Next Article in Journal
Effect of Myricetin on Lipid Metabolism in Primary Calf Hepatocytes Challenged with Long-Chain Fatty Acids
Previous Article in Journal
Mapping of Urinary Volatile Organic Compounds by a Rapid Analytical Method Using Gas Chromatography Coupled to Ion Mobility Spectrometry (GC–IMS)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nonalcoholic Fatty Liver Disease—A Concise Review of Noninvasive Tests and Biomarkers

1
Internal Medicine Department, Galilee Medical Center, Nahariya 2210001, Israel
2
Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311502, Israel
*
Author to whom correspondence should be addressed.
Metabolites 2022, 12(11), 1073; https://doi.org/10.3390/metabo12111073
Submission received: 3 October 2022 / Revised: 1 November 2022 / Accepted: 2 November 2022 / Published: 5 November 2022
(This article belongs to the Section Endocrinology and Clinical Metabolic Research)

Abstract

:
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide, with a continuously growing prevalence. The pathophysiology of the disease is complex and includes several mechanisms, with metabolic syndrome and insulin resistance playing a major role. It is crucial to diagnose NAFLD before it advances to nonalcoholic steatohepatitis (NASH), which can progress to cirrhosis, presented by its complications which include ascites, portal hypertension, bleeding varices and encephalopathy. Another important complication of NAFLD and cirrhosis is hepatocellular carcinoma (HCC), a cancer with increasing incidence and poor prognosis. Even with the growing prevalence of NAFLD, diagnosis via liver biopsies is unrealistic, considering the costs and complications. Noninvasive tests, including serum biomarkers and elastography, are cost-effective and convenient, thereby replacing liver biopsies in diagnosing and excluding liver fibrosis. However, currently, these noninvasive tests have several limitations, such as variability, inadequate accuracy and risk factors for error. The limitations and variability of these tests comet the investigator to propose combining them in diagnostic algorithms to produce more accurate tools. Identifying patients with significant fibrosis is important for targeted therapies to prevent disease progression. Effective screening using noninvasive tests can be crucial for patient risk stratification and early diagnosis.

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder, with an estimated global prevalence of 25% [1]. NAFLD has a bidirectional correlation with components of the metabolic syndrome. It often occurs together with type 2 diabetes, obesity, dyslipidemia, and hypertension, which constitute cardio-metabolic disease [1,2,3,4]. This led a panel of experts in 2020 to suggest that the nomenclature of NAFLD should be updated to metabolic-associated fatty liver disease (MAFLD) [3]. Patients with NAFLD can progress to the more severe form of non-alcoholic steatohepatitis (NASH). At this stage, the hepatocyte damage may lead to fibrosis and progress to cirrhosis in 20% of patients with NASH [5,6,7]. The exact mechanisms leading to NAFLD are not fully understood, but insulin resistance seems to play an important role [4].
NAFLD is defined by the presence of >5% of hepatic steatosis demonstrated, either radiographically or histologically, in the absence of significant alcohol consumption. The definitive diagnosis of NASH requires a liver biopsy with the histologic demonstration of >5% of hepatic steatosis in addition to evidence of hepatocyte inflammation in the form of ballooning degeneration [6,7]. Liver biopsy is considered the gold standard for the diagnosis of the global status of fibrosis of nonalcoholic fatty liver disease. Nevertheless, biopsies are invasive and carry the risk of complications and sampling errors, making biopsies impractical in being applied on a population level [8]. This has led to continuous searching for noninvasive biomarkers for diagnosing and staging liver fibrosis, especially in high-risk patients who benefit the most from early interventions to prevent progression to cirrhosis [9]. Among the imaging biomarkers used are Fibroscan and magnetic resonance imaging (MRI) [10]. Serum biomarkers include cytokeratin-18 (CK-18), a biomarker of apoptosis in NASH, liver enzymes and Pro-C3, a biomarker of fibrosis [11].
The aim of the study is to review the noninvasive tests used in the diagnosis and follow-up of patients with NAFLD.

2. NAFLD Represents a Wide Spectrum of Diseases

In recent decades, NAFLD prevalence has increased into pandemic dimensions, affecting about one-third of western populations, probably due to the increasing prevalence of obesity and metabolic disease [12,13]. NAFLD represents a spectrum of diseases ranging from simple steatosis to NASH and, finally, cirrhosis. Although approximately only 10–20% of patients with NAFLD progress to NASH, and 15–20% of patients with NASH progress to cirrhosis, the absolute numbers are not negligible due to the wide prevalence of NAFLD [14]. The complications of NAFLD can be divided into hepatic and extra-hepatic. The hepatic complications of NAFLD include progression to cirrhosis. Studies have shown that the progression from steatosis to fibrosis can take 14 years, while the progression from NASH to cirrhosis may take 7 years [15]. Decompensated cirrhosis which can be manifested as hepatic encephalopathy, ascites or gastrointestinal bleeding secondary to esophageal varices, occurs annually at a rate of 3–4% in patients with compensated cirrhosis [16]. A meta-analysis of 13 studies showed higher mortality rates in patients with cirrhosis than in patients without fibrosis [17]. Hepatocellular carcinoma (HCC) is another serious complication of NAFLD, with poor outcomes and limited therapeutic options. The majority of NAFLD-related HCC cases occur in the setting of cirrhosis. However, about 20% of the cases occur in non-cirrhotic livers. An important risk factor for developing HCC is the presence of diabetes and metabolic syndrome [18,19]. A recent study revealed that the lack of fibroblast growth factor 21 (FGF21), which has anti-inflammatory effects, enhances the progression from NASH to HCC [20]. In addition to HCC, there is a higher incidence of extra-hepatic malignancies, particularly colorectal and uterine cancers, in which the risk is increased twofold [21]. It is not surprising that cardiovascular risk is increased in the presence of NAFLD since both of them share many risk factors, including dyslipidemia, obesity, hypertension and diabetes. A key factor in the development of both diseases is insulin resistance which contributes to the lack of lipolysis suppression, thereby increasing free fatty acids (FFA) in the circulation, ending up accumulating in the liver and blood vessels [22,23,24]. Cardiovascular complications are the most common cause of death in NAFLD, accounting for nearly 40% of the cases [25]. NAFLD is a risk factor for severe disease and liver injury [26,27].

3. Serum Biomarkers of NASH

Since NASH is currently the target of pharmacological treatment, it is important to develop new serum biomarkers for NAFLD and NASH diagnosis, to aid in identifying high-risk patients who require further follow-up and treatment.

3.1. Alanine Aminotransferase (ALT)

ALT levels are usually elevated in patients with NAFLD. The mechanism of ALT elevation is thought to be associated with lipotoxicity and increased hepatic triglyceride content [6]. However, no correlation was found between the degree of ALT elevation and the severity of fibrosis or liver inflammation [6,28]. Liver biopsies of patients with elevated ALT revealed similar rates of advanced fibrosis as for patients with normal ALT levels. Therefore, ALT levels are poor biomarkers in assessing the degree of disease progression and should not be used to guide clinicians in deciding to perform liver biopsies [29].

3.2. PRO-C3

Liver fibrosis is characterized by the accumulation of collagen proteins. Fragments of collagen, called propeptides, are released during the process of fibrosis and may be useful as biomarkers of fibrotic tissue formation in the liver [14]. The N-terminal propeptide of type 3 procollagen (PRO-C3) is released by protease A disintegrin and metalloproteinase with thrombospondin motifs 2 (ADAMTS-2) [30,31]. Serum concentrations of N-protease cleaved PIIINP neo-epitope (PRO-C3) are associated with the degree of liver fibrosis and can be used to monitor the response to treatment with antifibrotic agents [32,33]. A recent study measured PRO-C3 levels in patients with severe obesity before and after bariatric surgery. Furthermore, they compared liver histology from biopsies obtained during the surgery. The study found that PRO-C3 levels were associated with advanced liver fibrosis in patients with severe obesity. The levels of PRO-C3 decreased after the bariatric surgery, and the decrease was correlated with improvement in metabolic and liver parameters [34].
A PRO-C3-based score, ADAPT (age, presence of diabetes, Pro-c3 and platelet count), which includes clinical and metabolic parameters, can predict the presence or absence of fibrosis in NAFLD and was shown to have high specificity and NPV. Recent studies have shown that the combination of ADAPT with liver stiffness measurements (LSM) can be reliably used to exclude advanced fibrosis in low-risk populations [33,35,36].

3.3. Cytokeratin-18 (CK-18)

CK-18 is an intermediate filament protein found in liver hepatocytes. It is cleaved and released from hepatocytes during apoptosis [37,38,39]. Therefore its serum concentration correlates with the degree of hepatocyte damage occurring during histological changes of NASH and NAFLD [20,21,22]. CK-18 concentrations are a marker of disease severity in NASH [40]. CK-18 was detected by ELISA, but a recent study suggests the use of new monoclonal antibodies raised against CK-18, which are more specific than the ELISA, in which the cut-off values were imprecise [41].

4. Noninvasive Fibrosis Scores in NAFLD

Noninvasive tests (NITs) can be used to exclude advanced fibrosis and to aid in risk stratification and specialist referral. NITs are considered ideal when they provide similar information to a biopsy, but none of these tests currently reach the accuracy of a liver biopsy. NITs of liver fibrosis have been suggested to be less accurate in type 2 diabetes mellitus (T2DM). A recent study by Boursier et al. compared the accuracy of NIT between 1051 NAFLD patients with and without T2DM. The study concluded that there is decreased accuracy of NIT in T2DM, which is partially attributed to the fact that T2DM modifies the level of some NAFLD biomarkers [42]. On the other hand, liver fibrosis biomarkers were found to be correlated positively with cardiovascular risk (CVR) scores. High CVR scores tend to be higher among patients with advanced fibrosis, supporting the association between liver fibrosis and cardiovascular risk [43].

4.1. Serum Scores

4.1.1. Fibrosis-4 (FIB-4)

FIB-4 is a score used to assess liver fibrosis based on easily available and inexpensive parameters. The formula includes age, platelet count, aspartate transaminase (AST) and alanine transaminase (ALT) levels. FIB-4 was initially developed to assess liver fibrosis in patients who were co-infected with the human immunodeficiency virus (HIV) and hepatitis C virus (HCV) [44]. Shah et al. tested and validated its use in patients with NAFLD, in addition to comparing its performance to other markers of fibrosis [8,45]. A cut-off value of ≤1.3 can exclude advanced fibrosis in 90% of patients with NAFLD in primary clinics (high NPV). Values higher than 1.3 require additional workup in referral centers [46]. A study by Yun Hwa Roh et al. found that cut-off values of 2.68 showed acceptable PPV in predicting advanced fibrosis; the incorporation of this value with sonographic results increased the diagnostic accuracy of ruling in patients with advanced fibrosis [44]. In accordance with this finding, FIB-4 was also used for the diagnosis of fibrosis and not only for ruling it out. Despite the wide use of this score in excluding fibrosis, several limitations should be considered, such as its reduced specificity in older aged patients and the need for an adjusted threshold in patients aged ≥65, and that this score was developed and tested in populations with a higher prevalence of advanced fibrosis and not as a first-line screening tool [7,47].
Tsung-Po Chen et al. emphasize that fatty liver is prevalent in the ambulatory elderly. Age is a risk factor for advanced fibrosis, with the disease likely progressing from a steatotic to a fibrotic picture with age. However, there is no significant association between high-risk FIB-4 and BMI [48].

4.1.2. Aspartate Aminotransferase/Platelet Ratio Index (APRI)

APRI is calculated as [(AST/ULN)/platelet count (109/L)] × 100. A cohort study of 145 patients with NAFLD showed that at the cutoff value of 1.0, APRI had a low sensitivity of 27% but a specificity of 89% [49]. This score has an excellent NPV for ruling out advanced fibrosis. Intermediate and high scores require further investigation, including liver biopsy [50]. A recent study by Yin-Lian Wu et al. (2021) evaluated the diagnostic performance of noninvasive scores, including APRI, in MAFLD. The definition of MAFLD was different from NAFLD and required no exclusion of other chronic liver diseases. Instead, MAFLD was defined as hepatic steatosis in the presence of metabolic disease. The NPV of APRI did not exceed 80%, and the PPV was as low as 50% at any cutoff value tested. These results led to the conclusion that APRI should not be used to evaluate advanced fibrosis in MAFLD [51].

4.1.3. The NAFLD Fibrosis Score (NFS)

NFS consists of diabetes/impaired fasting glucose, age, AST, ALT, platelets, BMI and albumin [52]. According to a recent meta-analysis that compared the performance of several scores in diagnosing advanced fibrosis, the AUROC for NFS was 0.84. NFS has a high NPV (>90%) for excluding advanced fibrosis [53,54].

4.1.4. Enhanced Liver Fibrosis (ELF)

ELF is a blood test that measures three markers of liver fibrosis: hyaluronic acid (HA), procollagen III amino-terminal peptide (PIIINP), and tissue inhibitor of matrix metalloproteinase 1 (TIMP-1) [55]. The cutoff value for diagnosing advanced fibrosis is ≥9.8, which demonstrated high specificity of >90% in diagnosing advanced fibrosis [56,57].

4.1.5. The Fatty Liver Index (FLI)

FLI was suggested in 2006 by Bedogni et al. to aid physicians in selecting patients with suspected NAFLD to proceed to liver ultrasonography to confirm steatosis [58]. FLI is based on body mass index (BMI), waist circumference, triglycerides and gamma-glutamyltransferase (GGT). FLI can be calculated using many available online calculators. The results range from 0 to 100. FLI < 30 was ruled out, and FLI ≥ 60 ruled in hepatic steatosis [59].

Elastography

Elastography is a noninvasive imaging modality used to evaluate the degree of liver fibrosis [60]. Liver fibrosis or stiffness is the excessive accumulation of extracellular matrix proteins, including collagen, that occurs in most types of chronic liver diseases. The principle of elastography involves the transmission of acoustic waves through the liver parenchyma. The velocity of wave propagation within the parenchyma reflects the degree of stiffness or, in other words, the degree of fibrosis.

Transient Elastography (TE) (Fibroscan)

Using a special probe, Fibroscan can provide two parameters, the degree of steatosis measured by the controlled attenuation parameter (CAP) and the degree of fibrosis reflected by the liver stiffness measurement [61]. The liver volume measured by Fibroscan is 100 times bigger than a biopsy sample, thereby providing more precise information about the liver parenchyma [62].
Transient elastography (TE) < 10 kPa can rule out compensated advanced chronic liver disease (cACLD). Values between 10 and 15 kPa are suggestive of cACLD. Values > 15 kPa are highly suggestive of cACLD. Patients with chronic liver disease and an LSM < 10 kPa by TE have a negligible 3-year risk (<1%) of decompensation and liver-related death. A rule of 5 for LSM by TE (10–15–20–25 kPa) should be used to denote progressively higher relative risks of decompensation and liver-related death independently of the etiology of chronic liver disease (Table 1) [63].

Ultrasonographic Fatty Liver Index (US-FLI)

US-FLI was proposed by Ballestri et al. in 2012. The diagnosis of fatty liver was based on several ultrasonographic parameters, including the presence of liver-kidney contrast, graded as mild/moderate (score 2) and severe (score 3). Additional criteria include the presence (score 1 each) or absence (score 0 each) of posterior attenuation of an ultrasound beam, vessel blurring, difficult visualization of the gallbladder wall, difficult visualization of the diaphragm and areas of focal sparing. The US-FLI score ranges from 2 to 8. US-FLI ≥ 2 showed a good diagnostic performance to detect the minimum steatosis amount of 10% on histology with high sensitivity (90.1%) and specificity (90%) [64]. A study performed later by Xavier et al. demonstrated the significant superiority of US-FLI in discriminating between different grades of steatosis compared to the FLI score [65].

5. Metabolic Profile of NAFLD

Much research on NAFLD has been done, but no accurate mechanism has been described yet. A good understanding of the metabolic and biological mechanisms of this situation could help us in developing good predictors and markers.
Several theories were suggested in the context of NASH development. The “double-hit” hypothesis suggests that the first insult or the “first hit” is characterized by lipid accumulation in the liver, resulting in steatosis, which enhances the liver’s susceptibility to the “second hit” characterized by inflammation and necrosis induced by oxidative stress and proinflammatory mediators [66,67]. The simplistic “double-hit” theory was substituted by a more complex theory, the “multiple-hit” hypothesis, which has become more accepted recently, representing the interaction of multiple metabolic and genetic factors leading to the development of NASH. Such “hits” include insulin resistance, hormones secreted from the adipose tissue, nutritional factors, gut microbiota and genetic and epigenetic factors [68].

5.1. The Role of Adipose Tissue in NAFLD

Obesity is associated with increased prevalence and severity of NAFLD. The adipose tissue functions as an endocrine organ secreting adipokines and cytokines [69]. Adiponectin is anti-inflammatory, and its levels are inversely associated with cardiovascular risk factors and visceral adiposity. It increases free fatty acid (FFA) oxidation and decreases gluconeogenesis resulting in decreased liver steatosis. Furthermore, it acts as a pro-inflammatory inhibitor, opposing the effects of tumor necrosis factor (TNF)-α and interleukin (IL)-6 [66,70,71]. Leptin secretion is directly proportional to the white fat mass. Circulating leptin levels are higher in patients with NAFLD than in healthy patients, and higher levels are associated with the severity [72]. Leptin has different effects depending on the stage of the disease. At early stages, it acts as an anti-steatotic and increases insulin sensitivity. At more advanced NAFLD stages, it has an unfavorable effect by promoting inflammation [73,74]. Pro-inflammatory cytokines are immunoregulatory cytokines that favor and enhance the process of inflammation [75]. Fat accumulation in the liver mediates cytokine production in hepatic cells. These cytokines induce inflammation, necrosis, cell apoptosis and mediated liver stiffness and fibrosis. Pro-inflammatory cytokines are also secreted by adipose tissue and are involved in the recruitment and activation of macrophages, a process that leads to chronic low-grade inflammation and the development of insulin resistance and cardiovascular diseases [73,74,75,76,77].

5.2. Lipotoxicity in NAFLD

Lipotoxicity is a leading factor in the development and progression of NAFLD and NASH. The total amount of triglycerides stored in the liver cells is not the main determinant of lipotoxicity [76,77]. Specific lipid classes, especially free fatty acids (FFA), which accumulate in hepatocyte cytoplasm, act as damaging agents via multiple mechanisms. These include activation of death receptors, modification of mitochondrial function and oxidative stress, which lead to the progression from NAFLD to NASH, its inflammatory form. Insulin resistance plays a major role in lipotoxicity by enhancing lipolysis in adipose tissue and increasing the amount of circulation FFAs [76,77].

5.3. The Role of Glucose in NAFLD

High-carbohydrate diets and NAFLD are strongly correlated [78]. Hyperglycemia is strongly associated with oxidative stress [79], leading to chronic inflammation and insulin resistance [80]. Insulin resistance in NAFLD is characterized by reductions in whole-body, hepatic, and adipose tissue insulin sensitivity. The mechanisms underlying the accumulation of fat in the liver may include excess dietary fat, increased delivery of free fatty acids to the liver, inadequate fatty acid oxidation, and increased de novo lipogenesis. Insulin resistance may enhance hepatic fat accumulation by increasing free fatty acid delivery and by hyperinsulinemia to stimulate anabolic processes. The impact of weight loss, metformin, and thiazolidinedione, all treatments aimed at improving insulin sensitivity, as well as other agents such as vitamin E, have been evaluated in patients with NAFLD and have shown some benefit. However, most intervention studies have been small and uncontrolled [81].

6. Risk Stratification

In light of the increased morbidity and mortality in NAFLD, and considering its wide prevalence, many efforts are put into identifying patients at risk for disease progression to enable early intervention and to prevent the development of cirrhosis [82].
Several algorithms were developed to screen NAFLD patients, each relying on different parameters (Figure 1) [83]. The European Association for Liver/-Diabetes/-Obesity Guidelines (EASL-EASD-EASO) uses the NFS and FIB-4 scores, while the German National Guidelines use LSM to guide the management of NAFLD patients [82].
NAFLD has almost reached epidemic proportions worldwide. Wide dissemination of current concepts on NAFLD and extensive collaboration between physicians, governments, non-governmental organizations, and the pharmaceutical industry is urgently needed to advance a NAFLD public health policies agenda that allows us to address this disease more holistically in a society-wide manner [80].

7. Future Perspective (Where the Field Should Go to Identify NAFLD)

Metabolomics and lipidomics studies help clinicians identify biomarkers associated with the pathophysiology of NAFLD and NASH. Impairment in amino acid metabolism in NAFLD is linked to insulin resistance and results in higher fasting concentrations of essential amino acids, as has also been observed in obesity. In patients with chronic liver disease, phenylalanine and its metabolite tyrosine are frequently found to be decreased, whereas BCAAs, especially leucine, isoleucine, valine and glutamate-glutamine are increased, mainly because of insulin resistance. However, as liver disease progresses, the opposite is often observed, with high amino acids (AAAs) and reduced BCAAs, especially in patients with chronic hepatic insufficiency. In addition, a reduction in hepatic glutathione levels and elevation of methionine levels has been associated with liver damage and the progression of NAFLD to NASH [82]. Another important observation is related to alterations in lipid metabolism. NASH is strongly associated with alterations in circulating fatty acid flux and intact lipids such as triglycerides and phospholipids, which is partially due to alterations in liver de novo lipogenesis (DNL), lipolysis rate and VLDL metabolism. Elevation in peripheral fatty acid flux and triglycerides containing fatty acids with low carbon number and double bonds, as well as a reduction in the levels of triglycerides containing polyunsaturated fatty acids (PUFAs), including ω-3 and ω-6 fatty acids, have been observed in NAFLD. Interestingly, this pattern of changes in triglycerides has also been reported in metabolic disorders, especially those associated with insulin resistance. Moreover, several studies have shown an increase in total bile acids in both liver and plasma of patients with NASH. In particular, a significant increase in fasting plasma concentration of bile acids, including glycocholic, taurocholic and taurochenodeoxycholic acid, in patients with NASH has been observed [84]. In order to derive robust metabolic signatures of NAFLD–NASH, the reported biomarkers will need to be consolidated and validated. Current efforts to improve metabolomics and lipidomics workflows are expected to improve our ability to effectively bridge the biomarker discovery stage, validation studies and translation to the clinic. Another area that is likely to lead to substantial advances in NASH biomarkers is the investigation of multiomic biomarkers. The gut microbiome is also a rich source of circulating metabolite biomarkers. Application of genome-scale metabolic modeling on shotgun metagenomics data from stool samples can predict which metabolites are being produced in specific diseases or physiological conditions. Such predictions can be validated by stool and serum metabolomics. Adding different layers of omics data is thus likely to lead to both improved sensitivity as well as specificity of NAFLD–NASH biomarkers [85].
Insulin resistance is the earliest detectable defect in the metabolic continuum leading to type 2 diabetes. Impaired glucose homeostasis is associated with obesity. This situation increased plasma-free fatty acids and ectopic lipids, which are connected to peripheral and hepatic insulin resistance. Branched-chain amino acids (BCAAs) are important nutrient signals that affect metabolism. Increased plasma levels of BCAAs are associated with a high risk of developing metabolic syndrome and insulin resistance. The significant down-regulation of genes related to BCAA catabolism and mitochondrial energy metabolism increased the expression of inflammation-related genes [84]. The branched-chain amino acids and microarray assay-detecting transcripts of these genes could be future tools for identifying NAFLD.
The FibroScan-AST (FAST) score provides an efficient way to non-invasively identify patients at risk of progressive NASH for clinical trials or treatments when they become available, thereby reducing unnecessary liver biopsy in patients unlikely to have significant disease. FAST score performance is good across the full range of validation cohorts. AUROC ranged from 0.74 to 0.95, with PPV up to 0.85 and NPV ranging from 0.73 to 1 using the dual cutoffs approach, with cutoffs derived in the derivation cohort [86].

8. Conclusions

NAFLD is a multi-system disease with increasing prevalence. Early diagnosis contributes greatly to risk stratification and management, preventing the disease’s complications. Much research was done to develop and improve noninvasive tests which can replace liver biopsies and guide the management of NAFLD patients. Combining several tests and scores and creating charts for risk stratification and management helps the primary physician in managing such patients and in referring them to specialized centers.
However, more research needs to be done to compare the best test combinations to increase the sensitivity of the diagnosis.

Author Contributions

Conceptualization, T.B. and N.A.; software, T.B.; validation, N.A., M.B. (Mariana Boulos) and M.B. (Maamoun Basheer); formal analysis, N.A.; investigation, T.B.; resources, T.B.; data curation, T.B.; writing—original draft preparation, T.B.; writing—review and editing, N.A.; visualization, M.B. (Mariana Boulos); supervision, N.A.; project administration, T.B.; funding acquisition, N.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Younossi, Z.M.; Koenig, A.B.; 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, 73–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Eslam, M.; Sanyal, A.J.; George, J.; on behalf of theInternational Consensus Panel. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014.e1991. [Google Scholar] [CrossRef]
  3. Tarantino, G.; Finelli, C. What about non-alcoholic fatty liver disease as a new criterion to define metabolic syndrome? World J. Gastroenterol. 2013, 19, 3375–3384. [Google Scholar] [CrossRef] [PubMed]
  4. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef] [PubMed]
  5. McGlinchey, A.J.; Govaere, O.; Geng, D.; Ratziu, V.; Allison, M.; Bousier, J.; Petta, S.; de Oliviera, C.; Bugianesi, E.; Schattenberg, J.M.; et al. Metabolic signatures across the full spectrum of non- alcoholic fatty liver disease. JHEP Rep. 2022, 4, 100477. [Google Scholar] [CrossRef] [PubMed]
  6. Sheka, A.C.; Adeyi, O.; Thompson, J.; Hameed, B.; Crawford, P.A.; Ikramuddin, S. Nonalcoholic steatohepatitis: A review. JAMA 2020, 323, 1175–1183. [Google Scholar] [CrossRef]
  7. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The Diagnosis and Management of Nonalcoholic Fatty Liver Disease: Practice Guidance From the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef] [Green Version]
  8. Piazzolla, V.A.; Mangia, A. Noninvasive Diagnosis of NAFLD and NASH. Cells 2020, 9, 1005. [Google Scholar] [CrossRef]
  9. Kanwal, F.; Shubrook, J.H.; Adams, L.A.; Pfotenhauer, K.; Wong, V.W.-S.; Wright, E.; Abdelmalek, M.F.; Harrison, S.A.; Loomba, R.; Mantzoros, C.S.; et al. Clinical Care Pathway for the Risk Stratification and Management of Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology 2021, 161, 1657–1669. [Google Scholar] [CrossRef]
  10. Ajmera, V.; Loomba, R. Imaging biomarkers of NAFLD, NASH, and fibrosis. Mol. Metab. 2021, 50, 101167. [Google Scholar] [CrossRef]
  11. Di Mauro, S.; Scamporrino, A.; Filippello, A.; Di Pino, A.; Scicali, R.; Malaguarnera, R.; Purrello, F.; Piro, S. Clinical and Molecular Biomarkers for Diagnosis and Staging of NAFLD. Int. J. Mol. Sci. 2021, 22, 11905. [Google Scholar] [CrossRef]
  12. Targher, G.; Corey, K.E.; Byrne, C.D.; Roden, M. The complex link between NAFLD and type 2 diabetes mellitus—Mechanisms and treatments. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 599–612. [Google Scholar] [CrossRef]
  13. Juanola, O.; Martínez-López, S.; Francés, R.; Gómez-Hurtado, I. Non-alcoholic fatty liver disease: Metabolic, genetic, epigenetic and environmental risk factors. Int. J. Environ. Res. Public Health 2021, 18, 5227. [Google Scholar] [CrossRef] [PubMed]
  14. Cardoso, A.C.; de Figueiredo-Mendes, C.; Villela-Nogueira, C.A. Current management of NAFLD/NASH. Liver Int. 2021, 41, 89–94. [Google Scholar] [CrossRef] [PubMed]
  15. Armandi, A.; Bugianesi, E. Natural history of NASH. Liver Int. 2021, 41 (Suppl. 1), 78–82. [Google Scholar] [CrossRef] [PubMed]
  16. Powell, E.E.; Wong, V.W.; Rinella, M. Non-alcoholic fatty liver disease. Lancet 2021, 397, 2212–2224. [Google Scholar] [CrossRef]
  17. Taylor, R.S.; Taylor, R.J.; Bayliss, S.; Hagström, H.; Nasr, P.; Schattenberg, J.M.; Ishigami, M.; Toyoda, H.; Wong, V.W.-S.; Peleg, N.; et al. Association Between Fibrosis Stage and Outcomes of Patients With Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Gastroenterology 2020, 158, 1611–1625.e12. [Google Scholar] [CrossRef] [Green Version]
  18. Ioannou, G.N. Epidemiology and risk-stratification of NAFLD-associated HCC. J. Hepatol. 2021, 75, 1476–1484. [Google Scholar] [CrossRef]
  19. Calzadilla Bertot, L.; Adams, L.A. The Natural Course of Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2016, 17, 774. [Google Scholar] [CrossRef] [Green Version]
  20. Zheng, Q.; Martin, R.C.; Shi, X.; Pandit, H.; Yu, Y.; Liu, X.; Guo, W.; Tan, M.; Bai, O.; Meng, X.; et al. Lack of FGF21 promotes NASH-HCC transition via hepatocyte-TLR4-IL-17A signaling. Theranostics 2020, 10, 9923–9936. [Google Scholar] [CrossRef]
  21. Allen, A.M.; Hicks, S.B.; Mara, K.C.; Larson, J.J.; Therneau, T.M. The risk of incident extrahepatic cancers is higher in non-alcoholic fatty liver disease than obesity—A longitudinal cohort study. J. Hepatol. 2019, 71, 1229–1236. [Google Scholar] [CrossRef] [PubMed]
  22. Lombardi, R.; Iuculano, F.; Pallini, G.; Fargion, S.; Fracanzani, A.L. Nutrients, Genetic Factors, and Their Interaction in Non-Alcoholic Fatty Liver Disease and Cardiovascular Disease. Int. J. Mol. Sci. 2020, 21, 8761. [Google Scholar] [CrossRef] [PubMed]
  23. Sakurai, Y.; Kubota, N.; Yamauchi, T.; Kadowaki, T. Role of Insulin Resistance in MAFLD. Int. J. Mol. Sci. 2021, 22, 4156. [Google Scholar] [CrossRef]
  24. Watt, M.J.; Miotto, P.M.; De Nardo, W.; Montgomery, M. The Liver as an Endocrine Organ—Linking NAFLD and Insulin Resistance. Endocr. Rev. 2019, 40, 1367–1393. [Google Scholar] [CrossRef] [PubMed]
  25. Maurice, J.; Manousou, P. Non-alcoholic fatty liver disease. Clin. Med. 2018, 18, 245–250. [Google Scholar] [CrossRef] [Green Version]
  26. Huang, R.; Zhu, L.; Wang, J.; Xue, L.; Liu, L.; Yan, X.; Huang, S.; Li, Y.; Yan, X.; Zhang, B.; et al. Clinical features of COVID-19 patients with non-alcoholic fatty liver disease. Hepatol. Commun. 2020, 4, 1758–1768. [Google Scholar] [CrossRef] [PubMed]
  27. Ji, D.; Qin, E.; Xu, J.; Zhang, D.; Cheng, G.; Wang, Y.; Lau, G. Non-alcoholic fatty liver diseases in patients with COVID-19: A retrospective study. J. Hepatol. 2020, 73, 451–453. [Google Scholar] [CrossRef] [PubMed]
  28. Maximos, M.; Bril, F.; Portillo Sanchez, P.; Lomonaco, R.; Orsak, B.; Biernacki, D.; Suman, A.; Weber, M.; Cusi, K. The role of liver fat and insulin resistance as determinants of plasma aminotransferase elevation in nonalcoholic fatty liver disease. Hepatology 2015, 61, 153–160. [Google Scholar] [CrossRef]
  29. Verma, S.; Jensen, D.; Hart, J.; Mohanty, S.R. Predictive value of ALT levels for non-alcoholic steatohepatitis (NASH) and advanced fibrosis in non-alcoholic fatty liver disease (NAFLD). Liver Int. 2013, 33, 1398–1405. [Google Scholar] [CrossRef]
  30. Frei, A.; Zimmermann, A.; Weigand, K. The N-Terminal Propeptide of Collagen Type III in Serum Reflects Activity and Degree of Fibrosis in Patients with Chronic Liver Disease. Hepatology 1984, 4, 830–834. [Google Scholar] [CrossRef]
  31. Karsdal, M.A.; Hjuler, S.T.; Luo, Y.I.; Rasmussen, D.G.; Nielsen, M.J.; Holm Nielsen, S.; Leeming, D.J.; Goodman, Z.; Arch, R.H.; Patel, K.; et al. Assessment of liver fibrosis progression and regression by a serological collagen turnover profile. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 316, G25–G31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Le Goff, C.; Somerville, R.P.; Kesteloot, F.; Powell, K.; Birk, D.E.; Colige, A.C.; Apte, S.S. Regulation of Procollagen Amino-Propeptide Processing during Mouse Embryogenesis by Specialization of Homologous ADAMTS Proteases: Insights on Collagen Biosynthesis and Dermatosparaxis. Development 2006, 133, 1587–1596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Daniels, S.J.; Leeming, D.J.; Eslam, M.; Hashem, A.M.; Nielsen, M.J.; Krag, A.; Karsdal, M.A.; Grove, J.I.; Neil Guha, I.; Kawaguchi, T.; et al. ADAPT: An Algorithm Incorporating PRO-C3 Accurately Identifies Patients With NAFLD and Advanced Fibrosis. Hepatology 2019, 69, 1075–1086. [Google Scholar] [CrossRef] [Green Version]
  34. Bel Lassen, P.; Nori, N.; Bedossa, P.; Genser, L.; Aron-Wisnewsky, J.; Poitou, C.; Surabattula, R.; Juul Nielsen, M.; Asser Karsdal, M.; Julie Leeming, D.; et al. Fibrogenesis Marker PRO-C3 Is Higher in Advanced Liver Fibrosis and Improves in Patients Undergoing Bariatric Surgery. J. Clin. Endocrinol. Metab. 2022, 107, e1356–e1366. [Google Scholar] [CrossRef] [PubMed]
  35. Karsdal, M.A.; Henriksen, K.; Nielsen, M.J.; Byrjalsen, I.; Leeming, D.J.; Gardner, S.; Goodman, Z.; Patel, K.; Krag, A.; Christiansen, C.; et al. Fibrogenesis assessed by serological type III collagen formation identifies patients with progressive liver fibrosis and responders to a potential antifibrotic therapy. Am. J. Physiol. Liver Physiol. 2016, 311, G1009–G1017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Eslam, M.; Wong, G.L.-H.; Hashem, A.M.; Chan, H.L.-Y.; Nielsen, M.J.; Leeming, D.J. A sequential algorithm combining ADAPT and liver stiffness can stage metabolic-associated fatty liver disease in hospital-based and primary care patients. Am. J. Gastroenterol. 2021, 116, 984–993. [Google Scholar] [CrossRef]
  37. Tsai, E.; Lee, T. Diagnosis And Evaluation Of Nonalcoholic Fatty Liver Disease/Nonalcoholic Steatohepatitis, Including Noninvasive Biomarkers And Transient Elastography. Clin. Liver Dis. 2018, 22, 73–92. [Google Scholar] [CrossRef]
  38. Lee, J.; Vali, Y.; Boursier, J.; Duffin, K.; Verheij, J.; Brosnan, M.J.; Zwinderman, K.; Anstee, Q.M.; Bossuyt, P.M.; Zafarmand, M.H. Accuracy of cytokeratin 18 (M30 and M65) in detecting non-alcoholic steatohepatitis and fibrosis: A systematic review and meta-analysis. PLoS ONE 2020, 15, e0238717. [Google Scholar]
  39. Chang, Y.-H.; Lin, H.-C.; Hwu, D.-W.; Chang, D.-M.; Lin, K.-C.; Lee, Y.-J. Elevated serum cytokeratin-18 concentration in patients with type 2 diabetes mellitus and non-alcoholic fatty liver disease. Ann. Clin. Biochem. Int. J. Lab. Med. 2018, 56, 141–147. [Google Scholar] [CrossRef]
  40. Zhou, J.-H.; Cai, J.-J.; She, Z.-G.; Li, H.-L. Noninvasive evaluation of nonalcoholic fatty liver disease: Current evidence and practice. World J. Gastroenterol. 2019, 25, 1307–1326. [Google Scholar] [CrossRef]
  41. Eguchi, A.; Iwasa, M.; Yamada, M.; Tamai, Y.; Shigefuku, R.; Hasegawa, H.; Hasegawa, H.; Hirokawa, Y.; Hayashi, A.; Okuno, K.; et al. A new detection system for serum fragmented cytokeratin 18 as a biomarker reflecting histologic activities of human nonalcoholic steatohepatitis. Hepatol. Commun. 2021, 13, e17622. [Google Scholar] [CrossRef] [PubMed]
  42. Boursier, J.; Canivet, C.M.; Costentin, C.; Lannes, A.; Delamarre, A.; Sturm, N.; Bail, B.L.; Michalak, S.; Oberti, F.; Hilleret, M.N.; et al. Impact of Type 2 Diabetes on the Accuracy of Noninvasive Tests of Liver Fibrosis With Resulting Clinical Implications. Clin Gastroenterol Hepatol. 2022; in press. [Google Scholar]
  43. Ballestri, S.; Mantovani, A.; Baldelli, E.; Lugari, S.; Maurantonio, M.; Nascimbeni, F.; Marrazzo, A.; Romagnoli, D.; Targher, G.; Lonardo, A. Liver Fibrosis Biomarkers Accurately Exclude Advanced Fibrosis and Are Associated with Higher Cardiovascular Risk Scores in Patients with NAFLD or Viral Chronic Liver Disease. Diagnostics 2021, 11, 98. [Google Scholar] [CrossRef] [PubMed]
  44. Roh, Y.H.; Kang, B.-K.; Jun, D.W.; Lee, C.-M.; Kim, M. Role of FIB-4 for reassessment of hepatic fibrosis burden in referral center. Sci. Rep. 2021, 11, 13616. [Google Scholar] [CrossRef]
  45. Shah, A.G.; Lydecker, A.; Murray, K.; Tetri, B.N.; Contos, M.J.; Sanyal, A.J. Comparison of noninvasive markers of fibrosis in patients with nonalcoholic fatty liver disease. Clin. Gastroenterol. Hepatol. 2009, 7, 1104–1112. [Google Scholar] [CrossRef] [Green Version]
  46. Castera, L.; Friedrich-Rust, M.; Loomba, R. Noninvasive Assessment of Liver Disease in Patients with Nonalcoholic Fatty Liver Disease. Gastroenterology 2019, 156, 1264–1281. [Google Scholar] [CrossRef] [Green Version]
  47. McPherson, S.; Hardy, T.; Dufour, J.F.; Petta, S.; Romero-Gomez, M.; Allison, M. Age as a confounding factor for the accurate non-invasive diagnosis of advanced NAFLD fibrosis. Am. J. Gastroenterol. 2017, 112, 740–751. [Google Scholar] [CrossRef] [Green Version]
  48. Chen, T.; Lai, M.; Lin, W.; Huang, K.; Yang, K. Metabolic profiles and fibrosis of nonalcoholic fatty liver disease in the elderly: A community-based study. J. Gastroenterol. Hepatol. 2020, 35, 1636–1643. [Google Scholar] [CrossRef]
  49. McPherson, S.; Stewart, S.F.; Henderson, E.; Burt, A.D.; Day, C.P. Simple non-invasive fibrosis scoring systems can reliably exclude advanced fibrosis in patients with non-alcoholic fatty liver disease. Gut 2010, 59, 1265–1269. [Google Scholar] [CrossRef] [Green Version]
  50. Lai, M.; Afdhal, N.H. Liver Fibrosis Determination. Gastroenterol. Clin. N. Am. 2019, 48, 281–289. [Google Scholar] [CrossRef]
  51. Wu, Y.L.; Kumar, R.; Wang, M.F.; Singh, M.; Huang, J.F.; Zhu, Y.Y.; Lin, S. Validation of conventional non-invasive fibrosis scoring systems in patients with metabolic associated fatty liver disease. World J. Gastroenterol. 2021, 27, 5753–5763. [Google Scholar] [CrossRef] [PubMed]
  52. Angulo, P.; Hui, J.M.; Marchesini, G.; Bugianesi, E.; George, J.; Farrell, G.C.; Enders, F.; Saksena, S.; Burt, A.D.; Bida, J.P.; et al. The NAFLD fibrosis score: A noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology 2007, 45, 846–854. [Google Scholar] [CrossRef] [PubMed]
  53. Xiao, G.; Zhu, S.; Xiao, X.; Yan, L.; Yang, J.; Wu, G. Comparison of laboratory tests, ultrasound, or magnetic resonance elastography to detect fibrosis in patients with nonalcoholic fatty liver disease: A meta-analysis. Hepatology 2017, 66, 1486–1501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Castera, L. Non-invasive tests for liver fibrosis in NAFLD: Creating pathways between primary healthcare and liver clinics. Liver Int. 2020, 40, 77–81. [Google Scholar] [CrossRef] [Green Version]
  55. Lichtinghagen, R.; Pietsch, D.; Bantel, H.; Manns, M.P.; Brand, K.; Bahr, M.J. The Enhanced Liver Fibrosis (ELF] score: Normal values, influence factors and proposed cut-off values. J. Hepatol. 2013, 59, 236–242. [Google Scholar] [CrossRef]
  56. Vilar-Gomez, E.; Chalasani, N. Non-invasive assessment of non-alcoholic fatty liver disease: Clinical prediction rules and blood-based biomarkers. J. Hepatol. 2018, 68, 305–315. [Google Scholar] [CrossRef]
  57. Fagan, K.J.; Pretorius, C.J.; Horsfall, L.U.; Irvine, K.M.; Wilgen, U.; Choi, K.; Fletcher, L.M.; Tate, J.; Melino, M.; Nusrat, S.; et al. ELF score ≥9.8 indicates advanced hepatic fibrosis and is influenced by age, steatosis and histological activity. Liver Int. 2015, 35, 1673–1681. [Google Scholar] [CrossRef]
  58. Bedogni, G.; Bellentani, S.; Miglioli, L.; Masutti, F.; Passalacqua, M.; Castiglione, A.; Tiribelli, C. The fatty liver index: A simple and accurate predictor of hepatic steatosis in the general population. BMC Gastroenterol. 2006, 6, 33. [Google Scholar] [CrossRef] [Green Version]
  59. Lonardo, A.; Ballestri, S.; Bedogni, G.; Bellentani, S.; Tiribelli, C. The Fatty liver Index (FLI) 15 years later: A reappraisal. Metab. Target Organ Damage 2021, 1, 10. [Google Scholar] [CrossRef]
  60. Barr, R.G. Shear Wave Liver Elastography. Abdom. Radiol. 2018, 43, 800–807. [Google Scholar] [CrossRef]
  61. Oeda, S.; Tanaka, K.; Oshima, A.; Matsumoto, Y.; Sueoka, E.; Takahashi, H. Diagnostic Accuracy of FibroScan and Factors Affecting Measurements. Diagnostics 2020, 10, 940. [Google Scholar] [CrossRef] [PubMed]
  62. Castera, L.; Forns, X.; Alberti, A. Non-invasive evaluation of liver fibrosis using transient elastography. J. Hepatol. 2008, 48, 835–847. [Google Scholar] [CrossRef] [PubMed]
  63. de Franchis, R.; Bosch, J.; Garcia-Tsao, G.; Reiberger, T.; Ripoll, C.; Baveno VII Faculty. Baveno VII-Renewing consensus in portal hypertension. J. Hepatol. 2021, 76, 959–974. [Google Scholar] [CrossRef] [PubMed]
  64. Ballestri, S.; Nascimbeni, F.; Baldelli, E.; Marrazzo, A.; Romagnoli, D.; Targher, G.; Lonardo, A. Ultrasonographic fatty liver indicator detects mild steatosis and correlates with metabolic/histological parameters in various liver diseases. Metabolism 2017, 72, 57–65. [Google Scholar] [CrossRef]
  65. Xavier, S.A.; Monteiro, S.O.; Arieira, C.M.; Castro, F.D.; Magalhães, J.T.; Leite, S.M.; Marinho, C.M.; Cotter, J.B. US-FLI Score–Is It Possible to Predict the Steatosis Grade with an Ultrasonographic Score? Mol. Genet. Metab. 2021, 132, 204–209. [Google Scholar] [CrossRef]
  66. Berlanga, A.; Guiu-Jurado, E.; Porras, J.A.; Auguet, T. Molecular Pathways in Non-Alcoholic Fatty Liver Disease. Clin. Exp. Gastroenterol. 2014, 7, 221–239. [Google Scholar]
  67. Abenavoli, L.; Milic, N.; Di Renzo, L.; Preveden, T.; Medić-Stojanoska, M.; De Lorenzo, A. Metabolic aspects of adult patients with nonalcoholic fatty liver disease. World J. Gastroenterol. 2016, 22, 7006–7016. [Google Scholar] [CrossRef]
  68. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef]
  69. Makri, E.; Goulas, A.; Polyzos, S.A. Epidemiology, Pathogenesis, Diagnosis and Emerging Treatment of Nonalcoholic Fatty Liver Disease. Arch. Med. Res. 2020, 52, 25–37. [Google Scholar] [CrossRef]
  70. Milić, S.; Lulić, D.; Štimac, D. Non-alcoholic fatty liver disease and obesity: Biochemical, metabolic and clinical presentations. World J. Gastroenterol. 2014, 20, 9330–9337. [Google Scholar]
  71. Shabalala, S.C.; Dludla, P.V.; Mabasa, L.; Kappo, A.P.; Basson, A.K.; Pheiffer, C.; Johnson, R. The Effect of Adiponectin in the Pathogenesis of Non-Alcoholic Fatty Liver Disease (NAFLD] and the Potential Role of Polyphenols in the Modulation of Adiponectin Signaling. Biomed. Pharmacother. 2020, 131, 110785. [Google Scholar] [CrossRef] [PubMed]
  72. Polyzos, S.A.; Aronis, K.; Kountouras, J.; Raptis, D.D.; Vasiloglou, M.; Mantzoros, C.S. Circulating leptin in non-alcoholic fatty liver disease: A systematic review and meta-analysis. Diabetologia 2015, 59, 30–43. [Google Scholar] [CrossRef] [PubMed]
  73. Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S. Obesity and nonalcoholic fatty liver disease: From pathophysiology to therapeutics. Metabolism 2019, 92, 82–97. [Google Scholar] [CrossRef] [PubMed]
  74. Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S. Leptin in nonalcoholic fatty liver disease: A narrative review. Metabolism 2015, 64, 60–78. [Google Scholar] [CrossRef] [PubMed]
  75. Stojsavljević, S.; Gomerčić Palčić, M.; Virović Jukić, L.; Smirčić Duvnjak, L.; Duvnjak, M. Adipokines and proinflammatory cytokines, the key mediators in the pathogenesis of nonalcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 18070–18091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Svegliati-Baroni, G.; Pierantonelli, I.; Torquato, P.; Marinelli, R.; Ferreri, C.; Chatgilialoglu, C.; Bartolini, D.; Galli, F. Lipidomic biomarkers and mechanisms of lipotoxicity in non-alcoholic fatty liver disease. Free Radic. Biol. Med. 2019, 144, 293–309. [Google Scholar] [CrossRef]
  77. Branković, M.; Jovanović, I.; Dukić, M.; Radonjić, T.; Oprić, S.; Klašnja, S.; Zdravković, M. Lipotoxicity as the Leading Cause of Non-Alcoholic Steatohepatitis. Int. J. Mol. Sci. 2022, 23, 5146. [Google Scholar] [CrossRef]
  78. Basaranoglu, M.; Basaranoglu, G.; Bugianesi, E. Carbohydrate intake and nonalcoholic fatty liver disease: Fructose as a weapon of mass destruction. Hepatobiliary Surg. Nutr. 2015, 4, 109–116. [Google Scholar]
  79. Gjorgjieva, M.; Mithieux, G.; Rajas, F. Hepatic stress associated with pathologies characterized by disturbed glucose production. Cell Stress 2019, 3, 86–99. [Google Scholar] [CrossRef] [Green Version]
  80. Xian, Y.X.; Weng, J.P.; Xu, F. MAFLD vs. NAFLD: Shared Features and Potential Changes in Epidemiology, Pathophysiology, Diagnosis, and Pharmacotherapy. Chin. Med. J. 2020, 134, 8–19. [Google Scholar] [CrossRef]
  81. Dyson, J.K.; McPherson, S.; Anstee, Q.M. Republished: Non-alcoholic fatty liver disease: Non-invasive investigation and risk stratification. Postgrad. Med. J. 2014, 90, 254–266. [Google Scholar] [CrossRef] [PubMed]
  82. Blank, V.; Petroff, D.; Beer, S.; Böhlig, A.; Heni, M.; Berg, T.; Bausback, Y.; Dietrich, A.; Tönjes, A.; Hollenbach, M.; et al. Current NAFLD guidelines for risk stratification in diabetic patients have poor diagnostic discrimination. Sci. Rep. 2020, 10, 18345. [Google Scholar] [CrossRef] [PubMed]
  83. Rinella, M.E.; Sanyal, A.J. Management of NAFLD: A stage-based approach. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 196–205. [Google Scholar] [CrossRef] [PubMed]
  84. Wiklund, P.; Zhang, X.; Pekkala, S.; Autio, R.; Kong, L.; Yang, Y.; Keinänen-Kiukaanniemi, S.; Alen, M.; Cheng, S. Insulin resistance is associated with altered amino acid metabolism and adipose tissue dysfunction in normoglycemic women. Sci. Rep. 2016, 6, 24540. [Google Scholar] [CrossRef] [Green Version]
  85. Masoodi, M.; Gastaldelli, A.; Hyötyläinen, T.; Arretxe, E.; Alonso, C.; Gaggini, M.; Brosnan, J.; Anstee, Q.M.; Millet, O.; Ortiz, P.; et al. Metabolomics and lipidomics in NAFLD: Biomarkers and non-invasive diagnostic tests. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 835–856. [Google Scholar] [CrossRef] [PubMed]
  86. Newsome, P.N.; Sasso, M.; Deeks, J.J.; Paredes, A.; Boursier, J.; Chan, W.K.; Yilmaz, Y.; Czernichow, S.; Zheng, M.H.; Wong, V.W.S.; et al. FibroScan-AST (FAST) score for the non-invasive identification of patients with non-alcoholic steatohepatitis with significant activity and fibrosis: A prospective derivation and global validation study. Lancet Gastroenterol. Hepatol. 2020, 5, 362–373. [Google Scholar] [CrossRef]
Figure 1. Management with NAFLD risks. Based on Raniella et al., 2016 [83].
Figure 1. Management with NAFLD risks. Based on Raniella et al., 2016 [83].
Metabolites 12 01073 g001
Table 1. Fibroscan parameters used to determine the degree of steatosis and fibrosis reflected by the liver stiffness measurement. Abbreviations: LSM: liver stiffness measure, cACLD: compensated advanced chronic liver disease, CSPH: clinically significant portal hypertension.
Table 1. Fibroscan parameters used to determine the degree of steatosis and fibrosis reflected by the liver stiffness measurement. Abbreviations: LSM: liver stiffness measure, cACLD: compensated advanced chronic liver disease, CSPH: clinically significant portal hypertension.
LSM (kPa)Liver Decompensation and Liver Related-Death
≤5Exclude cACLD
10–15+ Platelets ≥ 150 k → exclude CSPH ( avoid endoscopy)
15–20+ Platelets ≥ 150 k → assume cACLD ( avoid endoscopy)
20–25Highly assume cACLD
≥25CSPH
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bassal, T.; Basheer, M.; Boulos, M.; Assy, N. Nonalcoholic Fatty Liver Disease—A Concise Review of Noninvasive Tests and Biomarkers. Metabolites 2022, 12, 1073. https://doi.org/10.3390/metabo12111073

AMA Style

Bassal T, Basheer M, Boulos M, Assy N. Nonalcoholic Fatty Liver Disease—A Concise Review of Noninvasive Tests and Biomarkers. Metabolites. 2022; 12(11):1073. https://doi.org/10.3390/metabo12111073

Chicago/Turabian Style

Bassal, Tamara, Maamoun Basheer, Mariana Boulos, and Nimer Assy. 2022. "Nonalcoholic Fatty Liver Disease—A Concise Review of Noninvasive Tests and Biomarkers" Metabolites 12, no. 11: 1073. https://doi.org/10.3390/metabo12111073

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop