Abstract
Non-alcoholic fatty liver disease (NAFLD) refers to a spectrum of diseases involving the deposition of fat in the hepatocytes of people with little to no alcohol consumption. NAFLD is associated with hypertension, diabetes, obesity, etc. As their prevalence increases, the propensity and severity of NAFLD might increase. As per the recently developed multi-hit hypothesis, factors like oxidative stress, genetic predisposition, lipotoxicity, and insulin resistance have been found to play a key role in the development of NAFLD and its associated complications. This article focuses on NAFLD, its pathophysiology, risk factors, and the various genetic and epigenetic factors involved in its development along with possible treatment modalities.
We conducted an all-language literature search on Medline, Cochrane, Embase, and Google Scholar until October 2021. The following search strings and Medical Subject Heading (MeSH) terms were used: “NAFLD,” “NASH,” “Fibrosis,” and “Insulin Resistance.” We explored the literature on NAFLD for its epidemiology, pathophysiology, the role of various genes, and how they influence the disease and associated complications about the disease and its hepatic and extrahepatic complications. With its rapidly increasing prevalence rates across the world and serious complications like NASH and hepatocellular carcinoma, NAFLD is becoming a major public health issue and more research is needed to formulate better screening tools and treatment protocols.
Keywords: treatment choices, mitochondrial senescence, demographics, long-term prognosis, disease progression, human genetics and epigenetics, nonalcoholic fatty liver disease (nafld)
Introduction and background
Non-alcoholic fatty liver disease (NAFLD) refers to a spectrum of diseases caused by deposition of fats in more than 5% of hepatocytes, primarily in the form of triglycerides in the absence of alcohol consumption (over 21 drinks/week for men and 14 drinks/week for women), use of medications causing hepatic steatosis, or hereditary and autoimmune causes [1]. The histological presentation can range from the bland accumulation of triglycerides within the hepatocytes (fatty liver) to non-alcoholic steatohepatitis (NASH). Fatty liver is a benign non-progressive form of the disease, whereas NASH is associated with inflammation, hepatocyte ballooning, and deposition of collagen fibers that can eventually progress to cirrhosis and liver cancer [2].
After Mayo Clinic first described the occurrence of NAFLD in 1980, the pathogenesis has been widely studied, although the exact mechanism is still unknown [3,4]. According to previous research that describes the “two-hit hypothesis,” the first strike is represented by increased hepatic triglyceride content and resistance to insulin. This vulnerable liver is then subjected to a second hit, which comprises inflammatory cytokines, adipokines, mitochondrial dysfunction, and oxidative stress [5]. At present, the “multiple hit model” is the commonly accepted idea that involves the association with metabolic dysfunction and interactions of complex environmental and genetic influences. NAFLD is now considered the hepatic manifestation of metabolic syndrome and is closely associated with high triglyceride level content, low high-density lipoprotein (HDL), hypertension, and morbid obesity [6].
As it requires histological diagnosis, the gold standard for diagnosis of NAFLD is liver biopsy. But specific non-invasive modalities are acceptable, including hepatic ultrasonography (USG), abdominal computed tomography (CT) scan, and magnetic resonance imaging (MRI) [7]. Serum biomarkers such as alanine aminotransferase (ALT) and aspartate aminotransferase (AST) have been used for screening [8]. The prevalence of NAFLD in the general population in various nations is estimated to range from 3% to more than 24% using blood markers [8]. NASH affects around 6-8% of individuals in the United States (about 25% of those with NAFLD) [9]. If these studies are assumed to represent the wider population, it would imply cirrhosis in 1.5-2% of the US population owing to NAFLD [9].
While the progression to cirrhosis is rare, subsequent chances of development to primary liver cancer can be as high as 3%. The incidence of hepatocellular carcinoma and intrahepatic cholangiocarcinoma associated with NAFLD is gradually increasing [10]. Documentation of progressive hepatic fibrosis is used as an important surrogate marker for advanced liver disease. In recent studies, the annual fibrosis progression rate was found to be 0.09 times (95% CI: 0.06-0.12) in patients with baseline NAFLD [11]. In the United States, NASH is the second most common cause of liver disease among transplant recipients [12]. With the rise in the incidence of obesity, hypercaloric diet, and sedentary lifestyle, NAFLD has become the dominant form of chronic liver disease among the pediatric and adolescent age group, with cases detected as young as three years [13,14]. Over the last decade, many treatments have been developed and tested, but no effective treatment is currently available [15]. Thus, it becomes crucial to identify the demography at the highest risk and the elements that influence the advancement of NAFLD.
Review
Pathogenesis and molecular mechanisms of NAFLD
Typically, two pathologically different conditions are identified: non-alcoholic fatty liver (NAFL) and NASH. On the one hand, NAFL is characterized by simple liver steatosis; on the other hand, NASH is identified by the presence of macrovesicular fat accumulation, hepatic ballooning, and lobular inflammation with or without any fibrosis [16-18].
NAFLD is a multifactorial disease, and its pathogenesis and progression are not fully understood. It is a complex disease that interacts with other organs in the body, for instance, on the intestine-liver-adipose tissue axis for its development. Multiple pathways have been proposed over the past years on how NAFLD progresses [19]. Although the most famous theory to explain NAFLD is the “two-hit hypothesis,” a more accurate explanation of NAFLD pathogenesis explained that several molecular and metabolic changes occur together in its development and progression. This “multiple hit” hypothesis considers multiple insults acting in concert to cause NAFLD [19,20].
Lipotoxicity and NAFLD
Hepatic lipotoxicity happens when the liver's capacity to use, reserve, and export free fatty acids (FFAs) as triglycerides (TGs) is saturated by a massive FFA influx from the periphery, mainly the adipose tissue or by increased hepatic de novo lipogenesis [21-23].
FFAs are exported from the liver as very-low-density lipoprotein (VLDL) or stored in hepatic lipid droplets after being metabolized via mitochondrial β-oxidation or re-esterification into TGs inside the hepatocytes. Lipid droplets inside the hepatocytes undergo lipolysis and release fatty acids into the hepatocyte FFA pool [24-26]. Lipotoxic species formed from excess FFA inside liver cells cause endoplasmic reticulum inflammation, oxidative stress, and activation of inflammatory mediators [27-29].
Insulin Resistance and NAFLD
Insulin resistance plays a vital role in the pathogenesis of NAFLD [30]. The role of insulin in our body is to stimulate glucose utilization and increase lipid accumulation in insulin-sensitive organs such as skeletal muscle, adipose tissue, and liver. Insulin resistance leads to increased lipolysis in dysfunctional adipose tissue and reduced glucose uptake in skeletal muscle [27,28]. As a result, there is an increased FFA in the bloodstream and the liver as well. Higher hepatic uptake of FFA leads to the accumulation of TGs in the liver and the release of VLDL via fatty acid esterification.
In contrast, glucose uptake contributes to a lesser extent via de novo lipogenesis, leading to steatosis (NAFL) and dyslipidemia [26]. Studies have shown metabolic cross-talk between liver and adipose tissue [30]. Some peptides released by adipose tissue, such as adiponectin and interleukin 6 (IL6), have both proinflammatory and protective effects on the liver [31,32]. The enzyme dipeptidyl peptidase 4 (DPP-4) can promote insulin resistance. This enzyme, secreted by hepatocytes, has been seen to work on plasma factor Xa to stimulate macrophages within the visceral tissue of mice. This mechanism is known to promote insulin resistance, emphasizing the important cross-talk between the liver and other tissues underlying metabolic dysregulation in NAFLD [33].
Oxidative Stress
Oxidative stress plays an important role in the pathogenesis of NAFLD [34,35]. Oxidative stress means the imbalance between the production of reactive oxygen species (ROS) and the scavenging capacity of the antioxidant system [36]. Mitochondria are the most important source of ROS in NAFLD, mainly due to increased fatty acid oxidation (FAO) [37]. At high concentrations, ROS cause oxidative modifications to cellular macromolecules (DNA, lipids, proteins, etc.) and cause accumulation of damaged macromolecules, initiating liver injury [35].
ROS can initiate lipid peroxidation by targeting polyunsaturated fatty acids, resulting in highly reactive aldehyde products such as 4-hydroxy-2-nonenal (4-HNE) and malondialdehyde (MDA). These reactive compounds have longer half-lives than free radicals and can diffuse into the extracellular space to boost tissue damage. Multiple studies have reported that both MDA and 4-HNE are increased in experimental animal models of NASH and in NASH patients compared to patients with NAFL [35-38].
Cytochrome P450, lipoxygenase, and cyclooxygenase, well-characterized pro-oxidant systems, combined with free-radical products, have been involved in the early stages of NAFLD [39,40]. On the contrary, a reduction in the activity of antioxidant enzymes such as catalase, glutathione peroxidase, glutathione S-transferase, superoxide dismutase, as wells as ROS scavengers (ascorbic acid, glutathione, α-tocopherol, ubiquinone, thioredoxin, and bilirubin) is a characteristic of livers from NASH patients [38,39,41].
Immune Cells Involved in NAFLD
Multiple immune cells are involved in developing NAFLD, such as activated Kupffer cells, the liver's resident macrophages, in combination with the recruitment of proinflammatory, monocyte-derived macrophages and neutrophil leukocytes from circulation [42,43]. With the substantial accumulation of macrophages, their polarization appears “proinflammatory” at the expense of anti-inflammatory and repair macrophages, likely because of the high abundance of proinflammatory cytokines such as interferon-γ, tumor necrosis factor (TNF), and lipopolysaccharide in NASH [44].
Epidemiology and risk factors of NAFLD
NAFLD has been increasing remarkably worldwide in recent years. It is estimated that the current prevalence of NAFLD exceeds 24% of the population globally [11], which implicitly indicates the increase in liver-related morbidity and mortality in the next few years [45,46]. The main cause for concern of NAFLD (particularly NASH) is that it is one of the commonest causes of hepatocellular cancer (HCC) and is expected to supersede viral hepatitis as the most common etiology for HCC in the United States due to the rapidly growing epidemic of metabolic syndrome [47,48]. NASH is also considered an increasing cause of liver transplantation in the US, UK, and also in developing countries [49].
Although the NAFLD disease spectrum ends with cirrhosis, hepatocellular carcinoma (HCC), cardiac events, and chronic kidney disease (CKD) are the major causes of death among NAFLD patients [50]. In addition, one study suggested a relationship between multiple NAFLD comorbidities and the poor prognosis of coronavirus disease 2019 (COVID-19) in NAFLD-infected individuals [51]. Due to its wide-ranging effects on multiple body systems, it is important to initiate a non-invasive screening method to detect NAFLD early.
Tables 1, 2 show the estimated prevalence of NAFLD globally according to regions and race, respectively. The highest prevalence is in the Middle East and South America (around 30%) and the lowest in Africa (13%) [11]. The effect of gender on NAFLD progression is not clear [52], but recent studies showed that males are more likely to develop NASH [53,54].
Table 1. Prevalence of NAFLD in different regions.
Region | Estimated prevalence | Notes |
North America | 27%-34% [55] | N/A |
South America | 31% [11] | Brazil reported the highest NAFLD prevalence and Peru the lowest [56] |
Europe | 25% [11] | Varies by countries with a reported low prevalence rate of 8% in Romania to a reportedly high prevalence of 45% in Greece [57] |
Asia | 15%-20% [58] | These rates are higher in urban areas than those reported from rural areas indicating the highly increasing change of Asian lifestyle in urban areas [11] |
Africa | 13% [11] | Could be low due to a poor reporting system [59] |
Middle East | 32% [11] | N/A |
Table 2. Prevalence of NAFLD in different ethnicities.
Race | Estimated prevalence | Notes |
Hispanic Americans | 45% [60] | Highest prevalence |
African Americans | 24% [60] | Lowest prevalence |
European American | 33% [60] | N/A |
Asians American | 18% [61] | N/A |
Hispanics Mexicans | 33% | Higher prevalence of NAFLD than Hispanics of Dominican origin (16%), and Hispanics of Puerto Rican origin (18%) |
Risk Factors of NAFLD
There are many established risk factors for NAFLD development and progression. A strong association has been found between diabetes mellitus type 2 (DM2) and NAFLD as a large proportion of DM2 patients have NAFLD. It is thought that insulin resistance plays a critical role at the cellular level in the pathogenesis of both conditions [62]. Other risk factors are somehow related to DM2, as obese people, who are at higher risk of DM2 [63], have a higher prevalence of NAFLD.
A study showed that >95% of patients with morbid obesity undergoing bariatric surgery would develop NAFLD in their life [64]. Because of this strong relationship between obesity and NAFLD, waist circumference has been suggested as a simple indicator for the risk of NAFLD development [65]. In addition, weight reduction strategies are emphasized as a preventive method of NAFLD development and progression [66]. A sedentary lifestyle and poor eating habits (high fructose (sugar) drinks and fatty food) are shared risk factors for the development of NAFLD and DM2 [67,68].
Studies among the Asian population showed that sarcopenia, the loss of skeletal muscle mass, has also been associated with the increasing severity of NAFLD [69]. This association needs further validation in the western world [69,70]. Women with subclinical metabolic disturbances are at higher risk of developing NAFLD after menopause, highlighting the protective role of estrogen in women before menopause [71].
Some studies stated that NAFLD could pass through generations, for instance, the metabolic phenotype of obese pregnant women could be transmitted to offspring, making them more susceptible to obesity and NAFLD [53,54]. Regarding gender, it is widely accepted that the male sex has an increased incidence of NAFLD. However, the role of sex in NAFLD progression remains inconclusive. Some studies report sex as an independent predictive factor of NAFLD severity, while others suggest exposure to estrogen to be a better metric [72]. The effect of sex hormones needs to be investigated further to elucidate their role.
Factors for NASH Development
Most studies are in agreement with shared factors that lead to NASH development. Factors associated with NASH development are body mass index (BMI) > 27 and lower hip-to-waist ratios. In addition, specific laboratory investigations were statistically significant, as elevated levels of ALT, AST, and serum sphingolipids (particularly ceramide) with low levels of HDL cholesterol were all associated with NASH development and progression [73-77]. Recently, a study by Tasneem et al. came up with a scoring system called the GULAB (based on gender, US liver findings, lipid (fasting) levels, ALT level, and BMI) score. It consists of several non-invasive parameters with a cutoff point of 5 associated with NASH development [74]. Another study by Seko et al. considered persistent elevation of ALT despite treatment as a valid indicator of NASH histological progression. It recommended strict control of serum ALT to prevent NAFLD progression [77].
Factors Inducing Advanced Fibrosis in NAFLD Patients
NASH patients with advanced fibrosis have a higher risk of developing liver cirrhosis that ends with liver cell failure [78]. Established factors associated with advanced fibrosis in NAFLD patients were male gender, age > 40 years, Caucasian ethnicity, DM2, hypertension, and a lower hip-to-waist ratio. Also, AST/ALT ratio > 1 and raised gamma-glutamyl transferase (GGT) were also noted in advanced fibrotic liver patients [73,74,79]. Thus patients with NASH are advised to check for cirrhosis in case of the presence of these parameters. Patients with NASH are at high risk of developing fibrosis, while patients with NAFL were a point of conflict, as many studies considered their condition as benign that would not develop liver fibrosis [80,81]. On the other hand, more recent studies found that NAFL patients may end up with liver fibrosis, but the progression is slower than NASH patients [79,82].
Genetic and epigenetic factors
Family studies and twin studies estimate the heritability of NAFLD between 20% and 70%, suggesting a vital genetic component for its development and progression [83]. The evolution of genome-wide association studies (GWAS) and candidate gene studies have helped identify numerous genes involved in lipid handling, insulin signaling, and even innate immunity. A select number of genes are summarized in the table below (Table 3). The most robust evidence across multiple ethnicities is for the patatin-like phospholipase domain-containing protein 3 (PNPLA3) gene, substituting isoleucine with methionine [84,85]. It produces a dysfunctional lipase enzyme that interferes with triglyceride metabolism and increases the probability of hepatic steatosis [86]. In addition, the PNPLA3 variant is also associated with the development of fibrosis and a five-fold increase in the risk of HCC [87]. The membrane-bound O-acyl transferase 7 (MBOAT7) gene involved in phospholipid metabolism has also been identified to increase the risk of NAFLD development and progression, particularly in the European population [88-90]. This gene has also been associated with a two times increase in risk for HCC [91]. The transmembrane 6 superfamily member 2 (TM6SF2) gene has also been implicated in NAFLD incidence and hepatic fibrosis development in Europeans [92,93]. This gene is involved in liver secretion of very-low-density lipoprotein; a loss-of-function mutation increases hepatic steatosis while protecting cardiovascular disease (CVD) risk [94]. However, this association has not been replicated in other ethnicities [95]. Table 3 is a brief summary of the various genes associated with NAFLD development and progression.
Table 3. Genes associated with NAFLD progression with a short description of their biochemical processes.
Gene | Variant | Biochemical effects | Effect on NAFLD |
PNPLA3 | rs738409 | Triglyceride metabolism | Increases incidence, progression, and HCC risk [84-87] |
MBOAT7 | rs641738 | Phospholipid metabolism | Increases incidence, progression, and HCC risk [88-91] |
TM6SF2 | rs58542926 | VLDL secretion | Increases incidence and progression [92-93] |
Glucokinase regulator (GCKR) | rs780094 | De novo lipogenesis | Increases incidence and progression [96] |
17β-hydroxysteroid dehydrogenase 13 (HSD17B13) | rs72613567 | Possibly lipid-related inflammation | Decreases progression [97] |
Interleukin - 28B (IL28B) | rs12979860 | Innate immunity | Decreases fibrosis [98] |
Superoxide dismutase 2 (SOD2) | rs4880 | Oxidative stress protection | Increases fibrosis [99] |
Genetic risk scores are proposed to stratify the severity of NAFLD based on the identified variants [100]. A recent study has demonstrated a better prediction of HCC risk when using genetic risk scores combining the PNPLA3, TM6SF2, glucokinase regulator (GCKR), MBOAT7, and 17β-hydroxysteroid dehydrogenase 13 (HSD17B13) variants [101].
Recent GWAS studies have identified new candidate genes, which may contribute to the genetic risk of NAFLD. For example, associations between a variant in the sorting and assembly machinery component 50 (SAMM50) and NAFLD incidence are reported in Asian and Mexican populations [102]. In addition, variants in glycerol-3-phosphate acyltransferase, mitochondrial (GPAM) and apolipoprotein E (APOE) genes are also identified to be associated with liver fat in the European population [103]. Validation studies need to be performed to understand the effects of the identified variants, which may add to the development of more robust genetic risk scores.
Epigenetics
Epigenetics is described as genetic modification without changes to deoxyribonucleic acid (DNA) sequences to cause a phenotypic change. Its role in the progression of NAFLD via DNA methylation, histone modification, and changes in microRNA (miRNA) expression is gathering increasing attention [94]. DNA methylation refers to the transfer of a methyl group to form 5-methylcytosine at cytosine-guanine dinucleotide (CpG) islands to reduce gene transcription [104].
Changes in methylation of CpG islands in the PNPLA3 gene and fibrogenic genes such as platelet-derived growth factor (PDGF) and transforming growth factor beta (TGFβ) have been associated with the progression of fibrosis [105]. A recent study has demonstrated a change in expression of 288 genes in samples with fibrosis along with a change in cell composition driven by DNA methylation [106]. This highlights the potential of DNA methylation to be the driving factor of fibrosis in NAFLD. Methylation of mitochondrial genes such as NADH dehydrogenase 6 (MT-ND6) in the liver has also been associated with more severe forms of NAFLD [107].
Recent studies have shown a dietary component in DNA methylation as methylation is driven by folate-dependent methyltransferases [104]. It has also been demonstrated that a high-fat diet leads to hypermethylation of peroxisome proliferator-activated receptor gamma (PPARγ), which is linked to triglyceride accumulation [108]. Hence, dietary interventions could act as a potential therapy for the reversal of NASH.
Changes in acetylation of amino acid residues of histone tails driven by histone acetylases (HATs) and histone deacetylases (HDACs) have been shown to affect inflammatory responses, which drive the development of NAFLD [84]. For example, increased activity of HAT has been described to increase transcription of hepatocyte carbohydrate-responsive element-binding protein (chEBP), a well-described activator of genes involved in acute phase response and lipogenesis [109]. Sirtuins (SIRT), a type of HDAC, attenuates responses to metabolic stress [110]. SIRT1 is demonstrated to have a protective effect on NAFLD and other metabolic conditions and its deletion was associated with increasing severity of hepatic steatosis [110,111]. Lately, a murine knock-out model has shown that a lack of SIRT2 is associated with increased lipogenesis and fibrosis, suggesting a crucial role for histone modification in the NAFLD progression to fibrosis [112].
miRNAs are non-coding RNAs that target messenger RNAs for degradation and regulate their expression [94]. Modifications in the hepatic miRNA profile have been associated with NASH and HCC [113]. Mi-122, the most common miRNA in the liver, has been associated with tumorigenesis due to the deletion of its tumor suppressor effects [113]. It is also associated with NASH and liver fibrosis by activating tissue remodeling genes [94]. Upregulation of mi-192 is also associated with an increased incidence of NASH and has been proposed to be part of the miRNA panel to identify NASH [114]. In addition, mi-166 has shown an association with fibrosis in the Chinese population [115]. Other miRNAs such as mi-21, mi-34a, and mi-16 have also been associated with NASH [94]. Recently, miR-374a, miR-143, miR-1, miR-23a, and miR-423 have also been associated with NASH and target mitochondrial genes and genes involved in lipogenesis, apoptosis, and fibrogenesis [116]. Further research is necessary to validate these miRNAs to develop non-invasive biomarkers.
Mitochondrial senescence
Whatever the primary etiology of NASH, mitochondrial senescence (especially respiratory chain insufficiency) plays a major role in the physiopathology of the disease. Senescence is a form of cellular adaptation to various normal and abnormal cellular processes. It follows different forms of stresses such as changes in the structure of chromatin, stress involving oxidation, and activation of oncogenes [117]. Senescence in hepatocytes and other supporting cells of the liver such as sinusoidal endothelial cells, hepatic stellate cells, and Kupffer cells are closely related to the development and progression of NAFLD [118].
Mitochondrial changes in senescent cells include increased size and amount of tricarboxylic acid byproducts, an increase in the number of mitochondria, ROS, and proton leaks, and a decrease in the function and the membrane potential [119,120]. In 2001, a study conducted by Sanyal et al. found many morphological differences in the mitochondria of a normal liver and that of a NASH patient. NASH was linked with swollen and round mitochondria, clear matrix, loss of mitochondrial cristae, and multiple paracrystalline inclusions, although no indication of a widespread deficiency in fatty acid beta-oxidation was found in any of the groups [121]. Intramitochondrial paracrystalline bodies were associated with NASH with very strong specificity compared with normal controls, fatty liver, and hepatitis C patients.
Though the beta-oxidation of fatty acids was not much affected in the hepatocytes, the defective respiratory chain in the hepatocyte mitochondria generated increased ROS. In a lipid-rich microenvironment, ROS production increases lipid peroxidation, which is toxic to the hepatocytes. ROS harms the respiratory chain due to oxidative damage to the mitochondrial DNA, which starts a vicious cycle [122]. The involvement of mitochondrial senescence in the pathophysiology of NAFLD was reinforced when studies concluded the association between drugs causing steatohepatitis and mitochondrial dysfunction. Drugs like amiodarone or perhexiline cause impairment of electron transfer in the respiratory chain and inhibit beta-oxidation leading to dysfunctional hepatocyte and death [123].
When senescence occurs, its degree is directly proportional to the extent of NAFLD. Some markers support senescence in the liver with NAFLD, and this includes p53, p21, p16, and, more specifically, senescence marker protein-30 (SMP30) [124-126]. SMP30 is involved in ROS and glucose breakdown and maintains calcium balance, but levels of SMP30 reduce with age [127]. Studies have shown that SMP30 reduces further in NAFLD [127]. A decrease in the amount of SMP30 causes fat accumulation in the liver and eventual death of hepatocytes due to the disruption of its metabolism [128].
13C-ketoisocaproic acid (KICA) and methionine are markers of mitochondrial function. Breath tests using them as biomarkers showed a significant reduction in carboxylation in patients of NASH but not in patients with fatty liver [129]. Due to high localization in the mitochondria, the urea cycle metabolites have also been used as a non-invasive biomarker for staging and prognosis of NAFLD [130]. These novel methods should be used in the early detection and limitation of progression of NAFLD. However, it remains unclear whether the link between mitochondrial defects and NASH is the cause or an effect of the disease.
Complications and prognosis of NAFLD
Hepatic Complications
As previously mentioned, NAFLD is a spectrum of diseases mainly composed of patients with simple steatosis. In some patients, it can progress to a more advanced disease characterized by NASH and liver fibrosis and cirrhosis, or even HCC [131]. In a retrospective analysis of 619 patients with NAFLD, Angulo et al. concluded that the severity of liver fibrosis is associated with increased long-term mortality and other liver-related complications [132]. However, a collaborative cohorts study by Bhala et al. showed that patients with NAFLD (biopsy-confirmed) with advanced fibrosis had lower rates of hepatic complications and HCC in comparison with patients with hepatitis C virus (HCV) infection, but overall mortality was the same in both groups [133].
The prevalence of HCC among NAFLD patients is increasing, particularly in developed countries, as suggested by Kim and El-Serag [134]. Currently, limited studies have suggested exact mechanisms of HCC in NAFLD patients. On the other hand, in a cohort study consisting of 130 NAFLD patients, Kanwal et al. concluded that there was an increased risk of HCC and cirrhosis with stepwise addition of each metabolic trait. Their results showed that type 2 diabetes mellitus (T2DM) conferred the highest risk of progression to HCC and suggested T2DM is an essential target for secondary prevention [135].
As with the increasing incidence of NASH, the incidence of NASH-related liver transplantation is increasing. A retrospective cohort study was done by Cholankeril et al. concluding that NASH is the most rapidly growing indication and currently the second leading indication for liver transplant [136].
Extrahepatic Complications
NAFLD is a multisystem disease and is closely associated with many extra-hepatic complications such as CVD, CKD, and certain malignancies. Among all, CVD is the most common cause of death in patients with NAFLD [137,138]. A meta-analysis including 16 observational prospective and retrospective studies by Targher et al. showed that patients with NAFLD had a higher incidence of CVD events than patients without NAFLD [139]. In another meta-analysis involving 27 cross-section studies, Oni et al. concluded that there is a strong association of NAFLD with several CVD complications such as increased carotid wall thickness, impaired flow-mediated vasodilation, increased arterial stiffness, and subclinical atherosclerosis, which is independent of traditional CVD risk factors and metabolic syndromes [140].
The link between NAFLD and the development of T2DM has been well established by several studies in the past decade; however, the mechanism linking both conditions is multifactorial [141].
As previously mentioned, CKD is a well-established complication of NAFLD. Patients with NAFLD/NASH have a higher prevalence of CKD than patients without NAFLD/NASH. Although the exact pathogenic mechanism linking NAFLD and CKD is not well understood, it is believed that proinflammatory mediators released as a result of liver inflammation in patients with NAFLD cause an increase in pro-oxidative and pro-fibrotic molecules, which play a key role in the mechanism of CKD [142].
Several studies conducted in recent years have established an important link between NAFLD and colorectal carcinoma (CRC) and suggested NAFLD as an independent risk factor for the development of colon polyps and eventually colorectal adenocarcinoma. Inflammation of the liver due to NAFLD causes increased levels of proinflammatory and procoagulant factors and decreases levels of adipokines and adiponectin (protective factors), thus contributing to the pathogenesis of CRC in patients with NAFLD [143]. A correlation between NAFLD and psoriasis with similar pathogenesis (increased proinflammatory cytokines and reduced anti-inflammatory cytokines) has also been demonstrated [144].
A cross-sectional study by Gutierrez-Grobe et al. at the university hospital in Mexico City involving 197 women showed a significant association between NAFLD and polycystic ovarian syndrome (PCOS) and concluded that the prevalence of NAFLD was higher in postmenopausal women and women with PCOS than premenopausal women [145]. In the past few years, several experiments have suggested a possible connection between chronic intermittent hypoxia of obstructive sleep apnea syndrome (OSAS) and NAFLD. A meta-analysis done by Musso et al., including 18 cross-sectional studies, established a link between OSAS and a higher risk of NAFLD [146].
Treatment of NAFLD
Weight loss through lifestyle modifications such as hypocaloric diets and physical exercise remains the primary and most effective treatment for NAFLD of all stages [147]. The American Association for the Study of Liver Disease (AASLD) recommends a minimum of 3-5% loss of body weight to improve hepatic steatosis and a more significant percent weight loss (7-10%) to improve features of NASH [17]. Although there is a lack of global consensus on a specific dietary strategy, the Mediterranean diet, rich in omega-3-unsaturated polyunsaturated fatty acids, is recommended by multiple European organizations as the best solution [148].
Pharmacological intervention is indicated only in the case of NASH [17]. The use of several pharmacologic agents has been explored; however, the supporting data are limited and largely empirical with many of their clinical trials occurring in uncontrolled environments. Currently, pioglitazone, a thiazolidinedione, is the only insulin-sensitizing agent recommended by AASLD to patients with NASH [17]. Vitamin E, with its antioxidant anti-inflammatory and anti-apoptotic properties, is recommended by AASLD only in nondiabetic patients with biopsy-proven NASH [17]. Monitoring of older male patients is indicated during continuous use for more than three years, due to an increased risk of prostate cancer [149-151]. Lipid-lowering agents (statins, fibrates, and ezetimibe) can reduce FFAs and triglyceride deposition in hepatocytes, decreasing disease progression [152]. This exploits the positive correlation between subclinical atherosclerosis and progression and mortality in NASH patients [152]. Surgical management of NASH is performed in cases of complications such as cirrhosis or HCC [153]. NASH-related cirrhosis and HCC are increasingly common indications for liver transplantation in the West [154]. Bariatric surgery has been shown to reverse histological features of NASH; however, it is not recognized as a treatment for NASH and is only indicated in severe obesity to aid weight loss [17,155].
The use of obeticholic acid has been found to noticeably improve the histological changes in patients with early stages of fibrosis in NASH, but the outcome is unsatisfactory in advanced NAFLD. In individuals with T2DM and NAFLD, the use of sodium-glucose cotransporter-2 (SGLT-2) inhibitors, like empagliflozin and dapagliflozin, lowers liver fat and raises ALT levels [153,154]. Newer thiazolidinediones like saroglitazar (a dual peroxisome proliferator-activated receptor (PPAR) α/γ agonist, approved for diabetic dyslipidemia) and elafibranor (an agonist of PPAR-α and PPAR-δ) have shown improvement in insulin resistance and serum lipid normalization, which are two important causes of NASH progression. The safety profile is excellent, although it causes reversible serum creatinine increases, which may limit its use in individuals with a renal illness. It calls for the personalization of drug combinations, depending on the stage of NASH and associated morbidities for each patient [154].
Established medications are being explored as potential therapies for NAFLD. For example, liraglutide, a glucagon-like peptide 1, has been demonstrated to cause a resolution in the histological features of NASH and can be considered as a potential treatment [156]. Other naturally occurring substances such as silymarin, betaine, quercetin, and probiotics have shown mixed results and may be used along with other therapies [157]. Several new medications targeting pathways driving progression are also in development. Cenicriviroc, a chemokine antagonist targeting pro-inflammatory pathways, was investigated for NASH treatment [158]. Selonsertib, which modulates cell death via apoptosis signal-regulating kinase-1, was considered [159]. Drugs targeting fibrogenesis such as simtuzumab were also researched [160]. However, their viability as treatments remains limited, possibly due to the complexity of the targeted pathways. Development of new pharmacological agents and their validation through large-scale randomized controlled trials are required to offset the increase in NASH-indicated liver transplantations.
Conclusions
NAFLD is a very broad disease, with multiple risk factors acting in concert to manifest its numerous clinical consequences. The incidence of risk factors associated with this disease, such as diabetes mellitus, for instance, is increasing rapidly across the world, increasing the prevalence of NAFLD. Despite repeated efforts by multiple teams from across the world, there is still no cure for NAFLD. Therefore, its management is focused on the prevention and early detection of this disease.
The content published in Cureus is the result of clinical experience and/or research by independent individuals or organizations. Cureus is not responsible for the scientific accuracy or reliability of data or conclusions published herein. All content published within Cureus is intended only for educational, research and reference purposes. Additionally, articles published within Cureus should not be deemed a suitable substitute for the advice of a qualified health care professional. Do not disregard or avoid professional medical advice due to content published within Cureus.
Footnotes
The authors have declared that no competing interests exist.
References
- 1.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. Chalasani N, Younossi Z, Lavine JE, et al. Hepatology. 2012;55:2005–2023. doi: 10.1002/hep.25762. [DOI] [PubMed] [Google Scholar]
- 2.Nonalcoholic fatty liver disease: pathology and pathogenesis. Tiniakos DG, Vos MB, Brunt EM. Annu Rev Pathol. 2010;5:145–171. doi: 10.1146/annurev-pathol-121808-102132. [DOI] [PubMed] [Google Scholar]
- 3.Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Ludwig J, Viggiano TR, McGill DB, Oh BJ. https://pubmed.ncbi.nlm.nih.gov/7382552/ Mayo Clin Proc. 1980;55:434–438. [PubMed] [Google Scholar]
- 4.The natural history of nonalcoholic fatty liver disease—an evolving view. Lindenmeyer CC, McCullough AJ. Clin Liver Dis. 2018;22:11–21. doi: 10.1016/j.cld.2017.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pathogenesis of non-alcoholic fatty liver disease in children and adolescence: from "two hit theory" to "multiple hit model". Fang YL, Chen H, Wang CL, Liang L. World J Gastroenterol. 2018;24:2974–2983. doi: 10.3748/wjg.v24.i27.2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Non-alcoholic fatty liver disease and metabolic syndrome in adolescents: pathogenetic role of genetic background and intrauterine environment. Alisi A, Cianfarani S, Manco M, Agostoni C, Nobili V. Ann Med. 2012;44:29–40. doi: 10.3109/07853890.2010.547869. [DOI] [PubMed] [Google Scholar]
- 7.Diagnosis of nonalcoholic fatty liver disease in children and adolescents: position paper of the ESPGHAN Hepatology Committee. Vajro P, Lenta S, Socha P, et al. J Pediatr Gastroenterol Nutr. 2012;54:700–713. doi: 10.1097/MPG.0b013e318252a13f. [DOI] [PubMed] [Google Scholar]
- 8.Relationship between alanine aminotransferase levels and metabolic syndrome in nonalcoholic fatty liver disease. Chen ZW, Chen LY, Dai HL, Chen JH, Fang LZ. J Zhejiang Univ Sci B. 2008;9:616–622. doi: 10.1631/jzus.B0720016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.The epidemiology of nonalcoholic fatty liver disease in adults. Clark JM. https://pubmed.ncbi.nlm.nih.gov/16540768/ J Clin Gastroenterol. 2006;40:0–10. doi: 10.1097/01.mcg.0000168638.84840.ff. [DOI] [PubMed] [Google Scholar]
- 10.NAFLD, NASH and liver cancer. Michelotti GA, Machado MV, Diehl AM. Nat Rev Gastroenterol Hepatol. 2013;10:656–665. doi: 10.1038/nrgastro.2013.183. [DOI] [PubMed] [Google Scholar]
- 11.Global epidemiology of nonalcoholic fatty liver disease-meta-analytic assessment of prevalence, incidence, and outcomes. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Hepatology. 2016;64:73–84. doi: 10.1002/hep.28431. [DOI] [PubMed] [Google Scholar]
- 12.Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United States. Wong RJ, Aguilar M, Cheung R, Perumpail RB, Harrison SA, Younossi ZM, Ahmed A. Gastroenterology. 2015;148:547–555. doi: 10.1053/j.gastro.2014.11.039. [DOI] [PubMed] [Google Scholar]
- 13.Nutrition and physical activity in NAFLD: an overview of the epidemiological evidence. Zelber-Sagi S, Ratziu V, Oren R. World J Gastroenterol. 2011;17:3377–3389. doi: 10.3748/wjg.v17.i29.3377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Non-alcoholic fatty liver disease: a massive problem. Day CP. Clin Med (Lond) 2011;11:176–178. doi: 10.7861/clinmedicine.11-2-176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Nonalcoholic fatty liver disease: targeted therapy in children--what is the right way? Alisi A, Nobili V. Nat Rev Gastroenterol Hepatol. 2011;8:425–426. doi: 10.1038/nrgastro.2011.117. [DOI] [PubMed] [Google Scholar]
- 16.Molecular pathways of nonalcoholic fatty liver disease development and progression. Bessone F, Razori MV, Roma MG. Cell Mol Life Sci. 2019;76:99–128. doi: 10.1007/s00018-018-2947-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Chalasani N, Younossi Z, Lavine JE, et al. Hepatology. 2018;67:328–357. doi: 10.1002/hep.29367. [DOI] [PubMed] [Google Scholar]
- 18.AISF position paper on nonalcoholic fatty liver disease (NAFLD): updates and future directions. Italian Association for the Study of the Liver (AISF) Dig Liver Dis. 2017;49:471–483. doi: 10.1016/j.dld.2017.01.147. [DOI] [PubMed] [Google Scholar]
- 19.The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD) Buzzetti E, Pinzani M, Tsochatzis EA. Metabolism. 2016;65:1038–1048. doi: 10.1016/j.metabol.2015.12.012. [DOI] [PubMed] [Google Scholar]
- 20.Evolving concepts in the pathogenesis of NASH: beyond steatosis and inflammation. Peverill W, Powell LW, Skoien R. Int J Mol Sci. 2014;15:8591–8638. doi: 10.3390/ijms15058591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hepatic insulin resistance is associated with increased apoptosis and fibrogenesis in nonalcoholic steatohepatitis and chronic hepatitis C. García-Monzón C, Lo Iacono O, Mayoral R, et al. J Hepatol. 2011;54:142–152. doi: 10.1016/j.jhep.2010.06.021. [DOI] [PubMed] [Google Scholar]
- 22.Hepatocyte apoptosis and fas expression are prominent features of human nonalcoholic steatohepatitis. Feldstein AE, Canbay A, Angulo P, Taniai M, Burgart LJ, Lindor KD, Gores GJ. Gastroenterology. 2003;125:437–443. doi: 10.1016/s0016-5085(03)00907-7. [DOI] [PubMed] [Google Scholar]
- 23.Role of hepatocyte S6K1 in palmitic acid-induced endoplasmic reticulum stress, lipotoxicity, insulin resistance and in oleic acid-induced protection. Pardo V, González-Rodríguez Á, Muntané J, Kozma SC, Valverde ÁM. Food Chem Toxicol. 2015;80:298–309. doi: 10.1016/j.fct.2015.03.029. [DOI] [PubMed] [Google Scholar]
- 24.From NASH to diabetes and from diabetes to NASH: mechanisms and treatment options. Gastaldelli A, Cusi K. JHEP Rep. 2019;1:312–328. doi: 10.1016/j.jhepr.2019.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Interorgan metabolic crosstalk in human insulin resistance. Gancheva S, Jelenik T, Álvarez-Hernández E, Roden M. Physiol Rev. 2018;98:1371–1415. doi: 10.1152/physrev.00015.2017. [DOI] [PubMed] [Google Scholar]
- 26.Fatty acid sources and their fluxes as they contribute to plasma triglyceride concentrations and fatty liver in humans. Jacome-Sosa MM, Parks EJ. Curr Opin Lipidol. 2014;25:213–220. doi: 10.1097/MOL.0000000000000080. [DOI] [PubMed] [Google Scholar]
- 27.NAFLD and diabetes mellitus. Tilg H, Moschen AR, Roden M. Nat Rev Gastroenterol Hepatol. 2017;14:32–42. doi: 10.1038/nrgastro.2016.147. [DOI] [PubMed] [Google Scholar]
- 28.Nonalcoholic fatty liver disease as a nexus of metabolic and hepatic diseases. Samuel VT, Shulman GI. Cell Metab. 2018;27:22–41. doi: 10.1016/j.cmet.2017.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mechanisms of NAFLD development and therapeutic strategies. Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Nat Med. 2018;24:908–922. doi: 10.1038/s41591-018-0104-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Association between diabetes, family history of diabetes, and risk of nonalcoholic steatohepatitis and fibrosis. Loomba R, Abraham M, Unalp A, Wilson L, Lavine J, Doo E, Bass NM. Hepatology. 2012;56:943–951. doi: 10.1002/hep.25772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.A stress signaling pathway in adipose tissue regulates hepatic insulin resistance. Sabio G, Das M, Mora A, et al. Science. 2008;322:1539–1543. doi: 10.1126/science.1160794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.The role of cytokines in non-alcoholic fatty liver disease. Tilg H. Dig Dis. 2010;28:179–185. doi: 10.1159/000282083. [DOI] [PubMed] [Google Scholar]
- 33.Hepatocyte-secreted DPP4 in obesity promotes adipose inflammation and insulin resistance. Ghorpade DS, Ozcan L, Zheng Z, et al. Nature. 2018;555:673–677. doi: 10.1038/nature26138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Ozcan U, Cao Q, Yilmaz E, et al. Science. 2004;306:457–461. doi: 10.1126/science.1103160. [DOI] [PubMed] [Google Scholar]
- 35.Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. [ Jun; 2022 ];Chen Z, Tian R, She Z, Cai J, Li H. Free Radic Biol Med. 2020 152:116–141. doi: 10.1016/j.freeradbiomed.2020.02.025. [DOI] [PubMed] [Google Scholar]
- 36.Oxidative stress: from basic research to clinical application. Sies H. Am J Med. 1991;91:0–8. doi: 10.1016/0002-9343(91)90281-2. [DOI] [PubMed] [Google Scholar]
- 37.Mitochondrial adaptation in nonalcoholic fatty liver disease: novel mechanisms and treatment strategies. Sunny NE, Bril F, Cusi K. Trends Endocrinol Metab. 2017;28:250–260. doi: 10.1016/j.tem.2016.11.006. [DOI] [PubMed] [Google Scholar]
- 38.Oxidative stress and antioxidant biomarkers in clinical and experimental models of non-alcoholic fatty liver disease. Ore A, Akinloye OA. Medicina (Kaunas) 2019;55:26. doi: 10.3390/medicina55020026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Complex I-mediated reactive oxygen species generation: modulation by cytochrome c and NAD(P)+ oxidation-reduction state. Kushnareva Y, Murphy AN, Andreyev A. Biochem J. 2002;368:545–553. doi: 10.1042/BJ20021121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Oxidative stress as a critical factor in nonalcoholic fatty liver disease pathogenesis. Spahis S, Delvin E, Borys JM, Levy E. Antioxid Redox Signal. 2017;26:519–541. doi: 10.1089/ars.2016.6776. [DOI] [PubMed] [Google Scholar]
- 41.Molecular insights into the role of mitochondria in non-alcoholic fatty liver disease. Lee J, Park JS, Roh YS. Arch Pharm Res. 2019;42:935–946. doi: 10.1007/s12272-019-01178-1. [DOI] [PubMed] [Google Scholar]
- 42.Targeting hepatic macrophages to treat liver diseases. Tacke F. J Hepatol. 2017;66:1300–1312. doi: 10.1016/j.jhep.2017.02.026. [DOI] [PubMed] [Google Scholar]
- 43.Both bone marrow-derived and non-bone marrow-derived cells contribute to AIM2 and NLRP3 inflammasome activation in a MyD88-dependent manner in dietary steatohepatitis. Csak T, Pillai A, Ganz M, et al. Liver Int. 2014;34:1402–1413. doi: 10.1111/liv.12537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Macrophages in nonalcoholic steatohepatitis: friend or foe? Grunhut J, Wang W, Aykut B, Gakhal I, Torres-Hernandez A, Miller G. https://pubmed.ncbi.nlm.nih.gov/29930864/ Eur Med J Hepatol. 2018;6:100–109. [PMC free article] [PubMed] [Google Scholar]
- 45.Liver transplantation and non-alcoholic fatty liver disease. Zezos P, Renner EL. World J Gastroenterol. 2014;20:15532–15538. doi: 10.3748/wjg.v20.i42.15532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Nonalcoholic steatohepatitis: the new frontier for liver transplantation. Bzowej NH. Curr Opin Organ Transplant. 2018;23:169–174. doi: 10.1097/MOT.0000000000000502. [DOI] [PubMed] [Google Scholar]
- 47.Nonalcoholic fatty liver disease and hepatocellular carcinoma: a weighty connection. Starley BQ, Calcagno CJ, Harrison SA. Hepatology. 2010;51:1820–1832. doi: 10.1002/hep.23594. [DOI] [PubMed] [Google Scholar]
- 48.Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Zheng Y, Ley SH, Hu FB. Nat Rev Endocrinol. 2018;14:88–98. doi: 10.1038/nrendo.2017.151. [DOI] [PubMed] [Google Scholar]
- 49.Nonalcoholic fatty liver disease: a systematic review. Rinella ME. JAMA. 2015;313:2263–2273. doi: 10.1001/jama.2015.5370. [DOI] [PubMed] [Google Scholar]
- 50.Association of non-alcoholic fatty liver disease with chronic kidney disease: a systematic review and meta-analysis. Musso G, Gambino R, Tabibian JH, et al. PLoS Med. 2014;11:0. doi: 10.1371/journal.pmed.1001680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.COVID-19 and non-alcoholic fatty liver disease: two intersecting pandemics. Portincasa P, Krawczyk M, Smyk W, Lammert F, Di Ciaula A. Eur J Clin Invest. 2020;50:0. doi: 10.1111/eci.13338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gender and racial differences in nonalcoholic fatty liver disease. Pan JJ, Fallon MB. World J Hepatol. 2014;6:274–283. doi: 10.4254/wjh.v6.i5.274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Developmental origins of nonalcoholic fatty liver disease. Brumbaugh DE, Friedman JE. Pediatr Res. 2014;75:140–147. doi: 10.1038/pr.2013.193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Epidemiology and natural history of non-alcoholic fatty liver disease. Fazel Y, Koenig AB, Sayiner M, Goodman ZD, Younossi ZM. Metabolism. 2016;65:1017–1025. doi: 10.1016/j.metabol.2016.01.012. [DOI] [PubMed] [Google Scholar]
- 55.Prevalence of nonalcoholic fatty liver disease in the United States: the Third National Health and Nutrition Examination Survey, 1988-1994. Lazo M, Hernaez R, Eberhardt MS, et al. Am J Epidemiol. 2013;178:38–45. doi: 10.1093/aje/kws448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Epidemiology of non-alcoholic and alcoholic fatty liver diseases. Mitra S, De A, Chowdhury A. Transl Gastroenterol Hepatol. 2020;5:16. doi: 10.21037/tgh.2019.09.08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.The burden of liver disease in Europe: a review of available epidemiological data. Blachier M, Leleu H, Peck-Radosavljevic M, Valla DC, Roudot-Thoraval F. J Hepatol. 2013;58:593–608. doi: 10.1016/j.jhep.2012.12.005. [DOI] [PubMed] [Google Scholar]
- 58.NAFLD in Asia--as common and important as in the West. Farrell GC, Wong VW, Chitturi S. Nat Rev Gastroenterol Hepatol. 2013;10:307–318. doi: 10.1038/nrgastro.2013.34. [DOI] [PubMed] [Google Scholar]
- 59.Non-alcoholic fatty liver disease in Africa: a hidden danger. Paruk IM, Pirie FJ, Motala AA. Glob Health Epidemiol Genom. 2019;4:0. doi: 10.1017/gheg.2019.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.The epidemiology of nonalcoholic fatty liver disease: a global perspective. Lazo M, Clark JM. Semin Liver Dis. 2008;28:339–350. doi: 10.1055/s-0028-1091978. [DOI] [PubMed] [Google Scholar]
- 61.Racial disparities in diagnosis and prognosis of nonalcoholic fatty liver disease. Samji NS, Snell PD, Singal AK, Satapathy SK. Clin Liver Dis (Hoboken) 2020;16:66–72. doi: 10.1002/cld.948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Prevalence and associated factors of non-alcoholic fatty liver disease in patients with type-2 diabetes mellitus. Leite NC, Salles GF, Araujo AL, Villela-Nogueira CA, Cardoso CR. Liver Int. 2009;29:113–119. doi: 10.1111/j.1478-3231.2008.01718.x. [DOI] [PubMed] [Google Scholar]
- 63.Mechanism linking diabetes mellitus and obesity. Al-Goblan AS, Al-Alfi MA, Khan MZ. Diabetes Metab Syndr Obes. 2014;7:587–591. doi: 10.2147/DMSO.S67400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Nonalcoholic fatty liver disease: an emerging threat to obese and diabetic individuals. Masuoka HC, Chalasani N. Ann N Y Acad Sci. 2013;1281:106–122. doi: 10.1111/nyas.12016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Fatty liver index vs waist circumference for predicting non-alcoholic fatty liver disease. Motamed N, Sohrabi M, Ajdarkosh H, et al. World J Gastroenterol. 2016;22:3023–3030. doi: 10.3748/wjg.v22.i10.3023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Body mass index and risk of nonalcoholic fatty liver disease: two electronic health record prospective studies. Loomis AK, Kabadi S, Preiss D, et al. J Clin Endocrinol Metab. 2016;101:945–952. doi: 10.1210/jc.2015-3444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Role of diet and lifestyle changes in nonalcoholic fatty liver disease. Nseir W, Hellou E, Assy N. World J Gastroenterol. 2014;20:9338–9344. doi: 10.3748/wjg.v20.i28.9338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Association of sugar sweetened beverages consumption with non-alcoholic fatty liver disease: a systematic review and meta-analysis. Asgari-Taee F, Zerafati-Shoae N, Dehghani M, Sadeghi M, Baradaran HR, Jazayeri S. Eur J Nutr. 2019;58:1759–1769. doi: 10.1007/s00394-018-1711-4. [DOI] [PubMed] [Google Scholar]
- 69.Nonalcoholic fatty liver disease in the Rotterdam Study: about muscle mass, sarcopenia, fat mass, and fat distribution. Alferink LJM, Trajanoska K, Erler NS, et al. J Bone Miner Res. 2019;34:1254–1263. doi: 10.1002/jbmr.3713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Sarcopenia is an independent risk factor for non-alcoholic steatohepatitis and significant fibrosis. Koo BK, Kim D, Joo SK, et al. J Hepatol. 2017;66:123–131. doi: 10.1016/j.jhep.2016.08.019. [DOI] [PubMed] [Google Scholar]
- 71.NAFLD and NASH in postmenopausal women: implications for diagnosis and treatment. DiStefano JK. Endocrinology. 2020;161:0. doi: 10.1210/endocr/bqaa134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.NAFLD as a sexual dimorphic disease: role of gender and reproductive status in the development and progression of nonalcoholic fatty liver disease and inherent cardiovascular risk. Ballestri S, Nascimbeni F, Baldelli E, Marrazzo A, Romagnoli D, Lonardo A. Adv Ther. 2017;34:1291–1326. doi: 10.1007/s12325-017-0556-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Independent predictors of fibrosis in patients with nonalcoholic fatty liver disease. Hossain N, Afendy A, Stepanova M, et al. Clin Gastroenterol Hepatol. 2009;7:1224–1229. doi: 10.1016/j.cgh.2009.06.007. [DOI] [PubMed] [Google Scholar]
- 74.Factors predicting non-alcoholic steatohepatitis (NASH) and advanced fibrosis in patients with non-alcoholic fatty liver disease (NAFLD) Tasneem AA, Luck NH, Majid Z. Trop Doct. 2018;48:107–112. doi: 10.1177/0049475517742261. [DOI] [PubMed] [Google Scholar]
- 75.Sphingolipids in non-alcoholic fatty liver disease and hepatocellular carcinoma: ceramide turnover. Simon J, Ouro A, Ala-Ibanibo L, Presa N, Delgado TC, Martínez-Chantar ML. Int J Mol Sci. 2019;21:40. doi: 10.3390/ijms21010040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Non-alcoholic fatty liver disease: insights from sphingolipidomics. Montefusco DJ, Allegood JC, Spiegel S, Cowart LA. Biochem Biophys Res Commun. 2018;504:608–616. doi: 10.1016/j.bbrc.2018.05.078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Serum alanine aminotransferase predicts the histological course of non-alcoholic steatohepatitis in Japanese patients. Seko Y, Sumida Y, Tanaka S, et al. Hepatol Res. 2015;45:0–61. doi: 10.1111/hepr.12456. [DOI] [PubMed] [Google Scholar]
- 78.Non-alcoholic steatohepatitis: a review of its mechanism, models and medical treatments. Peng C, Stewart AG, Woodman OL, Ritchie RH, Qin CX. Front Pharmacol. 2020;11:603926. doi: 10.3389/fphar.2020.603926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Evidence of NAFLD progression from steatosis to fibrosing-steatohepatitis using paired biopsies: implications for prognosis and clinical management. McPherson S, Hardy T, Henderson E, Burt AD, Day CP, Anstee QM. J Hepatol. 2015;62:1148–1155. doi: 10.1016/j.jhep.2014.11.034. [DOI] [PubMed] [Google Scholar]
- 80.Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Vernon G, Baranova A, Younossi ZM. Aliment Pharmacol Ther. 2011;34:274–285. doi: 10.1111/j.1365-2036.2011.04724.x. [DOI] [PubMed] [Google Scholar]
- 81.Long term prognosis of fatty liver: risk of chronic liver disease and death. Dam-Larsen S, Franzmann M, Andersen IB, et al. Gut. 2004;53:750–755. doi: 10.1136/gut.2003.019984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Fibrosis progression in nonalcoholic fatty liver vs nonalcoholic steatohepatitis: a systematic review and meta-analysis of paired-biopsy studies. Singh S, Allen AM, Wang Z, Prokop LJ, Murad MH, Loomba R. Clin Gastroenterol Hepatol. 2015;13:643–654. doi: 10.1016/j.cgh.2014.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Genetic predisposition in nonalcoholic fatty liver disease. Sookoian S, Pirola CJ. Clin Mol Hepatol. 2017;23:1–12. doi: 10.3350/cmh.2016.0109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Romeo S, Kozlitina J, Xing C, et al. Nat Genet. 2008;40:1461–1465. doi: 10.1038/ng.257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Meta-analysis of the influence of I148M variant of patatin-like phospholipase domain containing 3 gene (PNPLA3) on the susceptibility and histological severity of nonalcoholic fatty liver disease. Sookoian S, Pirola CJ. Hepatology. 2011;53:1883–1894. doi: 10.1002/hep.24283. [DOI] [PubMed] [Google Scholar]
- 86.A sequence variation (I148M) in PNPLA3 associated with nonalcoholic fatty liver disease disrupts triglyceride hydrolysis. He S, McPhaul C, Li JZ, et al. J Biol Chem. 2010;285:6706–6715. doi: 10.1074/jbc.M109.064501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Carriage of the PNPLA3 rs738409 C >G polymorphism confers an increased risk of non-alcoholic fatty liver disease associated hepatocellular carcinoma. Liu YL, Patman GL, Leathart JB, et al. J Hepatol. 2014;61:75–81. doi: 10.1016/j.jhep.2014.02.030. [DOI] [PubMed] [Google Scholar]
- 88.The MBOAT7 variant rs641738 alters hepatic phosphatidylinositols and increases severity of non-alcoholic fatty liver disease in humans. Luukkonen PK, Zhou Y, Hyötyläinen T, et al. J Hepatol. 2016;65:1263–1265. doi: 10.1016/j.jhep.2016.07.045. [DOI] [PubMed] [Google Scholar]
- 89.The MBOAT7-TMC4 variant rs641738 increases risk of nonalcoholic fatty liver disease in individuals of European descent. Mancina RM, Dongiovanni P, Petta S, et al. Gastroenterology. 2016;150:1219–1230. doi: 10.1053/j.gastro.2016.01.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.rs641738C>T near MBOAT7 is associated with liver fat, ALT and fibrosis in NAFLD: a meta-analysis. Teo K, Abeysekera KW, Adams L, et al. J Hepatol. 2021;74:20–30. doi: 10.1016/j.jhep.2020.08.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.MBOAT7 rs641738 variant and hepatocellular carcinoma in non-cirrhotic individuals. Donati B, Dongiovanni P, Romeo S, et al. Sci Rep. 2017;7:4492. doi: 10.1038/s41598-017-04991-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Kozlitina J, Smagris E, Stender S, et al. Nat Genet. 2014;46:352–356. doi: 10.1038/ng.2901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.TM6SF2 rs58542926 influences hepatic fibrosis progression in patients with non-alcoholic fatty liver disease. Liu YL, Reeves HL, Burt AD, et al. Nat Commun. 2014;5:4309. doi: 10.1038/ncomms5309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Genetics and epigenetics of NAFLD and NASH: clinical impact. Eslam M, Valenti L, Romeo S. J Hepatol. 2018;68:268–279. doi: 10.1016/j.jhep.2017.09.003. [DOI] [PubMed] [Google Scholar]
- 95.PNPLA3 and TM6SF2 polymorphisms in Brazilian patients with nonalcoholic fatty liver disease. Lisboa QC, Nardelli MJ, Pereira PA, et al. World J Hepatol. 2020;12:792–806. doi: 10.4254/wjh.v12.i10.792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Genome-wide association analysis identifies variants associated with nonalcoholic fatty liver disease that have distinct effects on metabolic traits. Speliotes EK, Yerges-Armstrong LM, Wu J, et al. PLoS Genet. 2011;7:0. doi: 10.1371/journal.pgen.1001324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.A protein-truncating HSD17B13 variant and protection from chronic liver disease. Abul-Husn NS, Cheng X, Li AH, et al. N Engl J Med. 2018;378:1096–1106. doi: 10.1056/NEJMoa1712191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.IL28B and PNPLA3 polymorphisms affect histological liver damage in patients with non-alcoholic fatty liver disease. Petta S, Grimaudo S, Cammà C, et al. J Hepatol. 2012;56:1356–1362. doi: 10.1016/j.jhep.2012.01.007. [DOI] [PubMed] [Google Scholar]
- 99.The SOD2 C47T polymorphism influences NAFLD fibrosis severity: evidence from case-control and intra-familial allele association studies. Al-Serri A, Anstee QM, Valenti L, et al. J Hepatol. 2012;56:448–454. doi: 10.1016/j.jhep.2011.05.029. [DOI] [PubMed] [Google Scholar]
- 100.Promoting genetics in non-alcoholic fatty liver disease: combined risk score through polymorphisms and clinical variables. Vespasiani-Gentilucci U, Gallo P, Dell'Unto C, Volpentesta M, Antonelli-Incalzi R, Picardi A. World J Gastroenterol. 2018;24:4835–4845. doi: 10.3748/wjg.v24.i43.4835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Non-invasive stratification of hepatocellular carcinoma risk in non-alcoholic fatty liver using polygenic risk scores. Bianco C, Jamialahmadi O, Pelusi S, et al. J Hepatol. 2021;74:775–782. doi: 10.1016/j.jhep.2020.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Genetic variants in COL13A1, ADIPOQ and SAMM50, in addition to the PNPLA3 gene, confer susceptibility to elevated transaminase levels in an admixed Mexican population. Larrieta-Carrasco E, Flores YN, Macías-Kauffer LR, et al. Exp Mol Pathol. 2018;104:50–58. doi: 10.1016/j.yexmp.2018.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Exome-wide association study on alanine aminotransferase identifies sequence variants in the GPAM and APOE associated with fatty liver disease. Jamialahmadi O, Mancina RM, Ciociola E, et al. Gastroenterology. 2021;160:1634–1646. doi: 10.1053/j.gastro.2020.12.023. [DOI] [PubMed] [Google Scholar]
- 104.Epigenetics in non-alcoholic fatty liver disease. Lee J, Kim Y, Friso S, Choi SW. Mol Aspects Med. 2017;54:78–88. doi: 10.1016/j.mam.2016.11.008. [DOI] [PubMed] [Google Scholar]
- 105.Targeted-bisulfite sequence analysis of the methylation of CpG islands in genes encoding PNPLA3, SAMM50, and PARVB of patients with non-alcoholic fatty liver disease. Kitamoto T, Kitamoto A, Ogawa Y, et al. J Hepatol. 2015;63:494–502. doi: 10.1016/j.jhep.2015.02.049. [DOI] [PubMed] [Google Scholar]
- 106.Differential DNA methylation and changing cell-type proportions as fibrotic stage progresses in NAFLD. Johnson ND, Wu X, Still CD, et al. Clin Epigenetics. 2021;13:152. doi: 10.1186/s13148-021-01129-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Epigenetic modification of liver mitochondrial DNA is associated with histological severity of nonalcoholic fatty liver disease. Pirola CJ, Gianotti TF, Burgueño AL, et al. Gut. 2013;62:1356–1363. doi: 10.1136/gutjnl-2012-302962. [DOI] [PubMed] [Google Scholar]
- 108.Epigenetic regulation of peroxisome proliferator-activated receptor gamma mediates high-fat diet-induced non-alcoholic fatty liver disease. Hajri T, Zaiou M, Fungwe TV, Ouguerram K, Besong S. Cells. 2021;10:1355. doi: 10.3390/cells10061355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Lysine acetylation of CREBH regulates fasting-induced hepatic lipid metabolism. Kim H, Mendez R, Chen X, Fang D, Zhang K. Mol Cell Biol. 2015;35:4121–4134. doi: 10.1128/MCB.00665-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.SIRT1 is critical regulator of FOXO-mediated transcription in response to oxidative stress. Kobayashi Y, Furukawa-Hibi Y, Chen C, Horio Y, Isobe K, Ikeda K, Motoyama N. https://pubmed.ncbi.nlm.nih.gov/16012755/ Int J Mol Med. 2005;16:237–243. [PubMed] [Google Scholar]
- 111.Plasma levels of SIRT1 associate with non-alcoholic fatty liver disease in obese patients. Mariani S, Fiore D, Basciani S, et al. Endocrine. 2015;49:711–716. doi: 10.1007/s12020-014-0465-x. [DOI] [PubMed] [Google Scholar]
- 112.The role of sirtuin 2 in sustaining functional integrity of the liver. Park S, Chung MJ, Son JY, et al. Life Sci. 2021;285:119997. doi: 10.1016/j.lfs.2021.119997. [DOI] [PubMed] [Google Scholar]
- 113.Nonalcoholic steatohepatitis is associated with altered hepatic microRNA expression. Cheung O, Puri P, Eicken C, et al. Hepatology. 2008;48:1810–1820. doi: 10.1002/hep.22569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Disease-specific miR-34a as diagnostic marker of non-alcoholic steatohepatitis in a Chinese population. Liu XL, Pan Q, Zhang RN, et al. World J Gastroenterol. 2016;22:9844–9852. doi: 10.3748/wjg.v22.i44.9844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.A pilot study of serum microRNAs panel as potential biomarkers for diagnosis of nonalcoholic fatty liver disease. Tan Y, Ge G, Pan T, Wen D, Gan J. PLoS One. 2014;9:0. doi: 10.1371/journal.pone.0105192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Analysis of miRNAs profiles in serum of patients with steatosis and steatohepatitis. Vulf M, Shunkina D, Komar A, et al. Front Cell Dev Biol. 2021;9:736677. doi: 10.3389/fcell.2021.736677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.The essence of senescence. Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. Genes Dev. 2010;24:2463–2479. doi: 10.1101/gad.1971610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Evaluating causality of cellular senescence in non-alcoholic fatty liver disease. Meijnikman AS, Herrema H, Scheithauer TP, Kroon J, Nieuwdorp M, Groen AK. JHEP Rep. 2021;3:100301. doi: 10.1016/j.jhepr.2021.100301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Feedback between p21 and reactive oxygen production is necessary for cell senescence. Passos JF, Nelson G, Wang C, et al. Mol Syst Biol. 2010;6:347. doi: 10.1038/msb.2010.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. Passos JF, Saretzki G, Ahmed S, et al. PLoS Biol. 2007;5:0. doi: 10.1371/journal.pbio.0050110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Nonalcoholic steatohepatitis: association of insulin resistance and mitochondrial abnormalities. Sanyal AJ, Campbell-Sargent C, Mirshahi F, et al. Gastroenterology. 2001;120:1183–1192. doi: 10.1053/gast.2001.23256. [DOI] [PubMed] [Google Scholar]
- 122.Mitochondrial dysfunction in NASH: causes, consequences and possible means to prevent it. Begriche K, Igoudjil A, Pessayre D, Fromenty B. Mitochondrion. 2006;6:1–28. doi: 10.1016/j.mito.2005.10.004. [DOI] [PubMed] [Google Scholar]
- 123.Steatohepatitis-inducing drugs cause mitochondrial dysfunction and lipid peroxidation in rat hepatocytes. Berson A, De Beco V, Lettéron P, et al. Gastroenterology. 1998;114:764–774. doi: 10.1016/s0016-5085(98)70590-6. [DOI] [PubMed] [Google Scholar]
- 124.The p66shc-mediated regulation of hepatocyte senescence influences hepatic steatosis in nonalcoholic fatty liver disease. Zhang J, Li Y, Wang B, Luo Y, Shi J, Zhao B. Med Sci Monit. 2020;26:0. doi: 10.12659/MSM.921887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Cellular senescence drives age-dependent hepatic steatosis. Ogrodnik M, Miwa S, Tchkonia T, et al. Nat Commun. 2017;8:15691. doi: 10.1038/ncomms15691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Detection of DNA damage response in nonalcoholic fatty liver disease via p53-binding protein 1 nuclear expression. Akazawa Y, Nakashima R, Matsuda K, et al. Mod Pathol. 2019;32:997–1007. doi: 10.1038/s41379-019-0218-8. [DOI] [PubMed] [Google Scholar]
- 127.Study of cellular senescence and vitamin D deficiency in nonalcoholic fatty liver disease and the potential protective effect of vitamin D supplementation. Al-Ghamdi HA, Al Fayez FF, Bima AI, Khawaji TM, Elsamanoudy AZ. J Clin Exp Hepatol. 2021;11:219–226. doi: 10.1016/j.jceh.2020.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Senescence marker protein-30/superoxide dismutase 1 double knockout mice exhibit increased oxidative stress and hepatic steatosis. Kondo Y, Masutomi H, Noda Y, et al. FEBS Open Bio. 2014;4:522–532. doi: 10.1016/j.fob.2014.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Mitochondria matter: systemic aspects of nonalcoholic fatty liver disease (NAFLD) and diagnostic assessment of liver function by stable isotope dynamic breath tests. Di Ciaula A, Calamita G, Shanmugam H, et al. Int J Mol Sci. 2021;22:7702. doi: 10.3390/ijms22147702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Mitochondrial mutations and genetic factors determining NAFLD risk. Dabravolski SA, Bezsonov EE, Baig MS, Popkova TV, Nedosugova LV, Starodubova AV, Orekhov AN. Int J Mol Sci. 2021;22:4459. doi: 10.3390/ijms22094459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.A position statement on NAFLD/NASH based on the EASL 2009 special conference. Ratziu V, Bellentani S, Cortez-Pinto H, Day C, Marchesini G. J Hepatol. 2010;53:372–384. doi: 10.1016/j.jhep.2010.04.008. [DOI] [PubMed] [Google Scholar]
- 132.Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Angulo P, Kleiner DE, Dam-Larsen S, et al. Gastroenterology. 2015;149:389–397. doi: 10.1053/j.gastro.2015.04.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.The natural history of nonalcoholic fatty liver disease with advanced fibrosis or cirrhosis: an international collaborative study. Bhala N, Angulo P, van der Poorten D, et al. Hepatology. 2011;54:1208–1216. doi: 10.1002/hep.24491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.The epidemiology of hepatocellular carcinoma in the USA. Kim HS, El-Serag HB. Curr Gastroenterol Rep. 2019;21:17. doi: 10.1007/s11894-019-0681-x. [DOI] [PubMed] [Google Scholar]
- 135.Effect of metabolic traits on the risk of cirrhosis and hepatocellular cancer in nonalcoholic fatty liver disease. Kanwal F, Kramer JR, Li L, et al. Hepatology. 2020;71:808–819. doi: 10.1002/hep.31014. [DOI] [PubMed] [Google Scholar]
- 136.Liver transplantation for nonalcoholic steatohepatitis in the US: temporal trends and outcomes. Cholankeril G, Wong RJ, Hu M, et al. Dig Dis Sci. 2017;62:2915–2922. doi: 10.1007/s10620-017-4684-x. [DOI] [PubMed] [Google Scholar]
- 137.Non-alcoholic fatty liver disease - a global public health perspective. Younossi ZM. J Hepatol. 2019;70:531–544. doi: 10.1016/j.jhep.2018.10.033. [DOI] [PubMed] [Google Scholar]
- 138.Non-alcoholic fatty liver disease and its relationship with cardiovascular disease and other extrahepatic diseases. Adams LA, Anstee QM, Tilg H, Targher G. Gut. 2017;66:1138–1153. doi: 10.1136/gutjnl-2017-313884. [DOI] [PubMed] [Google Scholar]
- 139.Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: a meta-analysis. Targher G, Byrne CD, Lonardo A, Zoppini G, Barbui C. J Hepatol. 2016;65:589–600. doi: 10.1016/j.jhep.2016.05.013. [DOI] [PubMed] [Google Scholar]
- 140.A systematic review: burden and severity of subclinical cardiovascular disease among those with nonalcoholic fatty liver; should we care? Oni ET, Agatston AS, Blaha MJ, et al. Atherosclerosis. 2013;230:258–267. doi: 10.1016/j.atherosclerosis.2013.07.052. [DOI] [PubMed] [Google Scholar]
- 141.Systemic complications of nonalcoholic fatty liver disease: when the liver is not an innocent bystander. Vanni E, Marengo A, Mezzabotta L, Bugianesi E. Semin Liver Dis. 2015;35:236–249. doi: 10.1055/s-0035-1562944. [DOI] [PubMed] [Google Scholar]
- 142.CKD and nonalcoholic fatty liver disease. Targher G, Chonchol MB, Byrne CD. Am J Kidney Dis. 2014;64:638–652. doi: 10.1053/j.ajkd.2014.05.019. [DOI] [PubMed] [Google Scholar]
- 143.Nonalcoholic fatty liver disease - a multisystem disease? Mikolasevic I, Milic S, Turk Wensveen T, et al. World J Gastroenterol. 2016;22:9488–9505. doi: 10.3748/wjg.v22.i43.9488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Management of moderate to severe psoriasis in patients with metabolic comorbidities. Gisondi P, Galvan A, Idolazzi L, Girolomoni G. Front Med (Lausanne) 2015;2:1. doi: 10.3389/fmed.2015.00001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Prevalence of non alcoholic fatty liver disease in premenopausal, posmenopausal and polycystic ovary syndrome women. The role of estrogens. Gutierrez-Grobe Y, Ponciano-Rodríguez G, Ramos MH, Uribe M, Méndez-Sánchez N. Ann Hepatol. 2010;9:402–409. [PubMed] [Google Scholar]
- 146.Association of obstructive sleep apnoea with the presence and severity of non-alcoholic fatty liver disease. A systematic review and meta-analysis. Musso G, Cassader M, Olivetti C, Rosina F, Carbone G, Gambino R. Obes Rev. 2013;14:417–431. doi: 10.1111/obr.12020. [DOI] [PubMed] [Google Scholar]
- 147.Modern approach to the clinical management of non-alcoholic fatty liver disease. Del Ben M, Polimeni L, Baratta F, Pastori D, Loffredo L, Angelico F. World J Gastroenterol. 2014;20:8341–8350. doi: 10.3748/wjg.v20.i26.8341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.A fad too far? Dietary strategies for the prevention and treatment of NAFLD. Moore MP, Cunningham RP, Dashek RJ, Mucinski JM, Rector RS. Obesity (Silver Spring) 2020;28:1843–1852. doi: 10.1002/oby.22964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.The role of vitamin E in the treatment of NAFLD. Perumpail BJ, Li AA, John N, et al. Diseases. 2018;6:86. doi: 10.3390/diseases6040086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Vitamin E and vitamin C treatment improves fibrosis in patients with nonalcoholic steatohepatitis. Harrison SA, Torgerson S, Hayashi P, Ward J, Schenker S. Am J Gastroenterol. 2003;98:2485–2490. doi: 10.1111/j.1572-0241.2003.08699.x. [DOI] [PubMed] [Google Scholar]
- 151.Vitamin E and the risk of prostate cancer: the Selenium and Vitamin E Cancer Prevention Trial (SELECT) Klein EA, Thompson IM Jr, Tangen CM, et al. JAMA. 2011;306:1549–1556. doi: 10.1001/jama.2011.1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Lipid-lowering agents in nonalcoholic fatty liver disease and steatohepatitis: human studies. Nseir W, Mograbi J, Ghali M. Dig Dis Sci. 2012;57:1773–1781. doi: 10.1007/s10620-012-2118-3. [DOI] [PubMed] [Google Scholar]
- 153.Liver transplantation for nonalcoholic steatohepatitis. Asrani SK. Clin Gastroenterol Hepatol. 2014;12:403–404. doi: 10.1016/j.cgh.2013.11.014. [DOI] [PubMed] [Google Scholar]
- 154.NAFLD and liver transplantation: disease burden, current management and future challenges. Burra P, Becchetti C, Germani G. JHEP Rep. 2020;2:100192. doi: 10.1016/j.jhepr.2020.100192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Bariatric surgery in patients with non-alcoholic fatty liver disease - from pathophysiology to clinical effects. Laursen TL, Hagemann CA, Wei C, Kazankov K, Thomsen KL, Knop FK, Grønbæk H. World J Hepatol. 2019;11:138–149. doi: 10.4254/wjh.v11.i2.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Armstrong MJ, Gaunt P, Aithal GP, et al. Lancet. 2016;13:679–690. doi: 10.1016/S0140-6736(15)00803-X. [DOI] [PubMed] [Google Scholar]
- 157.Betaine and nonalcoholic steatohepatitis: back to the future? Mukherjee S. World J Gastroenterol. 2011;17:3663–3664. doi: 10.3748/wjg.v17.i32.3663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Cenicriviroc for the treatment of liver fibrosis in adults with nonalcoholic steatohepatitis: AURORA phase 3 study design. Anstee QM, Neuschwander-Tetri BA, Wong VW, et al. Contemp Clin Trials. 2020;89:105922. doi: 10.1016/j.cct.2019.105922. [DOI] [PubMed] [Google Scholar]
- 159.No anti-fibrotic effect of selonsertib in NASH. Dickson I. Nat Rev Gastroenterol Hepatol. 2020;17:260. doi: 10.1038/s41575-020-0297-5. [DOI] [PubMed] [Google Scholar]
- 160.Simtuzumab is ineffective for patients with bridging fibrosis or compensated cirrhosis caused by nonalcoholic steatohepatitis. Harrison SA, Abdelmalek MF, Caldwell S, et al. Gastroenterology. 2018;155:1140–1153. doi: 10.1053/j.gastro.2018.07.006. [DOI] [PubMed] [Google Scholar]