Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2020 Feb 27;15(2):e0219412. doi: 10.1371/journal.pone.0219412

Serum miR-379 expression is related to the development and progression of hypercholesterolemia in non-alcoholic fatty liver disease

Kinya Okamoto 1,*, Masahiko Koda 1,#, Toshiaki Okamoto 1,, Takumi Onoyama 1,, Kenichi Miyoshi 1,, Manabu Kishina 1,, Tomomitsu Matono 1,, Jun Kato 1,, Shiho Tokunaga 1,, Takaaki Sugihara 1,, Akira Hiramatsu 2,#, Hideyuki Hyogo 3,#, Hiroshi Tobita 4,#, Shuichi Sato 4,#, Miwa Kawanaka 5,#, Yuichi Hara 6,#, Keisuke Hino 6, Kazuaki Chayama 2, Yoshikazu Murawaki 1, Hajime Isomoto 1
Editor: Catherine Mounier7
PMCID: PMC7046274  PMID: 32106257

Abstract

Introduction

Non-alcoholic fatty liver disease (NAFLD) has a wide spectrum, eventually leading to cirrhosis and hepatic carcinogenesis. We previously reported that a series of microRNAs (miRNAs) mapped in the 14q32.2 maternally imprinted gene region (Dlk1-Dio3 mat) are related to NAFLD development and progression in a mouse model. We examined the suitability of miR-379, a circulating Dlk1-Dio3 mat miRNA, as a human NAFLD biomarker.

Methods

Eighty NAFLD patients were recruited for this study. miR-379 was selected from the putative Dlk1-Dio3 mat miRNA cluster because it exhibited the greatest expression difference between NAFLD and non-alcoholic steatohepatitis in our preliminary study. Real-time PCR was used to examine the expression levels of miR-379 and miR-16 as an internal control. One patient was excluded due to low RT-PCR signal.

Results

Compared to normal controls, serum miR-379 expression was significantly up-regulated in NAFLD patients. Receiver operating characteristic curve analysis suggested that miR-379 is a suitable marker for discriminating NAFLD patients from controls, with an area under the curve value of 0.72. Serum miR-379 exhibited positive correlations with alkaline phosphatase, total cholesterol, low-density-lipoprotein cholesterol and non-high-density-lipoprotein cholesterol levels in patients with early stage NAFLD (Brunt fibrosis stage 0 to 1). The correlation between serum miR-379 and cholesterol levels was lost in early stage NAFLD patients treated with statins. Software-based predictions indicated that various energy metabolism–related genes, including insulin-like growth factor-1 (IGF-1) and IGF-1 receptor, are potential targets of miR-379.

Conclusions

Serum miR-379 exhibits high potential as a biomarker for NAFLD. miR-379 appears to increase cholesterol lipotoxicity, leading to the development and progression of NAFLD, via interference with the expression of target genes, including those related to the IGF-1 signaling pathway. Our results could facilitate future research into the pathogenesis, diagnosis, and treatment of NAFLD.

Introduction

Non-alcoholic fatty liver disease (NAFLD) is an important cause of chronic liver injury, with an increasing incidence worldwide [1]. NAFLD, regarded as a hepatic manifestation of metabolic syndrome, is defined by significant lipid deposition in hepatocytes (excessive numbers of fat-laden hepatocytes are observed by light microscopy), unrelated to excessive alcohol consumption [2]. The prevalence of NAFLD is almost 25% worldwide and expected to increase with increasing incidence of obesity and metabolic diseases such as type 2 diabetes mellitus (T2DM) and hyperlipidemia [3].

The mechanism underlying the development of NAFLD has not been fully elucidated. Currently, the multiple parallel hit theory is the most widely accepted mechanism for the progression of NAFLD [4]. This theory suggests that the disease process begins with de novo lipogenesis (DNL) by increase fructose consumption by western style diet and the development of insulin resistance resulting from excessive energy intake [5, 6]. Fructose is a potent lipogenic carbohydrate contributing to hepatic steatosis. Fructose is taken into hepatocyte via glucose transporter 2 and converted into fructose-1-phosphate (F1P) by fructokinase. These physiological sequences are not controlled by insulin and induced by fructose [6]. Fructose-bisphosphate aldolase B (known as hepatic aldolase) converts F1P into glycogen, glucose, lactate, and acetyl-CoA. Fructose also upregulate key transcription factors for fatty acid synthesis such as sterol response element binding protein 1c and carbohydrate responsive element binding Protein [7]. Both acetyl-CoA oversupply and induce of lipogenic enzymes increase DNL in hepatocyte strongly. Insulin resistance in turn leads to hyperinsulinemia, resulting in upregulated hepatic DNL and adipose tissue lipolysis. These “primary hits” increase the susceptibility of hepatocytes to multiple pathogenetic factors, such as upregulated expression of pro-inflammatory cytokines and eicosanoids, Fas ligand, and Toll-like receptor ligands; increased reactive oxygen species (ROS) generation; and altered production of adipokines [8]. Whole-body organs such as adipose tissue, the gut, and gut microbiota are also involved in the pathologic process [9, 10]. Collectively, these factors promote hepatocyte apoptosis through mitochondrial dysfunction [11] and an endoplasmic reticulum stress reaction [12]. Such continuous liver tissue injury ultimately leads to fibrosis [13].

The clinical status of NAFLD patients is generally classified broadly into one of just two categories: non-alcoholic fatty liver (NAFL) or non-alcoholic steatohepatitis (NASH) [14]. NAFL encompasses most of the NAFLD spectrum and is a benign condition. NASH, on the other hand, is defined as the combination of steatosis with lobular inflammation and hepatocyte ballooning; it can progress to liver fibrosis and result in cirrhosis and cancerous malignancies [14]. In contrast to NAFL, NASH is a life-threatening disease. Indeed, a cohort study showed that 35% of NASH patients die during the 7.6-year average follow-up period, whereas no NAFL patients followed in that study died during the same period [15].

Considering the wide disease spectrum of NAFLD, which can result in significant differences in prognosis, it is likely that mechanisms that regulate one or more of these multiple-hit factors exist. Some risk factors for the development of liver fibrosis in NAFLD include age over 50 years, severe obesity, complications associated with T2DM, increased ferritin levels, and patatin-like phospholipase domain–containing 3 gene polymorphisms [16, 17]. However, more-sensitive and -reliable biomarkers are urgently needed to predict outcome in NAFLD patients and enable treatment to begin in the early stage.

MicroRNAs (miRNAs) are a class of endogenous, noncoding, small RNAs that regulate gene expression [18]. Mature miRNAs are introduced into RNA-induced silencing complexes (RISCs) [19]. A RISC bearing a miRNA binds to a partially complementary mRNA sequence and represses the translation of that mRNA. Because miRNAs cause incomplete base-pair matching with mRNAs, a single miRNA can inhibit the translation of hundreds to thousands of target genes [20]. As such, miRNAs play an important role in many cellular processes, including metabolism, inflammation, and fibrosis [21]. Accumulating evidence from both animal model and human patients indicates that miRNAs contribute to the pathogenesis and progression of NAFLD. For example, the expression levels of miR-29c, miR-34a, miR-155, and miR-200b in mouse model liver and miR-122 and miR-34a in human liver are thought to be involved in the development of NASH [2224]. Our previous study showed that a series of miRNAs mapped in the 14q32.2 maternally imprinted gene cluster region delineated by the delta-like homolog 1 and type III iodothyronine deiodinase genes (Dlk1-Dio3 mat) are related to NAFLD development and progression in a NAFL/NASH mouse model (fatty liver Shionogi [FLS] and mutated leptin gene transferred FLS ob/ob) [25]. Seven miRNAs in the Dlk1-Dio3 mat (miR-127, -136, -376c, -379, -409-3p, -411, and -495) are strongly upregulated in both FLS and FLS ob/ob liver tissues. In contrast to previously reported NAFLD-related miRNAs, the expression of these seven miRNAs was higher in NAFL model mice than NASH model mice.

Recent studies have clearly indicated that miRNAs are secreted into circulating body fluids from various tissues [26]. A considerable amount of secreted miRNAs are protected from enzymatic and physical degradation by binding to proteins or lipoproteins that are then stored in exosomes [27]. These observations suggest that serum miRNAs are potential biomarkers for NAFLD, as they could reflect various pathologic changes in miRNA expression in the liver. Indeed, our preliminary study in human NAFLD patients indicated that serum levels of the respective human homologs of the candidate Dlk1-Dio3 mat miRNAs are related to NAFLD progression [25]. The aim of the present study was to examine the suitability of circulating 14q32.2 mat miRNA as a human NAFLD biomarker.

Materials and methods

Ethics statement

This study was approved by the committee for ethics in medical experiments on human subjects of the medical faculty of Tottori University (protocol no. 2374) and all collaborative medical institutes: Hiroshima University Hospital, JA Hiroshima General Hospital, Kawasaki University Hospital, and Shimane University Hospital. The study was conducted in accordance with the declaration of Helsinki. Written informed consent was obtained from each patient before blood was collected.

Sample size calculation

The effective sample size has been calculated from the outcomes of our previous clinical study about serum Dlk1-Dio3 mat miRNA expression in NAFLD patients [25]. According to the criterion of our previous study, we set up the relevant difference of miR-379 expression level is 2log2. The associated standard deviation was estimated to be 1.6log2 based on the serum miR-379 expression of normal control in our previous study [25]. Given a statistical power of 80% and 0.05 level of significance, a sample size of 11 in each group will be sufficient to detect a clinically relevant difference between groups. To allow for dropout of patients (up to 20%) and further subgroup analysis, we aim to recruit 80 participants.

Patient recruitment and collection of blood and liver samples

Recruiting and obtaining liver tissue, blood sample and clinical data were performed at Tottori University Hospital and collaborative medical institutes: Hiroshima University Hospital, JA Hiroshima General Hospital, Kawasaki University Hospital, and Shimane University Hospital from July 2014 to March 2016. All participants were Japanese who live in the western part of Japanese main-island. NAFLD outpatients who underwent continuous clinical follow-up at Tottori University Hospital or collaborative medical institutes were recruited by interview with our study members. In interview, we gave clear exposition of the purpose, procedures, duration and potential adverse events of our study using the printed description which have been obtained the approval of the committee for ethics in medical experiments on human subjects of the medical faculty of Tottori University. Participants were given sufficient time to read the consent and have all questions answered before signing the consent form voluntary. Exclusion criteria included chronic hepatitis B or C virus infection (positive hepatitis B surface antigen or hepatitis C antibody), habitual alcohol consumption over 20 g/day, administration of liver steatotic drugs (such as glucocorticoids, tamoxifen, amiodarone, methotrexate, or valproate), primary biliary cholangitis (positive anti-mitochondrial antibody), or autoimmune liver disease (positive anti-nuclear antibody or anti-smooth muscle antibody). All NAFLD patients underwent liver biopsy to confirm the diagnoses of NAFLD, and the histologic grade and NAFLD stage was determined according to the Brunt system [28]. NAFL and NASH were defined by >5% fat-laden hepatocytes in biopsy samples and at least 6 months of continuous blood test results in which alanine aminotransferase (ALT) and aspartate aminotransferase (AST) remained at <2-fold of the normal range or in excess, respectively. Blood sample collection for serum miRNA isolation and clinical blood tests were performed at the same time and within 1 month of liver biopsy. Blood samples were collected in the fasted state. For each sample, blood serum was isolated by refrigerated centrifugation at 4°C and 1500 × g for 10 min and then stored at −80°C until use. Eighty NAFLD patients were enrolled in this study. One NAFLD patient was excluded from this study due to low RT-PCR signal, even after 60 PCR cycles. The NAFLD patients were divided into two subgroups as follows: 9 NAFL patients, and 70 NASH patients. In another analysis, NAFLD patients were also divided into early stage (n = 53) and advanced stage (n = 26) groups. Early stage was defined as Brunt fibrosis stage 0 or 1, and the advanced stage was defined as Brunt fibrosis stage 2 to 4. As normal control, 10 outpatients with asymptomatic gallbladder stones without liver function abnormalities and fatty liver changes by ultrasound imaging were recruited. The clinicopathologic features of each patient group are shown in Table 1. We assessed whether our NAFLD sample can be considered representative of a larger NAFLD population. Concerning age of the participants, the mean age of our NAFLD patients are 48.6 ± 15.4. The majority of NAFLD concerning studies indicate that the median age of NAFLD patients are in the range from 40 to 59 [1]. Meta-analytic assessment of prevalence and incidence of NAFLD showed that the prevalence of NAFLD increases with age, however, the prevalence rate are similar from age 40’s to 60’s (26.5% to 28.9%, respectively) [1]. Former large population studies support that the prevalence of NAFLD is higher in men than in women [29]. In our present study, male NAFLD patients are also more frequently observed than female patients (54 males and 25 females). Overweight is one of the strong risk factors for NAFLD [1]. Previous Japanese NAFLD patients study also showed a clear relationship between BMI and NAFLD development [30]. In our study, mean of body mass index of NAFLD patients excess 25 (29.3 ± 6.1), which indicate overweight. Development and resolution of NAFLD were both closely related to metabolic syndrome especially diabetes mellitus (DM) [30]. Hamaguchi et al. reported that 25–40% of Japanese NAFLD patients have established DM [30]. In our study, 24 cases (30%) of NAFLD patients had complication of DM. In subgroup, 1 of NAFL (11%) and 23 of NASH (33%), and 16 of NAFLD early stage (30%) and 8 of NAFLD advanced stage (31%) had DM. Because of these demographic features, we regarded that our NAFLD participants could be considered as the representative group of larger population of NAFLD.

Table 1. Clinicopathologic features of NAFLD patients and controls.

Control (CON) (n = 10) NAFL (n = 9) NASH (n = 70) p value NAFLD early stage (n = 53) NAFLD advanced stage (n = 26) p value
NAFL and CON NASH and CON NAFL and NASH Early stage and CON Advanced stage and CON Early stage and advanced stage
Age 59.3 ± 16.6 44 ± 10 50 ± 16 0.080 0.162 0.533 45.4 ± 14.7 55.2 ± 14.9 0.023* 0.742 0.021*
Gender M/F 4 / 6 7 / 2 47 / 23 0.170 0.161 0.710 38 / 15 16 / 10 0.071 0.460 0.261
BMI 21.9 ± 5.2 26.4 ± 2.2 29.8 ± 6.3 0.270 0.002* 0.259 29.8 ± 5.5 28.4 ± 7.2 0.003* 0.024* 0.628
Brunt Stage - 0.89 ± 0.33 1.58 ± 0.87 - - 0.041* 1.0 ± 0.2 2.6 ± 0.6 - - < 0.001*
Brunt Grade - 1.0 ± 0 1.58 ± 0.67 - - 0.021* 1.3 ± 0.6 1.9 ± 0.6 - - 0.001*
T-Bil. 0.8 ± 0.3 0.9 ± 0.3 1.0 ± 0.4 0.927 0.479 0.805 0.9 ± 0.4 1.2 ± 0.3 0.908 0.071 0.014
Alb 4.3 ± 0.4 4.6 ± 0.4 4.4 ± 0.4 0.123 0.175 0.701 4.5 ± 0.4 4.4 ± 0.4 0.378 0.880 0.470
PT (%) 96.7 ± 9.5 107.9 ± 12.5 99.2 ± 13.2 0.415 0.187 0.938 104.0 ± 12.6 92.4 ± 11.7 0.578 0.837 0.001*
AST (U/L) 27.8 ± 18.8 40 ± 19 49 ± 19 0.360 0.005* 0.404 45.5 ± 16.4 53.3 ± 23.1 0.021* 0.002* 0.198
ALT (U/L) 25.5 ± 15.1 72 ± 41 77 ± 40 0.028* 0.001* 0.923 78.3 ± 39.1 74.5 ± 41.6 0.001* 0.002* 0.910
ALP (U/L) 276.5 ± 91.7 259 ± 67 237 ± 84 0.886 0.350 0.752 240.5 ± 73.4 238.6 ± 100.2 0.434 0.451 0.995
GGT (U/L) 47.3 ± 45.6 65 ± 45 62 ± 45 0.667 0.598 0.980 63.7 ± 46.1 61.4 ± 41.6 0.542 0.676 0.976
LDH (U/L) 158.3 ± 45.6 215 ± 84 209 ± 47 0.244 0.226 0.958 216.3 ± 58.4 199.5 ± 32.6 0.144 0.391 0.362
Ch-E (U/L) 348.3 ± 66.2 351 ± 85 379 ± 82 0.997 0.511 0.634 388.9 ± 79.7 352.8 ± 84.8 0.310 0.988 0.150
BUN (mg/dL) 11.0 ± 2.4 13.8 ± 2.5 13.1 ± 2.4 0.216 0.301 0.766 13.1 ± 2.5 13.3 ± 1.9 0.296 0.276 0.955
Cr (mg/dL) 0.56 ± 0.17 0.79 ± 0.13 0.75 ± 0.15 0.054 0.092 0.638 0.76 ± 0.14 0.74 ± 0.16 0.068 0.109 0.920
UA (mg/dL) 5.7 ± 1.2 6.0 ± 1.1 6.3 ± 1.4 0.973 0.792 0.883 6.3 ± 1.4 6.2 ± 1.4 0.805 0.867 0.985
Ferritin 42.4 ± 33.0 142.1 ± 74.0 210.6 ± 174.5 0.723 0.338 0.477 190.6 ± 158.6 229.1 ± 186.6 0.439 0.287 0.614
FBS (mg/dL) 93.7 ± 9.7 104.0 ± 11.5 117.6 ± 45.6 0.849 0.204 0.621 117.7 ± 47.8 113.9 ± 33.8 0.220 0.394 0.923
HgbA1c (%) 6.3 ± 1.0 5.9 ± 0.6 6.3 ± 1.5 0.911 0.996 0.658 6.3 ± 1.5 6.2 ± 1.4 0.995 0.999 0.938
IRI (μU/mL) 17.1 ± 19.6 18.3 ± 13.5 - - 0.820 18.8 ± 15.0 17.2 ± 12.8 - - 0,897
HOMA-IR 4.6 ± 5.7 5.3 ± 6.7 - - 0.768 5.5 ± 7.4 4.9 ± 4.5 - - 0.921
T-Chol (mg/dL) 202 ± 44 199 ± 47 204 ± 35 0.978 0.988 0.913 206.6 ± 36.7 197.5 ± 36.3 0.936 0.940 0.936
LDL-C (mg/dL) 134.1 ± 37.4 130.3 ± 43.9 131.3 ± 33.2 0.974 0.978 0.996 135.1 ± 33.9 122.5 ± 34.9 0.997 0.709 0.288
HDL-C (mg/dL) 67.2 ± 34.3 50.9 ± 6.9 49.4 ± 9.0 0.033* 0.004* 0.930 49.1 ± 7.9 50.6 ± 10.6 0.003* 0.012* 0.853
Non-HDL -C (mg/dL) 150.2 ± 17.0 147.8 ± 47.0 154.1 ± 36.4 0.908 0.830 0.643 157.6 ± 5.2 144.5 ± 7.6 0.683 0.758 0.161
TG (mg/dL) 104.3 ± 64.8 112.1 ± 50.9 149.9 ± 69.0 0.967 0.139 0.255 154.5 ± 70.6 128.4 ± 60.7 0.104 0.629 0.255
Compl. DM 0 1 (11%) 23 (33%) - - 0.182 16 (30%) 8 (31%) - - 0.958

Value data are expressed as the mean ± standard deviation.

*: p < 0.05 in analysis of variance (ANOVA). NAFLD early stage was defined as Brunt fibrosis stage 0 or 1, and advanced stage was defined as Brunt fibrosis stage 2 to 4. T-Bil: total bilirubin, Alb: albumin, AST: alanine aminotransferase, ALT: aspartate aminotransferase, ALP: alkaline phosphatase, GGT: gamma-glutamyl transferase, LDH: lactate dehydrogenase, Ch-E: choline esterase, BUN: blood urea nitrogen, Cr: creatinine, UA: uric acid, FBS: fasting blood sugar, HgbA1c: hemoglobin A1c which was measured by national glycohemoglobin standardization program certified method, IRI: immunoreactive insulin, HOMA IR: homeostasis model assessment of insulin resistance, T-Chol: total cholesterol, LDL-C: low-density-lipoprotein cholesterol, HDL-C: high-density-lipoprotein cholesterol, Non-HDL-C: calculated by subtracting HDL-C from T-Chol levels, TG: triacylglycerol, Compl. DM: complication of diabetes mellitus, DM can be diagnosed if the patient has HgbA1c level of 6.3% or greater on two separate blood tests.

miRNA expression analysis with human serum

miRNA extract from the serum, Quantitative real-time polymerase chain reaction (RT-PCR) and data analysis were carried out at Tottori University. miR-379 was selected from the putative Dlk1-Dio3 mat miRNA cluster because it exhibited the greatest difference in serum expression between NAFL and NASH patients in our preliminary study [25]. Comparing to the normal controls, serum miR-379 overexpressed in simple steatosis (we named NAFL to simple steatosis in our previous report) group (3.3 ± 3.1 log2) and down regulated in NASH patients group (-7.4 ± 5.9 log2) in our previous study [25]. A miRNeasy serum/plasma kit (Qiagen Venlo, Nederland) was used to extract miRNAs from each 200-μL serum sample according to the manufacturer’s instructions. The miScript II reverse transcription kit (Qiagen) was used for reverse transcription of serum miRNA according to the manufacturer’s instructions. RT-PCR was used to examine the expression levels of miRNA, and data were analyzed using the ΔΔCT method of relative quantification. Applied Biosystems TaqMan® MicroRNA Assays (Applied Biosystems, Waltham, MA, USA) and an ABI7900HT system (Applied Biosystems) were used for quantitative RT-PCR amplification of serum miRNAs. The primer sequences of hsa-miR-379 was UGGUAGACUAUGGAACGUA. We selected miR-16 as an endogenous control. miR-16 is one of the most commonly used reference miRNAs in serum miRNA expression analyses [31, 32]. To the best of our knowledge, no previous reports have indicated a relationship between liver disease and miR-16. We also examined preliminary study about RT-PCR measurement in human serum between endogenous miR-16 and non-mammal spike in control miRNA. Serum samples were obtained from normal controls (n = 10). 1.6 x 108 copies of C. elegans (Ce)-miR-39-1 (Qiagen) and 1 μg bacteriophage MS2 RNA (Roche, Penzberg, Germany) as carrier RNA were added for each 200 μL serum sample. PT-PCR was performed using the same protocol of the present study. The primer sequence of hsa-miR-16 was UAGCAGCACGUAAAUAUUGGCG. The manufacturer does not disclose the primer sequence of Ce-miR-39-1. Both of miR-16 and Ce-miR-39-1 were stably expressed between samples and their standard deviations of threshold cycles were within ± 1 cycle (34.0 ± 0.8 and 23.9 ± 0.1, respectively). We concluded that endogenous miR-16 could be applied as internal control of RT-PCR in serum miRNA.

Predicting miRNA targets

The physiological roles of miR-379 in liver are still hardly identified. Few former studies revealed the relationship between liver function and miR-379. A comprehensive investigation for miR-379 function should be carried out. We adopted the computational miRNA target genes prediction. Software prediction of miRNA target genes is a popular and reliable method to estimate miRNA physiological functions [33]. We used web-based software DIANA microT-CDS 5.0 (http://diana.cslab.ece.ntua.gr/) for miR-379 putative target gene analysis. The threshold for the target prediction score in DIANA microT-CDS was set to 0.7. One miRNA can interference hundreds to thousands genes [20]. We selected gene ontology (GO) annotation to select NAFLD related genes. Database for Annotation, Visualization, and Integrated Discovery (DAVID) 6.8 (http://david.abcc.ncifcrf.gov/) was used for GO annotation, and the Kyoto Encyclopedia of Genes and Genomes (KEGG) was used for pathway enrichment analysis.

Statistical analysis

Statistical analysis was performed using JMP 11.2.1 software (SAS Institute Inc., Cary, NC, USA). Value data are expressed as the mean ± standard deviation. The statistical significance of differences between groups was determined using the Student's t test or ANOVA, followed by Dunnett's test for multiple comparisons. Receiver operating characteristic (ROC) curve analysis was performed to assess NAFLD, NAFL, and NASH diagnostic accuracy. Linear regression analysis was used to examine correlations between miRNA levels and clinicopathologic parameters. Fisher’s exact test and the chi-square test were selected depending on the sample size and used to determine distribution differences of categorical variable. Differences were considered statistically significant at a p value < 0.05.

Results

Serum miR-379 expression was up-regulated in NAFLD patients

Compared to controls, serum miR-379 expression was significantly up-regulated in NAFLD patients (Fig 1A). In a subgroup analysis of NAFL and NASH patients, serum miR-379 expression was significantly higher in NAFL patients than normal controls (Fig 1B). We also compared early stage NAFLD (Brunt fibrosis stage 0 to 1) and advanced-stage NAFLD (Brunt fibrosis stage 2 to 4) patients with controls. Patients with early stage NAFLD exhibited significantly higher miR-379 expression than controls (Fig 1C). Expression of miR-379 in NASH patients was also higher than in controls, but the difference was not significant (p = 0.061) (Fig 1C). There was no significant difference in miR-379 expression between NAFL and NASH patients or between those with early or advanced-stage NAFLD.

Fig 1. Relative expression of serum miR-379 in NAFLD patients.

Fig 1

A: serum miR-379 expression level between NAFLD and normal control. B: serum miR-379 expression level between NAFL, NASH. C: serum miR-379 expression level between NAFLD early stage, advanced stage and normal control. NAFLD early stage was defined as Brunt fibrosis stage 0 or 1, and advanced stage was defined as Brunt fibrosis stage 2 to 4. Value data are expressed as the mean ± standard deviation. Quantitative real-time PCR (qRT-PCR) was used to examine miRNA levels. All qRT-PCR data were normalized to that for serum miR-16, and fold-change was calculated relative to data from normal controls. *p < 0.05.

Serum miR-379 is a potential NAFLD diagnostic marker

ROC curve analysis revealed that miR-379 is a potential marker for discriminating NAFLD patients from controls (area under the ROC curve [AUROC]: 0.72) (Fig 2). AUROC values for discriminating NAFL, NASH, and early and advanced-stage NAFLD patients from controls were 0.76, 0.72, 0.74, and 0.67, respectively (Fig 2).

Fig 2. Receiver operating characteristic (ROC) curve analysis.

Fig 2

ROC curves for serum miR-379 expression level for the diagnosis of NAFLD (A) and NAFLD subgroups (B: NAFL, C: NASH, D: NAFLD early stage and E: NAFLD advanced stage). Serum miR-379 expressions were normalized to that for serum miR-16. NAFLD early stage was defined as Brunt fibrosis stage 0 or 1, and advanced stage was defined as Brunt fibrosis stage 2 to 4. AUROC means area under the ROC curve.

Positive correlations were observed between serum miR-379 and alkaline phosphatase (ALP) or cholesterol levels in patients with NAFL or early stage NAFLD

We analyzed the correlations between clinicopathologic parameters and serum miR-379 levels in NAFLD patients. No significant correlation was identified between serum miR-379 expression in NAFLD patients and any of the parameters examined (S1 Fig). However, positive correlations were observed between serum miR-379 expression and ALP, total cholesterol, low-density-lipoprotein cholesterol (LDL-C) and non-HDL-C Cholesterol (non-HDL-C) levels in patients with early stage NAFLD (Fig 3). In contrast, there was no correlation between these parameters and serum miR-379 levels in controls or patients with advanced-stage NAFLD (Fig 3, S3 Fig).

Fig 3. Correlation between miR-379 and T-Chol, LDL-C, non-HDL-C and ALP levels.

Fig 3

Linear regression analysis was used to examine correlations between serum miR-379 levels and clinicopathologic parameters. Left, middle, and right columns present the results for the NAFLD early stage, NAFLD advanced stage groups and normal control group, respectively. Rows A, B, C and D show miR-379 correlation between T-Chol, LDL-C, non-HDL-C and ALP levels, respectively. R2: coefficient of determination. *: p < 0.05. Non-HDL-C: calculated by subtracting HDL-C from T-Chol levels.

Statin treatment weakened the correlation between miR-379 and cholesterol level

Nine of 51 patients with early stage NAFLD were undergoing treatment for hypercholesterolemia with hydroxymethyl glutaryl coenzyme A reductase (HMG CoA-reductase) inhibitors; commonly called statins. Among statin-treated and non-treated patients with early stage NAFLD, serum levels of total cholesterol, LDL-C, and triglycerides were similar (Fig 4). miR-379 expression levels were higher in the statin-treated group than the non-treated group, but the difference was not significant (5.1 ± 4.4 and 3.2 ± 4.8 log2 folds, respectively. p = 0.29). Linear regression analysis showed the non-treated group exhibited a significant positive correlation between total cholesterol and serum miR-379 expression. This trend was also observed in the statin-treated group, but the correlation was not significant (p = 0.10) (Fig 4).

Fig 4. Statin treatment and serum miR-379 expression, and correlation with cholesterol levels.

Fig 4

Value data are expressed as the mean ± standard deviation. R2: coefficient of determination. *p < 0.05.

Software-based predictions of miR-379 target genes

We predicted potential target genes of miR-379 using web-based software. Based on the selection criteria, 1423 human genes were identified as candidates. The candidate genes were classified according to GO annotation in Homo sapiens (Fig 5). Simple gene counting of GO terms showed that cellular process, metabolic process and biological regulation had a large proportion amount to over 70%.

Fig 5. Simple aggregation of Gene Ontology (GO) terms among putative miR-379 target genes.

Fig 5

The predicted miR-379 target gene dataset were fed into DAVID, version 6.8. Pie chart slices represent the number of genes associated with each GO term.

We also examined GO term enrichment analysis. The analysis can identify GO terms, which are significantly over-represented for DAVID pre-built human genome backgrounds [34]. miR-379 predicted target genes were richly represented in 12 GO terms compared to DAVID pre-built human genome backgrounds significantly (Table 2). Among 12 enriched GO, 10 terms related to cellular process regulations.

Table 2. GO-term enrichment analysis of predicted miR-379 target genes.

Go Term Gene Count % Fold enrichment p value
Positive regulation of macromolecule biosynthetic process 176 12.4 1.5 > 0.001*
Positive regulation of RNA metabolic process 156 11.0 1.5 > 0.001*
Positive regulation of gene expression 178 12.5 1.4 0.001*
Positive regulation of nucleobase-containing compound metabolic process 175 12.3 1.4 0.001*
Positive regulation of cellular biosynthetic process 181 12.7 1.4 0.002*
Positive regulation of transcription, DNA-templated 148 10.4 1.5 0.002*
Regulation of cellular macromolecule biosynthetic process 365 25.7 1.3 0.002*
Positive regulation of RNA biosynthetic process 149 10.5 1.5 0.002*
Regulation of macromolecule biosynthetic process 370 26.0 1.2 0.006*
Regulation of gene expression 387 27.2 1.2 0.010*
Cellular protein modification process 342 24.0 1.2 0.034*
Protein modification process 342 24.0 1.2 0.034*

Percentages indicate the number of predicted target genes associated with a GO term category compared to all predicted genes examined in the GO-term analysis. Fold-enrichment shows the abundance ratios of predicted miR-379 target genes and DAVID pre-built human genome backgrounds among GO terms. Only statistically significant results (p < 0.05) are displayed.

Each of the enriched GO terms still had hundreds genes (Table 2). It is one of the common weak points of this method that the output of genes can be large [34]. To select putative target gene for NAFLD pathology, we examined extending analysis using backend annotation database. Our GO term analysis showed that miR-379 seemed to relate biological regulations largely (Table 2). Therefore we selected KEGG pathway as the backend database. Ontology annotation via KEGG pathway mapping showed that biological functions have been identified for 32.8% of the candidate genes (467 of 1423 genes). Function-labeled miR-379 candidate target genes were primarily enriched in clusters associated with nutrition and energy regulation (FOXO and mTOR signaling pathways), cancer (melanoma, prostate cancer, p53 signaling, Hippo signaling, and transcriptional misregulation in cancer), and multi-functional cellular mechanisms or signaling pathways (cGMP-PKG signaling, focal adhesion, Hippo signaling pathway, pluripotency regulation in stem cells, TGF-beta signaling, and ubiquitin-mediated proteolysis) (Table 3).

Table 3. Enriched KEGG pathways among putative miR-379 target genes.

KEGG pathway Gene count % Fold enrichment p value
FOXO signaling pathway 21 1.5 2.3 > 0.001*
TGF-beta signaling pathway 15 1.1 2.6 0.001*
Ubiquitin mediated proteolysis 20 1.4 2.2 0.002*
Hippo signaling pathway 19 1.3 1.8 0.013*
Prostate cancer 13 0.9 2.2 0.015*
Transcriptional misregulation in cancer 20 1.4 1.9 0.018*
Signaling pathways regulating pluripotency of stem cells 17 1.2 2.3 0.027*
p53 signaling pathway 10 0.7 1.8 0.036*
cGMP-PKG signaling pathway 18 1.3 1.6 0.038*
Focal adhesion 21 1.5 2.1 0.038*
mTOR signaling pathway 9 0.6 1.7 0.040*
Melanoma 10 0.7 2.2 0.048*

Percentages indicate the number of predicted miR-379 target genes associated with a KEGG pathway compared to all predicted genes explored in the KEGG pathway analysis. Fold-enrichment shows the abundance ratios of predicted miR-379 target genes and DAVID pre-built human genome backgrounds among KEGG terms. Only statistically significant results (p < 0.05) are displayed.

Finally, to identify probable miR-379 target genes related to the pathology of NAFLD, we conducted a keyword search of the U.S. National Library of Medicine database PubMed (https://www.ncbi.nlm.nih.gov/pubmed) using the terms “KEGG annotated putative target gene” and “NAFLD” or “NASH”. A total of 27 predicted genes were associated with NAFLD development or progression, including multi-functional cellular mechanisms or signaling pathways (HDAC2), fibrosis and inflammation (CAT, CTGF, IL10, PDGFA, PDGFRA, SMAD4, TGFBR1, and THBS1), cell survival and proliferation (Bcl2, CCNB1, HGF, PMAIP1, PTEN, and YAP1), and energy management, including gluconeogenesis and lipogenesis (CREB1, EIF4E, FOXO1, INSR, IGF1, IGF1R, ITPR2, PRKAA1 and 2, RICTOR, SOCS1, and TCF7L2) (Table 4) [3560].

Table 4. Keyword search of the U.S. National Library of Medicine database PubMed to identify KEGG annotated miR-379 putative target genes associated with NAFLD or NASH.

Gene Code Protein name Reference
Bcl2 B-cell lymphoma 2 Panasiuk et al. 2006
CAT Catalase Kumar et al. 2013
CCNB1 Cyclin B1 Gentric et al. 2015
CREB1 cAMP responsive element binding protein 1 Oh et al. 2013
CTGF Connective tissue growth factor Colak et al. 2012
EIF4E Eukaryotic translation initiation factor 4E Wang et al. 2014
FOXO1 Forkhead box o1 Pan et al. 2017
HDAC2 Histone deacetylase 2 Kolodziejczyk et al. 2019
HGF Hepatocyte growth factor Kosone et al. 2007
INSR Insulin receptor Wu et al. 2017
IGF1 Insulin like growth factor 1 Adamek et al. 2018
IGF1R Insulin like growth factor 1 receptor Go et al. 2014
IL10 Interleukin 10 Cintra et al. 2008
ITPR2 Inositol 1, 4, 5-trisphosphate receptor type 2 Khamphaya et al. 2018
PDGFA Platelet derived growth factor subunit A Hardy et al. 2017
PDGFRA Platelet derived growth factor receptor A Abderrahmani et al.
PMAIP1 Phorbol 12-myristate 13-acetate induced protein 1 Kung et al. 2016
PRKAA1 5’ AMP-activated protein kinase catalytic subunit alpha 1 Garcia et al. 2019
PRKAA2 5’ AMP-activated protein kinase catalytic subunit alpha 2 Garcia et al. 2019
PTEN Phosphatase and tensin homolog Matsuda et al. 2013
RICTOR Rapamycin-insensitive companion of mammalian target of rapamycin Sydor et al. 2017
SMAD4 Small worm phenotype and mothers against decapentaplegic 4 Qin et al. 2018
SOCS1 Suppressor of cytokine signaling 1 Wang et al. 2017
TCF7L2 Transcription factor 7-like 2 Musso et al. 2009
TGFBR1 Transforming growth factor beta receptor 1 Matsubara et al. 2012
THBS1 Thrombospondin 1 Li et al. 2017
YAP1 yes-associated protein 1 Chen et al. 2018

Discussion

The present study revealed significantly higher serum levels of miR-379 in NAFLD patients compared to controls. Our previous study indicated that miR-379 expression in liver tissues of an NAFLD mouse model is strongly upregulated (>4 log2 compared to the normal control group) [25]. miR-379 secretion from liver tissue, probably via exosome particles rich in miR-379, appears to be related, at least in part, to the high circulating level observed in NAFLD patients.

Relatively little is known regarding the mechanism regulating miR-379 expression. miR-379 has been mapped to the miRNA cluster in the Dlk1-Dio3 mat region. Major regulators of Dlk1-Dio3 locus expression include methylated regulatory regions such as the germline-derived intergenic differentially methylated region and somatic MEG3-differentially methylated region [61, 62]. Moreover, CpG islands that are embedded in or near miRNA-coding regions also regulate the expression of Dlk1-Dio3 mat miRNA [63]. Dai et al. reported that miR-379 expression is directly regulated by DNA methylation [64]. In addition, histone acetylation functions synergistically with DNA methylation to regulate the Dlk1-Dio3 locus [63].

With respect to non–DNA methylation regulation, Guia and colleagues reported that the miRNA cluster miR-379/410 is a direct transcriptional target of the glucocorticoid receptor, which promotes insulin resistance and systemic dyslipidemia [65]. Guia et al. also showed that miR-379 is upregulated in liver tissue of obese subjects and that hepatic miR-379 expression in patients with obesity is correlated with both serum cortisol and triacylglycerol (TG) levels [65]. However, in our present study, TG levels in NAFLD patients did not differ significantly from those of controls (Table 1), and serum miR-379 expression was not correlated with TG level (p = 0.738, S1 Fig). This discrepancy may be related to differences between obese patients and NAFLD patients whose diagnosis was confirmed by liver biopsy. The mechanism of serum miRNA expression may also be related to this discrepancy. For example, sorting and selection occur during incorporation of cytosolic miRNAs into exosomes [66]. Because the level of circulating miRNAs is the sum total of miRNAs secreted from tissues/organs throughout the body, other metabolism-related organs may affect the level of circulating miRNA. Chartoumpekis et al. reported that miR-379 is overexpressed in white adipose tissue in an obese mouse model [67].

ROC curve analyses showed that miR-379 provides fair diagnostic accuracy for NAFLD. The AUROC of serum miR-379 for NAFLD diagnosis was >0.7 and similar to other single serologic markers for non-invasive detection of NAFLD, such as tumor necrosis factor–alpha, interleukin-6, and ferritin [68]. Most non-invasive NAFLD markers exhibit higher values and diagnostic accuracy in patients with liver fibrosis and cirrhosis [69]. In contrast to the majority of NAFLD diagnostic markers, the serum miR-379 level was significantly increased relative to NAFL, but there was no difference between NAFL and NASH. This distinctive feature of serum miR-379 may confer an advantage for detecting NAFLD in the early stage. For instance, serum miR-379 is a candidate factor for use in NAFLD diagnosis algorithms combining multiple biomarkers as a means of increasing sensitivity for early stage diagnosis [70].

Our present study showed that the serum miR-379 level is positively correlated with ALP in early stage NAFLD. Serum ALP is the traditional marker of cholestasis. However, the other cholestasis markers, such as bilirubin and gamma-glutamyl transferase, were not significantly correlated with miR-379 (S2 Fig). ALP is a plasma membrane–bound enzyme that catalyzes the hydrolysis of phosphate esters [71]. Though found in most body tissues, ALP is particularly abundant in the liver, bone, kidneys, and intestinal mucosa, with liver and bone serving as the predominate organs supplying ALP to circulating body fluids [71]. Chronic liver diseases, including NAFLD, increase serum ALP levels [72, 73]. Moreover, previous reports indicated that the serum ALP level is an independent marker of NAFLD development and progression. Pantsari et al. showed that a subset of NAFLD patients (elderly females) exhibit isolated elevation in ALP rather than aminotransferases [74]. Kocabay et al. reported that serum levels of ALP, but not gamma-glutamyl transferase, are increased in NAFLD patients with early fibrosis stage (stage 1 to 2) [75]. ALP is richly expressed in the canalicular membrane side of hepatocytes, and previous studies suggested that ALP relates the transport of bile acid, which plays a major role in cholesterol metabolism and excretion [76]. However, details regarding the physiologic functions of ALP are unclear. miR-379 may be related to NAFLD development and progression by directly or indirectly modulating ALP expression.

Our present study also showed that the serum miR-379 level is positively correlated with serum cholesterol in early stage NAFLD. The contribution of hypercholesterolemia to the development of NAFLD has not been fully elucidated; however, previous studies showed that hepatic cholesterol synthesis and circulating total cholesterol and LDL are increased in NAFLD [77]. Disruption of hepatic cholesterol homeostasis and free cholesterol (FC) accumulation in liver tissue is related to the pathogenesis of NAFLD [78, 79]. Some studies have shown that hepatic cholesterol synthesis is up-regulated in NAFL and NASH patients due to increased activity of a major regulator of cholesterol synthesis, sterol regulatory element–binding protein 2 and its downstream effector HMG CoA-reductase, which catalyzes a rate-limiting step in cholesterol synthesis [8082]. Interestingly, Min et al. also reported that up-regulation of cholesterol synthesis was not observed in control obese subjects [80].

Regarding other cholesterol-related metabolic functions in the liver of NAFLD patients, cholesterol de-esterification is increased, and cholesterol catabolism to bile acid and cholesterol efflux via the bile duct are attenuated [80]. These NAFLD-specific changes in cholesterol metabolism are believed to increase FC levels in liver tissues. FC accumulation in hepatocytes induces mitochondrial dysfunction, which results in increased production of ROS and leads to the unfolded protein response in the endoplasmic reticulum, leading to localized stress and apoptosis [79]. Mari et al. also reported that FC loading (but not that of fatty acids or triglycerides) into hepatocyte mitochondria membranes sensitizes the hepatocyte to pro-inflammatory cytokines (e.g., tumor necrosis factor–alpha and Fas) in mouse models, resulting in steatohepatitis [83]. Moreover, FC accumulation in non-parenchymal cells in liver tissues such as Kupffer cells and stellate cells promotes activation of these cells [84, 85]. The activated Kupffer cells secrete pro-inflammatory cytokines such as interleukin-1β and tumor necrosis factor–alpha, and activated stellate cells differentiate into myofibroblasts, which exhibit a high ability to produce extracellular matrix and fibrogenic cytokines, such as transforming growth factor–β [84, 85]. It has been hypothesized that miR-379 promotes the development and progression of NAFLD as a result of continuous over-nutrition—manifested primarily as obesity—by increasing the lipotoxicity of cholesterol. Cirrhosis and hepatocellular carcinoma are the most common liver-related causes of morbidity associated with NAFLD [86]. However, cardiovascular disease is the most common cause of death in NAFLD patients without cirrhosis [15]. Therefore, some reviewers have recommended giving priority to the prevention of cardiovascular or renal diseases over liver-specific treatments in patients with non-aggressive NAFLD [87].

miR-379 has also been associated with the risk of cardiovascular disease in early stage NAFLD via up-regulation of the serum cholesterol level, which plays an important role in atherosclerosis development. In the present study, however, no significant correlation between serum miR-379 and cholesterol levels was observed in control subjects and NAFLD patients with advanced fibrosis (Brunt stage 2 to 4). This suggests that such a correlation is pertinent only under limited conditions, such as early stage NAFLD–specific pathophysiologic and nutritional states. The serum miR-379 level in controls was significantly lower than that in patients with early stage NAFLD. Normal levels of miR-379 may be insufficient to affect cholesterol metabolism. With respect to advanced-stage NAFLD, it is known that serum cholesterol levels decline with progression of liver fibrosis, independent of the etiology of chronic liver disease [88]. The effect of miR-379 on cholesterol metabolism may be attenuated by decreased hepatic parenchymal function.

The present study also demonstrated that the use of statins to treat hypercholesterolemia in NAFLD patients weakens the relationship between serum miR-379 and cholesterol levels. Statins target hepatocytes and inhibit HMG-CoA reductase, which catalyzes the rate-limiting step in the cholesterol biosynthesis pathway, known as the mevalonate pathway [89]. HMG-CoA reductase converts HMG-CoA into mevalonic acid, a cholesterol precursor. Stains have a higher binding affinity for HMG-CoA reductase than HMG-CoA and thus block access to the active site by the substrate [89]. Previous studies indicated that statins improve hepatic steatosis and reduce hepatic inflammation and fibrosis in NAFLD patients [90, 91]. Moreover, long-term observations of NAFLD patients indicated that continuous statin treatment reduces rates of liver-related death and liver transplantation [92]. Statins may attenuate the effect of miR-379 on cholesterol biosynthesis, resulting in reduced cholesterol lipotoxicity in NAFLD.

GO term annotation analyses showed enrichment of cellular biosynthesis and metabolism–related genes among predicted miR-379 targets. Aberrations in biosynthesis and metabolism play important roles in metabolic disorders such as NAFLD. miR-379 appears to affect the development and progression of NAFLD by interfering with these target genes.

KEGG pathway mapping of prospective miR-379 target genes extracted biological functions such as nutrition and energy regulation, the down-regulation of which leads to the development of NAFLD. Searches of PubMed combining keywords with the selected putative target genes identified in the KEGG pathway analysis and NAFLD identified a number of metabolism-, inflammation-, and fibrosis-related genes. Among the selected putative target genes, IGF1 and IGF1R were identified as targets of miR-379 interference in previous studies [93, 94]. IGF-1 is an insulin-like anabolic hormone primarily secreted by hepatocytes, and circulating IGF-1 levels reflect hepatic IGF-1 expression [95]. Previous studies reported that adults with growth hormone deficiency in which hepatic IGF-1 production is impaired exhibit an increased prevalence of NASH; IGF-1 substitution ameliorated NAFLD in a mouse model [96, 97]. In NAFLD patients without growth hormone deficiency, serum IGF-1 levels are also significantly reduced [95, 98]. The mechanism by which IGF-1 and its signaling pathways protect against NAFLD have been found to involve a variety of biological functions, such as improving insulin sensitivity, decreasing ROS production, and inducing senescence of hepatic stellate cells [99101]. With respect to lipid metabolism, it has been reported that IGF-1 accelerates lipid oxidation and lipolysis [99]. Moreover, several previous studies revealed that serum IGF-1 is inversely correlated with serum levels of total cholesterol and LDL-C [102]. IGF1 appears to be one of the most significant miR-379 target genes with regard to promoting the development and progression of NAFLD via the enhancement of cholesterol lipotoxicity. Among other keyword-selected putative target genes, B-cell lymphoma 2 (BCL2), catalase (CAT), and cAMP responsive element binding protein 1 (CREB1) are reportedly down-regulated in the liver in NAFLD [36, 103, 104]. BCL2 and CAT are major anti-apoptosis genes that function by protecting against mitochondrial outer membrane permeabilization and detoxifying ROS, respectively [36, 103]. Down-regulation of BCL2 and CAT expression in liver tissue drives hepatocyte apoptosis, which is an important pathologic event in the development and progression of NAFLD. CREB1 is a transcription factor that regulates energy balance by suppressing hepatic fatty acid generation and accumulation via downregulation of hepatic-specific peroxisome proliferator activated receptor–𝛾 and fatty acid transporter CD36 expression [104]. miR-379 may affect the development and progression of NAFLD by interfering with the expression of these target genes, which is reportedly down-regulated in NAFLD.

A relationship with NAFLD has also been reported for other miR-379 target genes. For example, 5’-AMP–activated protein kinase catalytic subunit alpha 2 (PRKAA2) is the catalytic subunit alpha 2 of AMPK, a key sensor of energy status in mammalian cells. In the liver, AMPK phosphorylates and inactivates both acetyl-coenzyme A carboxylase and HMG-CoA reductase [105]. Acetyl-coenzyme A carboxylase regulates the biosynthesis of malonyl-CoA, which is the initial committed intermediate in fatty acid biosynthesis. Malonyl-CoA can inhibit carnitine palmitoyl transferase 1, which controls mitochondrial fatty acid oxidation [106]. Therefore, AMPK downregulation increases fatty acid and cholesterol biosynthesis and inhibits fatty acid oxidation, resulting in hepatic lipid accumulation. Although AMPK appears to be related to NAFLD development, details regarding levels of AMPK in hepatocytes are controversial [107].

Previous studies reported the relationship between miR-379 and various diseases. The majority of these studies suggest that miR-379 plays tumor suppressive role in many types of carcinomas, including nasopharyngeal carcinoma, cervical cancer, lung cancer, gastric cancer, hepatocellular carcinoma, bladder cancer, and osteosarcoma [108113]. With regard to metabolic disorders as described above, de Guia et al. revealed a relationship between miR-379 and lipid homeostasis dysregulation [65]. Additionally, patients with a congenital disease known as maternal uniparental disomy for chromosome 14, which causes overexpression of miR-379 of the Dlk1-Dio3 mat miRNA cluster, exhibit characteristic weight gain in early childhood that results in truncal obesity [114].

Our study had some limitations associated with sample size and study design. We did not perform spike in control measurement in NAFLD patients. We cannot assess equalities in RNA extraction efficacies and RT-PCR accurately measurement between NAFLD serum samples especially the drop out case. We used software programs to predict target genes of the candidate miRNAs. Although this method is commonly used, it carries a risk of missing some real targets because the software is designed to assess the relative strength of partial sequence complementarity between mRNA and miRNA. Ontology selection was used to select putative targets that might be relevant to cellular functions. However, ontology selection can only identify proteins for which the function has been identified. Notably, our understanding of the detailed mechanisms that promote the development and progression of NAFLD to NASH is still developing, but new insights are being obtained regularly.

Moreover, we did not confirm whether miR-379 actually interfered with any of the predicted target genes in vivo (e. g. expression measurement in serum or liver tissue) or in vitro, such as direct binding experiments or miR-379 ectopic overexpression by gene transfection. Complex intracellular regulatory networks influence the tissue-specific function of miRNAs [115]. Therefore, further studies are needed to assess whether the predicted targets are actual targets of miR-379 in NAFLD.

Concerning the correlation between serum ALP and miR-379, we could not definitively conclude that the correlation reflects only liver tissue pathologic changes. Bone is another major ALP-secreting organ, and the serum level of the bone isozyme of ALP is elevated in children, adolescents, and elderly people due to bone tissue turnover [116, 117]. Regarding our study participants, all NAFLD patients and control subjects were adults (age ranging from 20 to 76 years), and there was no significant relationship between serum ALP level and age (R2 = 0.0286; p = 0.115). Additionally, no pregnant subjects were included. The number of patients in this study was small, at less than 100. Consequently, the statistical power of the human serum data was relatively limited.

In conclusion, the serum level of miR-379, a member of Dlk1-Dio3 mat miRNA cluster, exhibits high potential as a biomarker for NAFLD. miR-379 also appears to increase cholesterol lipotoxicity, which promotes the development and progression of NAFLD by interfering with the expression of target genes, including those of the IGF-1 signaling pathway. To confidently identify more associations between highly complex and interactive miRNAs with NAFLD, future longitudinal studies with greater sample sizes will be necessary.

Supporting information

S1 Fig. Linear regression analysis of relationships between serum miR-379 and clinical features of NAFLD patients.

Normalized relative to serum miR-16; miR-379 values represent fold-difference relative to the normal control.

(TIF)

S2 Fig. Linear regression analysis of the relationships between serum miR-379 and clinical features of early stage NAFLD patients (Brunt fibrosis stage 0 to 1).

Normalized relative to serum miR-16; miR-379 values represent fold-difference relative to the normal control.

(TIF)

S3 Fig. Linear regression analysis of the relationships between serum miR-379 and clinical features of advanced-stage NAFLD patients (Brunt fibrosis stage 2 to 4).

Normalized relative to serum miR-16; miR-379 values represent fold-difference relative to the normal control.

(TIF)

Data Availability

All relevant data are within the manuscript and its Supporting Information files.

Funding Statement

Initials of the authors who received award: MK Grant number: 26461005 The full name of funder: A grant-in-aid for scientific research category C from the Japan Society for the Promotion of Science URL of funder: websitehttp://www.jsps.go.jp/english/e-grants/index.html Did the sponsors or funders play any role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript?: NO

References

  • 1.Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73–84. Epub 2015/12/29. 10.1002/hep.28431 . [DOI] [PubMed] [Google Scholar]
  • 2.Willner IR, Waters B, Patil SR, Reuben A, Morelli J, Riely CA. Ninety patients with nonalcoholic steatohepatitis: insulin resistance, familial tendency, and severity of disease. The American journal of gastroenterology. 2001;96(10):2957–61. Epub 2001/11/06. 10.1111/j.1572-0241.2001.04667.x . [DOI] [PubMed] [Google Scholar]
  • 3.Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L, Eslam M, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nature reviews Gastroenterology & hepatology. 2018;15(1):11–20. Epub 2017/09/21. 10.1038/nrgastro.2017.109 . [DOI] [PubMed] [Google Scholar]
  • 4.Tilg H, Moschen AR. Evolution of inflammation in nonalcoholic fatty liver disease: the multiple parallel hits hypothesis. Hepatology. 2010;52(5):1836–46. 10.1002/hep.24001 . [DOI] [PubMed] [Google Scholar]
  • 5.Basaranoglu M, Basaranoglu G, Sabuncu T, Senturk H. Fructose as a key player in the development of fatty liver disease. World journal of gastroenterology: WJG. 2013;19(8):1166–72. 10.3748/wjg.v19.i8.1166 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ouyang X, Cirillo P, Sautin Y, McCall S, Bruchette JL, Diehl AM, et al. Fructose consumption as a risk factor for non-alcoholic fatty liver disease. Journal of hepatology. 2008;48(6):993–9. Epub 2008/04/09. 10.1016/j.jhep.2008.02.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Jegatheesan P, De Bandt JP. Fructose and NAFLD: The Multifaceted Aspects of Fructose Metabolism. Nutrients. 2017;9(3). Epub 2017/03/10. 10.3390/nu9030230 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Leclercq IA, Farrell GC, Field J, Bell DR, Gonzalez FJ, Robertson GR. CYP2E1 and CYP4A as microsomal catalysts of lipid peroxides in murine nonalcoholic steatohepatitis. The Journal of clinical investigation. 2000;105(8):1067–75. 10.1172/JCI8814 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Stojsavljevic S, Gomercic Palcic M, Virovic Jukic L, Smircic Duvnjak L, Duvnjak M. Adipokines and proinflammatory cytokines, the key mediators in the pathogenesis of nonalcoholic fatty liver disease. World journal of gastroenterology: WJG. 2014;20(48):18070–91. 10.3748/wjg.v20.i48.18070 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Miura K, Ohnishi H. Role of gut microbiota and Toll-like receptors in nonalcoholic fatty liver disease. World journal of gastroenterology: WJG. 2014;20(23):7381–91. 10.3748/wjg.v20.i23.7381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Begriche K, Massart J, Robin MA, Bonnet F, Fromenty B. Mitochondrial adaptations and dysfunctions in nonalcoholic fatty liver disease. Hepatology. 2013;58(4):1497–507. 10.1002/hep.26226 . [DOI] [PubMed] [Google Scholar]
  • 12.Wei Y, Wang D, Topczewski F, Pagliassotti MJ. Saturated fatty acids induce endoplasmic reticulum stress and apoptosis independently of ceramide in liver cells. American journal of physiology Endocrinology and metabolism. 2006;291(2):E275–81. 10.1152/ajpendo.00644.2005 . [DOI] [PubMed] [Google Scholar]
  • 13.Hauff P, Gottwald U, Ocker M. Early to Phase II drugs currently under investigation for the treatment of liver fibrosis. Expert Opin Investig Drugs. 2015;24(3):309–27. 10.1517/13543784.2015.997874 . [DOI] [PubMed] [Google Scholar]
  • 14.Sanyal AJ, Brunt EM, Kleiner DE, Kowdley KV, Chalasani N, Lavine JE, et al. Endpoints and clinical trial design for nonalcoholic steatohepatitis. Hepatology. 2011;54(1):344–53. 10.1002/hep.24376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Adams LA, Lymp JF, St Sauver J, Sanderson SO, Lindor KD, Feldstein A, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology. 2005;129(1):113–21. Epub 2005/07/14. 10.1053/j.gastro.2005.04.014 . [DOI] [PubMed] [Google Scholar]
  • 16.Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, Pennacchio LA, et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet. 2008;40(12):1461–5. Epub 2008/09/30. 10.1038/ng.257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Anstee QM, Targher G, Day CP. Progression of NAFLD to diabetes mellitus, cardiovascular disease or cirrhosis. Nature reviews Gastroenterology & hepatology. 2013;10(6):330–44. Epub 2013/03/20. 10.1038/nrgastro.2013.41 . [DOI] [PubMed] [Google Scholar]
  • 18.Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of novel genes coding for small expressed RNAs. Science. 2001;294(5543):853–8. Epub 2001/10/27. 10.1126/science.1064921 . [DOI] [PubMed] [Google Scholar]
  • 19.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97. Epub 2004/01/28. 10.1016/s0092-8674(04)00045-5 . [DOI] [PubMed] [Google Scholar]
  • 20.Gregory RI, Chendrimada TP, Cooch N, Shiekhattar R. Human RISC couples microRNA biogenesis and posttranscriptional gene silencing. Cell. 2005;123(4):631–40. Epub 2005/11/08. 10.1016/j.cell.2005.10.022 . [DOI] [PubMed] [Google Scholar]
  • 21.Szabo G, Bala S. MicroRNAs in liver disease. Nature reviews Gastroenterology & hepatology. 2013;10(9):542–52. Epub 2013/05/22. 10.1038/nrgastro.2013.87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Cheung O, Puri P, Eicken C, Contos MJ, Mirshahi F, Maher JW, et al. Nonalcoholic steatohepatitis is associated with altered hepatic MicroRNA expression. Hepatology. 2008;48(6):1810–20. 10.1002/hep.22569 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Pogribny IP, Starlard-Davenport A, Tryndyak VP, Han T, Ross SA, Rusyn I, et al. Difference in expression of hepatic microRNAs miR-29c, miR-34a, miR-155, and miR-200b is associated with strain-specific susceptibility to dietary nonalcoholic steatohepatitis in mice. Lab Invest. 2010;90(10):1437–46. Epub 2010/06/16. 10.1038/labinvest.2010.113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Alisi A, Da Sacco L, Bruscalupi G, Piemonte F, Panera N, De Vito R, et al. Mirnome analysis reveals novel molecular determinants in the pathogenesis of diet-induced nonalcoholic fatty liver disease. Lab Invest. 2011;91(2):283–93. Epub 2010/10/20. 10.1038/labinvest.2010.166 . [DOI] [PubMed] [Google Scholar]
  • 25.Okamoto K, Koda M, Okamoto T, Onoyama T, Miyoshi K, Kishina M, et al. A Series of microRNA in the Chromosome 14q32.2 Maternally Imprinted Region Related to Progression of Non-Alcoholic Fatty Liver Disease in a Mouse Model. PLoS One. 2016;11(5):e0154676 Epub 2016/05/03. 10.1371/journal.pone.0154676 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Chen X, Ba Y, Ma L, Cai X, Yin Y, Wang K, et al. Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008;18(10):997–1006. Epub 2008/09/04. 10.1038/cr.2008.282 . [DOI] [PubMed] [Google Scholar]
  • 27.Thery C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79. Epub 2002/08/03. 10.1038/nri855 . [DOI] [PubMed] [Google Scholar]
  • 28.Brunt EM, Janney CG, Di Bisceglie AM, Neuschwander-Tetri BA, Bacon BR. Nonalcoholic steatohepatitis: a proposal for grading and staging the histological lesions. The American journal of gastroenterology. 1999;94(9):2467–74. 10.1111/j.1572-0241.1999.01377.x . [DOI] [PubMed] [Google Scholar]
  • 29.Ballestri S, Nascimbeni F, Baldelli E, Marrazzo A, Romagnoli D, Lonardo A. NAFLD as a Sexual Dimorphic Disease: Role of Gender and Reproductive Status in the Development and Progression of Nonalcoholic Fatty Liver Disease and Inherent Cardiovascular Risk. Adv Ther. 2017;34(6):1291–326. Epub 2017/05/21. 10.1007/s12325-017-0556-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hamaguchi M, Kojima T, Takeda N, Nakagawa T, Taniguchi H, Fujii K, et al. The metabolic syndrome as a predictor of nonalcoholic fatty liver disease. Ann Intern Med. 2005;143(10):722–8. Epub 2005/11/17. 10.7326/0003-4819-143-10-200511150-00009 . [DOI] [PubMed] [Google Scholar]
  • 31.Ng EK, Chong WW, Jin H, Lam EK, Shin VY, Yu J, et al. Differential expression of microRNAs in plasma of patients with colorectal cancer: a potential marker for colorectal cancer screening. Gut. 2009;58(10):1375–81. Epub 2009/02/10. 10.1136/gut.2008.167817 . [DOI] [PubMed] [Google Scholar]
  • 32.Asaga S, Kuo C, Nguyen T, Terpenning M, Giuliano AE, Hoon DS. Direct serum assay for microRNA-21 concentrations in early and advanced breast cancer. Clin Chem. 2011;57(1):84–91. Epub 2010/11/03. 10.1373/clinchem.2010.151845 . [DOI] [PubMed] [Google Scholar]
  • 33.Liu W, Wang X. Prediction of functional microRNA targets by integrative modeling of microRNA binding and target expression data. Genome Biol. 2019;20(1):18 Epub 2019/01/24. 10.1186/s13059-019-1629-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009;37(1):1–13. Epub 2008/11/27. 10.1093/nar/gkn923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Panasiuk A, Dzieciol J, Panasiuk B, Prokopowicz D. Expression of p53, Bax and Bcl-2 proteins in hepatocytes in non-alcoholic fatty liver disease. World journal of gastroenterology: WJG. 2006;12(38):6198–202. Epub 2006/10/13. 10.3748/wjg.v12.i38.6198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kumar A, Sharma A, Duseja A, Das A, Dhiman RK, Chawla YK, et al. Patients with Nonalcoholic Fatty Liver Disease (NAFLD) have Higher Oxidative Stress in Comparison to Chronic Viral Hepatitis. J Clin Exp Hepatol. 2013;3(1):12–8. Epub 2013/03/01. 10.1016/j.jceh.2012.10.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gentric G, Maillet V, Paradis V, Couton D, L'Hermitte A, Panasyuk G, et al. Oxidative stress promotes pathologic polyploidization in nonalcoholic fatty liver disease. The Journal of clinical investigation. 2015;125(3):981–92. Epub 2015/01/27. 10.1172/JCI73957 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Oh KJ, Han HS, Kim MJ, Koo SH. CREB and FoxO1: two transcription factors for the regulation of hepatic gluconeogenesis. BMB Rep. 2013;46(12):567–74. Epub 2013/11/19. 10.5483/BMBRep.2013.46.12.248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Colak Y, Senates E, Coskunpinar E, Oltulu YM, Zemheri E, Ozturk O, et al. Concentrations of connective tissue growth factor in patients with nonalcoholic fatty liver disease: association with liver fibrosis. Dis Markers. 2012;33(2):77–83. Epub 2012/08/01. 10.3233/DMA-2012-0907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wang C, Hu L, Zhao L, Yang P, Moorhead JF, Varghese Z, et al. Inflammatory stress increases hepatic CD36 translational efficiency via activation of the mTOR signalling pathway. PloS one. 2014;9(7):e103071 Epub 2014/07/23. 10.1371/journal.pone.0103071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Pan X, Zhang Y, Kim HG, Liangpunsakul S, Dong XC. FOXO transcription factors protect against the diet-induced fatty liver disease. Sci Rep. 2017;7:44597 Epub 2017/03/17. 10.1038/srep44597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kolodziejczyk AA, Zheng D, Shibolet O, Elinav E. The role of the microbiome in NAFLD and NASH. EMBO Mol Med. 2019;11(2). Epub 2018/12/29. 10.15252/emmm.201809302 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kosone T, Takagi H, Horiguchi N, Ariyama Y, Otsuka T, Sohara N, et al. HGF ameliorates a high-fat diet-induced fatty liver. Am J Physiol Gastrointest Liver Physiol. 2007;293(1):G204–10. Epub 2007/03/31. 10.1152/ajpgi.00021.2007 . [DOI] [PubMed] [Google Scholar]
  • 44.Wu H, Zhang T, Pan F, Steer CJ, Li Z, Chen X, et al. MicroRNA-206 prevents hepatosteatosis and hyperglycemia by facilitating insulin signaling and impairing lipogenesis. Journal of hepatology. 2017;66(4):816–24. Epub 2016/12/28. 10.1016/j.jhep.2016.12.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Adamek A, Kasprzak A. Insulin-Like Growth Factor (IGF) System in Liver Diseases. Int J Mol Sci. 2018;19(5). Epub 2018/04/28. 10.3390/ijms19051308 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Go GW, Srivastava R, Hernandez-Ono A, Gang G, Smith SB, Booth CJ, et al. The combined hyperlipidemia caused by impaired Wnt-LRP6 signaling is reversed by Wnt3a rescue. Cell Metab. 2014;19(2):209–20. Epub 2014/02/11. 10.1016/j.cmet.2013.11.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Cintra DE, Pauli JR, Araujo EP, Moraes JC, de Souza CT, Milanski M, et al. Interleukin-10 is a protective factor against diet-induced insulin resistance in liver. Journal of hepatology. 2008;48(4):628–37. Epub 2008/02/13. 10.1016/j.jhep.2007.12.017 . [DOI] [PubMed] [Google Scholar]
  • 48.Khamphaya T, Chukijrungroat N, Saengsirisuwan V, Mitchell-Richards KA, Robert ME, Mennone A, et al. Nonalcoholic fatty liver disease impairs expression of the type II inositol 1,4,5-trisphosphate receptor. Hepatology. 2017. Epub 2017/10/13. 10.1002/hep.29588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hardy T, Zeybel M, Day CP, Dipper C, Masson S, McPherson S, et al. Plasma DNA methylation: a potential biomarker for stratification of liver fibrosis in non-alcoholic fatty liver disease. Gut. 2017;66(7):1321–8. Epub 2016/03/24. 10.1136/gutjnl-2016-311526 PubMed Central PMCID: PMC5031527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Abderrahmani A, Yengo L, Caiazzo R, Canouil M, Cauchi S, Raverdy V, et al. Increased Hepatic PDGF-AA Signaling Mediates Liver Insulin Resistance in Obesity-Associated Type 2 Diabetes. Diabetes. 2018;67(7):1310–21. Epub 2018/05/08. 10.2337/db17-1539 . [DOI] [PubMed] [Google Scholar]
  • 51.Kung CP, Leu JI, Basu S, Khaku S, Anokye-Danso F, Liu Q, et al. The P72R Polymorphism of p53 Predisposes to Obesity and Metabolic Dysfunction. Cell Rep. 2016;14(10):2413–25. Epub 2016/03/08. 10.1016/j.celrep.2016.02.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Garcia D, Hellberg K, Chaix A, Wallace M, Herzig S, Badur MG, et al. Genetic Liver-Specific AMPK Activation Protects against Diet-Induced Obesity and NAFLD. Cell Rep. 2019;26(1):192–208 e6. Epub 2019/01/04. 10.1016/j.celrep.2018.12.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Matsuda S, Kobayashi M, Kitagishi Y. Roles for PI3K/AKT/PTEN Pathway in Cell Signaling of Nonalcoholic Fatty Liver Disease. ISRN Endocrinol. 2013;2013:472432 Epub 2013/02/23. 10.1155/2013/472432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sydor S, Sowa JP, Megger DA, Schlattjan M, Jafoui S, Wingerter L, et al. Acid sphingomyelinase deficiency in Western diet-fed mice protects against adipocyte hypertrophy and diet-induced liver steatosis. Mol Metab. 2017;6(5):416–27. Epub 2017/05/04. 10.1016/j.molmet.2017.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Qin G, Wang GZ, Guo DD, Bai RX, Wang M, Du SY. Deletion of Smad4 reduces hepatic inflammation and fibrogenesis during nonalcoholic steatohepatitis progression. J Dig Dis. 2018;19(5):301–13. Epub 2018/04/27. 10.1111/1751-2980.12599 . [DOI] [PubMed] [Google Scholar]
  • 56.Wang H, Shao Y, Yuan F, Feng H, Li N, Zhang H, et al. Fish Oil Feeding Modulates the Expression of Hepatic MicroRNAs in a Western-Style Diet-Induced Nonalcoholic Fatty Liver Disease Rat Model. Biomed Res Int. 2017;2017:2503847 Epub 2017/07/12. 10.1155/2017/2503847 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Musso G, Gambino R, Pacini G, De Michieli F, Cassader M. Prolonged saturated fat-induced, glucose-dependent insulinotropic polypeptide elevation is associated with adipokine imbalance and liver injury in nonalcoholic steatohepatitis: dysregulated enteroadipocyte axis as a novel feature of fatty liver. Am J Clin Nutr. 2009;89(2):558–67. Epub 2009/01/15. 10.3945/ajcn.2008.26720 . [DOI] [PubMed] [Google Scholar]
  • 58.Matsubara T, Tanaka N, Sato M, Kang DW, Krausz KW, Flanders KC, et al. TGF-beta-SMAD3 signaling mediates hepatic bile acid and phospholipid metabolism following lithocholic acid-induced liver injury. J Lipid Res. 2012;53(12):2698–707. Epub 2012/10/05. 10.1194/jlr.M031773 PubMed Central PMCID: PMC3494264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Li Y, Turpin CP, Wang S. Role of thrombospondin 1 in liver diseases. Hepatol Res. 2017;47(2):186–93. Epub 2016/08/06. 10.1111/hepr.12787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Chen P, Luo Q, Huang C, Gao Q, Li L, Chen J, et al. Pathogenesis of non-alcoholic fatty liver disease mediated by YAP. Hepatol Int. 2018;12(1):26–36. Epub 2018/01/14. 10.1007/s12072-017-9841-y . [DOI] [PubMed] [Google Scholar]
  • 61.da Rocha ST, Edwards CA, Ito M, Ogata T, Ferguson-Smith AC. Genomic imprinting at the mammalian Dlk1-Dio3 domain. Trends Genet. 2008;24(6):306–16. Epub 2008/05/13. 10.1016/j.tig.2008.03.011 . [DOI] [PubMed] [Google Scholar]
  • 62.Takada S, Paulsen M, Tevendale M, Tsai CE, Kelsey G, Cattanach BM, et al. Epigenetic analysis of the Dlk1-Gtl2 imprinted domain on mouse chromosome 12: implications for imprinting control from comparison with Igf2-H19. Hum Mol Genet. 2002;11(1):77–86. Epub 2002/01/05. 10.1093/hmg/11.1.77 . [DOI] [PubMed] [Google Scholar]
  • 63.Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA, et al. Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell. 2006;9(6):435–43. Epub 2006/06/13. 10.1016/j.ccr.2006.04.020 . [DOI] [PubMed] [Google Scholar]
  • 64.Dai R, Lu R, Ahmed SA. The Upregulation of Genomic Imprinted DLK1-Dio3 miRNAs in Murine Lupus Is Associated with Global DNA Hypomethylation. PloS one. 2016;11(4):e0153509 Epub 2016/04/14. 10.1371/journal.pone.0153509 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.de Guia RM, Rose AJ, Sommerfeld A, Seibert O, Strzoda D, Zota A, et al. microRNA-379 couples glucocorticoid hormones to dysfunctional lipid homeostasis. EMBO J. 2015;34(3):344–60. Epub 2014/12/17. 10.15252/embj.201490464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Zhang J, Li S, Li L, Li M, Guo C, Yao J, et al. Exosome and exosomal microRNA: trafficking, sorting, and function. Genomics Proteomics Bioinformatics. 2015;13(1):17–24. Epub 2015/03/01. 10.1016/j.gpb.2015.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chartoumpekis DV, Zaravinos A, Ziros PG, Iskrenova RP, Psyrogiannis AI, Kyriazopoulou VE, et al. Differential expression of microRNAs in adipose tissue after long-term high-fat diet-induced obesity in mice. PloS one. 2012;7(4):e34872 Epub 2012/04/13. 10.1371/journal.pone.0034872 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Pearce SG, Thosani NC, Pan JJ. Noninvasive biomarkers for the diagnosis of steatohepatitis and advanced fibrosis in NAFLD. Biomark Res. 2013;1(1):7 Epub 2013/11/21. 10.1186/2050-7771-1-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Angulo P, Hui JM, Marchesini G, Bugianesi E, George J, Farrell GC, et al. The NAFLD fibrosis score: a noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology. 2007;45(4):846–54. Epub 2007/03/30. 10.1002/hep.21496 . [DOI] [PubMed] [Google Scholar]
  • 70.Yang M, Xu D, Liu Y, Guo X, Li W, Guo C, et al. Combined Serum Biomarkers in Non-Invasive Diagnosis of Non-Alcoholic Steatohepatitis. PloS one. 2015;10(6):e0131664 Epub 2015/06/30. 10.1371/journal.pone.0131664 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kaplan MM. Alkaline phosphatase. Gastroenterology. 1972;62(3):452–68. Epub 1972/03/01. . [PubMed] [Google Scholar]
  • 72.Poupon R. Liver alkaline phosphatase: a missing link between choleresis and biliary inflammation. Hepatology. 2015;61(6):2080–90. Epub 2015/01/22. 10.1002/hep.27715 . [DOI] [PubMed] [Google Scholar]
  • 73.Tomizawa M, Kawanabe Y, Shinozaki F, Sato S, Motoyoshi Y, Sugiyama T, et al. Triglyceride is strongly associated with nonalcoholic fatty liver disease among markers of hyperlipidemia and diabetes. Biomed Rep. 2014;2(5):633–6. Epub 2014/07/24. 10.3892/br.2014.309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Pantsari MW, Harrison SA. Nonalcoholic fatty liver disease presenting with an isolated elevated alkaline phosphatase. Journal of clinical gastroenterology. 2006;40(7):633–5. Epub 2006/08/19. 10.1097/00004836-200608000-00015 . [DOI] [PubMed] [Google Scholar]
  • 75.Kocabay G, Telci A, Tutuncu Y, Tiryaki B, Ozel S, Cevikbas U, et al. Alkaline phosphatase: can it be considered as an indicator of liver fibrosis in non-alcoholic steatohepatitis with type 2 diabetes? Bratisl Lek Listy. 2011;112(11):626–9. Epub 2011/12/21. . [PubMed] [Google Scholar]
  • 76.Hatoff DE, Hardison WG. Bile acid-dependent secretion of alkaline phosphatase in rat bile. Hepatology. 1982;2(4):433–9. Epub 1982/07/01. 10.1002/hep.1840020407 . [DOI] [PubMed] [Google Scholar]
  • 77.Barshop NJ, Sirlin CB, Schwimmer JB, Lavine JE. Review article: epidemiology, pathogenesis and potential treatments of paediatric non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2008;28(1):13–24. Epub 2008/04/10. 10.1111/j.1365-2036.2008.03703.x . [DOI] [PubMed] [Google Scholar]
  • 78.Simonen P, Kotronen A, Hallikainen M, Sevastianova K, Makkonen J, Hakkarainen A, et al. Cholesterol synthesis is increased and absorption decreased in non-alcoholic fatty liver disease independent of obesity. Journal of hepatology. 2011;54(1):153–9. Epub 2010/10/16. 10.1016/j.jhep.2010.05.037 . [DOI] [PubMed] [Google Scholar]
  • 79.Gan LT, Van Rooyen DM, Koina ME, McCuskey RS, Teoh NC, Farrell GC. Hepatocyte free cholesterol lipotoxicity results from JNK1-mediated mitochondrial injury and is HMGB1 and TLR4-dependent. Journal of hepatology. 2014;61(6):1376–84. Epub 2014/07/30. 10.1016/j.jhep.2014.07.024 . [DOI] [PubMed] [Google Scholar]
  • 80.Min HK, Kapoor A, Fuchs M, Mirshahi F, Zhou H, Maher J, et al. Increased hepatic synthesis and dysregulation of cholesterol metabolism is associated with the severity of nonalcoholic fatty liver disease. Cell Metab. 2012;15(5):665–74. Epub 2012/05/09. 10.1016/j.cmet.2012.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Beg ZH, Stonik JA, Brewer HB Jr., Phosphorylation of hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase and modulation of its enzymic activity by calcium-activated and phospholipid-dependent protein kinase. J Biol Chem. 1985;260(3):1682–7. Epub 1985/02/10. . [PubMed] [Google Scholar]
  • 82.Caballero F, Fernandez A, De Lacy AM, Fernandez-Checa JC, Caballeria J, Garcia-Ruiz C. Enhanced free cholesterol, SREBP-2 and StAR expression in human NASH. Journal of hepatology. 2009;50(4):789–96. Epub 2009/02/24. 10.1016/j.jhep.2008.12.016 . [DOI] [PubMed] [Google Scholar]
  • 83.Mari M, Caballero F, Colell A, Morales A, Caballeria J, Fernandez A, et al. Mitochondrial free cholesterol loading sensitizes to TNF- and Fas-mediated steatohepatitis. Cell Metab. 2006;4(3):185–98. Epub 2006/09/05. 10.1016/j.cmet.2006.07.006 . [DOI] [PubMed] [Google Scholar]
  • 84.Leroux A, Ferrere G, Godie V, Cailleux F, Renoud ML, Gaudin F, et al. Toxic lipids stored by Kupffer cells correlates with their pro-inflammatory phenotype at an early stage of steatohepatitis. Journal of hepatology. 2012;57(1):141–9. Epub 2012/03/20. 10.1016/j.jhep.2012.02.028 . [DOI] [PubMed] [Google Scholar]
  • 85.Teratani T, Tomita K, Suzuki T, Oshikawa T, Yokoyama H, Shimamura K, et al. A high-cholesterol diet exacerbates liver fibrosis in mice via accumulation of free cholesterol in hepatic stellate cells. Gastroenterology. 2012;142(1):152–64 e10. Epub 2011/10/15. 10.1053/j.gastro.2011.09.049 . [DOI] [PubMed] [Google Scholar]
  • 86.Ekstedt M, Hagstrom H, Nasr P, Fredrikson M, Stal P, Kechagias S, et al. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology. 2015;61(5):1547–54. Epub 2014/08/16. 10.1002/hep.27368 . [DOI] [PubMed] [Google Scholar]
  • 87.Sumida Y, Yoneda M. Current and future pharmacological therapies for NAFLD/NASH. Journal of gastroenterology. 2018;53(3):362–76. Epub 2017/12/17. 10.1007/s00535-017-1415-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Cicognani C, Malavolti M, Morselli-Labate AM, Zamboni L, Sama C, Barbara L. Serum lipid and lipoprotein patterns in patients with liver cirrhosis and chronic active hepatitis. Arch Intern Med. 1997;157(7):792–6. Epub 1997/04/14. . [PubMed] [Google Scholar]
  • 89.Stancu C, Sima A. Statins: mechanism of action and effects. J Cell Mol Med. 2001;5(4):378–87. Epub 2002/06/18. 10.1111/j.1582-4934.2001.tb00172.x . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Eslami L, Merat S, Malekzadeh R, Nasseri-Moghaddam S, Aramin H. Statins for non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Cochrane Database Syst Rev. 2013;(12):CD008623 Epub 2014/01/01. 10.1002/14651858.CD008623.pub2 . [DOI] [PubMed] [Google Scholar]
  • 91.Dongiovanni P, Petta S, Mannisto V, Mancina RM, Pipitone R, Karja V, et al. Statin use and non-alcoholic steatohepatitis in at risk individuals. Journal of hepatology. 2015;63(3):705–12. Epub 2015/05/20. 10.1016/j.jhep.2015.05.006 . [DOI] [PubMed] [Google Scholar]
  • 92.Angulo P, Kleiner DE, Dam-Larsen S, Adams LA, Bjornsson ES, Charatcharoenwitthaya P, et al. Liver Fibrosis, but No Other Histologic Features, Is Associated With Long-term Outcomes of Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology. 2015;149(2):389–97 e10. Epub 2015/05/04. 10.1053/j.gastro.2015.04.043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Li K, Wang Y, Zhang A, Liu B, Jia L. miR-379 Inhibits Cell Proliferation, Invasion, and Migration of Vascular Smooth Muscle Cells by Targeting Insulin-Like Factor-1. Yonsei Med J. 2017;58(1):234–40. Epub 2016/11/23. 10.3349/ymj.2017.58.1.234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Huang DJ, Huang JZ, Yang J, Li YH, Luo YC, He HY, et al. Bioinformatic identification of IGF1 as a hub gene in hepatocellular carcinoma (HCC) and in-vitro analysis of the chemosensitizing effect of miR-379 via suppressing the IGF1/IGF1R signaling pathway. Eur Rev Med Pharmacol Sci. 2016;20(24):5098–106. Epub 2017/01/05. . [PubMed] [Google Scholar]
  • 95.Garcia-Galiano D, Sanchez-Garrido MA, Espejo I, Montero JL, Costan G, Marchal T, et al. IL-6 and IGF-1 are independent prognostic factors of liver steatosis and non-alcoholic steatohepatitis in morbidly obese patients. Obes Surg. 2007;17(4):493–503. Epub 2007/07/05. 10.1007/s11695-007-9087-1 . [DOI] [PubMed] [Google Scholar]
  • 96.Molitch ME, Clemmons DR, Malozowski S, Merriam GR, Vance ML, Endocrine S. Evaluation and treatment of adult growth hormone deficiency: an Endocrine Society clinical practice guideline. The Journal of clinical endocrinology and metabolism. 2011;96(6):1587–609. Epub 2011/05/24. 10.1210/jc.2011-0179 . [DOI] [PubMed] [Google Scholar]
  • 97.Nishizawa H, Takahashi M, Fukuoka H, Iguchi G, Kitazawa R, Takahashi Y. GH-independent IGF-I action is essential to prevent the development of nonalcoholic steatohepatitis in a GH-deficient rat model. Biochem Biophys Res Commun. 2012;423(2):295–300. Epub 2012/06/05. 10.1016/j.bbrc.2012.05.115 . [DOI] [PubMed] [Google Scholar]
  • 98.Arturi F, Succurro E, Procopio C, Pedace E, Mannino GC, Lugara M, et al. Nonalcoholic fatty liver disease is associated with low circulating levels of insulin-like growth factor-I. The Journal of clinical endocrinology and metabolism. 2011;96(10):E1640–4. Epub 2011/08/06. 10.1210/jc.2011-1227 . [DOI] [PubMed] [Google Scholar]
  • 99.Ichikawa T, Nakao K, Hamasaki K, Furukawa R, Tsuruta S, Ueda Y, et al. Role of growth hormone, insulin-like growth factor 1 and insulin-like growth factor-binding protein 3 in development of non-alcoholic fatty liver disease. Hepatol Int. 2007;1(2):287–94. Epub 2007/06/01. 10.1007/s12072-007-9007-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Csiszar A, Labinskyy N, Perez V, Recchia FA, Podlutsky A, Mukhopadhyay P, et al. Endothelial function and vascular oxidative stress in long-lived GH/IGF-deficient Ames dwarf mice. Am J Physiol Heart Circ Physiol. 2008;295(5):H1882–94. Epub 2008/09/02. 10.1152/ajpheart.412.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Nishizawa H, Iguchi G, Fukuoka H, Takahashi M, Suda K, Bando H, et al. IGF-I induces senescence of hepatic stellate cells and limits fibrosis in a p53-dependent manner. Sci Rep. 2016;6:34605 Epub 2016/10/11. 10.1038/srep34605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lam CS, Chen MH, Lacey SM, Yang Q, Sullivan LM, Xanthakis V, et al. Circulating insulin-like growth factor-1 and its binding protein-3: metabolic and genetic correlates in the community. Arterioscler Thromb Vasc Biol. 2010;30(7):1479–84. Epub 2010/04/10. 10.1161/ATVBAHA.110.203943 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zaki SM, Abdel Fattah S, Hassan DS. The differential effects of high-fat and high fructose diets on the liver of male albino rat and the proposed underlying mechanisms. Folia Morphol (Warsz). 2018. Epub 2018/07/17. 10.5603/FM.a2018.0063 . [DOI] [PubMed] [Google Scholar]
  • 104.Liu Y, Cheng F, Luo Y, Zhan Z, Hu P, Ren H, et al. PEGylated Curcumin Derivative Attenuates Hepatic Steatosis via CREB/PPAR-gamma/CD36 Pathway. Biomed Res Int. 2017;2017:8234507 Epub 2017/08/05. 10.1155/2017/8234507 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Hardie DG. Regulation of fatty acid and cholesterol metabolism by the AMP-activated protein kinase. Biochim Biophys Acta. 1992;1123(3):231–8. Epub 1992/02/12. 10.1016/0005-2760(92)90001-c . [DOI] [PubMed] [Google Scholar]
  • 106.Kim KH. Regulation of acetyl-CoA carboxylase. Curr Top Cell Regul. 1983;22:143–76. Epub 1983/01/01. 10.1016/b978-0-12-152822-5.50009-9 . [DOI] [PubMed] [Google Scholar]
  • 107.Song Z, Deaciuc I, Zhou Z, Song M, Chen T, Hill D, et al. Involvement of AMP-activated protein kinase in beneficial effects of betaine on high-sucrose diet-induced hepatic steatosis. Am J Physiol Gastrointest Liver Physiol. 2007;293(4):G894–902. Epub 2007/08/19. 10.1152/ajpgi.00133.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Zhao X, Chu J. MicroRNA-379 suppresses cell proliferation, migration and invasion in nasopharyngeal carcinoma by targeting tumor protein D52. Exp Ther Med. 2018;16(2):1232–40. Epub 2018/08/18. 10.3892/etm.2018.6302 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 109.Shi X, Xiao X, Yuan N, Zhang S, Yuan F, Wang X. MicroRNA-379 Suppresses Cervical Cancer Cell Proliferation and Invasion by Directly Targeting V-crk Avian Sarcoma Virus CT10 Oncogene Homolog-Like (CRKL). Oncol Res. 2018;26(7):987–96. Epub 2018/01/04. 10.3727/096504017X15140534417184 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Xu M, Qin S, Cao F, Ding S, Li M. MicroRNA-379 inhibits metastasis and epithelial-mesenchymal transition via targeting FAK/AKT signaling in gastric cancer. Int J Oncol. 2017;51(3):867–76. Epub 2017/07/18. 10.3892/ijo.2017.4072 . [DOI] [PubMed] [Google Scholar]
  • 111.Wu D, Niu X, Tao J, Li P, Lu Q, Xu A, et al. MicroRNA-379-5p plays a tumor-suppressive role in human bladder cancer growth and metastasis by directly targeting MDM2. Oncol Rep. 2017;37(6):3502–8. Epub 2017/05/13. 10.3892/or.2017.5607 . [DOI] [PubMed] [Google Scholar]
  • 112.Xie X, Li YS, Xiao WF, Deng ZH, He HB, Liu Q, et al. MicroRNA-379 inhibits the proliferation, migration and invasion of human osteosarcoma cells by targetting EIF4G2. Biosci Rep. 2017;37(3). Epub 2017/04/07. 10.1042/BSR20160542 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 113.Chen JS, Li HS, Huang JQ, Dong SH, Huang ZJ, Yi W, et al. MicroRNA-379-5p inhibits tumor invasion and metastasis by targeting FAK/AKT signaling in hepatocellular carcinoma. Cancer Lett. 2016;375(1):73–83. Epub 2016/03/06. 10.1016/j.canlet.2016.02.043 . [DOI] [PubMed] [Google Scholar]
  • 114.Mitter D, Buiting K, von Eggeling F, Kuechler A, Liehr T, Mau-Holzmann UA, et al. Is there a higher incidence of maternal uniparental disomy 14 [upd(14)mat]? Detection of 10 new patients by methylation-specific PCR. Am J Med Genet A. 2006;140(19):2039–49. Epub 2006/08/15. 10.1002/ajmg.a.31414 . [DOI] [PubMed] [Google Scholar]
  • 115.Liang Y, Ridzon D, Wong L, Chen C. Characterization of microRNA expression profiles in normal human tissues. BMC genomics. 2007;8:166 10.1186/1471-2164-8-166 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Turan S, Topcu B, Gokce I, Guran T, Atay Z, Omar A, et al. Serum alkaline phosphatase levels in healthy children and evaluation of alkaline phosphatase z-scores in different types of rickets. J Clin Res Pediatr Endocrinol. 2011;3(1):7–11. Epub 2011/03/31. 10.4274/jcrpe.v3i1.02 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kuo TR, Chen CH. Bone biomarker for the clinical assessment of osteoporosis: recent developments and future perspectives. Biomark Res. 2017;5:18 Epub 2017/05/23. 10.1186/s40364-017-0097-4 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Catherine Mounier

21 Nov 2019

PONE-D-19-17509

Serum miR-379 expression is related to the development and progression of hypercholesterolemia in non-alcoholic fatty liver disease

PLOS ONE

Dear Dr, Okamoto

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

More specifically, you need to confirm the level of expression of miR-379 target genes.

We would appreciate receiving your revised manuscript by the 20th of February 2020. When you are ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter.

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). This letter should be uploaded as separate file and labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. This file should be uploaded as separate file and labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. This file should be uploaded as separate file and labeled 'Manuscript'.

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out.

We look forward to receiving your revised manuscript.

Kind regards,

Catherine Mounier

Academic Editor

PLOS ONE

Journal Requirements:

1. When submitting your revision, we need you to address these additional requirements.

Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

http://www.journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and http://www.journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. In your Methods section, please provide additional information about the participant recruitment method and the demographic details of your participants. Please ensure you have provided sufficient details to replicate the analyses such as: a) the recruitment date range (month and year), b) a description of any inclusion/exclusion criteria that were applied to participant recruitment, c) a statement as to whether your sample can be considered representative of a larger population, d) a description of how participants were recruited, and e) descriptions of where participants were recruited and where the research took place.

In your Methods section, please provide a sample size calculation.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Partly

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: I Don't Know

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: Okamoto and colleagues set out to examine the suitability of circulating serum miR-379 as a potential non-invasive biomarker for non-alcoholic fatty liver disease using real-time PCR platform. The manuscript was well-written and has merit. However, there are some points that need to be clarified.

1. The presence of inhibitors in serum samples may limit the ability to extract RNA or accurately measure miRNAs by real-time PCR. I was wondering whether a spiked-in control was also included in this study to ensure efficiency of RNA extraction?

2. It is not clear how reverse transcription was carried out prior to real-time PCR. Please refer to lines 175-178.

3. It would be interesting to find out whether non-HDL cholesterol (total cholesterol minus HDL cholesterol) is a predictor for non-alcoholic fatty liver disease. Please refer to Table 1, page 11.

4. What was the basis of choosing miR-379 from a series of potential miRNAs (miR-127, miR-136, miR-376c, miR-379, miR-409-3p, miR-411, miR-495) mapped in the 14q32.3 maternally imprinted gene region? Please refer to lines 29-33 and lines 105-111).

Reviewer #2: The manuscript of Okamoto and colleagues highlights the correlation between the expression of miR-379 and NASH development and progression. The study was performed with serum collected from patients affected by early and advanced NASH.

The study was performed in an extremely accurate manner and based its achievement on the correlation between expression of miR-379 and the clinical markers of NASH, especially cholesterol lipotoxicity.

The soundness of the study is supported by previous study published from the same groupd with NASH mouse model.

Some minor points need to be addressed:

The number of patients involved in the study should be corrected between the abstract, the methods and the results section. Please include in the Abstract the number of control samples included in the study. In the Methods section is stated that the patients are 90 and it does not correspond with the abstract. In the Results section, the authors explain that one patient was excluded from the study. This information should be included in the Methods.

In order to better follow the results described in the text, to add capital letters to differentiate the different panels of the figures when necessary. For example, Figure 1 showing three different graphs of different patient groups.

Reviewer #3: The authors report increased expression of miR-379 in patients with NAFLD as compared to 10 control patients. They further show that miR-379 may be used as marker of NAFLD with AUROC of 0.76. Computer based prediction of miR-379 targets was preformed and IGF1, IGF1R and INSR were suggested as its targets. These results are of potential interest as they are reporting on potential biomarker of NAFLD in human subjects. Unfortunately, the authors did not adequately describe their results and the Result section appears much shorter than the Introduction. Also, they did not make an attempt to validate downstream targets of miR-379, which is a major missed opportunity.

Major Remarks

It is unclear how many patients were enrolled in the study. The authors state that “Ninety patients were enrolled in this study. The patients were divided into three groups, as follows: 10 patients with asymptomatic gallbladder stones as disease controls, 9 NAFL patients, and 71 NASH patients. In another analysis, NAFLD patients were divided into early stage (n = 53) and advanced-stage (n = 26) groups. (Lane 135-137).” Was the second analysis done on the same set of pts, or was it done on NASH pts only reported in the first analysis. If it was done on NASH pts why are the number different? First cohort 71 NASH pts, while second analysis included 53+26 = 79 patients. To make this clearer, table one should list the number of pts in each group. Furthermore, it should be clearly stated if this was done on one group of pts with two separate analysis or if two cohorts of pts were used.

I could not readily find figure legends. Some description of figures was found in the Results section but it was short and limited. It did not sufficiently explain the figures, nor did it indicate how many pts were included in each group. Is +/- denoting SE or SD? Figure 2 has only title listed and no description of the figure at all!

It is important to better understand the role of miR-379 in liver physiology. The authors attempted to do this by identifying potential target genes of miR-379 using web-based software. More information needs to be provided about this process before we can make any conclusions about the results that they are showing us.

The authors found 27 predicted gene targets of miR-379 that were associated with NAFLD development or progression. They focused their discussion on insulin-like growth factor-1 (IGF-1) and IGF-1 receptor. It would be imperative to verify expression of IGF1, IGF1R and INSR in these patients and determine if their expression indeed correlates with expression of miR-379.

Minor remarks:

That authors state that NAFLD “disease process begins with the development of insulin resistance resulting from excessive energy intake [5]. (Lanes 66-67). While insulin resistance may be universally found in pts with NAFLD, its role in pathogenesis is still unclear. Competing theories exist, with one suggesting that hepatic insulin resistance initiates development of NAFLD, whereas the other argues that insulin resistance is merely a consequence of increased liver lipid deposition. If the authors choose to stand by their wording, a better reference is needed than #5, which mainly talks about the effects of fructose in pathogenesis of NAFLD.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: Yes: Michael O. Baclig

Reviewer #2: Yes: Pietro Di Fazio

Reviewer #3: Yes: Samir Softic

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files to be viewed.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2020 Feb 27;15(2):e0219412. doi: 10.1371/journal.pone.0219412.r002

Author response to Decision Letter 0


27 Jan 2020

Dear academic editor Catherine Mounier,

We greatly appreciate the opportunity to revise our manuscript entitled “Serum miR-379 expression is related to the development and progression of hypercholesterolemia in non-alcoholic fatty liver disease” (ID: PONE-D-19-17509).

We also thank the reviewers very much for their careful reading our manuscript and insightful comments. We revised our manuscript according to journal requirements and reviewers’ comments.

Our alterations as a result of the academic editor and the reviewer’s comments are as follows.

Sincerely,

Kinya Okamoto

Second Department of Internal Medicine

Tottori University School of Medicine

E-mail: kinyah.okamoto@kje.biglobe.ne.jp

Journal Requirements:

1. When submitting your revision, we need you to address these additional requirements.

Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

http://www.journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and http://www.journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

Reply: We revised our manuscript style in accordance with PLOS ONE style template.

2. In your Methods section, please provide additional information about the participant recruitment method and the demographic details of your participants. Please ensure you have provided sufficient details to replicate the analyses such as: a) the recruitment date range (month and year), b) a description of any inclusion/exclusion criteria that were applied to participant recruitment, c) a statement as to whether your sample can be considered representative of a larger population, d) a description of how participants were recruited, and e) descriptions of where participants were recruited and where the research took place.

Reply: According to advices of the academic editor, we revised materials and methods part in our manuscript.

a) The recruitment date range

Reply: We indicated the date range (from July 2014 to March 2016).

b) A description of any inclusion/exclusion criteria that were applied to participant recruitment

Reply: We revised inclusion/exclusion criteria to enter into detail.

c) A statement as to whether your sample can be considered representative of a larger population

Reply: We described the assessment of representativeness of our NAFLD participants as follows.

(Lines 231-250 in revised manuscript with track change)

We assessed whether our NAFLD sample can be considered representative of a larger NAFLD population. Concerning age of the participants, the mean age of our NAFLD patients are 48.6 ± 15.4. The majority of NAFLD concerning studies indicate that the median age of NAFLD patients are in the range from 40 to 59 (1). Meta-analytic assessment of prevalence and incidence of NAFLD showed that the prevalence of NAFLD increases with age, however, the prevalence rate are similar from age 40’s to 60’s (26.5 % to 28.9 %, respectively) (1). Former large population studies support that the prevalence of NAFLD is higher in men than in women (29). In our present study, male NAFLD patients are also more frequently observed than female patients (54 males and 25 females). Overweight is one of the strong risk factors for NAFLD (1). Previous Japanese NAFLD patients study also showed a clear relationship between BMI and NAFLD development (30). In our study, mean of body mass index of NAFLD patients excess 25 (29.3 ± 6.1), which indicate overweight. Development and resolution of NAFLD were both closely related to metabolic syndrome especially diabetes mellitus (DM) (30). Hamaguchi et al. reported that 25 - 40% of Japanese NAFLD patients have established DM (3). In our study, 24 cases (30%) of NAFLD patients, had complication of DM. In subgroup, 1 of NAFL (11%) and 23 of NASH (33%), and 16 of NAFLD early stage (30%) and 8 of NAFLD advanced stage (31%) had DM. Because of these demographic features, we regarded that our NAFLD participants could be considered as the representative group of larger population of NAFLD.

d) A description of how participants were recruited

Reply: We added detail of participants recruiting method as follows.

(Lines 168-180)

Recruiting and obtaining liver tissue, blood sample and clinical data were performed at Tottori University Hospital and collaborative medical institutes: Hiroshima University Hospital, JA Hiroshima General Hospital, Kawasaki University Hospital, and Shimane University Hospital. from July 2014 to March 2016. All participants were Japanese who live in the western part of Japanese main-island. NAFLD outpatients who underwent continuous clinical follow-up at Tottori University Hospital or collaborative medical institutes were recruited by interview with our study members. In interview, we gave clear exposition of the purpose, procedures, duration and potential adverse events of our study using the printed description which have been obtained the approval of the committee for ethics in medical experiments on human subjects of the medical faculty of Tottori University. Participants were given sufficient time to read the consent and have all questions answered before signing the consent form voluntary.

e) Descriptions of where participants were recruited and where the research took place:

Reply: We described the places of participants recruit and research made.

(Lines 168-172)

Recruiting and obtaining liver tissue, blood sample and clinical data were performed at Tottori University Hospital and collaborative medical institutes: Hiroshima University Hospital, JA Hiroshima General Hospital, Kawasaki University Hospital, and Shimane University Hospital. from July 2014 to March 2016.

(Lines 281-282)

miRNA extract from the serum, Quantitative real-time polymerase chain reaction (RT-PCR) and data analysis were carried out at Tottori University.

In your Methods section, please provide a sample size calculation.

Reply: According to advice of the academic editor, we provide a sample size calculation for our present study in methods paragraph as follows.

(Lines 156-165)

Sample size calculation

The effective sample size has been calculated from the outcomes of our previous clinical study about serum Dlk1-Dio3 mat miRNA expression in NAFLD patients (25). According to the criterion of our previous study, we set up the relevant difference of miR-379 expression level is 2log2. The associated standard deviation was estimated to be 1.6log2 based on the serum miR-379 expression of normal control in our previous study (25). Given a statistical power of 80% and 0.05 level of significance, a sample size of 11 in each group will be sufficient to detect a clinically relevant difference between groups. To allow for dropout of patients (up to 20%) and further subgroup analysis, we aim to recruit 80 participants.

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: Okamoto and colleagues set out to examine the suitability of circulating serum miR-379 as a potential non-invasive biomarker for non-alcoholic fatty liver disease using real-time PCR platform. The manuscript was well-written and has merit. However, there are some points that need to be clarified.

1. The presence of inhibitors in serum samples may limit the ability to extract RNA or accurately measure miRNAs by real-time PCR. I was wondering whether a spiked-in control was also included in this study to ensure efficiency of RNA extraction?

Reply: Thank you very much for your advice. We had only examined preliminary study about RT-PCR measurement in human serum between endogenous miR-16 and non-mammal spike in control miRNA. Serum samples were obtained from normal controls (n = 10). 1.6 x 108 copies of C. elegans (Ce)-miR-39-1 (Qiagen, Venlo, Nederland) and 1 μg bacteriophage MS2 RNA (Roche, Penzberg, Germany) as carrier RNA were added for each 200 μL serum sample. Quantitative RT-PCRs was performed using the same protocol of the present study. Both of miR-16 and Ce-miR-39-1 were stably expressed between samples and their standard deviations of threshold cycles were within ± 1 cycle (34.0 ± 0.8 and 23.9 ± 0.1, respectively). We concluded that our miRNA extraction and RT-PCR works steady and endogenous miR-16 could be applied as internal control. However, We did not perform spike in control measurement in NAFLD patients. As the reviewer kindly pointed, we cannot assess equalities in RNA extraction efficacies and RT-PCR accurately measurement between NAFLD serum samples. We disclosed the preliminary study about miR-16 and spike-in control in the materials and methods part. And we also describe the limitation of our study in the discussion part as follows.

(Lines 300-354 in revised manuscript with track change)

We also examined preliminary study about RT-PCR measurement in human serum between endogenous miR-16 and non-mammal spike in control miRNA. Serum samples were obtained from normal controls (n = 10). 1.6 x 108 copies of C. elegans (Ce)-miR-39-1 (Qiagen) and 1 μg bacteriophage MS2 RNA (Roche, Penzberg, Germany) as carrier RNA were added for each 200 μL serum sample. PT-PCR was performed using the same protocol of the present study. The primer sequence of hsa-miR-16 was UAGCAGCACGUAAAUAUUGGCG. The manufacturer does not disclose the primer sequence of Ce-miR-39-1. Both of miR-16 and Ce-miR-39-1 were stably expressed between samples and their standard deviations of threshold cycles were within ± 1 cycle (34.0 ± 0.8 and 23.9 ± 0.1, respectively). We concluded that endogenous miR-16 could be applied as internal control of RT-PCR in serum miRNA.

(Lines 763-765)

We did not perform spike in control measurement in NAFLD patients. We cannot assess equalities in RNA extraction efficacies and RT-PCR accurately measurement between NAFLD serum samples especially the drop out case.

2. It is not clear how reverse transcription was carried out prior to real-time PCR. Please refer to lines 175-178.

Reply: According to the reviewer’s advice, we added the following sentence to the Materials and Methods section of the manuscript.

(Lines 290-292)

The miScript II reverse transcription kit (Qiagen) was used for reverse transcription of serum miRNA according to the manufacturer’s instructions.

3. It would be interesting to find out whether non-HDL cholesterol (total cholesterol minus HDL cholesterol) is a predictor for non-alcoholic fatty liver disease. Please refer to Table 1, page 11.

Reply: Thank you very much for your specific advice. According to the reviewer’s advice, we examined the relationships between serum miR-379 expression and non-HDL cholesterol (non-HDL-C) in NAFLD patients. Positive correlation was observed between serum miR-379 expression and non-HDL-C levels in patients with early stage NAFLD. We added the result in abstract, main manuscript, Table 1 and Fig 3.

4. What was the basis of choosing miR-379 from a series of potential miRNAs (miR-127, miR-136, miR-376c, miR-379, miR-409-3p, miR-411, miR-495) mapped in the 14q32.3 maternally imprinted gene region? Please refer to lines 29-33 and lines 105-111).

Reply: Our explanation for miRNA selection from the miRNA cluster of 14q32.3 maternally imprinted gene region might have been insufficient; therefore, we revised and added the following text.

(Lines 282-288)

miR-379 was selected from the putative Dlk1-Dio3 mat miRNA cluster because it exhibited the greatest difference in serum expression between NAFL and NASH patients in our preliminary study (25). Comparing to the normal controls, serum miR-379 overexpressed in simple steatosis (we named NAFL to simple steatosis in our previous report) group (3.3 ± 3.1 log2) and down regulated in NASH patients group (-7.4 ± 5.9 log2) in our previous study (25).

Reviewer #2: The manuscript of Okamoto and colleagues highlights the correlation between the expression of miR-379 and NASH development and progression. The study was performed with serum collected from patients affected by early and advanced NASH.

The study was performed in an extremely accurate manner and based its achievement on the correlation between expression of miR-379 and the clinical markers of NASH, especially cholesterol lipotoxicity.

The soundness of the study is supported by previous study published from the same groupd with NASH mouse model.

Some minor points need to be addressed:

The number of patients involved in the study should be corrected between the abstract, the methods and the results section. Please include in the Abstract the number of control samples included in the study. In the Methods section is stated that the patients are 90 and it does not correspond with the abstract. In the Results section, the authors explain that one patient was excluded from the study. This information should be included in the Methods.

Reply: Thank you for your pointing out about our mistake. We corrected patient number; total 80, and indicated the data of excluded patient in abstract and materials and methods part.

In order to better follow the results described in the text, to add capital letters to differentiate the different panels of the figures when necessary. For example, Figure 1 showing three different graphs of different patient groups.

Reply: Thank you very much for your kind advice. We revised layouts of Fig1, 2 and 3 and adding capital letters.

Reviewer #3: The authors report increased expression of miR-379 in patients with NAFLD as compared to 10 control patients. They further show that miR-379 may be used as marker of NAFLD with AUROC of 0.76. Computer based prediction of miR-379 targets was preformed and IGF1, IGF1R and INSR were suggested as its targets. These results are of potential interest as they are reporting on potential biomarker of NAFLD in human subjects. Unfortunately, the authors did not adequately describe their results and the Result section appears much shorter than the Introduction. Also, they did not make an attempt to validate downstream targets of miR-379, which is a major missed opportunity.

Major Remarks

It is unclear how many patients were enrolled in the study. The authors state that “Ninety patients were enrolled in this study. The patients were divided into three groups, as follows: 10 patients with asymptomatic gallbladder stones as disease controls, 9 NAFL patients, and 71 NASH patients. In another analysis, NAFLD patients were divided into early stage (n = 53) and advanced-stage (n = 26) groups. (Lane 135-137).” Was the second analysis done on the same set of pts, or was it done on NASH pts only reported in the first analysis. If it was done on NASH pts why are the number different? First cohort 71 NASH pts, while second analysis included 53+26 = 79 patients. To make this clearer, table one should list the number of pts in each group. Furthermore, it should be clearly stated if this was done on one group of pts with two separate analysis or if two cohorts of pts were used.

Reply: Thank you very much for pointing out our mistake. We corrected patient number and indicated the patient numbers in table 1.

Our 2 NAFLD subgroup studies (NAFL or NASH and NAFLD early stage or advanced stage) were examined same NAFLD patients group (n = 79). We revised the statement of NAFLD subgroup in the materials and methods part.

(Lines 224-226 in revised manuscript with track change)

The NAFLD patients were divided into two subgroups as follows: 9 NAFL patients, and 70 NASH patients. In another analysis, NAFLD patients were also divided into early stage (n = 53) and advanced stage (n = 26) groups.

I could not readily find figure legends. Some description of figures was found in the Results section but it was short and limited. It did not sufficiently explain the figures, nor did it indicate how many pts were included in each group. Is +/- denoting SE or SD? Figure 2 has only title listed and no description of the figure at all!

Reply: Our figure legends might be insufficient to explain the images correctly. We revised figure legends according to the reviewer’s advice.

It is important to better understand the role of miR-379 in liver physiology. The authors attempted to do this by identifying potential target genes of miR-379 using web-based software. More information needs to be provided about this process before we can make any conclusions about the results that they are showing us.

Reply: Thank you very much for your advice. We added the information about our study design in exploring miR-379 physiological functions. We also added the explanations about the results of GO ontology annotation and GO enrichment analysis.

(Lines 357-375)

The physiological roles of miR-379 in liver are still hardly identified. Few former studies revealed the relationship between liver function and miR-379. A comprehensive investigation for miR-379 function should be carried out. We adopted the computational miRNA target genes prediction. Software prediction of miRNA target genes is a popular and reliable method to estimate miRNA physiological functions (33). We used web-based software DIANA microT-CDS 5.0 (http://diana.cslab.ece.ntua.gr/) for miR-379 putative target gene analysis. The threshold for the target prediction score in DIANA microT-CDS was set to 0.7. One miRNA can interference hundreds to thousands genes (20). We selected gene ontology (GO) annotation to select NAFLD related genes. Database for Annotation, Visualization, and Integrated Discovery (DAVID) 6.8 (http://david.abcc.ncifcrf.gov/) was used for GO annotation, and the Kyoto Encyclopedia of Genes and Genomes (KEGG) was used for pathway enrichment analysis.

(Lines 503-504)

Simple gene counting of GO terms showed that cellular process, metabolic process and biological regulation had a large proportion amount to over 70%.

(Lines 511-521)

We also examined GO term enrichment analysis. The analysis can identify GO terms, which are significantly over-represented for DAVID pre-built human genome backgrounds (34). miR-379 predicted target genes were richly represented in 12 GO terms compared to DAVID pre-built human genome backgrounds significantly (Table 2). Among 12 enriched GO, 10 terms related to cellular process regulations.

(Lines 530-535)

Each of the enriched GO terms still had hundreds genes (Table 2). It is one of the common weak points of this method that the output of genes can be large (34). To select putative target gene for NAFLD pathology, we examined extending analysis using backend annotation database. Our GO term analysis showed that miR-379 seemed to relate biological regulations largely (Table 2). Therefore we selected KEGG pathway as the backend database.

The authors found 27 predicted gene targets of miR-379 that were associated with NAFLD development or progression. They focused their discussion on insulin-like growth factor-1 (IGF-1) and IGF-1 receptor. It would be imperative to verify expression of IGF1, IGF1R and INSR in these patients and determine if their expression indeed correlates with expression of miR-379.

Reply: We agree with your opinion. We also recognize it to be the biggest weak point of our present study. However, we do not have enough patient serum and liver tissue to confirm mRNAs and protein expression levels any more.

We discussed about this problem in limitation part of discussion as follows.

(Lines 774-779)

We did not confirm whether miR-379 actually interfered with any of the predicted target genes in vivo (e. g. expression measurement in serum or liver tissue) or in vitro, such as direct binding experiments or miR-379 ectopic overexpression by gene transfection. Complex intracellular regulatory networks influence the tissue-specific function of miRNAs (115). Therefore, further studies are needed to assess whether the predicted targets are actual targets of miR-379 in NAFLD.

Minor remarks:

That authors state that NAFLD “disease process begins with the development of insulin resistance resulting from excessive energy intake [5]. (Lanes 66-67). While insulin resistance may be universally found in pts with NAFLD, its role in pathogenesis is still unclear. Competing theories exist, with one suggesting that hepatic insulin resistance initiates development of NAFLD, whereas the other argues that insulin resistance is merely a consequence of increased liver lipid deposition. If the authors choose to stand by their wording, a better reference is needed than #5, which mainly talks about the effects of fructose in pathogenesis of NAFLD.

Reply: According to the reviewer’s advice, we added sentences and references for the role of fructose in NAFLD development as follows.

(Lines 74-85)

This theory suggests that the disease process begins with de novo lipogenesis (DNL) by increase fructose consumption by western style diet and the development of insulin resistance resulting from excessive energy intake (5, 6). Fructose is a potent lipogenic carbohydrate contributing to hepatic steatosis. Fructose is taken into hepatocyte via glucose transporter 2 and converted into fructose-1-phosphate (F1P) by fructokinase. These physiological sequences are not controlled by insulin and induced by fructose (6). Fructose-bisphosphate aldolase B (known as hepatic aldolase) converts F1P into glycogen, glucose, lactate, and acetyl-CoA. Fructose also upregulate key transcription factors for fatty acid synthesis such as sterol response element binding protein 1c and carbohydrate responsive element binding Protein (7). Both acetyl-CoA oversupply and induce of lipogenic enzymes increase DNL in hepatocyte strongly.

Attachment

Submitted filename: Response to Reviewers.docx

Decision Letter 1

Catherine Mounier

11 Feb 2020

Serum miR-379 expression is related to the development and progression of hypercholesterolemia in non-alcoholic fatty liver disease

PONE-D-19-17509R1

Dear Dr. Kinya Okamoto,

We are pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it complies with all outstanding technical requirements.

Within one week, you will receive an e-mail containing information on the amendments required prior to publication. When all required modifications have been addressed, you will receive a formal acceptance letter and your manuscript will proceed to our production department and be scheduled for publication.

Shortly after the formal acceptance letter is sent, an invoice for payment will follow. To ensure an efficient production and billing process, please log into Editorial Manager at https://www.editorialmanager.com/pone/, click the "Update My Information" link at the top of the page, and update your user information. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to enable them to help maximize its impact. If they will be preparing press materials for this manuscript, you must inform our press team as soon as possible and no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

With kind regards,

Catherine Mounier

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #2: All comments have been addressed

Reviewer #3: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: To the best of my knowledge, all the comments have been carefully and adequately addressed by the authors.

The manuscript has met all the criteria for publication.

Reviewer #2: The authors revised the manuscript according to the reviewers' suggestions. The manuscript quality has consistently improved and, to my opinion, it is suitable for publication.

Reviewer #3: The authors have addressed all my comments successfully. Unfortunately they did not have enough serum to validate expression of IGF-1, IGF1R, and INSR but they did acknowledge that this is the biggest weakness of the manuscript. I do not have additional comments for the authors.

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: Yes: Michael O. Baclig

Reviewer #2: Yes: Pietro Di Fazio

Reviewer #3: Yes: Samir Softic

Acceptance letter

Catherine Mounier

14 Feb 2020

PONE-D-19-17509R1

Serum miR-379 expression is related to the development and progression of hypercholesterolemia in non-alcoholic fatty liver disease

Dear Dr. Okamoto:

I am pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please notify them about your upcoming paper at this point, to enable them to help maximize its impact. If they will be preparing press materials for this manuscript, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

For any other questions or concerns, please email plosone@plos.org.

Thank you for submitting your work to PLOS ONE.

With kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Catherine Mounier

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Linear regression analysis of relationships between serum miR-379 and clinical features of NAFLD patients.

    Normalized relative to serum miR-16; miR-379 values represent fold-difference relative to the normal control.

    (TIF)

    S2 Fig. Linear regression analysis of the relationships between serum miR-379 and clinical features of early stage NAFLD patients (Brunt fibrosis stage 0 to 1).

    Normalized relative to serum miR-16; miR-379 values represent fold-difference relative to the normal control.

    (TIF)

    S3 Fig. Linear regression analysis of the relationships between serum miR-379 and clinical features of advanced-stage NAFLD patients (Brunt fibrosis stage 2 to 4).

    Normalized relative to serum miR-16; miR-379 values represent fold-difference relative to the normal control.

    (TIF)

    Attachment

    Submitted filename: Response to Reviewers.docx

    Data Availability Statement

    All relevant data are within the manuscript and its Supporting Information files.


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES