Case Control Study Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 21, 2024; 30(27): 3290-3303
Published online Jul 21, 2024. doi: 10.3748/wjg.v30.i27.3290
Targeted metabolomics study of fatty-acid metabolism in lean metabolic-associated fatty liver disease patients
Pei-Qi Sun, Yi-Fu Yuan, Qin Cao, Xiao-Yan Chen, Yuan-Ye Jiang, Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
Wen-Min Dong, Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
Li-Li Guo, Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
ORCID number: Wen-Min Dong (0000-0001-9135-6974); Li-Li Guo (0009-0008-7885-4686); Yuan-Ye Jiang (0000-0002-4979-4206).
Co-first authors: Pei-Qi Sun and Wen-Min Dong.
Co-corresponding authors: Li-Li Guo and Yuan-Ye Jiang.
Author contributions: Guo LL and Jiang YY conceived and designed the study; Jiang YY supervised the work, Guo LL contributed to the writing of the revision draft, they are the co-corresponding authors of this manuscript; Sun PQ and Dong WM performed the research, they contributed to this study equally; Sun PQ acquired patient and samples; Dong WM contributed to the writing of the manuscript; Yuan YF, Cao Q, and Chen XY analyzed the data and wrote the manuscript; All authors have read and approved the final manuscript.
Supported by Shanghai Natural Science Foundation, No. 22ZR1455900; Shanghai Putuo District Health System Science and Technology Innovation Project Key Project, No. ptkwws202201; and Shanghai Putuo District Xinglin Excellent Youth Talent Training Program, No. ptxlyq2201.
Institutional review board statement: An ethics review form is available for this study (Approval No: PTEC-A-2018-49-1).
Informed consent statement: All study participants, or their legal guardian, provided informed written consent prior to study enrollment.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: We consent to data sharing.
STROBE statement: The authors have read the STROBE Statement-checklist of items, and the manuscript was prepared and revised according to the STROBE Statement-checklist of items.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4. 0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Yuan-Ye Jiang, Doctor, Doctor, Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, No. 164 Lanxi Road, Shanghai 200062, China. yuanye1014@126.com
Received: January 22, 2024
Revised: May 9, 2024
Accepted: June 6, 2024
Published online: July 21, 2024
Processing time: 171 Days and 0.4 Hours

Abstract
BACKGROUND

The annual incidence of metabolic-associated fatty liver disease (MAFLD) in China has been increasing and is often overlooked owing to its insidious characteristics. Approximately 50% of the patients have a normal weight or are not obese. They are said to have lean-type MAFLD, and few studies of such patients are available. Because MAFLD is associated with abnormal lipid metabolism, lipid-targeted metabolomics was used in this study to provide experimental evidence for early diagnosis and pathogenesis.

AIM

To investigate the serum fatty-acid metabolic characteristics in lean-type MAFLD patients using targeted serum metabolomic technology.

METHODS

Between January and June 2022, serum samples were collected from MAFLD patients and healthy individuals who were treated at Shanghai Putuo District Central Hospital for serum metabolomics analysis. Principal component analysis and orthogonal partial least squares-discriminant analysis models were developed, and univariate analysis was used to screen for biomarkers of lean-type MAFLD and analyze metabolic pathways. UPLC-Q-Orbitrap/MS content determination was used to determine serum palmitic acid (PA), oleic acid (OA), linoleic acid (LA), and arachidonic acid (AA) levels in lean-type MAFLD patients.

RESULTS

Urea nitrogen and uric acid levels were higher in lean-type MAFLD patients than in healthy individuals (P < 0.05). Alanine transaminase and cholinesterase levels were higher in lean-type MAFLD patients than in healthy individuals (P < 0.01). The expression of high-density lipoprotein and apolipoprotein A-1 were lower in lean-type MAFLD patients than in healthy individuals (P < 0.05) and the expression of triglycerides and fasting blood glucose were increased (P < 0.01). A total of 65 biomarkers that affected the synthesis and metabolism of fatty acids were found with P < 0.05 and variable importance in projection > 1”. The levels of PA, OA, LA, and AA were significantly increased compared with healthy individuals.

CONCLUSION

The metabolic profiles of lean-type MAFLD patients and healthy participants differed significantly, yielding 65 identified biomarkers. PA, OA, LA, and AA exhibited the most significant changes, offering valuable clinical guidance for prevention and treatment of lean-type MAFLD.

Key Words: Lean-type metabolic-associated fatty liver disease; Targeted serum metabolomics; Fatty acids; Principal component analysis; Orthogonal partial least squares-discriminant analysis

Core Tip: This study targeted the serum metabolomics of healthy individuals and metabolic-associated fatty liver disease (lean-type MAFLD), screened biomarkers and related metabolic pathways, and conducted targeted quantitative analysis of their specific biomarkers with the aim of providing experimental evidence for the early diagnosis and pathogenesis of lean-type MALFD.



INTRODUCTION

The annual prevalence of metabolic-associated fatty liver disease (MAFLD) in China has been increasing, with the current rate exceeding 30%[1-4]. Owing to subtle early manifestations that often go unnoticed, approximately 20% of MAFLD patients progress to metabolic-associated steatohepatitis, fibrosis, cirrhosis, and liver cancer[5,6]. MAFLD is closely associated with obesity and type 2 diabetes, but approximately 40.8% of MAFLD patients have a body mass index (BMI) that does not meet the criteria for overweight or obesity[7-9]. This condition is commonly referred to as lean or nonobese-type MAFLD. Compared with other ethnic groups, the prevalence of metabolic disorders is higher in Asians with lower BMIs[10].

Because studies of lean-type MAFLD are limited, its pathogenesis and optimal treatment are not clear. Patients with lean-type MAFLD are at increased risk of progressing to fatty liver inflammation and liver fibrosis, with an incidence of 30%, and it is closely associated with metabolic dysfunction[11]. Metabolomics is a high-throughput detection method widely used for disease diagnosis and in mechanistic investigations[12]. In this study, we used ultra-high-performance liquid chromatography-tandem mass spectrometry with an electrospray ionization quadrupole trap analyzer to identify serum metabolic markers that distinguished lean-type MAFLD patients from healthy individuals. We aimed to identify metabolic pathways specific to those markers and to conduct a targeted investigation of the metabolites and pathways that were significantly changed in lean-type NAFLD. This study aimed to provide experimental evidence for the early diagnosis and pathogenesis of lean-type MAFLD.

MATERIALS AND METHODS
Study participants

Between January 2022 and June 2022, 20 patients diagnosed with lean-type MAFLD were recruited from the gastroenterology department of the Central Hospital of Putuo District, Shanghai. The control group included 20 healthy volunteers who were recruited after physical examination. General information and clinical data, including a complete blood count, liver function, renal function, and a lipid profile, were collected for the analysis. The research protocol was approved by the Ethics Committee of the Central Hospital of Putuo District (Affiliated to the Putuo Hospital of Shanghai University of Traditional Chinese Medicine; Approval No. PTEC-R-2020-29-1). All the enrolled patients provided informed consent before participating in the study.

Diagnostic criteria

MAFLD was diagnosed following the clinical criteria included in the 2010 Guidelines for the Diagnosis and Treatment of Nonalcoholic Fatty Liver Disease of the Chinese Society of Hepatology, Chinese Medical Association[11]. The criteria for MAFLD were: (1) No history of excessive alcohol consumption or an equivalent ethanol intake of < 140 g per week for men (< 70 g per week for women); (2) Absence of specific diseases, including viral hepatitis, drug-induced liver disease, hepatolenticular degeneration, autoimmune liver diseases that can cause fatty liver, or use of total parenteral nutrition; and (3) Histopathological changes in liver biopsy specimens consistent with the pathological diagnosis criteria for fatty liver disease.

MAFLD was defined by: (1) Liver imaging findings consistent with the diagnosis of diffuse fatty liver and exclusion of other causes; and (2) Manifestations related to metabolic syndrome with persistently elevated levels of serum alanine transaminase (ALT), aspartate transaminase, or gamma-glutamyl transferase for > 6 months. Individuals with abnormal enzyme profiles and a fatty liver on imaging and return to normal or improve after weight loss and decreased insulin resistance fulfill the diagnostic criteria of MAFLD. The diagnostic criteria for MAFLD included a BMI of < 23 for lean-type, 23 ≤ BMI < 28 for overweight-type, and ≥ 28 for obese-type.

Inclusion criteria

Patients who met the following criteria were included in the study: (1) Between 16 years and 75 years of age, regardless of sex; (2) Meeting the diagnostic criteria for lean-type MAFLD according to Western medical standards[13]; (3) Having complete and reliable clinical data, biochemical tests, and specimen collection; and (4) Providing consent to participate.

Exclusion criteria

The exclusion criteria were: (1) Presence of concomitant liver-extrinsic fibrotic diseases, including systemic lupus erythematosus, rheumatic diseases, renal failure, chronic obstructive pulmonary disease; (2) Presence of severe primary diseases related to cardiovascular, cerebrovascular, urinary, renal, hematopoietic systems, malignant tumors, other serious complications, or psychiatric disorders; (3) Presence of thyroid disorders, including hyperthyroidism, hypothyroidism, subclinical hypothyroidism, and Hashimoto’s thyroiditis; and (4) Lack of complete clinical data relevant to the study.

Materials

Ultra-high-performance liquid chromatography (Ultimate 3000; Thermo Fisher Scientific, Waltham, MA, United States); high-resolution mass spectrometry (Orbitrap Elite; Thermo Fisher Scientific); cryogenic high-speed centrifuge (1730R; Mr. Gene GmbH, Regensburg, Germany); ultrapure water system (Milli-Q; Merck Biotechnology Shanghai Co., Ltd., Shanghai, China); multi-tube vortex oscillator (VX-II; Beijing Tajin Technology Co., Ltd., Beijing, China); methanol (HPLC grade, Batch No.: O0621152; China National Pharmaceutical Group Chemical Reagent Co., Ltd., Shanghai, China); methyl tert-butyl ether (analytical grade, Batch No. 20210227; China National Pharmaceutical Group Chemical Reagent Co., Ltd.); formic acid (chromatography grade, Batch No.: D1290265; Shanghai ANPEL Scientific Instrument Co., Ltd., Shanghai, China); ammonium acetate (chromatography grade, Batch No. BCBR1129V; Shanghai ANPEL Scientific Instrument Co., Ltd.); isopropanol (chromatography grade, Batch No. V589K144; Shanghai ANPEL Scientific Instrument Co., Ltd.); acetonitrile (chromatography grade, Batch No. K3021728l Shanghai ANPEL Scientific Instrument Co., Ltd.).

Metabolomics study

Serum handling: The study included 20 patients with lean-type MAFLD and 20 healthy volunteers. Morning fasting venous blood (10 mL) was collected and allowed to stand at 4 °C for 2 h before centrifuging at 3000 rpm for 15 minutes. Serum (50 μL) was collected from the upper layer. Serum samples were mixed with 200 μL methanol and vortexed for complete extraction. After low-temperature centrifugation at 14000 rpm and 4 °C for 10 minutes, the supernatant was transferred to a sample vial for analysis.

Chromatographic conditions: Column: C18 chromatographic column (Hypersil Gold C18, 100 mm × 2.1 mm, 1.9 μm); flow rate: 0.3 mL/min; column temperature: 40 °C; mobile phase composition: A: Pure water + 0.1% formic acid and B: Acetonitrile + 0.1% formic acid. Gradient elution program: 0-2 minutes, 95% A; 2-12 minutes, 5%-95% A; 12-15 minutes, 5%-95% A; 15-17 minutes, 5%-95% A.

Mass spectrometric conditions: Positive ion mode: Heater temperature: 300 °C; sheath gas flow rate: 45 psi; auxiliary gas flow rate: 5 L/min; sweep gas flow rate: 0.3 L/min; electrospray voltage: 3.0 kV; capillary temperature: 350 °C; S-Lens RF level: 30%. Negative ion mode: Heater temperature 300 °C; sheath gas flow rate: 45 psi; auxiliary gas flow rate: 5 L/min; sweep gas flow rate: 0.3 L/min; electrospray voltage: 3.2 kV; capillary temperature: 350 °C; S-Lens RF level: 60%.

Fatty-acid targeted metabolomics

Chromatographic conditions: Column: Acquity UPLC BEH C8 column (2.1 mm × 100 mm, 1.7 μm) (Waters Corp., Milford, MA, United States); column temperature: 40 °C; flow rate: 0. 35 mL/min; mobile phase: water (0.1% formic acid): acetonitrile (0.1% formic acid); gradient elution program: 1 minute, 50% B; 1-5 minutes, 50%-80% B; 5-6.5 minutes, 80%-95% B; 6.5-10 minutes, 95% B.

Tandem mass spectrometry (MS/MS detection): The serum concentration of fatty acids and their metabolites were determined using ultra-high-performance liquid chromatography (H-Class; Waters Corp.) coupled with triple quadrupole mass spectrometry (6500; AB SCIEX, Framingham, MA, United States). MS/MS data were collected using deuterated arachidonoylethanolamide (AEA-d8), deuterated oleoylethanolamide (OEA-d4), deuterated linoleoylethanolamide (LEA-d4), and deuterated oleic acid (OA-d9) as internal standards. Analytes, including AEA, 2-arachidonoyl glycerol ester, palmitoylethanolamide (PEA), OEA, LEA, 2-arachidonoylglycerol (2-AG), 1-palmitoyl glycerol (1-PG), 1-oleoyl glycerol (1-OG), and 1-linoleoyl glycerol (1-LG), were detected in the positive electrospray ionization mode, and arachidonic acid (AA), stearic acid, palmitic acid (PA), OA, and linoleic acid (LA) were detected in the negative mode. The optimized operational conditions were: ion spray voltage of + 5500 V in positive mode and -4500 V in negative mode, ion source temperature of 550 °C. Nitrogen was used as the collision gas. The ion pairs and related internal standards for multiple reaction monitoring are shown in Table 1.

Table 1 Ion pairs of analytes and internal standards.
ID
Q1
Q3
DP
CE
CXP
AEA-d8356.100294.00064.9419.0918.93
AEA348.00062.00056.0042.0010.00
2-AG379.000287.400163.8620.0513.09
OEA326.400309.200195.3221.3129.01
OEA-d4330.60066.100132.6621.5512.58
LEA324.40062.100130.2617.7611.28
PEA300.30062.100124.0019.8310.20
LEA-d4328.10066.000133.8020.988.59
1-LG355.300338.40032.2110.7318.49
1-OG357.300339.300145.0013.2024.00
1-PG331.200313.500155.0012.7019.00
LA325.200279.100-10.11-8.72-25.70
OA327.200281.300-47.00-10.50-10.00
PA301.200255.200-58.00-14.00-15.00
AA303.100259.000-73.90-16.14-13.02
OA-d9336.300290.300-14.34-9.32-7.94

Sample preparation: A volume of 30 μL of serum was combined with mixed internal standards, followed by successive addition of 500 cL of methyl tert-butyl ether, 150 μL methanol, and 140 μL ultrapure water. The mixture was vortexed for 1 minute and then centrifuged at 4 °C for 10 minutes (3000 rpm). The upper layer was collected, concentrated, and dried before reconstitution with 100 μL acetonitrile. The resulting supernatant were transferred to a sample vial for further analysis.

Standard and internal standard preparation: Precisely weighed amounts of AEA, LEA, PEA, OEA, 2-AG, 1-LG, 1-PG, 1-OG, AEA-d8, OEA-d4, and LEA-d4 were prepared at concentrations of 1250, 500, 250, 125, 50, 25, 12. 5, 5, 1, and 0. 5 ng, respectively. OA, PA, AA, and LA standards were prepared at concentrations of 5, 10, and 20 μg/mL, respectively, including internal standards AA-d8 (10 μg/mL) and OA-d9 (1 μg/mL).

Data processing and statistical methods: Peak alignment, retention time correction, and peak area were calculated using LC-MS software. Accurate molecular weights and MS/MS spectra were used for the identification and database retrieval of the metabolites. Unsupervised principal component analysis (PCA) and orthogonal partial least squares-discriminant analysis (OPLS-DA) were used for multidimensional statistical analysis. Enrichment analysis of significantly altered metabolic pathways was performed using the Kyoto Encyclopedia of Genes and Genomes database. All other data were analyzed using SPSS 22.0 (IBM Corp., Armonk, NY, United States). Normally distributed quantitative data were reported as means ± SD. Comparison of quantitative data among groups was performed with analysis of variance if the data satisfied the normality and homogeneity of variance assumptions; otherwise, the Wilcoxon non-parametric test was used. P < 0. 05 was considered statistically significant.

RESULTS
Clinical data analysis

Twenty lean-type MAFLD patients and 20 healthy individuals were included in this study. Differences in general characteristics, including sex, age, or BMI observed in lean-type MAFLD patients and healthy individuals were not significant (Table 2). Routine blood tests revealed a significant difference between the white blood cell counts of the lean-type MAFLD patients and healthy controls (P < 0.01). Renal function test results revealed significant differences in urea nitrogen, uric acid, and creatinine levels in the lean-type MAFLD patients and healthy controls (P < 0.01). The results of liver function tests, including cholinesterase and ALT levels in the lean-type MAFLD patients and healthy controls, were significantly different (P < 0.01). The glucose metabolism tests found a significant difference of fasting blood glucose level between lean-type MAFLD patients and healthy controls (P < 0.01). Blood lipid analysis revealed significant differences in high-density lipoprotein (HDL), triglycerides, and apolipoprotein A1 (APOA-1) level between lean-type MAFLD patients and healthy controls (P < 0.01).

Table 2 General characteristics and clinical indicators of lean metabolic-associated fatty liver disease patients.
Indicator
Lean MAFLD group, n = 20
Control group, n = 20
Statistical value
P value
Male/female sex5/150/20χ2 = 3.6570.056
Age in years52.75 ± 15.1744.95 ± 12.11t = 1.7980.08
Weight in kg60 ± 6.1254.8 ± 5.64t = 2.7930.008b
BMI in kg/m²22.65 (21.52, 22.80)21.25 (20.05, 21.80)Z = 3.1960.001b
White blood cells as × 109/L7.25 ± 2.085.31 ± 1.06t = 3.7150.001b
Red blood cells as × 1012/L4.49 ± 0.634.23 ± 0.34t = 1.6160.117
Hemoglobin in g/L134.20 ± 20.78125.70 ± 13.58t = 1.5310.134
Hematocrit in %39.22 ± 5.8436.67 ± 3.27t = 1.7030.097
Platelets as × 109/L261.25 ± 69.41228.80 ± 45.42t = 1.7490.088
Mean platelet volume in fL10.35 ± 1.2110.96 ± 1.06t = -1.6930.099
Neutrophils in %55.03 ± 11.4951.53 ± 6.33t = 1.1920.243
Lymphocytes in %35.01 ± 11.4938.25 ± 6.39t = -1.1040.278
Monocytes in %7.53 ± 1.87736.71 ± 0.93t = 1.750.091
Mean red cell volume in fL87.90 (84.5, 90.58)87.85 (86.08, 89.95)Z = -0.2980.766
Mean red cell hemoglobin in pg30.25 (28.63, 31.13)30.55 (29.25, 31.28)Z = -0.690.49
Mean red cell hemoglobin concentration in g/L341.00 (337.00, 349.75)346.00 (337.50, 351.00)Z = -0.8950.371
Red cell volume distribution width in %12.90 (12.33, 13.63)12.50 (12.13, 13.08)Z = -1.1780.239
Eosinophils in %1.80 (0.75, 2.40)1.95 (1.53, 3.33)Z = -0.9340.35
Basophils in %0.50 (0.40, 0.70)0.60 (0.40, 0.70)Z = -0.8490.396
Creatinine in μmol/L66.25 ± 17.6557.20 ± 6.99t = 2.1320.043a
Urea nitrogen in mmol/L5.55 (4.35, 6.98)4.65 (3.78, 5.38)Z = -2.2740.023a
Uric acid in μmol/L350.50 (296.25, 450.00)269.00 (236.75, 310.00)Z = -3.0440.002b
Total bilirubin in μmol/L9.00 (11.00, 13.00)9.00 (10.00, 11.00)Z = -1.1860.236
Direct bilirubin in μmol/L1.66 (1.88, 2.30)1.56 (1.62, 1.92)Z = -1.5560.120
Alkaline phosphatase in U/L69.00 (59.25, 89.75)61.50 (51.00, 82.50)Z = -1.4480.148
Cholinesterase in U/L9214.95 ± 1232.297059.20 ± 1260.51t = 5.4690.001b
Alanine aminotransferase in U/L17.00 (11.00, 35.75)9.50 (6.25, 16.00)Z = -3.0220.003b
Aspartate aminotransferase in U/L24.00 (20.00, 33.75)20.50 (17.25, 26.75)Z = -1.640.101
γ-glutamyl transferase in U/L36.00 (21.50, 45.50)20.50 (16.50, 34.75)Z = -1.7190.086
Total protein in g/L73.05 ± 5.9372.15 ± 4.04t = 0.5610.578
Albumin in μmol/L42.10 ± 3.3741.55 ± 2.98t = 0.5470.588
High-density lipoprotein in mmol/L1.07 (0.98, 1.26)1.40 (1.27, 1.59)Z = -3.8820.001b
Low-density lipoprotein in mmol/L3.48 ± 0.723.40 ± 1.10t = 0.2550.800
Triglycerides in mmol/L2.68 (1.37, 4.20)1.09 (0.94, 1.39)Z = -3.5980.001b
Total cholesterol in mmol/L5.19 ± 1.075.06 ± 1.28t = 0.3660.716
Apolipoprotein A1 in g/L1.35 (1.22, 1.56)1.55 (1.36, 1.74)Z = -2.1530.031a
Apolipoprotein B in g/L0.99 ± 0.220.94 ± 0.32t = 0.5590.579
Glycated albumin in %12.60 (10.90, 14.45)13.00 (12.53, 14.18)Z = -0.7170.473
Fasting blood glucose in mmol/L5.40 (5.03, 6.05)4.90 (4.63, 5.15)Z = -3.0520.002b
LC-MS quality control results

The total ion chromatogram of quality control samples of sera from lean-type MAFLD patients is shown in Figure 1. The overlapping chromatograms indicated excellent instrument stability and consistent retention times, validating the reliability of the analytical data.

Figure 1
Figure 1 Total ion chromatogram. A: Negative mode; B: Positive mode.
PCA analysis

Patient serum metabolomics data were analyzed using Compound Discover software, followed by normalization. An unsupervised PCA model was constructed using Simca-P 14.0 and used to compare the overall profiles of individual samples in both positive and negative detection modes. In both positive and negative modes, samples from lean-type MAFLD patients clustered together and were clearly differentiated from the control group samples (Figure 2). That indicated a pronounced differences in the levels of metabolites found in lean-type MAFLD patients and healthy controls. The PCA model of the positive mode had an R2X of 0. 698 and Q2 of 0. 497, and for the negative mode the R2X was 0. 6794 and a Q2 of 0. 566. These results confirmed that PCA data model was able to elucidate variations in metabolites among samples.

Figure 2
Figure 2 Principal component analysis. A: Positive mode; B: Negative mode. SG: Lean metabolic-associated fatty liver disease (metabolic-associated fatty liver disease); SZC: Healthy lean individuals.
OPLS-DA analysis

OPLS-DA was used to comprehensively analyze and compare differences in the metabolite data collected from lean-type MAFLD patients and healthy controls. In both positive and negative modes, the lean-type MAFLD patients and healthy controls were completely separated in the OPLS-DA plots (Figure 3). The corresponding R2Y and Q2 values in the positive and negative modes were 0.957 and 0.962 and 0.954 and 0.921, respectively. To further validate the robustness of the model, a permutation test with 200 iterations was conducted, and it revealed no signs of data overfitting. This confirmed the good fit and predictive ability of the model, with statistically significant results.

Figure 3
Figure 3 Orthogonal partial least squares discriminant analysis. A: Positive mode; B: Negative mode. SG: Lean-type metabolic-associated fatty liver disease (metabolic-associated fatty liver disease); SZC: Lean healthy individuals.
Differential metabolic pathway screening

Following OPLS-DA analysis, differential metabolites were selected by a combination of variable importance in projection (VIP) values and t-tests. The selection criteria were set to satisfy VIP > 1 and P < 0.05. The selected metabolites were cross-referenced against the HMDB database. Ultimately, 65 potential differentially expressed metabolites were identified in lean-type MAFLD patients. Of those, 33 were identified in the positive mode and 32 in the negative modes (Figure 4). Metabolic pathway enrichment analysis of the identified metabolites with MetaboAnalyst 5.0 found that the 65 metabolites were associated with pathways involving unsaturated fatty acid biosynthesis, LA metabolism, fatty acid degradation, and ether lipid metabolism. Significant differences were found in the pathways of unsaturated fatty-acid biosynthesis and LA metabolism (Figure 5). A targeted metabolomic analysis of fatty acids was performed to gain a deeper understanding of the significance of fatty-acid metabolism in lean-type MAFLD patients.

Figure 4
Figure 4 Heatmap of differential metabolites.
Figure 5
Figure 5 Metabolic pathway enrichment analysis.
Fatty-acid targeted metabolomics standard curve and chromatogram

To construct the standard curve, an increasing concentration (μg/mL) series of mixed fatty-acid standards was sequentially injected, with the peak area plotted on the Y-axis. The linear range and correlation coefficients (r) are listed in Table 3. The results show that the standard curve had excellent linearity and was suitable for the quantitative detection of fatty acids in the samples. The multiple reaction monitoring chromatograms of the samples and standards are shown in Figures 6 and 7, and the results indicate that there was no interference from other matrices in the content detection of the samples.

Figure 6
Figure 6 Standard material and sample total ion chromatograms. A: Standard material; B: Sample total.
Figure 7
Figure 7 Multiple reaction monitoring chromatograms of palmitic acid, oleic acid, linoleic acid, arachidonic acid, and internal standards. A: Standards; B: Samples. PA: Palmitic acid; OA: Oleic acid; LA: Linoleic acid; AA: Arachidonic acid; OA-d9: Deuterated oleic acid.
Table 3 Fatty-acid standard curve and linear range.
No.
Name
Linear
Range in μg/mL
r
1Arachidonic acidy = 0.0087x - 0.00530.1-50.9963
2Oleic acidy = 0.0272x - 0.04250.1-50.9914
3Palmitic acidy = 0.0074x + 0.00360.1-50.9940
4Linoleic acidy = 0.0341x - 0.08210.1-50.9919
Changes in fatty-acid content in lean-type MAFLD patients

Changes in the fatty-acid content of lean-type MAFLD patients and healthy individuals are shown in Table 4. The PA, OA, LA, and AA levels in lean-type MAFLD patients were 3.41 ± 0.84, 2.63 ± 1.45, 2.42 ± 1.18, and 2.45 ± 1.21 μg/mL, respectively, and were significantly higher than those in the control group (P < 0.05).

Table 4 Changes in fatty-acid content in lean metabolic-associated fatty liver disease patients, n = 6.
No.
Name
Lean MAFLD patients
Normal individuals
1Palmitic acid3.41 ± 0.84a1.53 ± 0.85
2Oleic acid2.63 ± 1.45a1.26 ± 0.65
3Linoleic acid2.42 ± 1.18a1.30 ± 0.49
4Arachidonic acid2.45 ± 1.21a1.24 ± 0.56
DISCUSSION

Obesity and type 2 diabetes were prevalent in MAFLD patients, and approximately 40.8% of MAFLD patients had BMIs below threshold for overweight or obese MAFLD (< 23 kg/m2), referred to as lean-type MAFLD. According to epidemiological surveys, lean-type MAFLD accounts for 25.8% of all MAFLD cases globally, but accounts for 44.3% of the MAFLD cases in China[14]. Owing to its subtle onset, mild, or nonspecific symptoms, the diagnosis of lean-type MAFLD is challenging and is often diagnosed during routine liver function testing or imaging examination. Current research predominantly focuses on obese-type MAFLD, with limited studies on lean-type patients. This study used serum metabolomics to investigate biomarkers characteristic of lean-type MAFLD patients and performed a quantitative analysis.

In this study, the lean-type MAFLD patients had BMIs of < 23 kg/m2, and had higher BMIs and body mass than healthy lean participants. Compared with healthy controls, lean-type MAFLD patients had significantly elevated white blood cell counts (P < 0.01). Additionally, significant differences were observed in liver and kidney function indexes and lipid profile components, such as urea nitrogen, uric acid, creatinine, cholinesterase, ALT, fasting blood glucose, HDL, triglycerides, and APOA-1 (P < 0.01).

ALT is present in hepatic cell cytoplasm, where increased intracellular triglycerides in provide sufficient reactive substrates for lipid peroxidation, thereby affecting the activity of antioxidant enzymes. This leads to increased oxidative stress. When liver cells are damaged, intracellular enzymes are released into the blood, causing ALT to spill over from liver cells into the extracellular space, resulting in increased ALT in the peripheral blood. Therefore, ALT levels reflect the integrity of liver cells. In this study, ALT and triglyceride levels were significantly higher in lean-type MAFLD patients than they were in healthy lean controls (P < 0.01). Accumulation of excess lipids in liver cells is a crucial factor that leads to hepatocyte degeneration and inflammation. Therefore, effective lipid transport and reduction in lipid synthesis may be crucial for the prevention and treatment of fatty liver disease. APOA-1, the primary apolipoprotein in HDL, is involved in cholesterol transport[15]. Mice with APOA-1 deficiency cannot form normal HDL particles, resulting in decreased cholesterol transport to liver tissue and cholesterol accumulation[16], which is consistent with our finding that APOA-1 levels were lower in lean-type MAFLD patients than in lean healthy controls, whereas triglyceride levels were significantly increased (P < 0.05). Cholinesterase is an enzyme secreted into the bloodstream by liver cells. A cross-sectional analysis of 5384 individuals found elevated serum cholinesterase concentration in patients with fatty livers, which is consistent with our results[17].

Metabolomics is a high-throughput detection method that reflects disease status by the overall biochemical phenotype. It allows the examination of changes in endogenous metabolites at the macroscopic level following exposure to biological stimuli. By analyzing comprehensive metabolic profiles, it is possible to identify disease-associated metabolites and reveal their metabolic pathways. In our study, serum metabolomics and liquid chromatography coupled with triple quadrupole mass spectrometry were used to investigate biomarkers specicic to lean-type MAFLD. Using a VIP > 1 and P < 0.05” as selection criteria, 65 biomarkers specific to lean-type MAFLD patients were identified in both positive and negative modes. These biomarkers were primarily concentrated in pathways related to fatty-acid, AA, and ether lipid metabolism.

Additionally, quantitative lipidomic analysis of four specific biomarkers revealed significant increases in the serum level of PA, OA, LA, and AA in lean-type MAFLD patients (P < 0.05), which were 2.23, 2.09, 1.86, and 1.98 times higher, respectively, than those in healthy individuals. FAs are components of triglycerides. Approximately 60 types of fatty acids have been identified in the plasma and tissues. However, only a few of them can be absorbed and used by the human body[18]. The homeostasis of body fatty acids ensures normal functioning.

FAs have a crucial role in lipid metabolism, but studies of their role in lean-type MAFLD patients are lacking. Current research indicates a close association between fatty acids and metabolic disorders, in which elevated levels of PA, palmitoleic acid, and LA are positively correlated with the onset and progression of MAFLD[19]. A study by Gambino et al[20] compared the changes in serum free FAs in MAFLD patients and in healthy controls, and reported significant increases in the serum level of free LA, OA, and AA. A plasma lipidomics study by Puri et al[21] reported significantly higher levels of PA, OA, and LA in MAFLD and nonalcoholic steatohepatitis patients, than in healthy individuals, but found no significant changes in AA. These findings align broadly with our research findings and, to a certain extent, reflect the serum levels of PA, OA, LA, and AA in lean-type MAFLD patients.

AA is a fatty-acid present in the cell membrane and is involved in cellular signal transduction during various inflammatory responses[22]. Abnormalities of FA metabolism disrupt the balance between the release and uptake of FAs in serum, leading to increased FA generation and reduced re-esterification capability. This eventually causes the accumulation of serum FAs, resulting in lipotoxicity and subsequent damage to the cardiovascular, endocrine, and digestive systems[23]. These studies indicate that abnormal FA metabolism in lean-type MAFLD patients significantly increases the likelihood of developing metabolic disorders. Based on existing research, we believe that elevated serum levels of PA and OA are directly associated with the occurrence of lean-type MAFLD, and that increased LA and AA levels are linked to the progression of MAFLD. In addition, PA promotes hepatic stellate cell activation, increases extracellular matrix deposition in MAFLD rats[24], induces podocyte apoptosis[25], and contributes to inflammation[26]. OA is a monounsaturated FA and is the preferred substrate for synthesizing triglycerides and cholesterol esters, and can induce hepatic cell steatosis and enhance tumor invasiveness[27]. LA is an essential FA that cannot be synthesized in the body and must be obtained from dietary sources. LA is converted to AA enzymes, such as Δ-6 desaturase (Figure 8).

Figure 8
Figure 8 Effects of fatty acids on metabolic-associated fatty liver disease. Through serum targeted metabolomics studies, we measured four specific biomarkers. The preliminary conclusion was that the transformation of lean control individuals to lean metabolic-associated fatty liver disease (MAFLD) may affect the Oleic acid, linoleic-arachidonic acid and palmitic acid pathways. Linoleic acid is converted to arachidonic acid by Δ-6 desaturase, as shown in the figure.
CONCLUSION

Serum-targeted metabolomics found that fatty-acid metabolism was impaired in lean-type MAFLD patients. The biomarkers identified in this study potentially provide insights into the treatment of lean-type MAFLD. The study results warrant further investigation but are limited by the small sample size.

ACKNOWLEDGEMENTS

Special thanks to each of the authors for their contributions to this manuscript.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade C, Grade C

Novelty: Grade A, Grade B, Grade B

Creativity or Innovation: Grade A, Grade B, Grade B

Scientific Significance: Grade B, Grade B, Grade B

P-Reviewer: Capone D, Italy; Soriano-Ursúa MA, Mexico; Sukocheva OA, Australia S-Editor: Wang JJ L-Editor: Filipodia P-Editor: Zheng XM

References
1.  Fan JG. Epidemiology of alcoholic and nonalcoholic fatty liver disease in China. J Gastroenterol Hepatol. 2013;28 Suppl 1:11-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 202]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
2.  Estes C, Anstee QM, Arias-Loste MT, Bantel H, Bellentani S, Caballeria J, Colombo M, Craxi A, Crespo J, Day CP, Eguchi Y, Geier A, Kondili LA, Kroy DC, Lazarus JV, Loomba R, Manns MP, Marchesini G, Nakajima A, Negro F, Petta S, Ratziu V, Romero-Gomez M, Sanyal A, Schattenberg JM, Tacke F, Tanaka J, Trautwein C, Wei L, Zeuzem S, Razavi H. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016-2030. J Hepatol. 2018;69:896-904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 776]  [Cited by in F6Publishing: 1075]  [Article Influence: 179.2]  [Reference Citation Analysis (0)]
3.  Hao XY, Zhang K, Huang XY, Yang F, Sun SY. Muscle strength and non-alcoholic fatty liver disease/metabolic-associated fatty liver disease. World J Gastroenterol. 2024;30:636-643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 1]  [Reference Citation Analysis (0)]
4.  Ke Y, Xu C, Lin J, Li Y. Role of Hepatokines in Non-alcoholic Fatty Liver Disease. J Transl Int Med. 2019;7:143-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
5.  Castera L, Pinzani M. Non-invasive assessment of liver fibrosis: are we ready? Lancet. 2010;375:1419-1420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 98]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
6.  Alkhouri N, Feldstein AE. Noninvasive diagnosis of nonalcoholic fatty liver disease: Are we there yet? Metabolism. 2016;65:1087-1095.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 47]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
7.  Kim D, Kim WR. Nonobese Fatty Liver Disease. Clin Gastroenterol Hepatol. 2017;15:474-485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 237]  [Article Influence: 33.9]  [Reference Citation Analysis (1)]
8.  Liu CJ. Prevalence and risk factors for non-alcoholic fatty liver disease in Asian people who are not obese. J Gastroenterol Hepatol. 2012;27:1555-1560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 150]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
9.  Alam S, Jahid Hasan M, Khan MAS, Alam M, Hasan N. Effect of Weight Reduction on Histological Activity and Fibrosis of Lean Nonalcoholic Steatohepatitis Patient. J Transl Int Med. 2019;7:106-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
10.  Leung JC, Loong TC, Wei JL, Wong GL, Chan AW, Choi PC, Shu SS, Chim AM, Chan HL, Wong VW. Histological severity and clinical outcomes of nonalcoholic fatty liver disease in nonobese patients. Hepatology. 2017;65:54-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 255]  [Article Influence: 36.4]  [Reference Citation Analysis (0)]
11.  Sookoian S, Pirola CJ. Systematic review with meta-analysis: risk factors for non-alcoholic fatty liver disease suggest a shared altered metabolic and cardiovascular profile between lean and obese patients. Aliment Pharmacol Ther. 2017;46:85-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 156]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
12.  Qiu S, Cai Y, Yao H, Lin C, Xie Y, Tang S, Zhang A. Small molecule metabolites: discovery of biomarkers and therapeutic targets. Signal Transduct Target Ther. 2023;8:132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 112]  [Article Influence: 112.0]  [Reference Citation Analysis (0)]
13.  Wang AY, Dhaliwal J, Mouzaki M. Lean non-alcoholic fatty liver disease. Clin Nutr. 2019;38:975-981.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 73]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
14.  Ye Q, Zou B, Yeo YH, Li J, Huang DQ, Wu Y, Yang H, Liu C, Kam LY, Tan XXE, Chien N, Trinh S, Henry L, Stave CD, Hosaka T, Cheung RC, Nguyen MH. Global prevalence, incidence, and outcomes of non-obese or lean non-alcoholic fatty liver disease: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 2020;5:739-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 459]  [Article Influence: 114.8]  [Reference Citation Analysis (0)]
15.  Ma D, Liu W, Wang Y. ApoA-I or ABCA1 expression suppresses fatty acid synthesis by reducing 27-hydroxycholesterol levels. Biochimie. 2014;103:101-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
16.  Li CX, Chen LL, Li XC, Ng KT, Yang XX, Lo CM, Guan XY, Man K. ApoA-1 accelerates regeneration of small-for-size fatty liver graft after transplantation. Life Sci. 2018;215:128-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 12]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
17.  Katoh S, Peltonen M, Wada T, Zeniya M, Sakamoto Y, Utsunomiya K, Tuomilehto J. Fatty liver and serum cholinesterase are independently correlated with HbA1c levels: cross-sectional analysis of 5384 people. J Int Med Res. 2014;42:542-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
18.  Tvrzicka E, Kremmyda LS, Stankova B, Zak A. Fatty acids as biocompounds: their role in human metabolism, health and disease--a review. Part 1: classification, dietary sources and biological functions. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2011;155:117-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 208]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
19.  Park H, Hasegawa G, Shima T, Fukui M, Nakamura N, Yamaguchi K, Mitsuyoshi H, Minami M, Yasui K, Itoh Y, Yoshikawa T, Kitawaki J, Ohta M, Obayashi H, Okanoue T. The fatty acid composition of plasma cholesteryl esters and estimated desaturase activities in patients with nonalcoholic fatty liver disease and the effect of long-term ezetimibe therapy on these levels. Clin Chim Acta. 2010;411:1735-1740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
20.  Gambino R, Bugianesi E, Rosso C, Mezzabotta L, Pinach S, Alemanno N, Saba F, Cassader M. Different Serum Free Fatty Acid Profiles in NAFLD Subjects and Healthy Controls after Oral Fat Load. Int J Mol Sci. 2016;17:479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 63]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
21.  Puri P, Wiest MM, Cheung O, Mirshahi F, Sargeant C, Min HK, Contos MJ, Sterling RK, Fuchs M, Zhou H, Watkins SM, Sanyal AJ. The plasma lipidomic signature of nonalcoholic steatohepatitis. Hepatology. 2009;50:1827-1838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 463]  [Cited by in F6Publishing: 484]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
22.  Tallima H, El Ridi R. Arachidonic acid: Physiological roles and potential health benefits - A review. J Adv Res. 2018;11:33-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 334]  [Article Influence: 47.7]  [Reference Citation Analysis (0)]
23.  Pavithra N, Bannikoppa PS, Uthappa S, Kurpad AV, Mani I. Plasma Fatty Acid Composition and Estimated Desaturase Activities Reflect Dietary Patterns in Subjects with Metabolic Syndrome. Indian J Clin Biochem. 2018;33:290-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
24.  Hanayama M, Yamamoto Y, Utsunomiya H, Yoshida O, Liu S, Mogi M, Matsuura B, Takeshita E, Ikeda Y, Hiasa Y. The mechanism of increased intestinal palmitic acid absorption and its impact on hepatic stellate cell activation in nonalcoholic steatohepatitis. Sci Rep. 2021;11:13380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
25.  Xiang XY, Liu T, Wu Y, Jiang XS, He JL, Chen XM, Du XG. Berberine alleviates palmitic acidinduced podocyte apoptosis by reducing reactive oxygen speciesmediated endoplasmic reticulum stress. Mol Med Rep. 2021;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
26.  Chen J, Li L, Zhou Y, Zhang J, Chen L. Gambogic acid ameliorates high glucose- and palmitic acid-induced inflammatory response in ARPE-19 cells via activating Nrf2 signaling pathway: ex vivo. Cell Stress Chaperones. 2021;26:367-375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
27.  Song H, Yang R, Zhang J, Sun P, Xing X, Wang L, Sairijima T, Hu Y, Liu Y, Cheng H, Zhang Q, Li L. Oleic acid-induced steatosis model establishment in LMH cells and its effect on lipid metabolism. Poult Sci. 2023;102:102297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]