Next Article in Journal
How Did the Dietary Behavior of Older Korean Adults Change During the COVID-19 Pandemic?
Previous Article in Journal
An Evaluation of Food Allergy Management Practices in a Sample of Canadian and American Schools
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Curcumin for Inflammation Control in Individuals with Type 2 Diabetes Mellitus and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Randomized Controlled Trial

by
Metha Yaikwawong
1,
Laddawan Jansarikit
1,
Siwanon Jirawatnotai
1,2,3 and
Somlak Chuengsamarn
4,*
1
Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
2
Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
3
Faculty of Pharmacy, Silpakorn University, Nakhon Prathom 73000, Thailand
4
Division of Endocrinology and Metabolism, Faculty of Medicine, HRH Princess Maha Chakri Sirindhorn Medical Center, Srinakharinwirot University, Nakhon Nayok 26120, Thailand
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(12), 1972; https://doi.org/10.3390/nu17121972
Submission received: 13 May 2025 / Revised: 4 June 2025 / Accepted: 8 June 2025 / Published: 10 June 2025
(This article belongs to the Section Nutrition and Obesity)

Abstract

:
Background: Curcumin, a bioactive polyphenol derived from turmeric, has demonstrated potential therapeutic effects in metabolic dysfunction-associated steatotic liver disease (MASLD) by modulating inflammation, oxidative stress, hepatic fat accumulation, and fibrosis. Objective: To evaluate the efficacy of curcumin in reducing hepatic steatosis and liver stiffness in patients with MASLD. Methods: In this randomized, double-blind, placebo-controlled trial, 78 patients with type 2 diabetes mellitus (T2DM) and MASLD were randomly assigned to receive either curcumin (1500 mg/day) or placebo for 12 months. The primary outcome was the change in tumor necrosis factor (TNF) levels. Secondary outcomes included changes in interleukin-1 beta (IL-1β), interleukin-6 (IL-6), antioxidant enzyme activities (glutathione peroxidase, superoxide dismutase), the oxidative stress marker malondialdehyde, non-esterified fatty acids, and hepatic parameters (hepatic steatosis and liver stiffness). Assessments were conducted at baseline and at 3, 6, 9, and 12 months. Results: All participants completed the study (curcumin group: n = 39; placebo group: n = 39). Curcumin significantly reduced TNF levels at all follow-up points compared to placebo (p < 0.001). IL-1β, IL-6, and malondialdehyde levels also declined significantly (p < 0.001), while antioxidant enzyme activities, including glutathione peroxidase and superoxide dismutase, increased significantly (p < 0.001), indicating improved oxidative balance. Furthermore, curcumin led to significant reductions in non-esterified fatty acids, total body fat, BMI, hepatic steatosis, and liver stiffness compared to placebo. Conclusions: Twelve months of curcumin supplementation improved glycemic control, reduced systemic inflammation and oxidative stress, and significantly improved hepatic steatosis and liver stiffness in patients with MASLD. These findings support curcumin as a promising adjunctive therapy for MASLD management.

Graphical Abstract

1. Introduction

Metabolic dysfunction-associated steatotic liver disease (MASLD) is defined by hepatic steatosis in conjunction with at least one condition, such as obesity, overweight status, type 2 diabetes mellitus (T2DM), or laboratory markers of metabolic dysregulation [1]. The term MASLD was introduced in 2023 by an international expert panel as a replacement for nonalcoholic fatty liver disease. Its incidence has surged alongside the global increase in obesity, T2DM, and metabolic syndrome, making MASLD the most prevalent chronic liver disease worldwide [2,3]. It is also a growing contributor to severe liver conditions, including cirrhosis, liver failure, and hepatocellular carcinoma [4].
The pathogenesis of MASLD is multifactorial, involving both environmental influences and genetic predispositions [5]. Hepatic steatosis develops primarily from the accumulation of free fatty acids, which originate from systemic circulation and dietary sources. This process is further exacerbated by de novo hepatic lipogenesis. Disease progression features hepatic stress responses, oxidative stress, mitochondrial dysfunction, stellate cell activation, and alterations in the intestinal microbiota, all of which perpetuate liver damage [5]. Historically, liver fibrosis severity has been gauged through liver biopsy, an invasive method prone to sampling errors. Emerging noninvasive imaging techniques, such as magnetic resonance elastography and transient elastography (FibroScan), are increasingly used to measure liver stiffness, a key indicator of fibrosis severity.
Curcumin, the main curcuminoid in turmeric (Curcuma longa), has well-documented antioxidant and anti-inflammatory properties [6,7,8]. In vitro and in vivo studies show that curcumin can inhibit hepatic stellate cell activation by blocking leptin signaling, regulating glucose and lipid metabolism, and maintaining the balance of extracellular matrix synthesis and degradation [9]. Various animal models have demonstrated their anti-inflammatory, antifibrotic, and antihyperlipidemic effects [10,11].
Several randomized clinical trials have demonstrated that curcumin possesses hepatoprotective, antioxidant, anti-inflammatory, antidiabetic, and lipid-lowering properties in humans. Notably, most of these studies were of relatively short duration, typically around 8 weeks. For instance, Rahmani et al. (2016) conducted an 8-week randomized, double-blind, placebo-controlled trial in patients with non-alcoholic fatty liver disease (NAFLD), finding that curcumin supplementation significantly reduced liver fat content and improved metabolic parameters compared to placebo [12]. Similarly, Panahi et al. (2017) reported that 8 weeks of phytosomal curcumin supplementation led to significant improvements in liver enzymes and ultrasonographic findings in NAFLD patients [13]. These findings suggest that curcumin may be beneficial in managing NAFLD.
Despite these promising findings, the number of randomized clinical trials on curcumin supplementation remains limited. This study aimed to evaluate the efficacy of curcumin on inflammatory and hepatic parameters in patients with MASLD. It also assessed curcumin’s antioxidant and weight management effects through a double-blind, placebo-controlled design to determine its potential as a therapeutic intervention.

2. Methods

2.1. Study Design and Participants

This randomized, double-blind, placebo-controlled trial was conducted at the HRH Princess Maha Chakri Sirindhorn Medical Center, Srinakharinwirot University, Nakhon Nayok, Thailand. Data analysis and reporting followed the Consolidated Standards of Reporting Trials guidelines [14] (see Supplemental Figure S2). Seventy-eight patients with MASLD were recruited from a cohort of individuals with T2DM based on predefined inclusion and exclusion criteria. A detailed enrollment flow chart is provided in Supplemental Figure S2.

2.2. Inclusion Criteria

Participants were eligible if they were at least 35 years old and had hepatic steatosis confirmed by FibroScan (Echosens, Paris, France) with a controlled attenuation parameter (CAP) greater than 248 dB/m [15]. Additional criteria included no history of significant alcohol consumption (fewer than 20 g/day for women and fewer than 30 g/day for men) and no other liver disorders, malignancies, or kidney disorders. Individuals with a recent history of substantial weight loss or bariatric surgery were also excluded. All participants received metformin for hyperglycemia management, and other antidiabetic medications were excluded at recruitment to reduce potential confounding effects.

2.3. Exclusion Criteria

Exclusion criteria consisted of pregnancy or breastfeeding, participation in another clinical trial, hypersensitivity to the study supplement, uncontrolled hypertension, uncontrolled dyslipidemia, or unwillingness to continue the trial. Individuals with T2DM who required antidiabetic medications other than metformin were excluded, and no participants received insulin injections. Antidiabetic treatment regimens were not adjusted during the study. Hypertension and dyslipidemia were managed with stable antihypertensive and antidyslipidemic medications, respectively, with no changes permitted. Details on specific medications are provided in Table 1.

2.4. Study Protocol

The study design featured a 12-month intervention preceded by a 3-month run-in period (month −3 to month 0). During the run-in, all participants received standardized education on diet and exercise. They were also given written lifestyle recommendations and attended a 20–30 min one-on-one workshop emphasizing healthy lifestyle habits. After this baseline phase, participants underwent the 12-month intervention. They were advised to follow medical nutrition therapy guidelines and engage in regular physical activity. Metformin was administered throughout the study to manage hyperglycemia.

2.5. Ethics

Ethical approval was obtained from the Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand (SWUEC/FB 4/2556). The trial was registered with the Thai Clinical Trials Registry (TCTR20140303003) and conducted in accordance with the Declaration of Helsinki. All participants provided written informed consent prior to enrollment

2.6. Dietary and Exercise Recommendations

Before the study, participants were counseled on dietary and exercise guidelines that emphasized consuming low-glycemic index foods, such as legumes and whole grains, and increasing dietary fiber. They were also instructed to engage in at least 150 min of moderate-intensity aerobic exercise per week [16,17]. Participants were encouraged to maintain consistent physical activity throughout the trial. To assess dietary intake, they completed a questionnaire at baseline and again at 12 weeks. The questionnaires were analyzed using the Computer Dietary Guidance System Software (version 3.0; Peking Union Medical College & West China Center of Medical Sciences, Beijing, China). A baseline questionnaire further evaluated dietary habits, including the frequency and quantity of meat, milk, egg, and vegetable consumption (Supplemental Table S1).

2.7. Randomization Procedures

Following participant screening, informed consent, and completion of diet and lifestyle training, a random allocation sequence was generated using computer-based methods or random number tables. The sequence was concealed from individuals responsible for enrolling participants to prevent selection bias. Participants were then randomly assigned to the intervention or control group according to the concealed allocation sequence. Regular monitoring and verification were conducted to confirm the maintenance of blinding and to ensure consistent CAP and liver stiffness. Comprehensive documentation of the randomization process, allocation sequence, and group assignments was securely maintained for subsequent analysis.

2.8. Blinding Procedures

To maintain blinding among participants and healthcare providers, placebo capsules were designed to match curcumin capsules in appearance, taste, and administration method. All participants received their assigned treatments according to a standardized protocol, with blinding preserved throughout the study. Healthcare providers applied uniform procedures for both groups and followed standardized administration protocols, minimizing potential bias. Treatment containers were labeled with codes accessible only to the trial coordinator, and providers were trained to avoid revealing intervention details inadvertently. Regular follow-up assessments monitored outcomes and adverse effects, while compliance checks ensured adherence to trial protocols. All outcome data were systematically collected to minimize loss to follow-up and maintain data completeness. To evaluate the effectiveness of blinding, both participants and investigators were asked to guess the assigned intervention group (curcumin or placebo) at the conclusion of the study. The accuracy of these guesses was then compared to the expected probability of correct guesses under random chance (50%) using a two-sided exact binomial test.

2.9. Intervention

Participants consumed three blinded capsules of either curcumin (250 mg of curcuminoids per capsule) or placebo twice daily, for a total of six capsules per day over 12 months. Both curcumin and placebo capsules were manufactured by the Government Pharmaceutical Organization of Thailand. Capsule compliance was evaluated during follow-up visits at 3, 6, 9, and 12 months by recording capsule counts (see Supplemental Table S2).

2.10. Preparation of Curcuminoid Capsules

Turmeric (Curcuma longa Linn.) rhizomes from Kanchanaburi Province, Thailand, were dried, ground, and extracted with ethanol. The resulting semisolid ethanol extract contained oleoresin and curcuminoids. Oleoresin was removed to yield a curcuminoid extract containing 75–85% total curcuminoids. High-performance thin-layer chromatography was employed to determine peak ratios of curcumin, demethoxycurcumin, and bisdemethoxycurcumin. Each capsule contained 250 mg of this curcuminoid extract, which was encapsulated according to Good Manufacturing Practice standards [18]. Extracted chemical fingerprints and detailed compositions are presented in Supplemental Figure S2.

2.11. Study Outcomes

The primary outcome was a significant reduction in TNF levels in the curcumin group compared with the placebo group. The secondary outcomes included changes in CAP and liver stiffness, measured noninvasively by transient elastography (FibroScan 520 Touch; Echosens, Paris, France). To optimize measurement consistency, investigators underwent rigorous training in transient elastography, and standardized protocols were established for patient preparation and positioning. Patients fasted for at least 2 h before the procedure and were positioned supine with the right arm maximally abducted to allow optimal probe placement in the right mid-axillary line. Measurements were taken at the right liver lobe through intercostal spaces, with the probe maintained perpendicular to the liver capsule to ensure reliable readings. These steps enhanced the reproducibility of CAP and liver stiffness assessments.
Additional secondary outcomes included changes in waist circumference, total body fat (TBF), malondialdehyde, superoxide dismutase, glutathione peroxidase, and high-sensitivity C-reactive protein. Adverse effects were defined as elevated creatinine (≥1.2 mg/dL), aspartate transaminase or alanine transaminase levels that were at least three times the upper limit of normal, and any self-reported symptoms [19].

2.12. Data Collection and Measurement Methods

Measurements were conducted at baseline and at 3, 6, 9, and 12 months postintervention. Baseline data included demographic information, medical history, medication use, body weight, height, waist circumference, and vital signs. Waist circumference, an indicator of abdominal obesity, was measured horizontally between the iliac crest and the costal margin [20].
Body fat percentage (BFP) was estimated using equations developed by Xin Liu et al. [21], which incorporates body mass index (BMI), age, and waist circumference. The following equations have been validated in independent population-based samples of Chinese men and women, enhancing their applicability to Asian populations. The formulas are as follows:
For men:
BFP = 0.567 × BMI + 0.101 × age + 0.23 × waist circumference − 5.4
For women:
BFP = 0.439 × BMI + 0.221 × age + 0.18 × waist circumference − 9.4
After calculating BFP, TBF in kilograms was derived using the following equation:
TBF = (BFP × total body weight [kg])/100.
Fasting plasma glucose, glycated hemoglobin (HbA1c), aspartate aminotransferase, alanine aminotransferase, and gamma-glutamyl transferase (GGT) were measured using standard procedures. Plasma triglyceride (TG) levels were analyzed with diagnostic kits (Randox Laboratories Ltd., Antrim, UK) on an automated analyzer (spACE; Schiapparelli Biosystems Inc., Columbia, MD, USA).
Serum levels of pro-inflammatory cytokines—interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor (TNF)—were successfully measured using validated enzyme-linked immunosorbent assays (ELISAs). Blood samples were collected, processed, and stored according to standard protocols to ensure sample integrity. IL-1β was measured using the Human IL-1β ELISA Kit (Abcam, Cambridge, UK, catalog No. ab214025), which has a sensitivity of 14.06 pg/mL and intra- and inter-assay coefficients of variation (CVs) of 4.8% and 5.6%, respectively. IL-6 was assessed using the Human IL-6 ELISA Kit (Abcam, Cambridge, UK, catalog No. ab178013), with a sensitivity of 2 pg/mL and CVs of 4.2% (intra-assay) and 6.4% (inter-assay). TNF levels were determined using the Human TNF ELISA Kit (Abcam, Cambridge, UK, catalog No. ab181421), with a sensitivity of 4.32 pg/mL and intra- and inter-assay CVs of 2.5% and 3.1%, respectively.
Total serum antioxidant status was determined by an automated method developed by Erel, which evaluates antioxidative capacity against hydroxyl radical-initiated reactions [22]. Superoxide dismutase and glutathione peroxidase activities were measured calorimetrically with RANSOD and RANSEL kits (Randox Laboratories Ltd., Crumlin, UK) on an Abbott Alcyon 300 automated analyzer (Abbott Laboratories, Abbott Park, IL, USA). Malondialdehyde, a marker of lipid peroxidation and oxidative stress, was quantified by reacting with thiobarbituric acid to form a fluorescent complex; fluorescence intensity was measured at 547 nm (excitation 525 nm) using a Kontron SFM 25A spectrofluorometer (Kontron, Milan, Italy) [23]. Non-esterified fatty acids (NEFA) were measured enzymatically using a commercial kit (Wako Chemicals GmbH, Neuss, Germany).
Lipid accumulation product (LAP) was calculated by the following formulas [24]:
For men:
LAP = (waist circumference − 65) × triglycerides (TG) [mmol/L]
For women:
LAP = (waist circumference − 58) × TG [mmol/L]
Hepatic steatosis index (HSI) was computed using this formula [25]:
HSI = 8 × (ALT/AST ratio) + BMI (+ 2 for women).
Finally, the fatty liver index (FLI) was determined using the following equation [26]:
e0.953×ln(TG)+0.139×BMI+0.718×ln(GGT)+0.053×WC−15.745/1 + e0.953×ln(TG)+0.139×BMI+0.718×ln(GGT)+0.053×WC−15.745 × 100
where BMI is body mass index, GGT is gamma-glutamyl transferase, TG is triglycerides, and WC is waist circumference.

2.13. Sample Size

The initial sample size calculation was based on detecting a 2 pg/mL difference in mean serum TNF levels, with 80% statistical power (β = 0.20), requiring 21 participants per group [27]. While TNF is the primary endpoint, secondary outcomes such as liver stiffness are also critical for evaluating liver fibrosis. Based on prior studies evaluating changes in liver stiffness, a sample size of 34 participants was estimated to provide 90% power to detect a 3.1 kPa reduction in liver stiffness [28]. This underscores the necessity of tailoring sample size calculations to the specific effect sizes and variability expected in the secondary outcome. Sample size calculations were performed using G*Power software (version 3.1) [29].

2.14. Statistical Analysis

Baseline demographic data are reported as means (SD) for continuous variables or as numbers (percentages) for categorical variables. Outcome variables are presented as medians (interquartile range) at baseline, 3, 6, and 9 months for both groups. A per-protocol analysis was conducted. Between-group comparisons used the independent samples t-test for normally distributed data and the Mann-Whitney U test for non-normally distributed data. Categorical variables were analyzed using Fisher’s exact test or chi-square tests. Normality was assessed analytically via the Shapiro-Wilk test.
To assess potential sex-specific responses to curcumin supplementation, we conducted sex-stratified linear regression analyses on the change (Δ) in key outcomes over the 12-month intervention period. Each model was adjusted for baseline values and age to control for potential confounding factors.
The assessment of normality and all other statistical analyses were conducted using R software version 4.3.2 (R Foundation for Statistical Computing, Vienna, Austria) [30].

3. Results

3.1. Baseline Characteristics

Supplemental Figure S1 illustrates the trial flow chart, and Table 1 presents the baseline characteristics of all 78 randomized participants. There were no significant differences between the two groups at baseline. The intervention outcomes were assessed in several domains, including anti-inflammatory and antioxidant effects, changes in non-esterified fatty acid levels, anthropometric measurements, glycemic control, hepatic steatosis, liver stiffness, and potential adverse effects.
Additionally, Supplemental Table S2 summarizes the mean daily nutrient intake—covering total energy intake (kcal/day), macronutrient distribution (percentage of energy from carbohydrates, protein, and fat), and fiber intake (g/day)—assessed at baseline and after 12 months using validated dietary questionnaires. No significant between-group differences were observed at either time point, indicating that dietary intake remained stable throughout the study. These findings support the comparability of lifestyle factors between groups and reduce the likelihood that dietary variation confounded the observed effects of the intervention.

3.2. Anti-Inflammatory Effects

Compared with placebo, curcumin significantly reduced pro-inflammatory cytokines, including TNF, IL-1β, and IL-6, at 3, 6, 9, and 12 months (Table 2).

3.3. Antioxidant Effects

Curcumin significantly increased glutathione peroxidase activity compared with placebo (Table 2). Superoxide dismutase activity was also significantly higher in the curcumin group at 3, 6, 9, and 12 months. In contrast, malondialdehyde, a marker of oxidative stress and a potential contributor to MASLD progression, was significantly lower in the curcumin group than in the placebo group at these same time points (Table 2).

3.4. Non-Esterified Fatty Acid Levels

Curcumin significantly reduced NEFA levels—a key driver of hepatic fat accumulation and MASLD pathogenesis—compared to placebo at 6 months (Table 2), suggesting enhanced liver metabolic function and reduced lipotoxicity.

3.5. Anthropometric Measurements and Weight Management

Waist circumference and TBF were significantly lower in the curcumin group than in the placebo group at 6, 9, and 12 months. BMI was also significantly reduced in the curcumin group at 12 months (Table 2).

3.6. Glycemic Control Effects

The median values of diabetes-related blood chemistries, including HbA1c and fasting plasma glucose, were significantly lower in the curcumin group than in the placebo group at 6, 9, and 12 months (Table 2).

3.7. CAP and Liver Stiffness

The CAP was significantly lower in the curcumin group than in the placebo group at 3, 6, 9, and 12 months (Table 2). Liver stiffness was also significantly reduced in the curcumin group compared with the placebo group at all follow-up visits (Table 2).

3.8. Fatty Liver-Associated Indicators

The FLI was significantly lower in the curcumin group at 3, 6, 9, and 12 months (Table 2). The LAP was also notably lower in the curcumin group at all time points. In addition, the HSI showed significant reductions in the curcumin group at 3, 6, 9, and 12 months (Table 2).

3.9. Sex-Stratified Analyses

Curcumin supplementation significantly improved inflammatory markers, oxidative stress, anthropometric indices, metabolic profiles, and hepatic parameters in both male and female participants. Comprehensive results are presented in the Supplemental Table S2.

3.10. Blinding Assessment

Both participants and investigators were asked to guess the assigned treatment group at the end of the study. As shown in the Supplemental Table S3, the accuracy of these guesses did not significantly differ from random chance. Among participants, 48.7% correctly identified their group allocation (p = 0.84), and among investigators, 52.6% guessed correctly (p = 0.73). These results suggest that blinding was effectively preserved throughout the trial.

3.11. Adverse Effects

To assess the potential adverse effects of curcumin, kidney and liver function tests were conducted (Supplementary Table S4). No significant differences were found between the groups in aspartate transaminase, alanine transaminase, or creatinine levels. Additionally, no episodes of hypoglycemia were reported in the curcumin group.

3.12. Overall Safety and Compliance

Curcumin extract appeared safe for at least 12 months of use. Capsule consumption rates were similar in both groups, indicating comparable compliance (Supplemental Table S5). Consequently, the observed effects do not appear to reflect differential adherence.

4. Discussion

MASLD affects approximately 30% of the global population and is a major contributor to cirrhosis and hepatocellular carcinoma [31]. This condition is especially common among overweight or obese individuals, with a global prevalence of nearly 50%. In patients with MASLD and metabolic-associated steatohepatitis, the prevalence of T2DM is about 22.5%. Moreover, up to 80% of individuals with MASLD may develop new-onset T2DM [32,33]. The synergistic relationship between MASLD and T2DM exacerbates adverse outcomes beyond what either condition alone would cause. Insulin resistance, which underlies T2DM, compromises glucose uptake in insulin-sensitive tissues such as the liver and adipose tissue [34]. This disruption in glycolipid homeostasis substantially contributes to MASLD progression [35].
Curcumin, known for its cardiovascular protective effects, is regarded as a promising nutraceutical [36]. This study focused on ethanol-extracted curcumin as an intervention to explore a safe, well-tolerated, and widely accessible option for atherosclerosis prevention in patients with T2DM [37].
This double-blind, placebo-controlled trial used the FibroScan 520 Touch, a noninvasive device that measures hepatic steatosis via the CAP [38]. Our findings revealed that 3 months of curcumin supplementation significantly decreased pro-inflammatory cytokines—TNF, IL-1β, and IL-6—compared with placebo. These results underscore curcumin’s pronounced anti-inflammatory effects. Additionally, curcumin’s antioxidant properties were assessed by measuring glutathione peroxidase and superoxide dismutase activities, as well as malondialdehyde levels, a marker of lipid peroxidation. Markers of abdominal obesity (waist circumference) and overall adiposity (TBF and BMI) were also evaluated.
To ensure accurate interpretation of curcumin’s effects, individuals with uncontrolled T2DM were excluded, as changes in their antihyperglycemic regimens could have confounded the findings. Notably, despite well-controlled T2DM, the placebo group exhibited elevated pro-inflammatory cytokine levels, reflecting the chronic inflammatory milieu characteristic of T2DM. This observation underscores curcumin’s potential for mitigating inflammation and metabolic dysregulation in this patient population.
Curcumin’s anti-inflammatory and antioxidant properties have been widely investigated in both in vitro and in vivo models [39,40,41]. Studies show that curcumin reduces IL-1β [42], IL-6 [43], and TNF [43], key cytokines involved in the pathogenesis of MASLD. Excessive levels of these cytokines exacerbate hepatic inflammation and lipid accumulation, thereby advancing MASLD [44,45]. Furthermore, MASLD is marked by increased oxidative stress, lipid peroxidation, and diminished antioxidant defenses [46]. Lipid peroxidation produces inflammatory mediators, including malondialdehyde, a well-established biomarker of oxidative stress [47].
In our study, a 3-month curcumin intervention significantly reduced TNF, IL-1β, and IL-6 levels in patients with MASLD. These results align with those reported by Saadati et al., who observed lower TNF levels after a 12-week curcumin supplementation in patients with non-alcoholic fatty liver disease [48]. Jazayeri-Tehrani et al. similarly noted decreased TNF and IL-6 levels following 3 months of nanocurcumin administration [49], while Ghaffari et al. documented reduced IL-6 after a 10-week turmeric regimen [50].
Our findings also demonstrated that curcumin supplementation substantially lowered malondialdehyde levels and bolstered antioxidant defenses, as evidenced by increased glutathione peroxidase and superoxide dismutase activities [51]. These observations corroborate the work of Ghaffari et al., who reported elevated total serum antioxidant capacity and diminished malondialdehyde levels after turmeric supplementation in non-alcoholic fatty liver disease patients [50].
Additionally, in vivo studies indicate that curcumin supplementation reduces NEFA levels [52,53]. In our study, curcumin significantly lowered NEFA levels after 6 months of intervention in patients with MASLD. These findings align with those of Li-Xin et al., who reported decreased total serum free fatty acid and malondialdehyde levels in curcumin-treated individuals with T2DM [54]. Elevated NEFAs contribute to hepatic lipid accumulation, promote lipotoxicity, and increase oxidative stress and inflammation in hepatocytes, thereby exacerbating metabolic dysfunction. NEFAs thus play a critical role in the metabolic disturbances that underlie MASLD pathogenesis [55].
Curcumin supplementation also led to substantial reductions in waist circumference, TBF, and BMI. Jazayeri-Tehrani et al. similarly observed reduced waist circumference with nanocurcumin, although they did not find a significant change in BMI [49]. In contrast, our study noted a clear BMI reduction, consistent with a meta-analysis of randomized controlled trials showing significant decreases in body weight and BMI following curcumin supplementation [56]. Although the exact mechanisms remain under investigation, curcumin’s anti-inflammatory properties may contribute to BMI reduction. While acute inflammation can lead to increased energy expenditure and weight loss.
While inflammation is typically associated with increased energy expenditure and weight loss in conditions like cachexia, obesity is characterized by chronic low-grade inflammation that contributes to insulin resistance and metabolic dysregulation, thereby hindering weight loss [57,58]. Curcumin’s anti-inflammatory properties may help restore metabolic balance by improving insulin sensitivity, modulating adipokines, and regulating lipid metabolism [59]. Clinical trials have demonstrated that curcumin supplementation can reduce body weight and BMI, even in settings with controlled lifestyle interventions [59,60].
Furthermore, participants receiving curcumin exhibited significant reductions in HbA1c and fasting plasma glucose at 6, 9, and 12 months compared with placebo. Chronic hyperglycemia activates the NF-κB pathway, leading to the production of pro-inflammatory cytokines such as TNF, IL-1β, and IL-6 [61]. Curcumin effectively suppressed TNF levels and improved glycemic control by lowering fasting glucose and insulin levels, independent of weight loss [62]. These findings suggest that curcumin may serve as a valuable adjunct for weight management and glycemic control in individuals with obesity. However, further studies are needed to clarify its mechanisms and optimize its therapeutic use.
Curcumin’s anti-inflammatory and antioxidant properties may underlie its beneficial metabolic effects, including reductions in waist circumference, TBF, visceral fat, and BMI. These changes could explain the observed decreases in CAP. In our study, 12 months of curcumin supplementation significantly lowered CAP. Similarly, Saadati et al. reported a marked reduction in hepatic steatosis following a 12-week curcumin intervention [48]. Our findings also showed that curcumin improved fatty liver-related indices—HSI, FLI, and LAP—after 3 months of treatment. These results align with those of Cicero et al., who documented improvements in HSI, FLI, and LAP after 56 days of curcumin therapy [63]. Naseri et al. likewise found reductions in these indices following 12 weeks of supplementation [64].
We observed significant reductions in liver stiffness. Although liver stiffness is a widely used non-invasive marker of hepatic fibrosis, it is also sensitive to hepatic inflammation. Therefore, elevated liver stiffness may reflect inflammatory activity rather than true fibrotic changes, and the observed reductions may primarily indicate improvements in inflammation rather than definitive fibrosis regression [65]. Curcumin’s well-established anti-inflammatory properties likely contributed to these changes [66]. However, as liver stiffness is influenced by both fibrosis and inflammation, the precise nature of the improvements remains uncertain without histological confirmation.
Regarding safety, curcumin has been well tolerated in humans at doses up to 8000 mg/day, with no significant toxicity observed [67]. This aligns with the 1500 mg/day dosage in our study, which did not cause severe adverse effects. Although multiple randomized clinical trials have highlighted curcumin’s potential for managing MASLD [48,49,50], most interventions lasted only 12 weeks and lacked rigorous safety evaluations. By extending the follow-up to 12 months and systematically assessing adverse effects, our study addressed these limitations. Our findings suggest that curcumin supplementation effectively attenuates inflammatory and oxidative stress pathways, evidenced by reduced pro-inflammatory cytokines and enhanced antioxidant enzyme activity.
It is noteworthy that our study included a higher proportion of women than is typically reported in MASLD research, a condition generally more prevalent in men [68,69]. This discrepancy may stem from the inclusion of postmenopausal women, who experience hormonal changes which may elevate MASLD prevalence to rates potentially exceeding those in men [70]. Despite the female-dominant cohort, sex-stratified analyses revealed that curcumin supplementation consistently improved inflammatory markers, oxidative stress, anthropometric measures, metabolic parameters, and hepatic steatosis in both sexes. These effects remained significant after adjusting for age, suggesting that curcumin’s therapeutic benefits are not sex-dependent. Nonetheless, future studies with larger, more sex-balanced populations are needed to validate these findings and explore potential sex-specific responses.
This study has several strengths, including an extended follow-up period and comprehensive assessments of inflammatory cytokines and antioxidant markers, both of which are vital for understanding metabolism and MASLD pathophysiology. Nevertheless, several limitations warrant discussion. First, MASLD was not histologically confirmed, as liver biopsies were not performed. While non-invasive methods like FibroScan provide quantitative measures of liver stiffness and steatosis, respectively, they may not fully capture subtle histological changes, potentially leading to misclassification, especially in cases where such changes are minimal yet clinically significant [71]. Second, the sample size was relatively small. Third, the single-dose design precluded analysis of dose–response relationships [72]. Lastly, as a single-center randomized controlled trial, the findings may not be generalizable to other populations and settings.

5. Conclusions

Curcumin, a bioactive compound in turmeric, exhibits promising hepatoprotective effects through its anti-inflammatory and antioxidant properties. Our findings support the use of curcumin supplementation as a potential therapeutic strategy for MASLD. Given the lack of FDA-approved treatments specifically for MASLD, incorporating curcumin into clinical management may offer a safe and effective alternative for disease prevention and treatment.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nu17121972/s1, Figure S1: CONSORT 2010 checklist for reporting a randomized trial; Figure S2: CONSORT flow diagram of participant enrollment and allocation; Figure S3: High–performance thin–layer chromatography profiles of curcuminoid extracts compared with standard curcuminoid chromatograms; Table S1: Mean daily nutrient intake of participants at baseline and 12 months by group; Table S2: Sex–stratified linear regression analysis of curcumin effect on change in outcomes at 12 months; Table S3: Accuracy of blinding guesses by participants and onvestigators; Table S4: Renal and hepatic adverse effects of curcumin and placebo at each follow–up visit; Table S5: Capsule consumption per 3–month interval and daily intake at follow–up visits. Reference [14] was cited in Supplementary Materials.

Author Contributions

S.C. conceived the study, screened and examined all recruited subjects, performed data analysis, and reviewed and edited the manuscript. M.Y. conducted data analysis and wrote the manuscript. L.J. contributed to data research. S.J. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a joint grant from the Thailand Research Fund (grant number 022/2012) and the Department for Development of Thai Traditional and Alternative Medicine, Ministry of Public Health (grant number 29/2013, awarded to S.C.).

Institutional Review Board Statement

This study commenced after obtaining approval from the approved by the Ethics Committee of the Faculty of Medicine of Srinakharinwirot University, Bangkok, Thailand (SWUEC/FB 4/2556, approval date: 22 February 2013). The study was registered in Thai clinical trials regentrify (TCTR ID: TCTR20140303003, registered date: 3 March 2014).

Informed Consent Statement

Informed written consent was obtained from participants after informing them about the benefits and risks of the study. Autonomy was maintained by study participants as participation in the trial.

Data Availability Statement

The original contributions presented in the study are included in the article, further inquiries can be directed to the corresponding author.

Acknowledgments

The authors thank the Thai Government Pharmaceutical Organization for providing the curcumin extract and placebo used in this study. We also extend our appreciation to all participants and the medical team at the outpatient clinic of HRH Princess Maha Chakri Sirindhorn Medical Center, Srinakharinwirot University, Nakhon Nayok, Thailand, for their invaluable contributions.

Conflicts of Interest

All authors have no conflicts of interest to declare. The funders had no role in the design.

Abbreviations

BFPbody fat percentage
BMIbody mass index
CAPcontrolled attenuation parameter
GGTgamma-glutamyl transferase
GPxglutathione peroxidase
FLIfatty liver index
HbA1cglycated hemoglobin
HSIhepatic steatosis index
IL-1βinterleukin-1 beta
IL-6interleukin-6
IQRinterquartile range
LAPlipid accumulation product
MASLDmetabolic dysfunction-associated steatotic liver disease
MDAmalondialdehyde
NEFAnon-esterified fatty acids
SODsuperoxide dismutase
T2DMtype 2 diabetes mellitus
TBFtotal body fat
TGtriglycerides
TNFtumor necrosis factor
WCwaist circumference

References

  1. Rinella, M.E.; Lazarus, J.V.; Ratziu, V.; Francque, S.M.; Sanyal, A.J.; Kanwal, F.; Romero, D.; Abdelmalek, M.F.; Anstee, Q.M.; Arab, J.P. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. Hepatology 2023, 78, 1966–1986. [Google Scholar] [CrossRef] [PubMed]
  2. El-Kassas, M.; Cabezas, J.; Coz, P.I.; Zheng, M.-H.; Arab, J.P.; Awad, A. Nonalcoholic fatty liver disease: Current global burden. Semin. Liver Dis. 2022, 42, 401–412. [Google Scholar] [CrossRef]
  3. Younossi, Z.M.; Golabi, P.; Paik, J.M.; Henry, A.; Van Dongen, C.; Henry, L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): A systematic review. Hepatology 2023, 77, 1335–1347. [Google Scholar] [CrossRef]
  4. Younossi, Z.M.; Henry, L. Epidemiology of non-alcoholic fatty liver disease and hepatocellular carcinoma. JHEP Rep. 2021, 3, 100305. [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. Dattani, J.J.; Rajput, D.K.; Moid, N.; Highland, H.N.; George, L.B.; Desai, K.R. Ameliorative effect of curcumin on hepatotoxicity induced by chloroquine phosphate. Environ. Toxicol. Pharmacol. 2010, 30, 103–109. [Google Scholar] [CrossRef]
  7. Tang, Y.; Zheng, S.; Chen, A. Curcumin eliminates leptin’s effects on hepatic stellate cell activation via interrupting leptin signaling. Endocrinology 2009, 150, 3011–3020. [Google Scholar] [CrossRef]
  8. Jang, E.M.; Choi, M.S.; Jung, U.J.; Kim, M.J.; Kim, H.J.; Jeon, S.M.; Shin, S.K.; Seong, C.N.; Lee, M.K. Beneficial effects of curcumin on hyperlipidemia and insulin resistance in high-fat-fed hamsters. Metabolism 2008, 57, 1576–1583. [Google Scholar] [CrossRef]
  9. Tang, Y. Curcumin targets multiple pathways to halt hepatic stellate cell activation: Updated mechanisms in vitro and in vivo. Dig. Dis. Sci. 2015, 60, 1554–1564. [Google Scholar] [CrossRef]
  10. Vizzutti, F.; Provenzano, A.; Galastri, S.; Milani, S.; Delogu, W.; Novo, E.; Caligiuri, A.; Zamara, E.; Arena, U.; Laffi, G.; et al. Curcumin limits the fibrogenic evolution of experimental steatohepatitis. Lab. Investig. 2010, 90, 104–115. [Google Scholar] [CrossRef]
  11. Ramirez-Tortosa, M.C.; Ramirez-Tortosa, C.L.; Mesa, M.D.; Granados, S.; Gil, A.; Quiles, J.L. Curcumin ameliorates rabbits’s steatohepatitis via respiratory chain, oxidative stress, and TNF-alpha. Free Radic. Biol. Med. 2009, 47, 924–931. [Google Scholar] [CrossRef] [PubMed]
  12. Rahmani, S.; Asgary, S.; Askari, G.; Keshvari, M.; Hatamipour, M.; Feizi, A.; Sahebkar, A. Treatment of non-alcoholic fatty liver disease with curcumin: A randomized placebo-controlled trial. Phytother. Res. 2016, 30, 1540–1548. [Google Scholar] [CrossRef] [PubMed]
  13. Panahi, Y.; Kianpour, P.; Mohtashami, R.; Jafari, R.; Simental-Mendía, L.E.; Sahebkar, A. Efficacy and safety of phytosomal curcumin in non-alcoholic fatty liver disease: A randomized controlled trial. Drug Res. 2017, 67, 244–251. [Google Scholar] [CrossRef] [PubMed]
  14. Schulz, K.F.; Altman, D.G.; Moher, D.; for the CONSORT Group. CONSORT 2010 Statement: Updated guidelines for reporting parallel group randomised trials. BMC Med. 2010, 8, 18. [Google Scholar] [CrossRef]
  15. Petta, S.; Wong, V.W.; Camma, C.; Hiriart, J.B.; Wong, G.L.; Marra, F.; Vergniol, J.; Chan, A.W.; Di Marco, V.; Merrouche, W.; et al. Improved noninvasive prediction of liver fibrosis by liver stiffness measurement in patients with nonalcoholic fatty liver disease accounting for controlled attenuation parameter values. Hepatology 2017, 65, 1145–1155. [Google Scholar] [CrossRef]
  16. American Diabetes Association. Executive summary: Standards of medical care in diabetes—2013. Diabetes Care 2013, 36 (Suppl. S1), S4–S10. [Google Scholar] [CrossRef]
  17. Brand, J.C.; Colagiuri, S.; Crossman, S.; Allen, A.; Roberts, D.C.; Truswell, A.S. Low-Glycemic Index Foods Improve Long-Term Glycemic Control in NIDDM. Diabetes Care 1991, 14, 95–101. [Google Scholar] [CrossRef]
  18. World Health Organization. Annex 3: Supplementary Guidelines on Good Manufacturing Practices for the Manufacture of Herbal Medicines. In WHO Technical Report Series, No. 937; World Health Organization: Geneva, Switzerland, 2006. [Google Scholar]
  19. Chainani-Wu, N. Safety and anti-inflammatory activity of curcumin: A component of tumeric (Curcuma longa). J. Altern. Complement. Med. 2003, 9, 161–168. [Google Scholar] [CrossRef]
  20. Ross, R.; Neeland, I.J.; Yamashita, S.; Shai, I.; Seidell, J.; Magni, P.; Santos, R.D.; Arsenault, B.; Cuevas, A.; Hu, F.B. Waist circumference as a vital sign in clinical practice: A Consensus Statement from the IAS and ICCR Working Group on Visceral Obesity. Nat. Rev. Endocrinol. 2020, 16, 177–189. [Google Scholar] [CrossRef]
  21. Liu, X.; Sun, Q.; Sun, L.; Zong, G.; Lu, L.; Liu, G.; Rosner, B.; Ye, X.; Li, H.; Lin, X. The development and validation of new equations for estimating body fat percentage among Chinese men and women. Br. J. Nutr. 2015, 113, 1365–1372. [Google Scholar] [CrossRef]
  22. Erel, O. A novel automated direct measurement method for total antioxidant capacity using a new generation, more stable ABTS radical cation. Clin. Biochem. 2004, 37, 277–285. [Google Scholar] [CrossRef] [PubMed]
  23. Del Rio, D.; Pellegrini, N.; Colombi, B.; Bianchi, M.; Serafini, M.; Torta, F.; Tegoni, M.; Musci, M.; Brighenti, F. Rapid fluorimetric method to detect total plasma malondialdehyde with mild derivatization conditions. Clin. Chem. 2003, 49, 690–692. [Google Scholar] [CrossRef] [PubMed]
  24. Bedogni, G.; Kahn, H.S.; Bellentani, S.; Tiribelli, C. A simple index of lipid overaccumulation is a good marker of liver steatosis. BMC Gastroenterol. 2010, 10, 98. [Google Scholar] [CrossRef] [PubMed]
  25. Lee, J.H.; Kim, D.; Kim, H.J.; Lee, C.H.; Yang, J.I.; Kim, W.; Kim, Y.J.; Yoon, J.H.; Cho, S.H.; Sung, M.W.; et al. Hepatic steatosis index: A simple screening tool reflecting nonalcoholic fatty liver disease. Dig. Liver Dis. 2010, 42, 503–508. [Google Scholar] [CrossRef]
  26. Bedogni, G.; Bellentani, S.; Miglioli, L.; Masutti, F.; Passalacqua, M.; Castiglione, A.; Tiribelli, C. The Fatty Liver Index: A simple and accurate predictor of hepatic steatosis in the general population. BMC Gastroenterol. 2006, 6, 33. [Google Scholar] [CrossRef]
  27. Eslamparast, T.; Poustchi, H.; Zamani, F.; Sharafkhah, M.; Malekzadeh, R.; Hekmatdoost, A. Synbiotic supplementation in nonalcoholic fatty liver disease: A randomized, double-blind, placebo-controlled pilot study123. Am. J. Clin. Nutr. 2014, 99, 535–542. [Google Scholar] [CrossRef]
  28. Cho, H.; Lee, Y.B.; Ha, Y.; Chon, Y.E.; Kim, M.N.; Lee, J.H.; Park, H.; Rim, K.S.; Hwang, S.G. Changes in liver stiffness values assessed using transient elastography in chronic hepatitis B patients treated with tenofovir disoproxil fumarate: A prospective observational study. BMC Gastroenterol. 2023, 23, 210. [Google Scholar] [CrossRef]
  29. Kang, H. Sample size determination and power analysis using the G* Power software. J. Educ. Eval. Health Prof. 2021, 18, 17. [Google Scholar] [CrossRef]
  30. R Core Team. R: A Language and Environment for Statistical Computing; R Foundation for Statistical Computing: Vienna, Austria, 2013. [Google Scholar]
  31. Paik, J.M.; Henry, L.; Younossi, Y.; Ong, J.; Alqahtani, S.; Younossi, Z.M. The burden of nonalcoholic fatty liver disease (NAFLD) is rapidly growing in every region of the world from 1990 to 2019. Hepatol. Commun. 2023, 7, e0251. [Google Scholar] [CrossRef]
  32. Diaconu, C.T.; Guja, C. Nonalcoholic Fatty Liver Disease and Its Complex Relation with Type 2 Diabetes Mellitus-From Prevalence to Diagnostic Approach and Treatment Strategies. J. Clin. Med. 2022, 11, 5144. [Google Scholar] [CrossRef]
  33. Liang, Y.; Chen, H.; Liu, Y.; Hou, X.; Wei, L.; Bao, Y.; Yang, C.; Zong, G.; Wu, J.; Jia, W. Association of MAFLD With Diabetes, Chronic Kidney Disease, and Cardiovascular Disease: A 4.6-Year Cohort Study in China. J. Clin. Endocrinol. Metab. 2022, 107, 88–97. [Google Scholar] [CrossRef] [PubMed]
  34. Lee, S.H.; Park, S.Y.; Choi, C.S. Insulin Resistance: From Mechanisms to Therapeutic Strategies. Diabetes Metab. J. 2022, 46, 15–37. [Google Scholar] [CrossRef] [PubMed]
  35. Yki-Jarvinen, H.; Luukkonen, P.K.; Hodson, L.; Moore, J.B. Dietary carbohydrates and fats in nonalcoholic fatty liver disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 770–786. [Google Scholar] [CrossRef] [PubMed]
  36. Li, H.; Sureda, A.; Devkota, H.P.; Pittalà, V.; Barreca, D.; Silva, A.S.; Tewari, D.; Xu, S.; Nabavi, S.M. Curcumin, the golden spice in treating cardiovascular diseases. Biotechnol. Adv. 2020, 38, 107343. [Google Scholar] [CrossRef]
  37. Chuengsamarn, S.; Rattanamongkolgul, S.; Phonrat, B.; Tungtrongchitr, R.; Jirawatnotai, S. Reduction of atherogenic risk in patients with type 2 diabetes by curcuminoid extract: A randomized controlled trial. J. Nutr. Biochem. 2014, 25, 144–150. [Google Scholar] [CrossRef]
  38. Eddowes, P.J.; Sasso, M.; Allison, M.; Tsochatzis, E.; Anstee, Q.M.; Sheridan, D.; Guha, I.N.; Cobbold, J.F.; Deeks, J.J.; Paradis, V.; et al. Accuracy of FibroScan Controlled Attenuation Parameter and Liver Stiffness Measurement in Assessing Steatosis and Fibrosis in Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology 2019, 156, 1717–1730. [Google Scholar] [CrossRef]
  39. Kumashiro, N.; Erion, D.M.; Zhang, D.; Kahn, M.; Beddow, S.A.; Chu, X.; Still, C.D.; Gerhard, G.S.; Han, X.; Dziura, J. Cellular mechanism of insulin resistance in nonalcoholic fatty liver disease. Proc. Natl. Acad. Sci. USA 2011, 108, 16381–16385. [Google Scholar] [CrossRef]
  40. Radwan, A.M.; Fatoh, S.A.; Massoud, A.; Tousson, E. Effectiveness of curcumin nanoparticles in rat liver fibrosis caused by thioacetamide. Environ. Toxicol. 2024, 39, 388–397. [Google Scholar] [CrossRef]
  41. Bruck, R.; Ashkenazi, M.; Weiss, S.; Goldiner, I.; Shapiro, H.; Aeed, H.; Genina, O.; Helpern, Z.; Pines, M. Prevention of liver cirrhosis in rats by curcumin. Liver Int. 2007, 27, 373–383. [Google Scholar] [CrossRef]
  42. Afrin, R.; Arumugam, S.; Rahman, A.; Wahed, M.I.I.; Karuppagounder, V.; Harima, M.; Suzuki, H.; Miyashita, S.; Suzuki, K.; Yoneyama, H.; et al. Curcumin ameliorates liver damage and progression of NASH in NASH-HCC mouse model possibly by modulating HMGB1-NF-κB translocation. Int. Immunopharmacol. 2017, 44, 174–182. [Google Scholar] [CrossRef]
  43. Jain, S.K.; Rains, J.; Croad, J.; Larson, B.; Jones, K. Curcumin supplementation lowers TNF-alpha, IL-6, IL-8, and MCP-1 secretion in high glucose-treated cultured monocytes and blood levels of TNF-alpha, IL-6, MCP-1, glucose, and glycosylated hemoglobin in diabetic rats. Antioxid. Redox Signal. 2009, 11, 241–249. [Google Scholar] [CrossRef] [PubMed]
  44. Henao-Mejia, J.; Elinav, E.; Jin, C.; Hao, L.; Mehal, W.Z.; Strowig, T.; Thaiss, C.A.; Kau, A.L.; Eisenbarth, S.C.; Jurczak, M.J. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012, 482, 179–185. [Google Scholar] [CrossRef] [PubMed]
  45. Duan, Y.; Pan, X.; Luo, J.; Xiao, X.; Li, J.; Bestman, P.L.; Luo, M. Association of Inflammatory Cytokines With Non-Alcoholic Fatty Liver Disease. Front. Immunol. 2022, 13, 880298. [Google Scholar] [CrossRef] [PubMed]
  46. Hardwick, R.N.; Fisher, C.D.; Canet, M.J.; Lake, A.D.; Cherrington, N.J. Diversity in antioxidant response enzymes in progressive stages of human nonalcoholic fatty liver disease. Drug Metab. Dispos. 2010, 38, 2293–2301. [Google Scholar] [CrossRef]
  47. Hadizadeh, F.; Faghihimani, E.; Adibi, P. Nonalcoholic fatty liver disease: Diagnostic biomarkers. World J. Gastrointest. Pathophysiol. 2017, 8, 11. [Google Scholar] [CrossRef]
  48. Saadati, S.; Sadeghi, A.; Mansour, A.; Yari, Z.; Poustchi, H.; Hedayati, M.; Hatami, B.; Hekmatdoost, A. Curcumin and inflammation in non-alcoholic fatty liver disease: A randomized, placebo controlled clinical trial. BMC Gastroenterol. 2019, 19, 133. [Google Scholar] [CrossRef]
  49. Jazayeri-Tehrani, S.A.; Rezayat, S.M.; Mansouri, S.; Qorbani, M.; Alavian, S.M.; Daneshi-Maskooni, M.; Hosseinzadeh-Attar, M.J. Nano-curcumin improves glucose indices, lipids, inflammation, and Nesfatin in overweight and obese patients with non-alcoholic fatty liver disease (NAFLD): A double-blind randomized placebo-controlled clinical trial. Nutr. Metab. 2019, 16, 8. [Google Scholar] [CrossRef]
  50. Ghaffari, A.; Rafraf, M.; Navekar, R.; Asghari-Jafarabadi, M. Effects of turmeric and chicory seed supplementation on antioxidant and inflammatory biomarkers in patients with non-alcoholic fatty liver disease (NAFLD). Adv. Integr. Med. 2018, 5, 89–95. [Google Scholar] [CrossRef]
  51. 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]
  52. Rukkumani, R.; Sri Balasubashini, M.; Menon, V.P. Protective effects of curcumin and photo-irradiated curcumin on circulatory lipids and lipid peroxidation products in alcohol and polyunsaturated fatty acid-induced toxicity. Phytother. Res. 2003, 17, 925–929. [Google Scholar] [CrossRef]
  53. Yan, C.; Zhang, Y.; Zhang, X.; Aa, J.; Wang, G.; Xie, Y. Curcumin regulates endogenous and exogenous metabolism via Nrf2-FXR-LXR pathway in NAFLD mice. Biomed. Pharmacother. 2018, 105, 274–281. [Google Scholar] [CrossRef] [PubMed]
  54. Na, L.X.; Li, Y.; Pan, H.Z.; Zhou, X.L.; Sun, D.J.; Meng, M.; Li, X.X.; Sun, C.H. Curcuminoids exert glucose-lowering effect in type 2 diabetes by decreasing serum free fatty acids: A double-blind, placebo-controlled trial. Mol. Nutr. Food Res. 2013, 57, 1569–1577. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, Z.; Tian, R.F.; She, Z.G.; Cai, J.J.; Li, H.L. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic. Biol. Med. 2020, 152, 116–141. [Google Scholar] [CrossRef]
  56. Mousavi, S.M.; Milajerdi, A.; Varkaneh, H.K.; Gorjipour, M.M.; Esmaillzadeh, A. The effects of curcumin supplementation on body weight, body mass index and waist circumference: A systematic review and dose-response meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 2020, 60, 171–180. [Google Scholar] [CrossRef]
  57. Argilés, J.M.; Busquets, S.; García-Martínez, C.; López-Soriano, F.J. Mediators involved in the cancer anorexia-cachexia syndrome: Past, present, and future. Nutrition 2005, 21, 977–985. [Google Scholar] [CrossRef]
  58. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef]
  59. Yaikwawong, M.; Jansarikit, L.; Jirawatnotai, S.; Chuengsamarn, S. Curcumin extract improves beta cell functions in obese patients with type 2 diabetes: A randomized controlled trial. Nutr. J. 2024, 23, 119. [Google Scholar] [CrossRef]
  60. Di Pierro, F.; Bressan, A.; Ranaldi, D.; Rapacioli, G.; Giacomelli, L.; Bertuccioli, A. Potential role of bioavailable curcumin in weight loss and omental adipose tissue decrease: Preliminary data of a randomized, controlled trial in overweight people with metabolic syndrome. Preliminary study. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 4195–4202. [Google Scholar]
  61. Jobin, C.; Bradham, C.A.; Russo, M.P.; Juma, B.; Narula, A.S.; Brenner, D.A.; Sartor, R.B. Curcumin blocks cytokine-mediated NF-κB activation and proinflammatory gene expression by inhibiting inhibitory factor I-κB kinase activity. J. Immunol. 1999, 163, 3474–3483. [Google Scholar] [CrossRef]
  62. Lee, Y.M.; Kim, Y. Is Curcumin Intake Really Effective for Chronic Inflammatory Metabolic Disease? A Review of Meta-Analyses of Randomized Controlled Trials. Nutrients 2024, 16, 1728. [Google Scholar] [CrossRef]
  63. Cicero, A.F.G.; Sahebkar, A.; Fogacci, F.; Bove, M.; Giovannini, M.; Borghi, C. Effects of phytosomal curcumin on anthropometric parameters, insulin resistance, cortisolemia and non-alcoholic fatty liver disease indices: A double-blind, placebo-controlled clinical trial. Eur. J. Nutr. 2020, 59, 477–483. [Google Scholar] [CrossRef] [PubMed]
  64. Naseri, K.; Saadati, S.; Yari, Z.; Askari, B.; Mafi, D.; Hoseinian, P.; Asbaghi, O.; Hekmatdoost, A.; de Courten, B. Curcumin Offers No Additional Benefit to Lifestyle Intervention on Cardiometabolic Status in Patients with Non-Alcoholic Fatty Liver Disease. Nutrients 2022, 14, 3224. [Google Scholar] [CrossRef] [PubMed]
  65. Hammerich, L.; Tacke, F. Hepatic inflammatory responses in liver fibrosis. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 633–646. [Google Scholar] [CrossRef]
  66. Rivera-Espinoza, Y.; Muriel, P. Pharmacological actions of curcumin in liver diseases or damage. Liver Int. 2009, 29, 1457–1466. [Google Scholar] [CrossRef]
  67. Hsieh, C. Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions. Anticancer Res. 2001, 21, e2900. [Google Scholar]
  68. van Son, K.C.; Te Nijenhuis-Noort, L.C.; Boone, S.C.; Mook-Kanamori, D.O.; Holleboom, A.G.; Roos, P.R.; Lamb, H.J.; Alblas, G.; Coenraad, M.J.; Rosendaal, F.R.; et al. Prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) in a middle-aged population with overweight and normal liver enzymes, and diagnostic accuracy of noninvasive proxies. Medicine 2024, 103, e34934. [Google Scholar] [CrossRef]
  69. Ji, H.; Cheng, S.; Heart-Liver Axis Research, C. Sex differences in prevalence and prognosis of steatotic liver disease phenotypes: Biological sex matters. J. Hepatol. 2024, 80, e68–e69. [Google Scholar] [CrossRef]
  70. Joo, S.K.; Kim, W. Sex differences in metabolic dysfunction-associated steatotic liver disease: A narrative review. Ewha Med. J. 2024, 47, e17. [Google Scholar] [CrossRef]
  71. Patel, K.; Sebastiani, G. Limitations of non-invasive tests for assessment of liver fibrosis. JHEP Rep. 2020, 2, 100067. [Google Scholar] [CrossRef]
  72. Sahebkar, A. Dual effect of curcumin in preventing atherosclerosis: The potential role of pro-oxidant–antioxidant mechanisms. Nat. Prod. Res. 2015, 29, 491–492. [Google Scholar] [CrossRef]
Table 1. Baseline characteristics of study participants.
Table 1. Baseline characteristics of study participants.
VariablePlaceboCurcuminp Value *
Mean ± SD
(n = 39)
Mean ± SD
(n = 39)
Sex (M:F ratio)14/25 (0.56)11/28 (0.39)0.62
Age60.28 ± 9.49 57.33 ± 9.390.17
BMI (kg/m2)27.50 ± 3.2427.24 ± 3.110.93
Systolic blood pressure (mmHg)128.34 ± 1.45128.56 ± 1.520.86
Diastolic blood pressure (mmHg)74.56 ± 1.2774.32 ± 1.510.81
TNF (pg/mL)4.63 ± 1.614.74 ± 1.440.73
IL-1β (pg/mL)0.42 ± 0.260.47 ± 0.240.31
IL-6 (pg/mL)8.75 ± 1.108.94 ± 10.560.48
Glutathione peroxidase (U/L)6050 ± 2045.66596 ± 2898.470.43
Superoxide dismutase (U/mL)241.33 ± 55.11231.87 ± 37.140.58
Malondialdehyde (μmol/L)2.01 ± 0.491.98 ± 0.450.84
Total body fat (%)25.27 ± 6.5824.90 ± 7.350.72
Waist circumference (cm)95.33 ± 9.0794.49 ± 10.160.60
Non-esterified fatty acid (μmol/L)0.92 ± 0.461.01 ± 0.390.30
Liver stiffness (kPa)7.29 ± 3.676.65 ± 2.410.77
CAP (db/m)281.97 ± 29.04273.67 ± 21.510.17
Fatty liver index59.38 ± 24.2159.86 ± 24.860.16
Hepatic steatosis index36.73 ± 4.7136.46 ± 4.450.85
Lipid accumulation product60.07 ± 29.5557.47 ± 35.880.63
Creatinine (mg/dL)0.82 ± 0.040.85 ± 0.030.75
AST (U/L)25.31 ± 0.7825.34 ± 0.740.58
ALT (U/L)26.78 ± 1.4828.09 ± 1.350.08
History of cerebrovascular disease ††2(5.1%)1(2.6%)1.00
History of coronary artery disease ††3(7.7%)3(7.7%)1.00
History of hypertension ††27(69.2%)24(61.5%)0.63
History of diabetic nephropathy ††5(12.8%)7(17.9%)0.75
History of dyslipidemia ††28(71.8%)29(74.4%)1.00
Antihypertensive medications ††
Angiotensin receptor blockers27(69.2)29(74.4)0.80
Calcium channel blockers9(23.1)6(15.4)0.57
Beta blockers7(17.9)6(15.4)1.00
Antidyslipedemic medications ††
Statins21(53.8)17(43.6)0.50
* Data were evaluated by the Mann–Whitney U test, except for sex (M:F ratio); Chi-square test; †† Values expressed as number (percentage). ALT = alanine transaminase; AST = aspartate aminotransferase; BMI = body mass index; CAP = controlled attenuation parameter; IL-1β = Interleukin-1 beta; IL-6 = Interleukin-6; M:F = Male to Female; TNF = tumor necrosis factor.
Table 2. Comparison of body composition, biochemical markers, and hepatic parameters between groups.
Table 2. Comparison of body composition, biochemical markers, and hepatic parameters between groups.
OutcomesFollow-Up Period (mo)PlaceboCurcuminp Values *
Median (IQR)Min–MaxMedian (IQR)Min–Max
TNF (pg/mL)05.28 (3.52)2.64–7.045.28 (3.08)2.64–7.04NS
36.16 (2.20)2.64–7.044.46 (2.64)2.64–6.16<0.001
66.33 (2.71)2.18–14.264.01 (2.50)2.13–6.60<0.001
96.69 (2.53)3.30–14.363.99 (2.55)2.10–6.55<0.001
127.06 (3.55)2.75–15.373.28 (1.58)1.35–6.55<0.001
IL-1β (pg/mL)00.43 (0.44)0.03–0.860.43 (0.43)0.02–0.88NS
30.51 (0.46)0.02–0.760.38(0.40)0.02–0.840.021
60.89 (0.27)0.20–1.440.42 (0.26)0.12–0.89<0.001
90.92 (0.18)0.20–1.450.42 (0.19)0.13–0.64<0.001
120.93 (0.17)0.32–1.460.31 (0.18)0.12–0.54<0.001
IL-6 (pg/mL)08.80 (1.76) 7.04–10.558.80 (1.76)7.04–10.56NS
39.27 (2.64)7.04–10.568.28 (2.64)7.04–10.560.002
612.99 (4.50)7.55–17.998.49 (4.72)3.50–12.40<0.001
913.24 (3.35)7.65–18.007.60 (5.57)3.21–13.24<0.001
1214.39 (3.85)4.33–18.506.35 (5.44)3.10–12.40<0.001
GPx (U/L) 06243 (2549)1083–92206717 (2939)1124–11,969NS
36044 (2938)3540–19,4846591 (3303)4252–13,425<0.001
66189 (1643)2550–18,3867987 (2807)4549–17,679<0.001
95367 (1817)3769–89659898 (2143) 6089–15,436<0.001
124653 (1231)3576–798012,468 (3586)5874–17,790<0.001
SOD (U/mL)0231 (76)153–379218 (50)192–348NS
3214 (79)153–420236 (43)192–3480.027
6211 (58)150–420257 (63)195–3560.01
9206 (20)150–246269 (63)243–356<0.001
12180 (30)120–211310 (58.5)269–399<0.001
MDA (μmol/L)01.99 (0.73)1.20–3.331.90 (0.58)1.30–3.23NS
32.19 (0.52)1.21–3.421.92 (0.77)1.12–3.170.027
62.28 (0.47)0.77–3.541.87 (0.82)1.22–2.64<0.001
92.32 (0.5901.23–3.901.70 (0.65)0.93–2.50<0.001
122.39 (0.83)1.20–3.961.35 (0.43)0.93–2.20<0.001
TBF (%)024.57(9.55)13.77–36.8923.16(9.64)11.98–40.77NS
325.71(8.08)13.03–38.5421.53(9.18)12.19–39.10NS
625.88(7.88)14.91–39.1420.76(9.16)11.76–41.160.001
926.77(8.95)15.75–39.7421.20(8.26)11.39–39.620.003
1227.08(8.93)16.48–42.8220.19(7.8411.25–36.900.001
WC (cm)095 (11)76.00–112.0095 (12)74.00–120.00NS
398 (10)75.00–113.0094 (9)75.00–117.00NS
6100 (9.5)78.00–115.0093 (9)75.00–135.000.01
999 (11)78.00–117.0092 (10)74.00–117.000.003
1299 (10.5)76.00–118.0091 (11)73.00–114.00<0.001
NEFA (μmol/L)00.85 (0.57)0.20–1.980.84 (0.5)0.30–1.73NS
31.15 (0.66)0.34–1.861.14 (0.57)0.34–1.79NS
61.18 (0.78)0.32–1.890.87 (0.58)0.34–1.850.001
91.19 (0.58)0.45–1.980.87 (0.48)0.34–1.790.007
121.25 (0.75)0.40–1.950.88 (0.53)0.20–1.920.002
BMI (kg/m2)027.06 (4.79)19.05–34.7727.68 (4.87)20.43–36.58NS
327.63 (4.25)17.89–33.6927.10 (4.45)19.22–36.20NS
627.78 (3.68)18.07–33.4626.33 (4.45)18.80–35.160.031
927.46 (4.77)17.89–33.0326.08 (4.29)19.22–36.720.017
1227.70 (4.16)17.72–33.3225.97 (4.05)20.55–35.550.002
Glucose (mg/dL)0126 (26.5)91–181122 (22)79–163NS
3128 (36.5)102–195124 (28)80–171NS
6131 (37.5)98–214123 (27)79–1710.029
9130 (27.0)105–185120 (18)75–150<0.001
12131 (26.5)98–187117 (20)70–151<0.001
HbA1C (%)06.2 (0.85)5.1–8.96.3 (0.70)4.8–7.8NS
36.6 (0.70)5.4–8.96.3 (0.70)4.8–8.0NS
66.6 (1.00)5.4–9.06.2 (1.05)4.5–8.30.030
96.7 (1.10)5.6–10.36.1 (1.05)4.3–8.20.003
126.6 (1.20)5.3–9.95.9 (0.90)4.2–8.40.003
Liver stiffness (kPa)06.2 (3.60)3.20–19.606.6 (3.15)2.80–14.20NS
36.6 (2.70)3.90–14.306.0 (2.00)3.00–9.50<0.001
67.2 (2.15)4.20–12.105.9 (2.16)3.00–9.46<0.001
97.9 (3.20)4.80–13.505.5 (1.90)3.00–8.11<0.001
126.9 (3.45)5.00–13.504.2 (1.60)2.90–7.60<0.001
CAP (dB/m)0279 (34.5)248–387264 (30)248.00–323NS
3258 (49)184–365287 (34)147.00–319<0.001
6287 (34)203–353263 (53)100.00–305<0.001
9280 (67.5)202–293250 (58) 102.00–293<0.001
12292 (64.5)199–35223 (51)107.00–274<0.001
FLI064.09 (35.29)12.68–92.3960.84 (35.35)10.64–98.71NS
366.37 (31.92)14.42–92.4157.89 (39.49)13.99–98.660.035
669.92 (32.71)16.64–95.3646.64(27.37)10.21–98.860.001
971.45 (40.01)15.77–96.1445.46(31.39)10.82–98.350.002
1276.43 (36.25)17.36–97.5539.64 (35.92)9.21–97.44<0.001
HSI036.50 (7.28)28.39–47.1536.46 (7.19)28.39–43.63NS
337.91 (6.43) 29.26–50.3736.62 (5.41)27.11–48.410.042
638.15 (6.03)27.95–49.9236.49 (5.27)26.58–47.820.033
938.30 (5.67)29.26–52.9136.22 (5.39)26.02–47.460.033
1238.95 (6.09)29.99–54.4435.92 (5.61)23.57–48.380.010
LAP053.68 (34.94)12.61–139.1745.86 (31.25)13.01–176.37NS
364.38 (39.28)25.32–128.4744.73 (31.45)12.87–173.440.003
672.86 (42.36)29.14–184.2243.38 (27.71)13.42–148.200.003
959.78 (45.73)29.62–207.0040.72 (19.80)12.04–144.610.004
1265.24 (53.58)30.90–191.5939.85 (23.38)12.19–136.90<0.001
* p values were evaluated by the Mann–Whitney U test. BMI = body mass index; CAP = controlled attenuated parameter; GPx = glutathione peroxidase; FLI = fatty liver index; HbA1C = glycated hemoglobin; HSI = hepatic steatosis Index; IL-1β = Interleukin-1 beta; IL-6 = interleukin-6; IQR = Interquartile range; LAP = lipid accumulation product; MDA = malondialdehyde; mo = months; NEFA = non-esterified fatty acid; NS = not significant; SOD = superoxide dismutase; TBF = total body fat; TNF = tumor necrosis factor; WC = Waist circumference
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

Yaikwawong, M.; Jansarikit, L.; Jirawatnotai, S.; Chuengsamarn, S. Curcumin for Inflammation Control in Individuals with Type 2 Diabetes Mellitus and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Randomized Controlled Trial. Nutrients 2025, 17, 1972. https://doi.org/10.3390/nu17121972

AMA Style

Yaikwawong M, Jansarikit L, Jirawatnotai S, Chuengsamarn S. Curcumin for Inflammation Control in Individuals with Type 2 Diabetes Mellitus and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Randomized Controlled Trial. Nutrients. 2025; 17(12):1972. https://doi.org/10.3390/nu17121972

Chicago/Turabian Style

Yaikwawong, Metha, Laddawan Jansarikit, Siwanon Jirawatnotai, and Somlak Chuengsamarn. 2025. "Curcumin for Inflammation Control in Individuals with Type 2 Diabetes Mellitus and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Randomized Controlled Trial" Nutrients 17, no. 12: 1972. https://doi.org/10.3390/nu17121972

APA Style

Yaikwawong, M., Jansarikit, L., Jirawatnotai, S., & Chuengsamarn, S. (2025). Curcumin for Inflammation Control in Individuals with Type 2 Diabetes Mellitus and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Randomized Controlled Trial. Nutrients, 17(12), 1972. https://doi.org/10.3390/nu17121972

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