Observational Study Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jun 28, 2015; 21(24): 7529-7544
Published online Jun 28, 2015. doi: 10.3748/wjg.v21.i24.7529
Liver fat deposition and mitochondrial dysfunction in morbid obesity: An approach combining metabolomics with liver imaging and histology
Nahum Calvo, Raúl Beltrán-Debón, Esther Rodríguez-Gallego, Anna Hernández-Aguilera, Maria Guirro, Roger Mariné-Casadó, Lidón Millá, Josep M Alegret, Jordi Camps, Jorge Joven, Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Campus of International Excellence Southern Catalonia, 43201 Reus, Spain
Fàtima Sabench, Daniel del Castillo, Servei de Cirurgia General i de l’Aparell Digestiu, Hospital Universitari de Sant Joan de Reus, Universitat Rovira i Virgili, 43201 Reus, Spain
María Vinaixa, Miguel Àngel Rodríguez, Xavier Correig, Metabolomics Platform, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, IISPV, Universitat Rovira i Virgili, 43007 Tarragona, Spain
Roberto García-Álvarez, Research and Collaborations, General Electric Healthcare, 78530 Buc, France
Javier A Menendez, Metabolism and Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology, Girona Biomedical Research Institute, 17007 Girona, Spain
Author contributions: Calvo N and Beltrán-Debón R contributed equally and were responsible for acquired data from MRI and MRS; all authors contributed to conception and design, acquisition of data, or analysis and interpretation of data; the article was written and edited by Joven J and Menendez JA and all authors revised and approved the final version; García-Álvarez R established the acquisition protocols in MRI and MRS and defined the methods of analysis; Sabench F and del Castillo D were responsible for the correct management of the participants; and Vinaixa M, Rodríguez MA and Correig X analysed by 1H-NMR the metabolites extracted in liver biopsies.
Supported by Universitat Rovira I Virgili and the Hospital de Sant Joan de Reus; Some aspects have been funded by grants from the Carlos III Health Institute, Madrid, Spain and the European Fund for Regional Development, No. PI08/1381 and No. PI11/00130.
Ethics approval: The local ethics committee, which acts as the institutional review board of the Hospital de Sant Joan de Reus.
Clinical trial registration: EPINOLS/12-03-29/3proj6; OBESPAD/14-07-31/7proj3.
Informed consent: The study protocol and procedures were approved, and written informed consent was obtained from the participants.
Conflict-of-interest: The authors declare that there are no conflicts of interest associated with this manuscript.
Data sharing: All data associated with this manuscript, conveniently anonymized, are available under request from the corresponding author, who takes responsibility for the integrity and the accuracy of data analysis.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Jorge Joven, MD, PhD, Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d’Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Campus of International Excellence Southern Catalonia, c/ Sant Llorenç 21, 43201 Reus, Spain. jorge.joven@urv.cat
Telephone: +34-977-310300 Fax: +34-977-59386
Received: November 27, 2014
Peer-review started: November 28, 2014
First decision: December 26, 2014
Revised: January 20, 2015
Accepted: February 12, 2015
Article in press: February 13, 2015
Published online: June 28, 2015

Abstract

AIM: To explore the usefulness of magnetic resonance imaging (MRI) and spectroscopy (MRS) for assessment of non-alcoholic fat liver disease (NAFLD) as compared with liver histological and metabolomics findings.

METHODS: Patients undergoing bariatric surgery following procedures involved in laparoscopic sleeve gastrectomy were recruited as a model of obesity-induced NAFLD in an observational, prospective, single-site, cross-sectional study with a pre-set duration of 1 year. Relevant data were obtained prospectively and surrogates for inflammation, oxidative stress and lipid and glucose metabolism were obtained through standard laboratory measurements. To provide reliable data from MRI and MRS, novel procedures were designed to limit sampling variability and other sources of error using a 1.5T Signa HDx scanner and protocols acquired from the 3D or 2D Fat SAT FIESTA prescription manager. We used our previously described 1H NMR-based metabolomics assays. Data were obtained immediately before surgery and after a 12-mo period including histology of the liver and measurement of metabolites. Values from 1H NMR spectra obtained after surgery were omitted due to technical limitations.

RESULTS: MRI data showed excellent correlation with the concentration of liver triglycerides, other hepatic lipid components and the histological assessment, which excluded the presence of non-alcoholic steatohepatitis (NASH). MRI was sufficient to follow up NAFLD in obese patients undergoing bariatric surgery and data suggest usefulness in other clinical situations. The information provided by MRS replicated that obtained by MRI using the -CH3 peak (0.9 ppm), the -CH2- peak (1.3 ppm, mostly triglyceride) and the

-CH=CH- peak (2.2 ppm). No patient depicted NASH. After surgery all patients significantly decreased their body weight and steatosis was virtually absent even in patients with previous severe disease. Improvement was also observed in the serum concentrations of selected variables. The most relevant findings using metabolomics indicate increased levels of triglyceride and monounsaturated fatty acids in severe steatosis but those results were accompanied by a significant depletion of diglycerides, polyunsaturated fatty acids, glucose-6-phosphate and the ATP/AMP ratio. Combined data indicated the coordinated action on mitochondrial fat oxidation and glucose transport activity and may support the consideration of NAFLD as a likely mitochondrial disease. This concept may help to explain the dissociation between excess lipid storage in adipose tissue and NAFLD and may direct the search for plasma biomarkers and novel therapeutic strategies. A limitation of our study is that data were obtained in a relatively low number of patients.

CONCLUSION: MRI is sufficient to stage NAFLD in obese patients and to assess the improvement after bariatric surgery. Other data were superfluous for this purpose.

Key Words: Fatty liver disease, Magnetic resonance imaging, Lipids, Magnetic resonance spectroscopy, Metabolomics, Mitochondrial function, Morbid obesity, Non-alcoholic fatty liver disease, Non-alcoholic steatohepatitis

Core tip: Despite remaining obese, non-alcoholic fatty liver disease (NAFLD) regressed in patients undergoing bariatric surgery. Magnetic resonance imaging was sufficient to assess these findings and spectroscopy and measurement of liver fat content were superfluous for this purpose. Patients were free of hepatocyte ballooning or fibrosis and results may limit further the indication for liver biopsy to patients with known factors affecting the progression of liver disease. Additional information on the dissociation between lipid storage in adipose tissue and NAFLD suggests the presence of mitochondrial and inflammatory disease and may focus on the unmet need for plasma biomarkers of circulating metabolites produced during altered cellular fatty oxidation and glycolysis.


  • Citation: Calvo N, Beltrán-Debón R, Rodríguez-Gallego E, Hernández-Aguilera A, Guirro M, Mariné-Casadó R, Millá L, Alegret JM, Sabench F, del Castillo D, Vinaixa M, Rodríguez M&, Correig X, García-Álvarez R, Menendez JA, Camps J, Joven J. Liver fat deposition and mitochondrial dysfunction in morbid obesity: An approach combining metabolomics with liver imaging and histology. World J Gastroenterol 2015; 21(24): 7529-7544
  • URL: https://www.wjgnet.com/1007-9327/full/v21/i24/7529.htm
  • DOI: https://dx.doi.org/10.3748/wjg.v21.i24.7529

INTRODUCTION

Non-alcoholic fatty liver disease (NAFLD) is an important cardiovascular and metabolic risk factor defined as a higher than 5% intra-cytoplasmic fat deposition in the liver in the absence of alcohol consumption, toxin exposure or viral disease[1,2]. Because a progressive course is possible in this condition, a benign prognosis should not be assumed. This is particularly disturbing because the prevalence of NAFLD is constantly increasing (now > 30% in adults in developed countries). This has been attributed to the increased lifespan and the upsurge in obesity but these associations are not clinically well substantiated. For example, the presence of moderate NAFLD is common in young patients with morbid obesity[3,4]. The effect of weight loss on NAFLD is beneficial in most patients but difficult to achieve with simple advice on changes in lifestyle. In patients with morbid obesity, bariatric surgery is an alternative and effective treatment to obtain a rapid and sustained weight loss. Additionally, other beneficial effects for associated metabolic conditions, apparently unrelated to the decrease in body weight, have been described[5]. Accurate mechanisms of action, however, remain poorly understood.

There are no reliable non-invasive markers to assess the beneficial effects of surgical procedures on NAFLD and other obesity-associated morbidities. The sensitivity and accuracy of ultrasound are too low to detect NAFLD in obese patients and liver biopsy is the only method to assess the presence of non-alcoholic steatohepatitis (NASH) and/or fibrosis, which characterize the progression in NAFLD. Therefore, the early diagnosis of subclinical NAFLD is uncommon in clinical practice because physicians and/or patients usually find liver biopsy to be a difficult undertaking, which requires specific training so as to ensure the lowest rate of complications[6,7]. The consequence is that NAFLD, a condition that may lead to eventual liver-related mortality, is frequently understated[8].

Determination of the hepatic triglyceride concentration[9] may be useful but we have based our hypothesis on previous findings in animal models[10] indicating that cholesterol and fatty acids in the liver are better indicators of inflammation, and consequently 1H NMR metabolic profiling of multiple lipid metabolites in liver extracts may extend our understanding of underlying mechanisms. We also hypothesized that this approach could add useful information to magnetic resonance imaging (MRI) and spectroscopy (MRS) findings in order to explore hepatic lipid mobilization. Finally, we reasoned that correction of possible confounding factors may improve the clinical application of MRI in the assessment of NAFLD in obese patients and its follow-up after bariatric surgery.

MATERIALS AND METHODS
Study population

This is an observational, prospective, single-site, cross-sectional study with a pre-set duration of 1 year involving the recruitment of 47 patients. Only 19 patients were finally included. We discarded 3 patients due to the confirmation of clandestine high consumption of alcohol or prescribed medication that could alter liver function. Other patients refused either the MRI procedure or the biopsy after surgery (n = 8). Other patients (n = 9) simply did not fit into the apparatus. Finally, the obtained spectra were considered suboptimal in 8 patients due to excessive proximity between the coil and the gantry. Subsequent studies indicate that to obtain 100% of valid spectra, the width at the level of iliac crests should be less than 100 cm. The local ethics committee, which acts as the institutional review board of the Hospital de Sant Joan de Reus, approved the study protocol and procedures, and written informed consent was obtained from the participants (EPINOLS/12-03-29/3proj6; OBESPAD/14-07-31/7proj3). Investigators did not participate in the statistical analyses and those responsible for the acquisition of different data were blinded to each other until discussion of the manuscript. Data were collected from selected patients referred to the surgery department of our hospital for the assessment of morbid obesity [body mass index (BMI) ≥ 40], who fulfilled the indications for the performance of laparoscopic sleeve gastrectomy in this period of time[11]. Liver viral diseases were ruled out via standard laboratory analyses. Selected patients were clinically assessed immediately (< 24 h) before surgery and after a 12-mo period. Results for the standard techniques used in this study are illustrated in Figure 1.

Figure 1
Figure 1 Representative results obtained with the methods described in patients with low (A) and severe fatty liver disease (B). The resulting spectra were drawn in blue and black, respectively. Scale bar in pictures under Histology title represents 100 μm (magnification × 100).
Liver biopsy

We strictly followed the designated guidelines and recommendations set by the American Association for the Study of Liver Diseases[12]. Portions of the liver were obtained during the surgical procedure to limit risks and the variability based on location of biopsy as well as to be certain of the accurate location of extraction. The procedure was performed with needle devices rather than wedge resection to avoid overestimates of fibrosis due to its proximity to the capsule. Liver biopsies after surgery (12 mo) were undertaken with a real-time image-guided percutaneous method immediately after MRI assessment. The above-mentioned guidelines include the accepted pathological features of fatty liver disease, especially the indices of inflammation (steatohepatitis) and fibrosis. Trained pathologists provided the relevant data using current histological scoring systems. Due to the low number of included patients and the need for parametric comparisons, we used percentage of fat for further analyses as previously described[1]. Patients were considered free of steatosis when percentage of fat was ≤ 5%[1] as evaluated using image analysis software (AnalySIS, Soft Imaging System, Munster, Germany)[4].

MRI and MRS

MR data were acquired on a 1.5T Signa HDx MRI Scanner (General Electric Healthcare, Waukesha, WI) and included pre-surgical information regarding vascular structures. Patients fasted overnight and studies were performed in supine position using a whole-body radiofrequency coil for signal excitation and a 16-channel torso phased array coil for reception (General Electric Coils, Cleveland, OH). Imaging protocols[13] consisted of: (1) coronal view (slice thickness 6 mm; gap 1 mm; flip angle 75°; matrix size 256 × 192; field of view 48 cm); (2) axial view (slice thickness 7 mm; gap 1 mm; flip angle 75°; matrix 256 × 192; field of view 48 cm); (3) axial T2 fast recovery Spin-Echo sequence with fat suppression (slice thickness 8 mm; gap 1 mm; matrix size 320 × 224; field of view 48 cm); (4) axial T1-weighted dual phase gradient-echo sequence (repetition time in ms: 120/2.1 in opposed phase and 120/4.2 when in phase; flip angle 80°; slice thickness 7 mm; gap 1 mm; matrix 256 × 224; field of view 48 cm); and (5) a single-voxel breath-hold spectroscopy sequence. The Point-RESolved Spectroscopy[14] sequence was used to record spin echos: acquisition with TR = 1500 ms, TE = 35 ms and 128 signal averages per data-frame with an eight-phase cycling scheme (2048 data points per spectroscopy frame with a spectral band width of 2500 Hz) resulting in an acquisition time of approximately 12 min. Chemical shift-selective radio frequency pulses (n = 3) suppressed the water signal. To discriminate the peaks corresponding to metabolites from artefacts, each spectrum represents the average of 16 data-frame acquisitions (2 without water suppression)[15]. To ensure the accurate biopsy site, the voxel was placed manually to provide accessibility and to avoid motion artefacts by the heart and flow artefacts from vascular structures[13,16]. The biopsy procedure was carefully supervised according to this information. Values for signal intensity (SI) were obtained from pre-designed regions of interest (ROI) in the liver, spleen (both 20 mm × 20 mm) and paravertebral musculature (5 mm × 5 mm) and calculations were performed to avoid the possible T2 effect caused by ferric deposits. The hepatic fat fraction was measured according to the formula: SIin-phase - SIout-of-phase/2SIin-phase[14,16-18]. The visceral and subcutaneous adipose tissues were quantified in out-of-phase images at pre-designed ROI. The MRS spectra were transformed using specific software via the SAGE (Spectroscopy Analysis General Electric) single voxel PROBE-Q reconstruction and normalized by water signal. The amplitude and area under the curve of each detectable peak were calculated as described[19].

1H NMR based metabolomics assays and laboratory procedures

Portions of hepatic tissue obtained from liver biopsy were flash-frozen and homogenized[20]. For NMR measurements hydrophilic and lipophilic metabolites were separately lyophilized overnight to remove water and reconstituted as described[10,21]. One- and two-dimensional 1H NMR spectra were obtained at a 600.20 MHz frequency using an Avance III-600 Bruker spectrometer equipped with an inverse TCI 5 mm cryoprobe (Bruker, Rheinstetten, Germany). The frequency spectra were baseline corrected, and then calibrated using TopSpin software (version 2.1, Bruker). Resonance assignments were performed as described[10,22]. Selected peaks were integrated using the AMIX 3.8 software package (Bruker) and the absolute concentration of single metabolites measured as described[23] (Table 1). The integral at 0.87 corresponding to ω-CH3 was used as a reference for total fatty acid chains to estimate the molar percentage of fatty acid signals. Common laboratory measurements were performed with commercially available reagents and well-established methods[24,25]. The liver-tissue content of cholesterol and triglyceride was also determined after lipid extraction with the isopropyl alcohol-hexane method[26]. Cytokines and adipokines were measured using enzyme-linked immunosorbent assays following the manufacturer’s instructions (RD Systems, Minneapolis, United States).

Table 1 Absolute concentration of single metabolites obtained by 1H NMR metabolomic profiles.
MeanSDKruskall-Wallis (P)None
Low
Moderate
Severe
U Mann-Withney (P)
MeanSDMeanSDMeanSDMeanSDN vs LL vs MM vs SL vs S
Lipid-soluble metabolites (mmol/g liver)
Total cholesterol5.191.712.740.515.032.155.890.685.991.06
Free cholesterol1.320.401.090.181.520.411.360.421.000.27
Sterified cholesterol3.871.651.660.703.501.824.541.014.991.31
Triglycerides141.3392.950.00813.716.3388.8259.32184.6126.11259.2967.090.0170.020
Diglycerides8.043.2210.357.428.402.378.552.704.910.960.039
Monoglycerides0.730.200.690.250.800.280.770.120.570.06
Total phospholipids22.755.860.04629.447.7424.281.5623.751.6513.596.030.0250.020
PC15.904.0620.055.7016.711.3317.130.739.504.210.0250.020
PE10.592.3112.372.5811.130.3311.520.796.792.750.0250.020
Phosphatidylinositol1.990.850.0281.540.652.760.711.770.601.130.220.0280.020
Phosphatidylserine0.130.070.070.010.140.080.150.070.100.03
Lysophosphatidylcholine1.070.750.620.070.820.201.591.221.020.250.045
Sphingomyelin1.970.392.180.532.130.222.030.241.410.370.020
Plasmalogen1.010.130.970.231.010.061.100.080.880.17
Lipid-soluble metabolites (%mol)
Omega-3 fatty acids2.141.320.0094.570.402.491.151.440.100.980.320.0280.020
Oleic acid (18:1 ω-9)24.534.3716.421.7024.344.2926.621.1526.812.950.045
Arachidonic (20:4 ω-6) + eicosapentaenoic (20:5 ω-3) acids3.513.090.0099.822.893.962.161.940.321.050.360.0450.0250.020
Docosahexaenoic acid (22:6 ω-3)0.720.650.0121.970.410.850.560.390.090.200.140.0280.027
Linoleic acid (18:2 ω-6)5.911.810.029.630.236.230.845.020.984.270.850.0450.020
PUFA20.4117.580.00955.7312.6023.4312.911.551.715.573.110.0280.020
MUFA, with PUFA93.1511.470.008112.963.5397.795.2787.685.9479.816.570.0450.0280.020
PUFA/MUFA ratio0.200.140.0150.490.090.230.110.130.020.070.030.020
PC/PE ratio1.480.091.610.121.500.081.480.061.380.06
Aqueous-soluble metabolites (mmol/g liver)
Glucose-6-phosphate32.6513.341.6932.9635.637.9434.274.4617.9612.1
Glycogen2.192.342.570.621.700.812.954.111.661.67
UDPG1.220.501.740.751.250.141.190.540.890.73
3-pyruvate1.720.682.241.211.930.491.750.450.920.63
3-hidroxybutirate0.760.351.040.860.710.250.690.250.780.40
Lactate20.486.6317.375.6023.753.2320.956.5615.2510.9
Fumarate0.300.100.320.060.360.120.280.060.210.050.038
Free glycerol1.640.941.530.281.480.261.470.392.352.29
NAD/NADH/NADPH0.390.130.500.180.460.070.350.100.260.110.038
ATP/ADP/AMP1.680.702.431.301.860.461.550.591.020.520.038
UTP/UDP/UMP0.870.290.850.211.030.230.970.200.430.110.0250.020
Uracil0.040.020.030.010.050.020.040.010.020.02
Carnitine1.160.592.160.821.110.351.020.500.830.56
Cholines1.500.361.620.051.590.271.580.311.070.50
Acetates0.480.110.450.020.540.080.490.130.390.12
Ascorbic acid1.360.571.800.401.300.381.360.621.181.01
Leucine0.550.100.500.020.590.090.590.100.440.020.0250.020
Valine0.600.130.550.010.660.090.650.120.420.070.0250.020
Alanine6.072.385.684.187.811.325.830.833.222.540.0280.038
Isoleucine0.250.050.230.010.260.040.290.060.190.020.025
Lysine1.260.261.210.081.380.131.340.330.920.140.020
Glutamine3.961.595.831.904.480.893.601.192.271.86
Glutamate4.421.630.0384.700.015.631.783.920.822.650.980.020
Methionine0.800.240.760.240.840.230.700.180.900.42
Phenylalanine0.260.050.250.020.290.030.270.040.170.050.020
Threonine1.460.502.130.591.580.451.360.080.940.53
Tyrosine0.200.050.0260.160.040.240.020.210.040.140.050.0450.020
Histidine0.160.070.250.130.170.060.130.040.120.04
Taurine12.534.0717.031.5513.684.1411.571.468.815.38
Gluthathione oxidized0.730.401.310.460.630.290.570.380.780.42
Creatine3.161.244.390.783.740.732.890.911.611.370.038
Creatinine0.150.040.0100.150.010.190.020.120.020.100.050.0450.0060.020
Statistical analysis

Unless otherwise stated, quantitative variables were expressed as mean and interquartile range and categorical variables as absolute or relative frequencies. Statistical significance was considered if P values were less than 0.05 and all analyses were conducted using the Statistical Package for Social Sciences (SPSS-18.0, Chicago, IL). Differences between groups were assessed with a Student’s t-test, Kruskal-Wallis or Mann Whitney U-test. Spearman or Pearson correlation coefficients were used to evaluate the degree of association between variables. To assess differences before and after surgery, calculations were performed with Welch’s t-tests and/or Wilcoxon’s rank sum tests as well as ANOVA for repeated measures. To correct for multiple testing we used the False Discovery Rate estimated using the Q-value. The choice of tests was based on normality of values and the sample size, which varies with losses in observations as indicated in results.

RESULTS
Baseline and follow-up characteristics of participants

No acute illness or surgical complications were recorded during the study. Most patients were female (79%), with ages between 34 and 55 years. The reference BMI was calculated immediately before surgery (range: 42.1-51.2 kg/m2) and changes were negligible in the previous 6 mo. The body weight decreased in all patients. As a group, the mean decrease was 41.2 kg, approximately 35%. There were changes in parallel with weight loss, especially improvements in glucose and lipid metabolism, and in the circulating levels of leptin and adiponectin and the paraoxonase activity. Some inflammatory cytokines also decreased with weight loss, including C-reactive protein, but others did not. Contrarily, changes in the activities of enzymes traditionally considered as surrogates of liver function improved after surgery but independently of weight loss (Table 2, n = 19).

Table 2 Selected variables to explore metabolic changes and anti-oxidative or anti-inflammatory effects prior to and one year after surgery.
Before surgeryAfter surgeryP value
BMI, kg/m245.4 (44.1-47.0)32.3 (30.1-35.4)< 0.001
Liver steatosis, %30 (5-55)6 (0-15)< 0.001
Glucose, mmol/L7.9 (6.7-10.1)5.4 (4.6-6.2)< 0.001
Insulin, pmol/L114.2 (86.3-166.8)66.9 (58.0-70.1)< 0.001
HOMA2-IR4.73 (1.85-10.67)1.95 (1.30-5.80)< 0.001
Total cholesterol, mmol/L4.93 (4.59-5.24)4.28 (4.05-4.87)< 0.001
Triglycerides, mmol/L1.76 (1.12-2.42)1.08 (0.85-1.38)< 0.001
HDL-cholesterol, mmol/ L1.06 (0.94-1.23)1.31 (1.02-1.44)< 0.001
NEFAs, mmol/L1.08 (0.85-1.48)0.80 (0.61-0.96)< 0.050
Bilirubin, mmol/L8.2 (5.1-9.1)9.1 (7.2-8.5)NS
ALP, μKat/L0.82 (0.70-1.1)0.65 (0.54-0.92)< 0.050
AST, μKat/L1.03 (0.37-1.24)0.82 (0.24-1.14)NS
ALT, μKat/L0.52 (0.43-1.25)0.45 (0.33-0.97)NS
GGT, μKat/L0.34 (0.24-1.08)0.30 (0.20-0.95)NS
Amylase, μKat/L0.67 (0.54-0.84)0.42 (0.33-0.65)< 0.050
Paraoxonase, U/L214 (160-410)360 (200-405)< 0.050
Leptin, ng/mL32.5 (21.2-40.1)9.2 (6.3-11.1)< 0.001
Adiponectin, ng/mL4.2 (2.1-5.9)7.5 (4.9-8.2)< 0.001
Interleukin 6, pg/mL0.52 (0.21-0.75)0.43 (0.12-0. 59)NS
Interleukin 8, pg/mL1.86 (1.45-2.15)1.42 (1.22-1.65)NS
TNF-α, pg/mL6.21 (4.35-7.82)5.12 (3.95-7.36)< 0.050
CCL2, pg/mL510 (425-590)395 (280-430)< 0.001
Hs-CRP, mg/L4.6 (1.5-7.2)1.1 (0.3-1.67)< 0.001
Bariatric surgery and remission of NAFLD

Despite morbid obesity, NAFLD was not a universal finding and severity was modest as assessed histologically (< 15% of triglyceride accumulation in 30% of the patients). In some it was negligible (< 5%) and in others severe (> 70%). This distribution was not explained by metabolic derangements. We discarded the presence of NASH as we did not observe hepatocyte ballooning, lobular inflammation or significant fibrosis. The correlation between histological percentages of triglyceride accumulation and hepatic fat fraction obtained by MRI was considered excellent (Figure 2A). One year after surgery, NAFLD decreased in all patients and was virtually absent (0%-7%) (Figure 2B; Table 2). In this second observation, with reduced values, the association between methods was also excellent. Sensitivity and specificity to stage the disease were sufficient (both > 90%). These data are probably the result of the continuous cooperation between radiologists and surgeons and the correction for T2 shortening due to liver iron deposition. The mathematical expression most likely will change among laboratories to ensure correct stage in severe cases. Additional corrections in spleen and muscles were apparently unnecessary (Figure 3). Further, the lack of sampling variability is paramount in comparisons because the heterogeneous distribution of NAFLD is relatively common in these patients. This fact has been illustrated in Figure 4.

Figure 2
Figure 2 Correlation between hepatic steatosis measured by magnetic resonance imaging and by histological analysis before surgery (A); non-alcoholic fatty liver disease was considerably reduced after one year in all patients (B).
Figure 3
Figure 3 Magnetic resonance imaging signal was further corrected by the spleen (A) or muscle (B) intensity signals, but this did not improve the quality of the correlation with hepatic steatosis assessed by histological analysis.
Figure 4
Figure 4 Liver fat distribution is commonly non-homogeneous in non-alcoholic fat liver disease, and this fact could influence the diagnosis of steatosis.
Results obtained by MRS were similar to those observed by MRI

Contrary to our hypothesis, MRS did not provide additional and relevant information in the staging of NAFLD. Moreover, in some patients without NAFLD, some peaks were undetected and inter-individual variability of other peaks was too high to be considered. However, there were at least 3 peaks with reliable information to compare values before and after surgery (the 0.9-ppm, 1.3-ppm peak and 2.2 ppm peak, Figure 5) most likely indicating the presence of -CH3 (0.9 ppm), -CH2- (1.3 ppm) and -CH=CH- (2.2 ppm) (Figure 6). The calculations of either amplitude or the area under the curve (AUC) yield similar results with fair-to-good correlation with histologically assessed values but less robust than those observed with MRI (Figure 7). Characteristically, the 1.3-ppm and 2.2-ppm peaks are considered as representative of triglyceride and unsaturated fatty acids, but actual values obtained by biochemical measurements (see below) suggest the combination of multiple signals from other compounds.

Figure 5
Figure 5 Hepatic steatosis was evaluated in patients before and after surgery in liver biopsy (A, B); the decrease in hepatic fat content also could be visualized by magnetic resonance spectroscopy (C, D), where the peaks that offer more information were those at 0. 9, 1.3 and 2.2 ppm (E). Scale bar in A and B represents 100 μm (magnification × 100).
Figure 6
Figure 6 Representative spectrum of identifiable metabolites with magnetic resonance spectroscopy in vivo: 0. 9 ppm - terminal carbon (-CH3); 1.3 ppm - total fatty acyl chains (-CH2-); 2.0-2.2 ppm - poly-unsaturated fatty acids (-CH2=CH2-); 2.6 ppm -Citrate; 3.2 ppm - choline containing compounds; 5.3-5.6 ppm - mono+polyunsaturated fatty acids. Water relative resonance frequency appears at 4.7 ppm as major compound. In this case, water is suppressed for facilitate the visualization of other detectable compounds.
Figure 7
Figure 7 Similar results were obtained using the following methods of calculation: Amplitude of the peaks measurement (left open-circle plots) and area under the curve (AUC, right black-circle plots). The most important peaks associated with fat are at 1.3 ppm (A) and 2.2 ppm (B). 1.3 ppm peak is mainly associated with triglycerides (C) while 2.2 ppm peak is associated with unsaturations (D).
Metabolomics: Liver fat composition and the assessment of steatosis

The correlation between steatosis and liver triglyceride content was not different from that obtained between steatosis and MRI (r = 0.89, P = 1.2 × 10-5, n = 16) indicating that the invasive measurement of liver triglyceride content is not a valuable diagnostic tool in these patients. Data provided by 1H NMR spectroscopic profiles are given only for the biopsy obtained during surgery because in some samples obtained in the second biopsy the concentrations of most metabolites were lower than a predesignated detection level. Hence, this portion of the liver was used only for clinical purposes. 1H NMR spectra of aqueous soluble liver tissue extracts showed resonances associated with low molecular weight metabolites, and 1H NMR spectra from lipid soluble extracts depicted several dominating regions with major peaks attributable to double bonds mainly from protons belonging to di- or triacylated glycerols, the phospholipids polar head groups, methylene, methyl groups of the fatty acyl chains and unsaturated fatty acyl moieties, and they were all associated with liver fat deposition (Table 1). Interestingly, in patients with no or very moderate NAFLD, the levels of triglycerides and oleic acid were low and there were increased hepatic levels of polyunsaturated fatty acids (PUFA)ω-3 fatty acyls, docosahexanoic 22:6 (n-3) (DHA), arachidonic 20:4 (n-6) (ARA) + eicosapentaenoic 20:5 (n-3) (EPA), and the ratio between poly- and mono-unsaturated fatty acids (PUFA/MUFA). These changes paralleled the increased levels of glycerol, lactate, glucose-6-phosphate (G-6-P), carnitine and taurine. Conversely, in moderate to severe steatosis, the levels of triglyceride, oleic acid, and some amino acids were high but there was a significant depletion of PUFA. This is illustrated by significantly positive or inverse correlations found in these metabolites with the stage of NAFLD (Figure 8). Importantly, in severe NAFLD there were significant reductions in diglycerides (DAG), G-6-P, fumarate, NAD/NADH, ATP/AMP, and UTP/UMP (Table 1) most likely indicating a dysfunction in the coordination of mitochondrial fat oxidation and glucose transport activity.

Figure 8
Figure 8 Several lipidic metabolites measured using 1H NMR-based metabolomics were clearly positively associated with hepatic steatosis (A-D); Others showed the opposite behavior (E-J). ARA: Arachidonic acid; EPA: Eicosapentaenoic acid; DHA: Docosahexaenoic acid; PUFA: Poly-unsaturated fatty acids; MUFA: Mono-unsaturated fatty acids.
Dissociation between excess lipid storage in adipose tissue, NAFLD and insulin resistance

Our data indicate that even in patients with morbid obesity the incidence of NAFLD is not universal. Bariatric surgery was accompanied by a reduction in BMI and practically eliminated the ectopic fat in the liver but patients remained obese (BMI > 30). There was no correlation between BMI and steatosis (r = 0.227). Additionally, body fat distribution was not associated with the development of fatty liver as the correlations between the subcutaneous and visceral adipose tissues, measured by MRI, and fatty liver severity were not different from 0 (Figure 9). Moreover, the degree of impaired glucose metabolism of patients was not associated with the presence of NAFLD. There were insulin-sensitive individuals with a high percentage of ectopic liver fat and there were participants with insulin resistance remaining free of NAFLD. This is counterintuitive because the overflow of energy storage from adipose tissue to the liver is apparently unavoidable. Our findings reinforce the concept that the composition of ectopic lipid is important in searching for metabolites most likely mediating defects in insulin signalling and/or mitochondrial function[6,10,27-32].

Figure 9
Figure 9 Body fat content was measured using magnetic resonance imaging. Both visceral (A) and subcutaneous adipose tissue (B) were not related to the development of non-alcoholic fatty liver disease.
DISCUSSION

The metabolic syndrome and obesity are strong predictors of NAFLD but mechanisms to explain an association between NAFLD and morbid obesity remain unclear. Some, most likely 20%-30%, of obese patients are currently identified as “metabolically healthy”[33] but this is irrelevant because it is not translated into a lower risk of mortality[34]. NAFLD is not a benign condition and consequently should be carefully assessed. Our MRI data indicate that in patients with morbid obesity this technique provides diagnostic accuracy > 90% with sufficient sensitivity and specificity (both > 90%) to stage the disease and to assess the effect of surgery. The main limitations include the cost, the requirement for considerable efforts to minimize sampling variability and the size of the patients undergoing bariatric surgery. The latter is also the cause of limitations in our study, which should probably be extended to a more protracted time of observation.

In this group of patients, our data indicate that MRS, liver biopsy and the measurement of liver fat content do not provide clinically useful information and were considered either superfluous or redundant because the likelihood to find NASH was low. Further research, however, should include technical improvements in the design of the apparatus and in the ability to obtain more detailed spectra. The advantage of this non-invasive technique is obvious but our study design precludes the possibility to test its value in differentiating NAFLD from NASH and/or significant fibrosis, which is important to assess prognosis and indicate management[2,3,35]. However, we consistently observe superior effects on weight loss, NAFLD and lipid and glucose metabolic derangements using bariatric surgery as compared with intensive but conservative medical therapy. Further, bariatric surgery improves insulin sensitivity, lipid metabolism, inflammation, oxidative stress and most likely other factors that might contribute to NAFLD. Previous data suggest the speculation that rapid and sustained weight loss may also improve the histologic characteristics of NASH[6,36,37] but even modest weight reduction (7%-10%) results in significant improvement of NASH[38,39]. In addition, we provide further evidence that inflammatory factors, oxidative stress, and the liberation of cytokines from adipose tissue are likely modulators[40].

Our data limit further the clinical indication of liver biopsy in obese patients for those with the additional presence of known factors affecting the progression of liver disease such as alcohol or drugs consumption, toxin exposure or viral disease. However, they also indicate that liver biopsy is critical to obtain research material to uncover the precise molecular mechanisms that remain elusive. Particularly, we observed a significant mobilization in metabolites according to the severity of NAFLD. We used 1H NMR spectra to obtain signals with technology similar to both MRI and MRS[10] but future studies should include analytical methods that increase the number of reliably detected metabolites[4,22]. This is important because research may result in alternative therapeutic strategies. Of note, we have found that severe NAFLD is accompanied by the progressive inhibition of fatty acid oxidation, which increases glucose oxidation and decreases glucose formation in fatty liver, indicating both mitochondrial dysfunction and the existence of metabolites that cause defects in insulin signalling or glucose transport activity[4,31,41]. The consideration of NAFLD as a likely mitochondrial disease[42,43] is important and confirms our previous findings in animal models characterized by the accumulation of damaged mitochondria and mitochondrial fragmentation[30,44], apparently involved in response to liver inflammation, excess of energy intake and autophagy[45,46]. The implications of a change in paradigm might be also of clinical value. Obesity down-regulates liver autophagy and the therapeutic target should be the restoration of liver autophagy. This is also important due to the clear dissociation between lipid storage in adipose tissue, NAFLD and insulin resistance. This is in line with the Randle cycle, which draws attention to competition between glucose and fatty acids for their oxidation, and pharmacological models of mitochondrial impairment, which stimulates insulin sensitivity and protects against diet-induced obesity, NAFLD and insulin resistance[47,48]. However, an important limitation of our study is the relatively low amount of valid data and we cannot exclude the possibility of other important metabolic factors. It may also be useful to focus the search for serum biomarkers among metabolites involved in energy metabolism[4,49]. It is likely that a unique technique may not suffice and serum biomarkers should be combined with MRI or other techniques (e.g., elastography) to provide the optimal diagnostic discrimination that is required in the fight against obesity-associated disorders[15]. Interestingly, these data are partially confirmative of findings indicating that metabolic changes are weight-dependent. However, in the assessment of NAFLD progression these may be limited to the morbidly obese patient[50].

To sum up, MRI is a sufficient, non-invasive technique, adequate to accurately stage NAFLD in obese patients and to assess the clinical follow-up of bariatric surgery. In uncomplicated patients, when procedures are performed to avoid sampling variability and confounding factors, data from MRS, liver biopsy and the measurement of liver fat content may be considered superfluous. Data from metabolomics may be useful to explain the dissociation between excess lipid storage in adipose tissue, NAFLD and insulin resistance. Additionally, they may support the consideration of NAFLD as a likely mitochondrial disease, which is important to fully understand the pathogenesis and to focus the search for biomarkers and therapeutic strategies.

ACKNOWLEDGMENTS

We thank too many to cite for expert technical assistance, excellent care and samples provided, as well as for criticisms in the design of the study and in writing the manuscript.

COMMENTS
Background

The prevalence of non-alcoholic fatty liver disease (NAFLD) is constantly increasing and is associated with an upsurge in overweight and obesity. Early detection is important because fat deposition in the liver is a recognized risk for cardiovascular health and may progress to severe liver disease. On the other hand, the reversal of this condition is beneficial.

Research frontiers

The metabolic mechanisms leading to NAFLD are not well understood. In obese patients, weight loss modifies its course considerably but to achieve this effect is currently a considerable clinical challenge. Under certain conditions bariatric surgery is an effective treatment to obtain a rapid and sustained weight loss. Unfortunately, liver biopsy remains the only available method to obtain useful information. The consequence is that early diagnosis of NAFLD is difficult. In this study, the authors explore the safety and specificity of MRI in providing accurate diagnosis.

Innovations and breakthroughs

The previous application of non-invasive procedures to ascertain some clinical aspects of NAFLD has been considered suboptimal. The authors devised imaging protocols taking into account all technical limitations and they found them useful especially in bariatric surgery, which requires pre-surgical information regarding vascular structures. These resulted in excellent correlations between histological percentages of triglyceride accumulation and hepatic fat fraction obtained by magnetic resonance imaging (MRI). Using some sophisticated spectroscopic analyses, they have concluded that beneficial effects in associated metabolic conditions achieved by bariatric surgery are apparently unrelated to the decrease in body weight. Some data also indicate the relevance of mitochondrial function in the pathogenesis of ectopic fat deposition and potentially new therapeutic perspectives.

Applications

MRI is an adequate non-invasive technique, to accurately stage fatty liver disease in the clinical follow-up of bariatric surgery. Associated technologies may be used in the assessment of other clinical conditions and to clarify the actual mechanisms of fatty liver disease.

Terminology

Fat should be stored in adipose tissue. The presence in the liver represents a serious metabolic disturbance. The authors have used nuclear magnetic resonance, a physical phenomenon in which nuclei in a magnetic field absorb and re-emit electromagnetic radiation, to obtain images and relevant data on the composition of tissues.

Peer-review

This is a good observational study in which the authors evaluated the efficacy of MRI in NAFLD. The results are interesting and suggest new metabolic insights. An important limitation is the relatively low number of patients included. Further research should be directed to confirm these findings and to generalize its clinical use in obesity-related conditions.

Footnotes

P- Reviewer: Akyuz U, Koch TR, Mihaila RG S- Editor: Qi Y L- Editor: Logan S E- Editor: Ma S

References
1.  Tiniakos DG, Vos MB, Brunt EM. Nonalcoholic fatty liver disease: pathology and pathogenesis. Annu Rev Pathol. 2010;5:145-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 586]  [Cited by in F6Publishing: 605]  [Article Influence: 43.2]  [Reference Citation Analysis (0)]
2.  Adams LA, Lymp JF, St Sauver J, Sanderson SO, Lindor KD, Feldstein A, Angulo P. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology. 2005;129:113-121.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Söderberg C, Stål P, Askling J, Glaumann H, Lindberg G, Marmur J, Hultcrantz R. Decreased survival of subjects with elevated liver function tests during a 28-year follow-up. Hepatology. 2010;51:595-602.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Rodríguez-Gallego E, Guirro M, Riera-Borrull M, Hernández-Aguilera A, Mariné-Casadó R, Fernández-Arroyo S, Beltrán-Debón R, Sabench F, Hernández M, del Castillo D. Mapping of the circulating metabolome reveals α-ketoglutarate as a predictor of morbid obesity-associated non-alcoholic fatty liver disease. Int J Obes (Lond). 2015;39:279-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 72]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
5.  Sjöström L, Lindroos AK, Peltonen M, Torgerson J, Bouchard C, Carlsson B, Dahlgren S, Larsson B, Narbro K, Sjöström CD. Lifestyle, diabetes, and cardiovascular risk factors 10 years after bariatric surgery. N Engl J Med. 2004;351:2683-2693.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Ratziu V, Charlotte F, Heurtier A, Gombert S, Giral P, Bruckert E, Grimaldi A, Capron F, Poynard T. Sampling variability of liver biopsy in nonalcoholic fatty liver disease. Gastroenterology. 2005;128:1898-1906.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Czaja AJ, Carpenter HA. Optimizing diagnosis from the medical liver biopsy. Clin Gastroenterol Hepatol. 2007;5:898-907.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Ekstedt M, Franzén LE, Mathiesen UL, Thorelius L, Holmqvist M, Bodemar G, Kechagias S. Long-term follow-up of patients with NAFLD and elevated liver enzymes. Hepatology. 2006;44:865-873.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Jiménez-Agüero R, Emparanza JI, Beguiristain A, Bujanda L, Alustiza JM, García E, Hijona E, Gallego L, Sánchez-González J, Perugorria MJ. Novel equation to determine the hepatic triglyceride concentration in humans by MRI: diagnosis and monitoring of NAFLD in obese patients before and after bariatric surgery. BMC Med. 2014;12:137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
10.  Vinaixa M, Rodríguez MA, Rull A, Beltrán R, Bladé C, Brezmes J, Cañellas N, Joven J, Correig X. Metabolomic assessment of the effect of dietary cholesterol in the progressive development of fatty liver disease. J Proteome Res. 2010;9:2527-2538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 115]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
11.  Terra X, Auguet T, Guiu-Jurado E, Berlanga A, Orellana-Gavaldà JM, Hernández M, Sabench F, Porras JA, Llutart J, Martinez S. Long-term changes in leptin, chemerin and ghrelin levels following different bariatric surgery procedures: Roux-en-Y gastric bypass and sleeve gastrectomy. Obes Surg. 2013;23:1790-1798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 85]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
12.  Rockey DC, Caldwell SH, Goodman ZD, Nelson RC, Smith AD. Liver biopsy. Hepatology. 2009;49:1017-1044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1449]  [Cited by in F6Publishing: 1419]  [Article Influence: 94.6]  [Reference Citation Analysis (0)]
13.  Neamat-Allah J, Wald D, Hüsing A, Teucher B, Wendt A, Delorme S, Dinkel J, Vigl M, Bergmann MM, Feller S. Validation of anthropometric indices of adiposity against whole-body magnetic resonance imaging--a study within the German European Prospective Investigation into Cancer and Nutrition (EPIC) cohorts. PLoS One. 2014;9:e91586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 62]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
14.  Cassidy FH, Yokoo T, Aganovic L, Hanna RF, Bydder M, Middleton MS, Hamilton G, Chavez AD, Schwimmer JB, Sirlin CB. Fatty liver disease: MR imaging techniques for the detection and quantification of liver steatosis. Radiographics. 2009;29:231-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 213]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
15.  Fitzpatrick E, Dhawan A. Noninvasive biomarkers in non-alcoholic fatty liver disease: current status and a glimpse of the future. World J Gastroenterol. 2014;20:10851-10863.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Anderson PJ, Chan JC, Chan YL, Tomlinson B, Young RP, Lee ZS, Lee KK, Metreweli C, Cockram CS, Critchley JA. Visceral fat and cardiovascular risk factors in Chinese NIDDM patients. Diabetes Care. 1997;20:1854-1858.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Yokoo T, Bydder M, Hamilton G, Middleton MS, Gamst AC, Wolfson T, Hassanein T, Patton HM, Lavine JE, Schwimmer JB. Nonalcoholic fatty liver disease: diagnostic and fat-grading accuracy of low-flip-angle multiecho gradient-recalled-echo MR imaging at 1.5 T. Radiology. 2009;251:67-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 263]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
18.  Guiu B, Petit JM, Loffroy R, Aho S, Ben Salem D, Masson D, Robin I, Vergès B, Hillon P, Cercueil JP. Liver methylene fraction by dual- and triple-echo gradient-echo imaging at 3.0T: Correlation with proton MR spectroscopy and estimation of robustness after SPIO administration. J Magn Reson Imaging. 2011;33:119-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
19.  Bondarenko AS. Analysis of large experimental datasets in electrochemical impedance spectroscopy. Anal Chim Acta. 2012;743:41-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 48]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
20.  Tous M, Ferré N, Rull A, Marsillach J, Coll B, Alonso-Villaverde C, Camps J, Joven J. Dietary cholesterol and differential monocyte chemoattractant protein-1 gene expression in aorta and liver of apo E-deficient mice. Biochem Biophys Res Commun. 2006;340:1078-1084.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Rull A, Rodríguez F, Aragonès G, Marsillach J, Beltrán R, Alonso-Villaverde C, Camps J, Joven J. Hepatic monocyte chemoattractant protein-1 is upregulated by dietary cholesterol and contributes to liver steatosis. Cytokine. 2009;48:273-279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
22.  Rull A, Vinaixa M, Angel Rodríguez M, Beltrán R, Brezmes J, Cañellas N, Correig X, Joven J. Metabolic phenotyping of genetically modified mice: An NMR metabonomic approach. Biochimie. 2009;91:1053-1057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 18]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
23.  Serkova N, Fuller TF, Klawitter J, Freise CE, Niemann CU. H-NMR-based metabolic signatures of mild and severe ischemia/reperfusion injury in rat kidney transplants. Kidney Int. 2005;67:1142-1151.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Simó JM, Castellano I, Ferré N, Joven J, Camps J. Evaluation of a homogeneous assay for high-density lipoprotein cholesterol: limitations in patients with cardiovascular, renal, and hepatic disorders. Clin Chem. 1998;44:1233-1241.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Marsillach J, Camps J, Ferré N, Beltran R, Rull A, Mackness B, Mackness M, Joven J. Paraoxonase-1 is related to inflammation, fibrosis and PPAR delta in experimental liver disease. BMC Gastroenterol. 2009;9:3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 71]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
26.  Tous M, Ferré N, Camps J, Riu F, Joven J. Feeding apolipoprotein E-knockout mice with cholesterol and fat enriched diets may be a model of non-alcoholic steatohepatitis. Mol Cell Biochem. 2005;268:53-58.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Joven J, Rull A, Rodriguez-Gallego E, Camps J, Riera-Borrull M, Hernández-Aguilera A, Martin-Paredero V, Segura-Carretero A, Micol V, Alonso-Villaverde C. Multifunctional targets of dietary polyphenols in disease: a case for the chemokine network and energy metabolism. Food Chem Toxicol. 2013;51:267-279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
28.  Kral JG, Schaffner F, Pierson RN, Wang J. Body fat topography as an independent predictor of fatty liver. Metabolism. 1993;42:548-551.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Stranges S, Dorn JM, Muti P, Freudenheim JL, Farinaro E, Russell M, Nochajski TH, Trevisan M. Body fat distribution, relative weight, and liver enzyme levels: a population-based study. Hepatology. 2004;39:754-763.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Omagari K, Kadokawa Y, Masuda J, Egawa I, Sawa T, Hazama H, Ohba K, Isomoto H, Mizuta Y, Hayashida K. Fatty liver in non-alcoholic non-overweight Japanese adults: incidence and clinical characteristics. J Gastroenterol Hepatol. 2002;17:1098-1105.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Hernández-Aguilera A, Rull A, Rodríguez-Gallego E, Riera-Borrull M, Luciano-Mateo F, Camps J, Menéndez JA, Joven J. Mitochondrial dysfunction: a basic mechanism in inflammation-related non-communicable diseases and therapeutic opportunities. Mediators Inflamm. 2013;2013:135698.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 102]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
32.  Shulman GI. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. N Engl J Med. 2014;371:1131-1141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 640]  [Cited by in F6Publishing: 650]  [Article Influence: 65.0]  [Reference Citation Analysis (0)]
33.  Samocha-Bonet D, Dixit VD, Kahn CR, Leibel RL, Lin X, Nieuwdorp M, Pietiläinen KH, Rabasa-Lhoret R, Roden M, Scherer PE. Metabolically healthy and unhealthy obese--the 2013 Stock Conference report. Obes Rev. 2014;15:697-708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 129]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
34.  Kramer CK, Zinman B, Retnakaran R. Are metabolically healthy overweight and obesity benign conditions?: A systematic review and meta-analysis. Ann Intern Med. 2013;159:758-769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 662]  [Cited by in F6Publishing: 654]  [Article Influence: 59.5]  [Reference Citation Analysis (0)]
35.  Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, Charlton M, Sanyal AJ. The diagnosis and management of non-alcoholic fatty liver disease: practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology. 2012;55:2005-2023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2413]  [Cited by in F6Publishing: 2449]  [Article Influence: 204.1]  [Reference Citation Analysis (0)]
36.  Schauer PR, Bhatt DL, Kirwan JP, Wolski K, Brethauer SA, Navaneethan SD, Aminian A, Pothier CE, Kim ES, Nissen SE. Bariatric surgery versus intensive medical therapy for diabetes--3-year outcomes. N Engl J Med. 2014;370:2002-2013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1202]  [Cited by in F6Publishing: 1125]  [Article Influence: 112.5]  [Reference Citation Analysis (0)]
37.  Schauer PR, Kashyap SR, Wolski K, Brethauer SA, Kirwan JP, Pothier CE, Thomas S, Abood B, Nissen SE, Bhatt DL. Bariatric surgery versus intensive medical therapy in obese patients with diabetes. N Engl J Med. 2012;366:1567-1576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1639]  [Cited by in F6Publishing: 1509]  [Article Influence: 125.8]  [Reference Citation Analysis (0)]
38.  Masana L, Camprubi M, Sarda P, Sola R, Joven J, Turner PR. The Mediterranean-type diet: is there a need for further modification? Am J Clin Nutr. 1991;53:886-889.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Promrat K, Kleiner DE, Niemeier HM, Jackvony E, Kearns M, Wands JR, Fava JL, Wing RR. Randomized controlled trial testing the effects of weight loss on nonalcoholic steatohepatitis. Hepatology. 2010;51:121-129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 973]  [Cited by in F6Publishing: 884]  [Article Influence: 63.1]  [Reference Citation Analysis (0)]
40.  Giby VG, Ajith TA. Role of adipokines and peroxisome proliferator-activated receptors in nonalcoholic fatty liver disease. World J Hepatol. 2014;6:570-579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 77]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
41.  Randle PJ. Regulatory interactions between lipids and carbohydrates: the glucose fatty acid cycle after 35 years. Diabetes Metab Rev. 1998;14:263-283.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Sanyal AJ. Mechanisms of Disease: pathogenesis of nonalcoholic fatty liver disease. Nat Clin Pract Gastroenterol Hepatol. 2005;2:46-53.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Joven J, Guirro M, Mariné-Casadó R, Rodríguez-Gallego E, Menéndez JA. Autophagy is an inflammation-related defensive mechanism against disease. Adv Exp Med Biol. 2014;824:43-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
44.  Rodríguez-Gallego E, Riera-Borrull M, Hernández-Aguilera A, Mariné-Casadó R, Rull A, Beltrán-Debón R, Luciano-Mateo F, Menendez JA, Vazquez-Martin A, Sirvent JJ. Ubiquitous transgenic overexpression of C-C chemokine ligand 2: a model to assess the combined effect of high energy intake and continuous low-grade inflammation. Mediators Inflamm. 2013;2013:953841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
45.  O’Neill LA, Hardie DG. Metabolism of inflammation limited by AMPK and pseudo-starvation. Nature. 2013;493:346-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 873]  [Cited by in F6Publishing: 836]  [Article Influence: 76.0]  [Reference Citation Analysis (0)]
46.  Kwanten WJ, Martinet W, Michielsen PP, Francque SM. Role of autophagy in the pathophysiology of nonalcoholic fatty liver disease: a controversial issue. World J Gastroenterol. 2014;20:7325-7338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 75]  [Cited by in F6Publishing: 76]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
47.  Zheng YZ, Edwards MC. Expression of resistance to barley stripe mosaic virus in barley and oat protoplasts. J Gen Virol. 1990;71:1865-1868.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
48.  Hue L, Taegtmeyer H. The Randle cycle revisited: a new head for an old hat. Am J Physiol Endocrinol Metab. 2009;297:E578-E591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 482]  [Cited by in F6Publishing: 487]  [Article Influence: 32.5]  [Reference Citation Analysis (0)]
49.  Kitano H. Towards a theory of biological robustness. Mol Syst Biol. 2007;3:137.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Barr J, Caballería J, Martínez-Arranz I, Domínguez-Díez A, Alonso C, Muntané J, Pérez-Cormenzana M, García-Monzón C, Mayo R, Martín-Duce A. Obesity-dependent metabolic signatures associated with nonalcoholic fatty liver disease progression. J Proteome Res. 2012;11:2521-2532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 167]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]