activation and oxidative stress. In NASH patients, dark chocolate but not milk chocolate can improve endothelial function.
Rafie et al. [57]2020ExperimentalGinger group: 23
Control group: 23
Ginger group: 50.04 ±
10.26
Control group: 47.95
± 9.24
Both gendersGinger
powder
Natural: ginger
powder
1500
mg
ginger
powder
daily
12 weeksPlaceboIndividuals with NAFLD responded more effectively to dietary recommendations and lifestyle modifications.
Daneshi-Maskooni et al. [58]2019ExperimentalGinger group: 43
Control group: 44
Ginger group: 45.5 ± 8.
9
Control group: 45.0 ±
7.7
Both gendersCapsules
s (green cardamom (GC)
from ginger
family)
Synthetic: capsules
(GC
from ginger
family)
Two 500
mg capsules
(GC
from ginger
family) three
times per day
3 monthsPlaceboIt is likely that, by increasing serum Sirt1 and irisin levels, GC supplementation reduced fatty liver grade, blood glucose indices, and lipid profiles in NAFLD patients with overweight or obesity.
Rahimlou et al. [59]2016ExperimentalGinger group: 23
Control group: 21
Ginger group: 45.45 ± 2.31
Control group: 45 ± 2.14
Both gendersGinger
capsule
Synthetic: ginger
capsule
Two 500 mg
ginger
capsules
twice
daily (2 g daily)
12 weeksPlaceboSupplementation with ginger was shown to improve certain NAFLD features, as well as decrease hs-CRP, TNF-α, ALT, GGT, hepatic steatosis, and HOMA-IR.
Anty, R et al. [60]2012Experimental19539 ± 12.3 yearsBoth gendersCoffee-containing
beverages: espresso; double
espresso; filtered (regular);
made with Italian coffee
machine; decaffeinated
coffee
Natural: coffee-containing
beverages: espresso; double
espresso; filtered (regular);
made with Italian coffee
machine; decaffeinated
coffee
-20 monthsPlaceboRegarding individuals who routinely drank coffee, the quantity of caffeine consumed and the severity of fibrosis were negatively correlated. Consuming coffee on a regular basis is one independent way to prevent fibrosis. There is no evidence linking the consumption of espresso coffee to the onset of fibrosis.
Zelber-Sagi,
S et al. [61]
2014Experimental34750.86 ± 10.35 yearsBoth gendersAll types of caffeinated coffeeNatural: All types of caffeinated coffeeFrom <once per
month to >5 per day
15 months-Steatosis development was unaffected by coffee drinking; however, the cross-sectional group showed an inverse association between coffee consumption and liver fibrosis.
ALT—alanine transaminase; AST—aspartate transaminase; CR—calorie restricted; GGT—gamma-glutamyl transferase; GTE—green tea extract; HOMA-IR—Homeostatic Model Assessment of Insulin Resistance; IR—insulin resistance; MD—Mediterranean diet; NAFLD—non-alcoholic fatty liver disease; NASH—non-alcoholic steatohepatitis; n-3 PUFA—omega-3 polyunsaturated fatty acids; UDCA—ursodeoxycholic acid; T2DM—type 2 diabetes mellitus; FGE—garlic extract; FMD—flow-mediated dilation; NOX2—NADPH oxidase 2; FLD—fatty liver disease; GC—green cardamom; hs-CRP—high-sensitivity C-reactive protein; TNF-α—tumor necrosis factor alpha.
Table 4. Summarized data of animal studies.
Table 4. Summarized data of animal studies.
StudyYearSample SizeAgeGenderInterventionForm of Intervention (Natural or Synthetic)Dose/DayDuration ControlStatus after Intervention
Ni X et al. [62]201630 (in 3 groups)7 weeksMale C57BL/6J miceObeticholic acid or silymarinSynthetic Obeticholic acid (30 mg/kg);
silymarin (30 mg/kg)
4 weeksNaïve control group: vehicle (1% HPMC + 1% Tween 20)
Vehicle group: vehicle (1% HPMC + 1% Tween 20)
Silymarin significantly reduces liver steatosis.
Meng-sha T. et al. [63]202130 (in 5 groups)Not mentioned Male Wistar ratsSilymarin SyntheticSilymarin (200 mg/kg)
/silymarin (400 mg/kg)
8 weeksNormal control group: standard chow + water
Silymarin control group: water + silymarin (400 mg/kg/day)
Fructose control group: 20% fructose solution
Treatment with a high dose of silymarin improves liver function and ameliorates steatosis.
Sun R. et al. [64]202030 (in 5 groups)6–8 weeks Male C57BL/6J miceSilybin + sodium tauroursodeoxycholateSyntheticSilybin (50 or 100 mg/kg/day)/
sodium tauroursodeoxycholate (50 mg/kg/day)
4 weeksVehicle group: standard normal diet
HFD group: high-fat/high-cholesterol diet
Silybin and sodium tauroursodeoxycholate treatment both reduced hepatic lipid accumulation.
Zhu SY et al. [65]201840 (in 4 groups)2 monthsMale
ICR mice
Silybum marianum oil (SMO)Natural (intragastric)Silybum marianum oil 5 mL/kg
Silybum marianum oil 10 mL/kg
8 weeksControl group: normal diet + distilled water
HFD group: high-fat
diet (70.5% normal diet + 10% lard + 10% yolk powder + 8%
sucrose + 1.5% cholesterol) + distilled water
Silybum marianum oil can significantly reduce fat accumulation and improve lipid metabolism.
Ou Q et al. [66]201818 (in 3 groups)6 weeks oldMale C57BL/6 miceSilybinSynthetic: oral105 mg/kg/day8 weeksControl group: methionine/choline-deficient (MCD) diet (DL-methionine (3 g/kg) + choline bitartrate (2 g/kg))
NASH group: methionine/choline-deficient (MCD) diet
Silybin prevents an increase in steatosis, fibrosis, and liver inflammation.
Ye JH et al. [67]201624 7 weeksMale C57BL/6J micePentoxifylline (PTX)Synthetic: intraperitoneal100 mg/kg8 weeksControl group:
normal diet
HFG group: high-fat diet + hyperglycemia (60 kcal% fat)
Pentoxifylline improves liver function and prevents fat accumulation by upregulating fatty acid β-oxidation.
Massart J. et al. [68]201210–12 mice in 4 groups5 weeksMale C57BL/6J-ob/ob mice and C57BL/6J-+/+ micePentoxifylline (PTX)Synthetic: oral 100 mg·kg−1·d−14 days or 3 weeksLean and obese untreated mice Pentoxifylline can exacerbate fatty liver due to the activation of liver lipogenesis.
Saeed A et al. [69]202116 (in
2 groups)
8–10 weeks/10–12 weeks oldC57BL/6J mice
Leptinob mutant (JAX ob/ob)
Vitamin ASynthetic 20 IU/g12 or 20 weeksControls (C57BL/6J) group: chow dietTreatment with vitamin A may accelerate the progression of the disease because vitamin A accumulates in hepatocytes.
Ipsen DH et al. [70]202160
chow (n = 5)
baseline euthanasia (n = 6)
HFD (n = 10) (ASA, n = 13)
(PTX, n = 13) (ASA + PTX,
n = 13)
Not mentioned Female Hartley guinea pigsAcetylsalicylic acid/pentoxifylline/acetylsalicylic + pentoxifyllineSynthetic 46.3 and 45.6 mg/d for acetylsalicylic acid and pentoxifylline8 weeksHealthy control group:
chow diet
HFD group: high-fat diet
NASH or liver fibrosis did not improve in guinea pigs administered acetylsalicylic and pentoxifylline alone or in combination.
Oliveira CP et al. [71]200318 (in 3 groups)Not mentioned Male Wistar ratsVitamin C or vitamin ESynthetic Vitamin E (200 mg/day)
Vitamin C (30 mg/kg/day)
4 weeks Control group: choline-deficient diet + vehicleVitamin E can prevent the development of steatosis, while vitamin C cannot.
Liu CW et al. [72]202032
(Ctrl) group (n = 5)
NASH groups (n = 9)
NASH-resv + EX527 group (n = 9)
NASH-rest (n = 9)
8 weeksC57BL/6 miceResveratrolSynthetic NASH-resv group (resveratrol
(30mg/kg/day))/
NASH-resv + EX527 group (resveratrol and EX527 (1mg/kg/day))
6 weeksControl group: normal chow
NASH groups: normal chow
Chronic treatment with resveratrol
significantly improves hepatic steatosis.
Karimian G. et al. [73]201518 (in 2 groups)5–6 weeks oldC57BL/6JolaHsd male miceVitamin E Synthetic: vitamin E supplements 20 mg/day1 week Control groups: choline-deficient L-amino acid or choline-sufficient L-amino acid After partial hepatectomy, vitamin E can be used to prevent the progression of steatosis.
Presa N. et al. [74]201920 (5 animals per group)Not mentioned Male Pemt +/+ and Pemt −/− miceVitamin E Synthetic:
vitamin E supplements
133 IU/kg/day (0.5 g/kg)3 weeksControl group: semi-synthetic HFD (60% kcal fat)Vitamin E dietary supplements can be a suitable treatment to prevent the progression of hepatic steatosis to more advanced stages of the disease.
Lee SW et al. [75]20214412 to 15 weeks oldMale C57BL/6 SMP30 KO mice and wild-type (WT) miceVitamin CSynthetic: vitamin C supplements1.5 g/L11 weeksWild-type mice: 60% high-fat dietChronic vitamin C deficiency due to de novo lipogen suppression can decrease the progression of MAFLD.
Zeng Q. et al. [76]2020606–8 weeksMale C57BL/6 miceVitamin CSynthetic Low-dose vitamin C group (15 mg/kg per day)
Medium-dose vitamin C group (30 mg/kg per day)
High-dose vitamin C group (90 mg/kg per day)
Prophylactic group: 12 weeks
Therapeutic group: 6 weeks
Control group: normal chow or HFD (25 kcal% fat +1.1% cholesterol and 0.5% cholate ad libitum)Different doses of vitamin C can have different effects on MAFLD.
Milton-Laskibar I. et al. [77]2021506 weeks Male Wistar ratsPterostilbene and resveratrolSynthetic: powdered diets Resveratrol30 group (30 mg/kg/d resveratrol)
Pterostilbene group (15 mg/kg/d pterostilbene) Pterostilbene group (30 mg/kg/d pterostilbene)
8 weeksControl group: standard diet
HFHF group: high-fat–high-fructose diet
Changes in gut microbiota that cause disease progression are poorly ameliorated by pterostilbene and resveratrol.
Sun M et al. [78]202146 (in 2 groups)5 weeks oldMale C57BL/6J miceCocoa powderNatural: cocoa powder80 mg/g 10 weeksControl group: high-fat diet Cocoa supplementation improves steatosis.
Reda D. et al. [79]2023329 weeksMale albino ratsCholecalciferol (vitamin D3)Synthetic 1000 IU/kg BW (3 days per week)10 weeksNegative control: standard rat chow
Positive group: high-fat diet (20%) + 25% fructose water
Vitamin D control: intramuscular vitamin D (1000 IU/kg BW)
Vitamin D can ameliorative lipid levels in liver and serum.
El-Din SH et al. [80]201490Not mentioned Male Sprague Dawley ratsGarlic and onion or combined
garlic and onion
Natural: onion oilGarlic (500 mg/kg
b.w)
Onion (100 mg/kg body b.w.)
8 weeksControl group:
standard chow diet
MAFLD group: high-fat diet (25
% fat + 1% cholesterol + 0.25% bile salts)
NAFLD group: switched to
regular diet
When administered together, garlic and onion significantly reduced liver steatosis compared to when administered separately.
Wang JH et al. [81]202130 6 weeks oldMale C57BL/6j mice Cynanchum atratumSynthetic: Cynanchum atratum powder 100, 200 mg/kg/day6 weeksPositive control group: metformin
Normal group: chow diet
HFD group: high-fat diet (60% kcal% fat) + fructose (20% w/v, dissolved in distilled water)
Cynanchum atratum can ameliorate fatty liver by enhancing fatty acid oxidation.
Li S. et al. [82]2014835 weeksMale Sprague Dawley ratsUrsolic acidSynthetic Group 1: 0.125% ursolic acid (low ursolic acid)
Group 2: 0.25% ursolic acid (medium ursolic acid)
Group 3: 0.5% ursolic acid (high ursolic acid)
6 weeksControl group: normal-fat diet
HFD group: high-fat diet
Ursolic acid effectively improves hepatic steatosis.
Wang Q. et al. [83]202030 (in 5 groups)/
15 (in 3 groups)
8 weeks oldMale C57BL/6 mice/
NLRP3 −/− mice
Naringenin Synthetic 50 or 100 mg·kg−1·day−17 days Control group: chow diet (0.35% (w/w) methionine + 0.1% (w/w) choline + 0.5% sodium carboxymethyl cellulose)
MCD groups: methionine/choline-deficient diet (0% methionine + 0% choline, 0.5% sodium carboxymethyl cellulose)
NGN 100 group: 100 mg·kg−1·day−1 naringenin
Naringenin has significant effects on the recovery of liver inflammation and liver steatosis.
Zhou J. et al. [84]2021254 weeks Male C57BL/6 miceMatcha green teaNatural 0.1%, 0.5%, and 1% matcha6 weeksNormal chow diet (NCD)
A high-fat diet (HFD)
Matcha supplementation ameliorates lipid accumulation.
Chao J. et al. [85]20143110 weeksMale C57BL/6 miceGallic acidSynthetic50 and 100 mg/kg/day16 weeksNormal group:
normal chow diet
HFD group: high-fat diet (5.24 kcal/g, 60% kcal from lard/soybean)
Gallic acid presents as a natural dietary compound capable of ameliorating NAFLD and other metabolic disorders.
Gao X. et al. [86]202350 (in 5 groups)58 weeksMale C57BL/6J miceLycopeneSyntheticLow-dose lycopene group (LLY) (20 mg/kg/d lycopene)
High-dose lycopene group (HLY) (60 mg/kg/d lycopene)
8 weeksNormal control group: normal chow diet (10% calories from fat) + saline solution
High-fat and high-fructose diet group: HFD (45% calories from fat) + 10% fructose solution
Resveratrol positive control group: high-fat diet + 10% fructose solution +50 mg/kg/d resveratrol
Lycopene dietary supplementation significantly prevented the accumulation of fat in liver.
Gu M et al. [87]2019216 weeksFemale C57BL/6J miceBetulinic acidSynthetic 100 mg·kg−1·day−16 weeks Chow group: 10% of calories derived from fat
High-fat group: HF (60% of calories derived from fat)
Betulinic acid supplementation ameliorated hepatic steatosis and inflammation.
Luo D. et al. [88]202232 (6–8 per 4 groups)3 to 4 weeks Male C57BL/6J Narl miceTianhuang formulaSynthetic 100 mg/kg/day6 weeks Model group (vehicle): high-fat diet (60 fat, 20 proteins, 20% carbohydrate)
Atorvastatin group: (1.5 mg/kg/day atrovastamin)
Control group: (not mentioned)
The Chinese Tianhuang formula improves MAFLD by regulating intestinal microbiota through its hepatoprotective effect.
Acedo SC et al. [89]2015206 weeksSwiss male micePentoxifyllineSynthetic: intraperitoneal100 mg/kg per day2 weeks Lean non-treated group: AIN-93 (control diet; 15% kcal of fat)
Lean treated mice: AIN-93 (control diet 15% kcal from fat + 100 mg/kg per day of pentoxifylline)
Non-treated obese mice: high-fat diet (HFD; 60% kcal from fat)
Pentoxifylline can improve obesity-related NAFLD by reducing liver inflammation and adipose tissue inflammation.
Panchal K et al. [90]2012408–9 weeks Male Wistar rats QuercetinSynthetic 0.8 g/kg8 weeksCorn starch-rich (C) diet group: 68% carbohydrates as polysaccharides + ~0.7% fat
High-carbohydrate, high-fat (H) diet group: 68% carbohydrates fructose + sucrose+ ~24% fat from beef tallow
Quercetin is effective against metabolic syndrome symptoms such as liver complications with debilitating steatosis.
Marcolin E. et al. [91]201264 (in 4 groups)Not mentioned C57BL/6 miceQuercetinNatural 50 mg/kg2 or 4 weeksControl group (CO): control diet + vehicle (sodium carboxymethylcellulose 1%)
Control + quercetin group (CO + Q): control diet + 50 mg/kg quercetin
NASH group (NASH): with methionine/choline-deficient diet + vehicle (sodium carboxymethylcellulose 1%)
Quercetin improves steatohepatitis by affecting proinflammatory and profibrotic gene pathways.
Panchal SK et al. [92]201172 (in 6 groups)8–9 weeks oldMale Wistar ratsRutinSynthetic 1.6 g/kg food8 weeksCorn starch-rich diet group: 68% carbohydrates + 0.7% fat (2 groups)
High-carbohydrate, high-fat diet group: 50% carbohydrate (mainly fructose) + 24% fat (mainly beef tallow) + 25% fructose in drinking water (total 68% carbohydrate (2 groups)
Rutin reduces fat deposition in hepatocytes.
Suguro R. et al. [93]2019 64 (in 8 groups)Not mentioned Male wild-type (WT) C57Bl/6 miceSilybin (SB) and tangeretin (TG)Synthetic 1.SB (300 or 150 mg/kg)
2.TG (150 mg/kg)
3.SB + TG at low dose (each 75 mg/kg)
4.SB + TG at middle dose (each 150 mg/kg)
5.SB + TG at high dose (each 300 mg/kg)
8 weeksControl group:
vehicle (35% PEG400: 15% Cremophor EL: 5% ethanol: 45% saline)
Model group:
vehicle (35% PEG400: 15% Cremophor EL: 5% ethanol: 45% saline)
Tangeretin and silybin co-treatment can inhibit de novo synthesis of fatty acids, thus improving lipid profiles.
Kuzu N. et al. [94]200821Not mentioned Male Sprague Dawley ratsEpigallocatechin gallate (EGCG)Synthetic 1 g/L epigallocatechin gallate in drinking water 6 weeks1. Standard rat diet
2. High-fat diet (HFD)
Epigallocatechin gallate significantly attenuates intrahepatic fat accumulation, inflammation, and fibrosis.
Mao QQ et al. [95]2021140 (in 14 groups)8 weeksMale C57BL/6J mice12 types of teasSynthetic: intragastric200 mg/kg 15 weeksControl group: normal diet (3.6 kcal/g, 12% calories from fat)
HFD model group: high-fat diet (5.0 kcal/g, 60% calories from fat)
Different teas had different effects, some of which reduced triglyceride content in the liver and improved liver steatosis.
Zhou DD et al. [96]2022120 (in 12 groups)8 weeksMale C57BL/6J mice10 types of green teaSynthetic: intragastric200 mg/kg 15 weeksControl group: normal diet (12% calories from fat)
HFD model group: high-fat diet (60% calories from fat)
In addition to lowering visceral fat buildup and oxidative stress, a number of green teas can also improve lipid profiles and treat hepatic steatosis.
Liu B. et al. [97]201950 (in 5 groups)8 weeksSPF-grade C57BL/6N mice (half male and half female)Raw bowl teaNatural: intragastric50 and 100 mg/kg once a day12 weeks Normal group: normal foods + drinking water
Model group: high-fat diet
Bezafibrate group: bezafibrate (100 mg/kg daily)
Lipopolysaccharides can play a role in the accumulation of fat in the liver.
Therefore, raw bowl tea can reduce fat accumulation by reducing lipopolysaccharides.
Li HY et al. [98]2023404 weeksC57BL/6J male miceTheabrowninNatural: intragastric2300 mg/kg/day14 weeksBlank control group: CD group
TB control group: CD + TB group NAFLD with obesity group: HFD group
Theabrownin intervention can effectively alleviate intestinal bacterial dysbiosis and strongly prevent hepatic steatosis.
Li B. et al. [99]202280 (in 8 groups)8 weeks C57BL/6J male miceSix types of tea (different black and dark teas)Synthetic: intragastric200 mg/kg b.w./d 15 weeksControl group: normal diet (3.6 kcal/g + 12% calories from fat)
Model group (HFD group): high-fat diet (HFD, 5.0 kcal/g+ 60% calories from fat)
Different extracts of black and dark tea taken as supplements may help prevent aberrant lipid deposition in the liver and liver steatosis.
Torres LF et al. [100]201930 (in 3 groups)12 weeks
Male C57Bl/6 miceGreen teaNatural 500 mg/kg b.w.12 weeksControl group: chow diet (32% protein, 55% carbohydrates + 13% lipids (2.88 kcal/g))
High-fat diet group: hyperlipidemic diet (20% protein, 36% carbohydrates + 34% lipids (5.31 kcal/g))
Green tea supplementation decreases lipid uptake and accumulation, as well as cholesterol synthesis, through miR-34a and miR-194 modulation in the liver.
Noori M. et al. [101]201618Not mentioned Male Sprague Dawley ratsPomegranate juiceNatural: drinking liquid60 ± 5 mL/day7 weeksControl group: standard chow diet (10% of energy derived from fat + 30% from
protein + and 60% from carbohydrates)
Model group:
high-fat, high-sugar diet (59% of energy derived from fat + 30% from carbohydrates + 11% from protein)
Regular drinking of pomegranate juice can downregulate hepatic proinflammatory and profibrotic conditions and prevent progression to NASH.
El-Lakkany NM et al. [102]2016108 (in 9 groups)Not mentioned Male Sprague Dawley ratsMetformin and N-acetylcysteineSynthetic: oral MTF: 150 mg/kg
NAC: 500 mg/kg,
8 weeks1. Normal control group: standard chow diet
2. NAFLD induced with high-fat diet (HFD): group 25% fat + 1% cholesterol + 0.25% bile salts
3. NAFLD switched to regular diet
In the co-treated group with metformin and N-acetylcysteine, hepatic steatosis significantly decreased, and hepatic fat accumulation reversed.
El-Din SHS et al. [103]201572 (in 9 groups)Not mentioned Male Sprague Dawley ratsRosuvastatin (RSV) and/or β-carotene (βC)Synthetic: oral Rosuvastatin:
10 mg/kg/day
β-carotene: 70 mg/kg
4 weeks1. Normal control: standard chow diet
2. NAFLD induced with high-fat diet (HFD) group
3. NAFLD switched to a regular diet group
The combined treatment of rosuvastatin and beta-carotene had a better effect on reducing fat accumulation in the liver than each drug alone.
MAFLD—metabolic dysfunction-associated fatty liver disease; HPMC—hydroxypropyl methylcellulose; HFD—high-fat diet; SMo—Silybum marianum oil; MCD—methionine/choline deficient; NASHA—non-alcoholic steatohepatitis; PTX—pentoxifylline; HFG—high-fat hyperglycemic diet; PEMT—phosphatidylethanolamine N-methyltransferase; WT—wild type; HFHF—high-fat–high-fructose diet; BW—body weight; NCD—normal chow diet; NGN100—naringenin 100; KCs—Kupffer cells; LLy—low-dose lycopene group; HLY—high-dose lycopene group; HF—high fat; AIN-93 diet—American Institute of Nutrition-93 diet; (C) diet—corn starch-rich diet; (H) diet—high-carbohydrate, high-fat diet; CO—control group; CO + Q—control + quercetin group; SB—silybin; TG—tangeretin; EGCG—epigallocatechin gallate; RBTP—raw bowl tea; CD group—blank control group; TB—theabrownin; MTF—metformin; NAC-N—acetylcysteine; RSV—rosuvastatin; βC-β—carotene.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mohammadian, K.; Fakhar, F.; Keramat, S.; Stanek, A. The Role of Antioxidants in the Treatment of Metabolic Dysfunction-Associated Fatty Liver Disease: A Systematic Review. Antioxidants 2024, 13, 797. https://doi.org/10.3390/antiox13070797

AMA Style

Mohammadian K, Fakhar F, Keramat S, Stanek A. The Role of Antioxidants in the Treatment of Metabolic Dysfunction-Associated Fatty Liver Disease: A Systematic Review. Antioxidants. 2024; 13(7):797. https://doi.org/10.3390/antiox13070797

Chicago/Turabian Style

Mohammadian, Kiana, Fatemeh Fakhar, Shayan Keramat, and Agata Stanek. 2024. "The Role of Antioxidants in the Treatment of Metabolic Dysfunction-Associated Fatty Liver Disease: A Systematic Review" Antioxidants 13, no. 7: 797. https://doi.org/10.3390/antiox13070797

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