metricas
covid
Buscar en
Annals of Hepatology
Toda la web
Inicio Annals of Hepatology Administration of silymarin in NAFLD/NASH: A systematic review and meta-analysis
Información de la revista
Vol. 29. Núm. 2.
(marzo - abril 2024)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
3860
Vol. 29. Núm. 2.
(marzo - abril 2024)
Original article
Acceso a texto completo
Administration of silymarin in NAFLD/NASH: A systematic review and meta-analysis
Visitas
3860
Shudi Lia, Fei Duanb, Suling Lib, Baoping Luc,
Autor para correspondencia
lbp1921@sohu.com

Corresponding author.
a The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou 450000, China
b The First Affiliated Hospital of Henan University of TCM Zhengzhou 450000, China
c Henan University of Chinese Medicine, Zhengzhou 450046, China
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (8)
Mostrar másMostrar menos
Tablas (1)
Table 1. Characteristics of included studies
Material adicional (1)
Graphical abstract

Keywords:
NAFLD
Energy metabolism
Liver injury
Liver histological changes
Anthropometric parameters
Abbreviations:
NAFLD
NASH
RCTs
PRISMA
TC
TG
HDL-C
LDL-C
FBG
FI
HOMA-IR
ALT
AST
BMI
WC
HC
RoB
SMD
OR
Texto completo
1Introduction

Nonalcoholic fatty liver disease (NAFLD) is a chronic pathological liver disease caused by excessive buildup of fat in liver cells [1]. NAFLD can range from simple fat accumulation in the liver to nonalcoholic steatohepatitis (NASH) alone or with fibrosis, which may subsequently lead to cirrhosis and liver cancer [2]. NAFLD is found in 25.2% of the population worldwide and poses serious health risks [3]. It is the hepatic manifestation of metabolic syndromes, such as obesity, dyslipidemia, insulin resistance, glucose intolerance and type 2 diabetes mellitus [4,5]. There are currently no pharmacological agents that are approved for NAFLD or NASH treatment [6]. Therefore, the development of treatments for NAFLD, especially the incurable NASH, is an unmet and urgent medical need.

A growing body of work indicates that adhering to the Mediterranean diet, supplementing with probiotics, antioxidants, polyphenols or specific nutrients with hepatoprotective effects may improve liver enzyme levels and hepatic steatosis in NAFLD, or slow its progression [7–10]. Silymarin is a potent liver-tropic antioxidant extracted from milk thistle (Silybum marianum). This extract is comprised of several antioxidant substances, the most abundant of which are silibine A and B and the flavonoid taxifolin [11]. Silymarin has multiple hepatoprotective effects, including antioxidant activity, cell regeneration, increased protein synthesis, and anti-inflammatory and antifibrotic properties [12]. As a result, silymarin has been proposed as a therapeutic approach for NAFLD [13].

NAFLD has a complex pathogenesis and can be caused by impaired metabolism and inflammation. Studies have shown that NAFLD is the hepatic manifestation of metabolic syndromes such as obesity, dyslipidemia, hypertension and type 2 diabetes mellitus (T2DM) [14]. In addition, NAFLD can also result from increased oxidative stress, which increases lipid peroxidation and reactive oxygen species production within hepatocytes and thereby leads to mitochondrial dysfunction and lipotoxicity [15,16]. Some studies showed that silymarin can improve insulin resistance by inhibiting the production of pro-inflammatory cytokines through inhibition of NF-κB signaling [17]. The anti-inflammatory and anti-fibrotic effects of silymarin are mediated by upregulated adiponectin expression, downregulated resistin expression, inhibition of NF-κB and related pathways, reduced activation of hepatic stellate cells, and decreased expression of interleukins (IL-2 and IL-4), TNF-α and IFN-γ[18].

Previous studies have suggested that silymarin should be initiated as early as possible in patients with fatty liver disease when the regenerative potential of the liver is still high [19,20]. Silymarin is a promising botanical treatment for NAFLD patients, and its efficacy has been investigated in several randomized-controlled trials (RCTs). However, a systematic evaluation of the efficacy of silymarin in NAFLD is currently lacking. This meta-analysis aims to systematically evaluate the efficacy of silymarin in NAFLD patients by characterizing its effects on energy metabolism, liver injury, liver histology and anthropometric parameters in order to provide support for its clinical application in NAFLD.

2Materials and Methods

This study was conducted according to the Preferred Reporting Items for Systemic Reviews and Meta-Analysis (PRISMA) statement [21] and was registered with PROSPERO (CRD42023398590).

2.1Databases

Relevant articles were retrieved from PubMed, Embase, the Cochrane Library, Web of Science, clinicaltrails.gov, and China National Knowledge Infrastructure.

2.2Search strategy

Search terms related to “silymarin” and “nonalcoholic fatty liver disease” were used to retrieve records from the databases. The search strategy and results are detailed in Supplementary Table S1.

2.3Study selection

Studies were searched and selected independently by three researchers. The titles and abstracts of the identified studies were first screened, then the full texts of potential studies were further assessed based on the eligibility criteria. The selected full texts were compared among the three researchers, and any disagreement or discrepancy was resolved by discussion and voting.

2.4Inclusion and exclusion criteria

Clinical studies were included if they meet the following criteria: (1) Study design: RCTs; (2) Participants: NAFLD patients, without age, gender, or race restrictions; (3) Intervention: Silymarin or silymarin complex; (4) Comparators: Same treatment as the intervention group except for silymarin; (5) Outcomes: 1) Energy metabolism indicators: Total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), fasting blood glucose (FBG), fasting insulin (FI), and homeostatic model assessment of IR (HOMA-IR); 2) Liver injury indicators: Alanine aminotransferase (ALT) and aspartate aminotransferase (AST); 3) Liver histological indicators: Fatty liver index, fatty liver score, and hepatic steatosis grade; 4) Anthropometric indicators: Body mass index (BMI), waist circumference (WC), and hip circumference (HC).

Exclusion criteria: (1) Alcoholic steatohepatitis, alcoholic fatty liver, cirrhosis or liver cancer; (2) Received additional medication(s) or has genetic predisposition (single nucleotide polymorphisms); (3) Underwent liver transplantation; (4) Conference papers, abstracts, non-original research, case reports, and non-peer-reviewed articles (e.g., conference materials and thesis).

2.5Quality assessment

The titles and abstracts of retrieved records were independently assessed by two authors to exclude irrelevant studies. Full texts of selected articles were then assessed individually based on the eligibility criteria. The risk of bias in eligible RCTs was evaluated using the Cochrane Collaboration's Risk of Bias (RoB) tool.

2.6Data synthesis and analysis

Statistical analyses were performed by RevMan v5.3 (The Cochrane Collaboration, Copenhagen, Denmark). Given the different methods used to measure outcomes in each original study, continuous and categorical variables were pooled using standard mean difference (SMD) and odds ratio (OR), respectively. If data were available and sufficient, subgroup analyses were also performed according to the type of disease (NAFLD/NASH), type of intervention (silymarin, silymarin complex), and duration (<12; ≥12 to ≤24; >24). Heterogeneity was assessed using the Cochran's Q test (I2 statistic). A fixed-effects model was used when the I2 is <50%; otherwise, a random-effects model was used. Source of heterogeneity was identified by sensitivity and subgroup analyses. All statistical tests are two-tailed with a significance level of 0.05. Sensitivity analysis was performed to identify the stability of the results. Publication bias was assessed using a funnel plot if a given outcome measure is reported in over 10 studies.

3Results3.1Study selection

A total of 1,039 articles (up until February 26, 2023) were initially retrieved according to the search strategy, and 26 RCTs were included in the meta-analysis (Figure 1).

Fig. 1.

PRISMA flow diagram.

(0,48MB).
3.2Basic characteristics and quality assessment

The 26 included studies [22–41] involved a total of 2,375 patients. The characteristics of the included studies are summarized in Table 1. The risk of bias assessment showed that the included studies had medium to low overall risk (Fig. 2). Since allocation concealment and blinding methods were unclear in most included studies, a clear judgement could not be made in these areas, resulting in reduced quality of evidence.

Table 1.

Characteristics of included studies

Author year  E/C  Male/female (E;C)  Age (E/C)  BMI (E/C)  Disease  Intervention  Control  Duration (weeks)  Outcomes 
Abenavoli 2015  10/10  8/26/4  47.10±19.78/54.89±16.34  29/32  NAFLD  Silymarin+A mediterranean hypocaloric diet  A mediterranean hypocaloric diet  24  TC, TG, HDL-C, LDL-C, FBG, FI, HOMA-IR, ALT, AST, Fatty liver index, BMI, WC, HC, 
Anushiravani 2019  30/30  NA  NA  25.7±3.3/26.1±3.1  NAFLD  Silymarin+Lifestyle modification  Placebo+Lifestyle modification  12  TC, TG, HDL-C, LDL-C, FBG, ALT, AST, BMI, WC, 
Ataee 2021  24/26  19/521/5  41.35±11.82/40.35±11.76  28.46 ± 5.03/29.07 ± 3.69  NAFLD  Silymarin  placebo  12  TC, TG, HDL-C, LDL-C, FBG, ALT, AST, Hepatic steatosis grade 
Chan 2017  49/50  24/2522/28  38.46±7.11/38.41±9.21/38.42±10.09  49.6±12.7/50.1±10.2  NASH  Silymarin  placebo  48  TC, TG, HDL-C, LDL-C, FBG, HOMA-IR, ALT, AST 
Chiurazzi 2022  36/32  16/2016/16  56.1±9.9/59.8±10.9  36.0±8.6/33.9±6.6  NAFLD  Silymarin +A mediterranean hypocaloric diet  A mediterranean hypocaloric diet  12  TC, TG, HDL-C, LDL-C, FBG, ALT, AST, BMI, WC, HC, Hepatic steatosis grade 
Curcio 2020  41/40  25/1625/15  56.9±14.2/67.6±11.3  NA  NAFLD  Silymarin complex+Lifestyle modification  Lifestyle modification  12  TC, TG, HDL-C, LDL-C, ALT, AST, Hepatic steatosis grade 
Famouri 2017  20/20  NA  11.8±3/10.5±3.2  25.8±4.1/24.3±4.5  NAFLD  Silymarin+Lifestyle modification  Lifestyle modification  12  Hepatic steatosis grade, BMI 
Hashemi 2009  50/50  28/2229/21  39.28±11.17/39.0±10.70  26.75±2.65/27.80±3.75  NASH  Silymarin  placebo  24  TC, TG, HDL-C, LDL-C, FBG, ALT, AST, BMI 
Mirhashemi 2022  27/25  NA  37.81±9.93/38.08±10.01  47.20±6.98/48.24±6.95  NAFLD  Silymarin+Lifestyle modification  Lifestyle modification  ALT, AST, BMI, Hepatic steatosis grade, 
Navarro 2019  26/27/25  13/139/18/11/14  47.3±10.8/48.2±11.4/49.5±10.9  35.3±4.8/33.5±4.3/33.4±4.8  NAFLD  Silymarin  placebo  12  Hepatic steatosis grade 
Rajendra 2022  29/30  NA  40.2±10.1/36.6±7.5  27.1±1.6/26.5±1.5  NAFLD  Silymarin  placebo  12  TC, TG, HDL-C, LDL-C, HOMA-IR, ALT, AST, FLI, 
Shaikh 2021  100/100  NA  49.5±7.3/49.7±10.5  29.4±3.57/29.2±3.93  NAFLD  Silymarin  placebo  12  ALT, AST 
Solhi 2014  33/31  NA  43.6±8.3/39.36±10.5  27.4±1.7/27.5±1.9  NASH  Silymarin +Lifestyle modification  Lifestyle modification  ALT, AST 
Chen 2012  30/30  NA  NA  NA  NAFLD  Silymarin+Lifestyle modification  lifestyle modification  12  TC, TG, ALT, AST 
Cui 2020  44/44  28/1629/15  45.34±4.98/44.91±5.33)  NA  NAFLD  Silymarin+Diisopropylamine Dichloroacetate  Diisopropylamine Dichloroacetate  TC, TG, ALT, AST 
Jiang 2020  46/46  26/2025/21  52.03±14.79/52.51±11.69  21.36±2.34/20.95±3.12  NAFLD  Silymarin+Bifidobacterium triple live powder  Bifidobacterium triple live powder  24  TC, TG, ALT, AST 
Li 2018  48/48  33/1532/16  56.03±10.82/5.86±1.26  NA  NAFLD  Silymarin+Diisopropylamine Dichloroacetate  Diisopropylamine Dichloroacetate  TC, TG, ALT, AST 
Li 2010  30/20  NA  NA  NA  NAFLD  Silymarin+Lifestyle modification  lifestyle modification  12  TG, HDL-C, LDL-C, ALT, AST 
Lisheng 2018  50/48  38/3233/15  43.1±6.4/43.7±6.0  NA  NAFLD  Silymarin+Polyene phosphatidylcholine  Polyene phosphatidylcholine  12  TC, TG, ALT, AST 
Liang 2015  45/45  24/2120/25  55. 7 ± 11. 9/55.2 ± 12.2  NA  NAFLD  Silymarin+Lovastatin capsules  Lovastatin capsules  12  TC, TG, HDL-C, LDL-C 
Liu 2012  42/34  NA  NA  27.09±1.67/27.11±1.65  NAFLD  Silymarin+Pioglitazone  Pioglitazone  12  TC, TG, ALT, Fatty liver score, BMI, 
Wang 2013  65/65  NA  40.0±3.0/40.0±4.043.14±9.57  NA  NAFLD  Silymarin+Lipitor capsules  Lipitor capsules  12  TC, TG, ALT, AST, Fatty Liver score 
Xiao 2019  150/150  94/5698/52  55.8±4.2/55.2±4.3  NA  NAFLD  Silymarin+Polyene phosphatidylcholine  Polyene phosphatidylcholine  12  TC, TG, ALT, AST 
Yang 2020  70/70  29/4130/40  46.05±13.89/45.82±14.05  NA  NAFLD  Silymarin+Polyene phosphatidylcholine  Polyene phosphatidylcholine  12  TC, TG, HDL-C, LDL-C, ALT, AST, FI, FBG, HOMA-IR 
Zhang 2018  50/50  29/2128/22  43.8±9.7/43.1±9.3  NA  NAFLD  Silymarin+Polyene phosphatidylcholine  Polyene phosphatidylcholine  24  TC, TG, ALT, AST 
Zhao 2008  50/34  NA  NA  NA  NAFLD  Silymarin+Polyene phosphatidylcholine  Polyene phosphatidylcholine  12  TC, ALT, AST 
Fig. 2.

Risk of bias of the included studies

(0,6MB).
3.3Effect of silymarin on energy metabolism3.3.1TC

TC was reported in 20 studies involving 1,891 patients. As shown in Fig. 3A, silymarin significantly decreased TC level in NAFLD patients (I2 = 93%, random-effects model; SMD = -0.85 (95% CI [-1.23, -0.47], P<0.0001). Subgroup analysis of the included studies revealed significant differences in TC levels between types of intervention [silymarin (95%CI = -1.03[-1.49, -0.58]) vs. silymarin complex (95%CI = -0.32[-0.64, 0.01]), P = 0.01], types of disease [NAFLD (SMD[95%CI] = -0.93[-1.33, -0.53]) vs. NASH (95%CI = -0.12[-0.40, 0.16]), P = 0.001], and treatment duration [<12 weeks (95%CI = -1.04[-1.43, -0.66]) vs. ≥12 to ≤24 weeks (95%CI = -0.86[-1.30, -0.42]) vs. >24weeks (95%CI = -0.22[-0.61, 0.18]), P = 0.010] (Figures S1-S3).

Fig. 3.

A: Meta-analysis of total cholesterol; B: Meta-analysis of triglycerides; C: Meta-analysis of high-density lipoprotein cholesterol; D: Meta-analysis of low-density lipoprotein cholesterol

(0,91MB).
3.3.2TG

Meta-analysis of TG (20 studies, n = 1,857) showed that TG was significantly different between the two groups [SMD = -0.62, (95% CI [-1.14, -0.10], P = 0.02, I2 = 96%, random-effects model]. Subgroup analysis revealed a significant difference in TG level between treatment duration [<12 weeks (95%CI = -2.47[-2.86, -2.09]) vs. ≥12 to ≤24 weeks (95%CI = -0.41[-0.95, 0.13]) vs. >24 weeks (95%CI = -0.51[-0.91, -0.11]), P<0.00001] (Figures S4-S6). These results suggested that silymarin can effectively reduce TG in NAFLD patients (Fig. 3B).

3.3.3HDL-C

Eleven studies involving 818 patients assessed HDL-C. Meta-analysis showed that HDL-C was significantly different between the two groups [SMD = 0.46 (95%CI [0.03, 0.89], P = 0.03, I2 = 88%, random-effects model]. Subgroup analysis demonstrated a significant difference in HDL-C level between types of intervention [silymarin (95%CI = -1.03[-1.49, -0.58]) vs. silymarin complex (95%CI = -0.32[-0.64, 0.01]), P = 0.03] (Figures S7-S9). These findings suggest that silymarin can increase HDL-C in NAFLD patients (Fig. 3C).

3.3.4LDL-C

Meta-analysis of 11 studies involving 817 patients showed that LDL-C level was significantly lower in the experimental group than in the control group [SMD = -0.81, (95% CI [-1.31, -0.31], P = 0.002, I2 = 91%, random-effects model)], suggesting that silymarin reduces LDL-C in NAFLD patients (Fig. 3D).

3.3.5FBG

FBG was reported in 7 studies (n = 537) and was significantly lower in the experimental group than in the control group [SMD = -0.09, (95% CI [-0.25, 0.08], P = 0.33, I2 = 0%, random-effects model)] (Fig. 4A).

Fig. 4.

A: Meta-analysis of fasting blood glucose; B: Meta-analysis of fasting insulin; C: Meta-analysis of homeostatic model assessment of IR

(0,53MB).
3.3.6FI

Meta-analysis showed that the pooled FI (2 studies, 160 patients) was significantly lower in the experimental group than in the control group [SMD = -0.59, (95% CI [-0.91, -0.28], P = 0.0002, I2 = 0%, random-effects model)], suggesting that silymarin can reduce FI level in NAFLD patients (Fig. 4B).

3.3.7HOMA-IR

HOMA-IR was pooled from 4 studies (n = 318) and was found to be significantly lower in the experimental group than in the control group [SMD = -0.37, (95% CI [-0.77, 0.04], P = 0.08, I2 = 64%, random-effects model)] (Fig. 4C).

3.4Effect of silymarin on liver injury3.4.1ALT

ALT was pooled from 23 studies (n = 2,138) and was found to be significantly lower in the experimental group than in the control group [SMD = -12.39 (95% CI [-19.69, -5.08], P<0.00001, I2 = 98%, random-effect model)], which implies that silymarin can reduce ALT level in NAFLD patients (Fig. 5A).

Fig. 5.

A: Meta-analysis of ALT; B: Meta-analysis of AST

(0,9MB).
3.4.2AST

Twenty-two studies with 2,091 patients reported AST. Meta-analysis showed that the experimental group had significantly lower AST level than the control group [SMD = -10.97 (95% CI [-15.51, -6.43], P<0.00001, I2 = 96%, random-effects model)], which demonstrates that silymarin lowers AST level in NAFLD patients (Fig. 5B).

3.5Effect on liver histological changes3.5.1Fatty liver index

Two studies containing 79 patients evaluated fatty liver index. Meta-analysis revealed a significantly lower fatty liver index in the experimental group than in the control group [SMD = -6.64 (95% CI [-10.59, -2.69], P = 0.0010, I2 = 16%, fixed-effect model)], which affirms that silymarin can reduce FLI in NAFLD patients (Fig. 6A).

Fig. 6.

A: Meta-analysis of fatty liver index; B: Meta-analysis of fatty liver score; C: Meta-analysis of hepatic steatosis grade

(0,49MB).
3.5.2Fatty liver score

Two studies involving 206 patients reported the fatty liver score, and meta-analysis revealed a significantly lower fatty liver score in the experimental group than in the control group [SMD = -0.51 (95% CI [-0.69, -0.33], P<0.00001, I2 = 0%, fixed-effects model)]. This suggests that silymarin can lower fatty liver score in NAFLD patients (Fig. 6C).

3.5.3Hepatic steatosis grade

Hepatic steatosis grade was pooled from 7 studies (n = 492) and was found to be significantly higher in the control group than in the experimental group [OR = 3.25, (95% CI [1.80, 5.87], P<0.0001, I2 = 0%, random-effects model)]. This result shows that silymarin can improve hepatic steatosis in NAFLD patients (Fig. 6D).

3.6Effect on anthropometric parameters3.6.1BMI

Seven studies with 416 patients assessed BMI. Meta-analysis showed that BMI was significantly lower in the experimental group than in the control group [SMD = -0.19 (95%CI [-0.39, 0.00], P = 0.05, I2 = 33%, fixed-effects model)], suggesting that silymarin can lower the BMI of NAFLD patients (Fig. 7A).

Fig. 7.

A: BMI; B: Meta-analysis of WC; C: Meta-analysis of HC

(0,52MB).
3.6.2WC

Meta-analysis of 3 studies (n = 148) showed that WC was similar between the two groups [SMD = 0.02 (95% CI [-0.31, 0.34], P = 0.92, I2 = 0%, fixed-effects model)] (Fig. 7B).

3.6.3HC

Similarly, HC (2 studies, 88 patients) was similar between the two groups [SMD = -0.16, (95% CI [-0.58, 0.26], P = 0.46, I2 = 0%, fixed-effects model)] (Fig. 7D).

3.7Sensitivity analysis and publication bias

Sensitivity analysis of all parameters showed that the results were robust. The funnel plots of TC, TG, HDL-C, HDL-C, ALT and AST were all symmetrical, indicating the absence of publication bias (Figures S10-S15).

4Discussion

The present study evaluated the effects of silymarin on energy metabolism, liver injury, liver histology and anthropometric parameters in NAFLD patients. Our results showed that silymarin plays a role in regulating energy metabolism, attenuating liver damage, and improving liver histology, and may, therefore, be a potential treatment for NAFLD.

NAFLD is a hepatic manifestation of dyslipidemia [4]. Elevated TC and TG levels are important risk factors for NAFLD [42], which is characterized by low HDL-C and high LDL-C levels [43,44]. Consistent with previous studies, we found that silymarin can improve blood HDL-C level and reduce blood TC, TG, LDL-C levels in NAFLD patients [45–48]. Insulin resistance and glucose metabolism dysfunction are typical clinical symptoms of NAFLD and metabolic syndrome [49], and elevated blood glucose can be found in 70-80% of NAFLD patients [1]. It was shown that silymarin can lower blood glucose, insulin and HOMA-IR [50,51], but the exact mechanism by which silymarin affects glucose levels is unclear. However, since silymarin is a powerful antioxidant, its effect on glucose levels may be mediated by inhibiting lipid peroxidation [52]. In addition, silymarin acts as an inhibitor of aldose reductase and can reduce insulin levels by inhibiting insulin secretion in response to glucose stimulation [53]. Silymarin was found to be effective in ameliorating insulin resistance in NAFLD mainly by reducing visceral fat, enhancing lipolysis, and suppressing gluconeogenesis [54]. We also found that silymarin can lower FI level and potentially FBG and HOMA-IR. The absence of significant differences in FBG and HOMA-IR may be related to the small number of included studies, and greater emphasis should be placed on the changes in FBG and HOMA-IR in subsequent studies to ascertain these findings. Collectively, our data indicate that silymarin may be involved in the regulation of energy metabolism of NAFLD patients. Although changes in energy metabolism have been suggested to affect the anthropometric parameters of NAFLD patients, we did not observe any significant differences in BMI, WC and HC between the experimental and control groups.

Silymarin also has a protective effect on the liver, as indicated by reduced AST and ALT levels in the experimental group. Elevated ALT and AST levels are biomarkers for liver injury and were shown to be associated with the occurrence and development of NAFLD. Furthermore, a higher ALT level is correlated with a higher incidence of NAFLD progression to cirrhosis or hepatocellular carcinoma [55,56]. It was reported that silymarin is a beneficial treatment for cirrhotic diabetic patients due to its ability to lower AST and ALT levels [57]. There are two mechanisms by which silymarin protects liver cells. First, silymarin protects intact liver cells or cells not yet irreversibly damaged by reducing oxidative stress and consequent cytotoxicity [58]. It can stabilize membrane permeability by suppressing lipid peroxidation, thereby maintaining the level of the antioxidant glutathione in the liver [59]. Second, silymarin exerts anti-inflammatory effects by inhibiting NF-κB to reduce inflammatory cytokine production in the liver parenchyma and interacting with protein kinase to downregulate cyclooxygenase-2 [58,60].

Furthermore, we found that silymarin can decrease fatty liver index and fatty liver score and improve hepatic steatosis grade in NAFLD patients. NAFLD is a clinicopathologic syndrome characterized by hepatic steatosis. Fatty liver index, fatty liver score and hepatic steatosis grade are often used to assess the severity of disease [61,62]. Oxidative stress, increased lipid peroxidation and decreased antioxidant status can all contribute to NAFLD progression. Silymarin was reported to protect the liver from oxidative stress, inflammation, steatosis, and fibrosis [63,64]. Because of the limited histological data, hepatic fibrosis was neither considered nor evaluated. In addition, silymarin downregulates the mRNA expression of enzymes responsible for de novo lipogenesis such as sterol-regulatory element binding protein (SREBP1c), fatty acid synthetase (FAS), and acetyl-CoA carboxylase 1 (ACC1), which consequently phosphorylates AMP-activated protein kinases in diabetic obese mice with NAFLD [65,66]. However, because of the limited histological data, hepatic fibrosis was neither considered nor evaluated.

Our subgroup analysis showed that TC and HDL-C levels are significantly different between different interventions. Silymarin alone was superior to silymarin complex in reducing TC and increasing HDL-C levels in NAFLD patients. Drug interactions may impair the efficacy of silymarin, and hence more consideration should be given to silymarin in the treatment of NAFLD. When using silymarin complex, care should be taken to ensure that the dosage of silymarin is sufficient and that other ingredients do not affect its efficacy. In addition, subgroup analysis based on type of disease revealed that silymarin has a greater effect on TC reduction in patients with NAFLD than in patients with NASH. NASH is a more severe form of NAFLD characterized not only by hepatic steatosis, but also by hepatic lobule inflammation, balloon-like changes in the liver cells, and fibrosis. Combined with the results of this study, silymarin should be initiated as early as possible to maximize its effect on delaying NAFLD progression [67]. Furthermore, TC and TG levels were also significantly different between different durations of treatment. Notably, the duration of treatment was 12-24 weeks in most included studies, and there were only few studies that examined a duration of <12 weeks or >24 weeks, which may result in bias in the results.

There are several limitations in this study. First, the types, the doses of silymarin and lifestyle management of the patients were different among the included RCTs. Second, the design of a few RCTs was not well standardized, which may affect the effectiveness of the evaluation. Furthermore, because of the limited histological data, hepatic fibrosis was neither considered nor evaluated. Last, different measurement methods used in each study may introduce bias in the results, such as when pooling the data for hepatic steatosis grade.

5Conclusions

Silymarin can regulate energy metabolism, attenuate liver damage and improve liver histology in NAFLD, and is thus a promising treatment for NAFLD. However, the results of this study should be interpreted with caution due to its limitations. Further studies with more adequate and proper methodological design are warranted to confirm these findings.

Availability of data and materials

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Funding

This study was supported by National famous traditional Chinese medicine Maudxi inherit- ance studio construction project (China Medical Office teaching Letter [2018] 119), Henan Province Chinese medicine scientific research special topic (2022ZY1167), National Clinical Research Base of Traditional Chinese Medicine, Henan Provincial Health Commission (2021 JDZX2014). The fund was not involved in any study design, data collection, analysis and interpretation, report writing, and article submission for publication.

CRediT authorship contribution statement

Shudi Li: Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Writing – original draft, Writing – review & editing. Fei Duan: Project administration, Resources, Supervision, Validation, Writing – original draft, Writing – review & editing. Suling Li: Conceptualization, Data curation, Software, Visualization, Writing – original draft, Writing – review & editing. Baoping Lu: Funding acquisition, Investigation, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing.

References
[1]
Younossi ZM, Golabi P, de Avila L, Paik JM, Srishord M, Fukui N, et al. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: a systematic review and meta-analysis. 2019;71(4):793-801.
[2]
Eslam M, Sarin SK, Wong VW-S, Fan J-G, Kawaguchi T, Ahn SH, et al. The Asian Pacific Association for the Study of the Liver clinical practice guidelines for the diagnosis and management of metabolic associated fatty liver disease. 2020;14:889-919.
[3]
Brunt EMJNrG, hepatology. Pathology of nonalcoholic fatty liver disease. 2010;7(4):195-203.
[4]
Vanni E, Bugianesi E, Kotronen A, De Minicis S, Yki-Järvinen H, Svegliati-Baroni GJD, et al. From the metabolic syndrome to NAFLD or vice versa? 2010;42(5):320-30.
[5]
Tarantino G, Finelli CJWjogW. What about non-alcoholic fatty liver disease as a new criterion to define metabolic syndrome? 2013;19(22):3375.
[6]
P Golabi, M Sayiner, Y Fazel, A Koenig, L Henry, ZM. Younossi.
Current complications and challenges in nonalcoholic steatohepatitis screening and diagnosis.
Expert Rev Gastroenterol Hepatol, 10 (2016), pp. 63-71
[7]
Gillessen A, Schmidt HH-JJAit. Silymarin as supportive treatment in liver diseases: a narrative review. 2020;37(4):1279-301.
[8]
L Abenavoli, M Milanović, N Milić, F Luzza, AM. Giuffrè.
Olive oil antioxidants and non-alcoholic fatty liver disease.
Expert Rev Gastroenterol Hepatol, 13 (2019), pp. 739-749
[9]
L Abenavoli, E Scarpellini, R Pellicano, S Fagoonee, T Larussa, F. Luzza.
Mediterranean diet and probiotics supplementation to treat non-alcoholic fatty liver disease.
Minerva Med, 111 (2020), pp. 526-528
[10]
L Abenavoli, N Milic, F Luzza, L Boccuto, A. De Lorenzo.
Polyphenols treatment in patients with nonalcoholic fatty liver disease.
J Transl Internal Med, 5 (2017), pp. 144-147
[11]
A-A Hajaghamohammadi, A Ziaee, R. Rafiei.
The efficacy of silymarin in decreasing transaminase activities in non-alcoholic fatty liver disease: a randomized controlled clinical trial.
Hepatitis Monthly, (2008),
[12]
Saller R, Brignoli R, Melzer J, Meier RJCMR. An updated systematic review with meta-analysis for the clinical evidence of silymarin. 2008;15(1):9-20.
[13]
Lazebnik L, Radchenko V, Golovanova YV, Zvenigorodskaya L, Konev YV, Seliverstov P, et al. Nonalcoholic fatty liver disease: clinic, diagnostics, treatment (guidelines for the specialists on internal medicine, 2nd version). 2017(3):6-23.
[14]
MM Ramírez-Mejía, X Qi, L Abenavoli, M Romero-Gómez, M Eslam, N. Méndez-Sánchez.
Metabolic dysfunction: the silenced connection with fatty liver disease.
[15]
Benedict M, Zhang XJWjoh. Non-alcoholic fatty liver disease: An expanded review. 2017;9(16):715.
[16]
Kim EJ, Kim B-h, Seo HS, Lee YJ, Kim HH, Son H-H, et al. Cholesterol-induced non-alcoholic fatty liver disease and atherosclerosis aggravated by systemic inflammation. 2014;9(6):e97841.
[17]
McCarty MFJMH. Potential utility of natural polyphenols for reversing fat-induced insulin resistance. 2005;64(3):628-35.
[18]
AJ. Sanyal.
AGA technical review on nonalcoholic fatty liver disease.
Gastroenterology, 123 (2002), pp. 1705-1725
[19]
R Aller, O Izaola, S Gómez, C Tafur, G González, E Berroa, et al.
Effect of silymarin plus vitamin E in patients with non-alcoholic fatty liver disease.
Random Clin Pilot Study, 19 (2015),
[20]
Edholm D, Kullberg J, Karlsson FA, Haenni A, Ahlström H, Sundbom MJSfo, et al. Changes in liver volume and body composition during 4 weeks of low calorie diet before laparoscopic gastric bypass. 2015;11(3):602-06.
[21]
Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. 2021;88:105906.
[22]
L Abenavoli, M Greco, I Nazionale, V Peta, N Milic, F Accattato, et al.
Effects of Mediterranean diet supplemented with silybin-vitamin E-phospholipid complex in overweight patients with non-alcoholic fatty liver disease.
Expert Rev Gastroenterol Hepatol, 9 (2015), pp. 519-527
[23]
A Anushiravani, N Haddadi, M Pourfarmanbar, V. Mohammadkarimi.
Treatment options for nonalcoholic fatty liver disease: A double-blinded randomized placebo-controlled trial.
Eur J Gastroenterol Hepatol, 31 (2019), pp. 613-617
[24]
Ataee Z, Vahabzadeh M, Kiapey SSM, Rahimi HR, Mozaffari HM, Ziaee MJHMJ. Triple therapy with garlic, silymarin and curcumin in non-alcoholic fatty liver disease: a randomized, placebo-controlled clinical trial. 2021;6(2):50-64.
[25]
Kheong CW, Mustapha NRN, Mahadeva SJCG, Hepatology. A randomized trial of silymarin for the treatment of nonalcoholic steatohepatitis. 2017;15(12):1940-49. e8.
[26]
Chiurazzi M, Cacciapuoti N, Di Lauro M, Nasti G, Ceparano M, Salomone E, et al. The synergic effect of a nutraceutical supplementation associated to a mediterranean hypocaloric diet in a population of overweight/obese adults with NAFLD. Nutrients 2022;14(22) doi: 10.3390/nu14224750
[27]
A Curcio, A Romano, S Cuozzo, A Di Nicola, O Grassi, D Schiaroli, et al.
Silymarin in combination with vitamin C, vitamin E, coenzyme Q10 and selenomethionine to improve liver enzymes and blood lipid profile in NAFLD patients.
[28]
F Famouri, M-M Salehi, N Rostampour, E Hashemi, A. Shahsanaee.
The effect of silymarin on non-alcoholic fatty liver disease of children.
J Herbmed Pharmacol, 6 (2016), pp. 16-20
[29]
Hashemi SJ, Hajiani E, Sardabi EH. A placebo-controlled trial of silymarin in patients with nonalcoholic fatty liver disease. 2009
[30]
Mirhashemi SH, Hakakzadeh A, Yeganeh FE, Oshidari B, Rezaee SP. Effect of 8 Weeks milk thistle powder (silymarin extract) supplementation on fatty liver disease in patients candidates for bariatric surgery. Metab Open 2022;14 doi: 10.1016/j.metop.2022.100190
[31]
VJ Navarro, SH Belle, M D'Amato, N Adfhal, EM Brunt, MW Fried, et al.
Silymarin in non-cirrhotics with non-alcoholic steatohepatitis: a randomized, double-blind, placebo controlled trial.
[32]
VKP Rajendra, S Kurapati, SK Balineni, Gogineni NTTJFFiH.
Disease. A blend of Sphaeranthus indicus flower head and Terminalia chebula fruit extracts reduces fatty liver and improves liver function in non-alcoholic.
Overweight Adults, 12 (2022),
[33]
KR Shaikh, S Shaikh, MA Ata, A Memon, UA Soomro, SS. Siddidui.
Therapeutic efficacy of Silymarin on liver aminotransferases in patients with nonalcoholic fatty liver disease.
Rawal Med J, 46 (2021), pp. 761-764
[34]
H Solhi, R Ghahremani, AM Kazemifar, Z Hoseini Yazdi.
Silymarin in treatment of non-alcoholic steatohepatitis: a randomized clinical trial.
Caspian J Internal Med, 5 (2014), pp. 9-12
[35]
L. Chen.
Effect of silybin in the treatment of non-alcoholic fatty liver disease.
China Med Eng, 20 (2012), pp. 141-145
[36]
Z Cui, C.
Effect of silybin combined with compound diisopropylamine dichloroacetate on nonalcoholic fatty liver.
Henan Med Res, 29 (2020), pp. 3172-3173
[37]
Y J, Y G, X H, Q L, X Z.
Clinical trial of bifid triple viable powder combined with silibinin capsules in the treatment of patients with non - alcoholic fatty liver disease.
Chin J Clin Pharmacol, 36 (2020), pp. 2212-2215
[38]
CL. Li.
Silybin combined with compound diisopropylamine dichloroacetate on the liver of patients with nonalcoholic fatty liver Effects of changes in function and lipid levels.
J North Pharm, 15 (2018), pp. 85-86
[39]
HQ Li, Y. Si.
Analysis of silybin in the treatment of noII—alcoholic steatohepatitis.
Chin J Prac Nerv Dis, 13 (2010), pp. 37-38
[40]
SL. Li.
To explore the therapeutic effect of polyene phosphatidylcholine combined with silybin in the treatment of nonalcoholic fatty liver.
J North Pharm, 15 (2018), pp. 65
[41]
CP Liang, QM Liu, YL. Zhong.
Efficacy of silybin combined with lovastatin in the treatment of nonalcoholic fatty liver.
J Mudanjiang Med Univ, 36 (2015), pp. 76-77
[42]
Zheng R-D, Zhuang Q-Y, Chen J-N, Chen J, Lu Y-HJZgZBzzZGZCJoH. Risk factors analysis of nonalcoholic fatty liver disease in Chinese men. 2013;21(1):62-65.
[43]
Alberti KG, Eckel RH, Grundy SM, Zimmet PZ, Cleeman JI, Donato KA, et al. Harmonizing the metabolic syndrome: a joint interim statement of the international diabetes federation task force on epidemiology and prevention; national heart, lung, and blood institute; American heart association; world heart federation; international atherosclerosis society; and international association for the study of obesity. 2009;120(16):1640-45.
[44]
Liangpunsakul S, Chalasani NJTAjotms. Unexplained elevations in alanine aminotransferase in individuals with the metabolic syndrome: results from the third National Health and Nutrition Survey (NHANES III). 2005;329(3):111-16.
[45]
Ebrahimpour-Koujan S, Gargari BP, Mobasseri M, Valizadeh H, Asghari-Jafarabadi MJP. Lower glycemic indices and lipid profile among type 2 diabetes mellitus patients who received novel dose of Silybum marianum (L.) Gaertn.(silymarin) extract supplement: A Triple-blinded randomized controlled clinical trial. 2018;44:39-44.
[46]
Fallahzadeh MK, Dormanesh B, Sagheb MM, Roozbeh J, Vessal G, Pakfetrat M, et al. Effect of addition of silymarin to renin-angiotensin system inhibitors on proteinuria in type 2 diabetic patients with overt nephropathy: a randomized, double-blind, placebo-controlled trial. 2012;60(6):896-903.
[47]
CHOU CH, CHEN YC, HSU MC, TSAI WL, CHANG CY, CHIU CHJJoFB. Effect of silymarin on lipid and alcohol metabolism in mice following long-term alcohol consumption. 2012;36(3):369-77.
[48]
Ozkaya M, Cakal E, Ustun Y, Engin-Ustun YJF, Sterility. Effect of metformin on serum visfatin levels in patients with polycystic ovary syndrome. 2010;93(3):880-84.
[49]
Deivanayagam S, Mohammed BS, Vitola BE, Naguib GH, Keshen TH, Kirk EP, et al. Nonalcoholic fatty liver disease is associated with hepatic and skeletal muscle insulin resistance in overweight adolescents. 2008;88(2):257-62.
[50]
Ghalandari K, Shabani M, Khajehlandi A, Mohammadi AJAoP, Biochemistry. Effect of aerobic training with silymarin consumption on glycemic indices and liver enzymes in men with type 2 diabetes. 2023;129(1):76-81.
[51]
Hussain SA-RJJomf. Silymarin as an adjunct to glibenclamide therapy improves long-term and postprandial glycemic control and body mass index in type 2 diabetes. 2007;10(3):543-47.
[52]
Maddux BA, See W, Lawrence Jr JC, Goldfine AL, Goldfine ID, Evans JLJD. Protection against oxidative stress—induced insulin resistance in rat L6 muscle cells by micromolar concentrations of α-lipoic acid. 2001;50(2):404-10.
[53]
F Lirussi, A Beccarello, G Zanette, A De Monte, V Donadon, M Velussi, et al.
Silybin-beta-cyclodextrin in the treatment of patients with diabetes mellitus and alcoholic liver disease.
Efficacy Study N Preparation Anti-Oxidant Agent, 15 (2002), pp. 222-231
[54]
Yao J, Zhi M, Gao X, Hu P, Li C, Yang XJBJoM, et al. Effect and the probable mechanisms of silibinin in regulating insulin resistance in the liver of rats with non-alcoholic fatty liver. 2013;46:270-77.
[55]
Natarajan Y, Kramer JR, Yu X, Li L, Thrift AP, El-Serag HB, et al. Risk of cirrhosis and hepatocellular cancer in patients with NAFLD and normal liver enzymes. 2020;72(4):1242-52.
[56]
Sogabe M, Okahisa T, Kurihara T, Takehara M, Kagemoto K, Okazaki J, et al. Differences among patients with and without nonalcoholic fatty liver disease having elevated alanine aminotransferase levels at various stages of metabolic syndrome. 2020;15(8):e0238388.
[57]
Velussi M, Cernigoi AM, Dapas F, Caffau C, Zilli MJJoh. Long-term (23 months) treatment with an anti-oxidant drug (silymarin) is effective on hyperinsulinemia, exogenous insulin need and malondialdehyde levels in cirrhotic diabetic patients. 1997;26(4):871-79.
[58]
Dehmlow C, Erhard J, de Groot HJH. Inhibition of Kupffer cell functions as an explanation for the hepatoprotective properties of silibinin. 1996;23(4):749-54.
[59]
Jung YS, Kim SJ, Kim YS, Choi DW, Kim YCJPm. Alterations in sulfur amino acid metabolism in mice treated with silymarin: a novel mechanism of its action involved in enhancement of the antioxidant defense in liver. 2013;79(12):997-1002.
[60]
Köksal E, Gülçin I, Beyza S, Sarikaya O, Bursal EJJoei, Chemistry m. In vitro antioxidant activity of silymarin 2009;24(2):395-405.
[61]
Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. 2020;73(1):202-09.
[62]
Powell EE, Wong VW-S, Rinella MJTL. Non-alcoholic fatty liver disease. 2021;397(10290):2212-24.
[63]
U Ahmad, J Akhtar, SP Singh, FJ Ahmad, S. Siddiqui.
Silymarin nanoemulsion against human hepatocellular carcinoma: development and optimization.
Artif Cells, Nanomed Biotechnol, 46 (2018), pp. 231-241
[64]
Ezhilarasan D, Karthikeyan S, Vivekanandan PJEt, Pharmacology. Ameliorative effect of silibinin against N-nitrosodimethylamine-induced hepatic fibrosis in rats. 2012;34(3):1004-13.
[65]
Sahin E, Bagci R, Bektur Aykanat NE, Kacar S, Sahinturk VJJofb. Silymarin attenuated nonalcoholic fatty liver disease through the regulation of endoplasmic reticulum stress proteins GRP78 and XBP-1 in mice. 2020;44(6):e13194.
[66]
Solomone F, Barbagallo I, Godos JJN. Silibinin restores NAD+ levels and induces the SIRT1/AMPK pathway in non-alcoholic fatty liver. 2017;9(10):1086.
[67]
Quek J, Chan KE, Wong ZY, Tan C, Tan B, Lim WH, et al. Global prevalence of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in the overweight and obese population: a systematic review and meta-analysis. 2023
Copyright © 2023. Fundación Clínica Médica Sur, A.C.
Opciones de artículo
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos