Next Article in Journal
Altered Metabolism in Glioblastoma: Myeloid-Derived Suppressor Cell (MDSC) Fitness and Tumor-Infiltrating Lymphocyte (TIL) Dysfunction
Next Article in Special Issue
Redox Signaling and Sarcopenia: Searching for the Primary Suspect
Previous Article in Journal
In Sickness and in Health: The Immunological Roles of the Lymphatic System
Previous Article in Special Issue
Mitochondrial Functioning and the Relations among Health, Cognition, and Aging: Where Cell Biology Meets Cognitive Science
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mitochondrial Mutations and Genetic Factors Determining NAFLD Risk

by
Siarhei A. Dabravolski
1,*,
Evgeny E. Bezsonov
2,3,
Mirza S. Baig
4,
Tatyana V. Popkova
5,
Ludmila V. Nedosugova
6,
Antonina V. Starodubova
7,8 and
Alexander N. Orekhov
2,3
1
Department of Clinical Diagnostics, Vitebsk State Academy of Veterinary Medicine [UO VGAVM], 7/11 Dovatora Str., 210026 Vitebsk, Belarus
2
Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia
3
Laboratory of Angiopathology, The Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
4
Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol 453552, India
5
V.A. Nasonova Institute of Rheumatology, 34A Kashirskoye Shosse, 115522 Moscow, Russia
6
I. M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubenskaya Street, 119991 Moscow, Russia
7
Federal Research Centre for Nutrition, Biotechnology and Food Safety, 2/14 Ustinsky Passage, 109240 Moscow, Russia
8
Pirogov Russian National Research Medical University, 1 Ostrovitianov Street, 117997 Moscow, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(9), 4459; https://doi.org/10.3390/ijms22094459
Submission received: 6 April 2021 / Revised: 21 April 2021 / Accepted: 22 April 2021 / Published: 24 April 2021
(This article belongs to the Special Issue Mitochondrial Dysfunction in Ageing and Diseases: Partie Deux)

Abstract

:
NAFLD (non-alcoholic fatty liver disease) is a widespread liver disease that is often linked with other life-threatening ailments (metabolic syndrome, insulin resistance, diabetes, cardiovascular disease, atherosclerosis, obesity, and others) and canprogress to more severe forms, such as NASH (non-alcoholic steatohepatitis), cirrhosis, and HCC (hepatocellular carcinoma). In this review, we summarized and analyzed data about single nucleotide polymorphism sites, identified in genes related to NAFLD development and progression. Additionally, the causative role of mitochondrial mutations and mitophagy malfunctions in NAFLD is discussed. The role of mitochondria-related metabolites of the urea cycle as a new non-invasive NAFLD biomarker is discussed. While mitochondria DNA mutations and SNPs (single nucleotide polymorphisms) canbe used as effective diagnostic markers and target for treatments, age and ethnic specificity should be taken into account.

1. Introduction

NAFLD is one of the most common chronic liver diseases worldwide. This disease is multifactorial and complex, involving many aetiological parameters, such as diet, genetic predisposition, and lifestyle, and it is often associated with DM (diabetes milieus), MetS (metabolic syndrome), obesity, and IR (insulin resistance). The main feature is the surplus fat accumulation in the liver (often called hepatosteatosis), not caused by trauma, considerable alcohol consumption, or inflammation. Without medical attention, NAFLD may further progress to NASH, which can be distinguished by chronic inflammation, fibrosis, damaged hepatocytes, and a higher risk of developing HCC and cirrhosis [1]. The NASH stage can develop over months and years; during this period, normal functional tissue is damaged and slowly replaced by scar tissue (fibrosis). Liver performance is decreasing gradually, which eventually often leads to complete failure, at which point liver transplantation may be required [2]. End-stage liver disease such as HCC can develop in NAFLD patients with rather low prevalence (less than 15%); however, such disease’s progression can be independent of cirrhosis status [3].
Today, NAFLD is present in about 24% of the world’s population, among high-risk groups: >80% of obese, 60% of diabetic, and 20% of lean people [4]. However, the worldwide prognosis for NAFLD is negative, suggesting further growth in the number of cases, the increased death rate from cirrhosis, and HCC [5].
Despite its critical importance, the effective treatment for NAFLD is still missing. However, many drugs are ongoing clinical trials [6]. Early and precise diagnostic criteria are crucial for effective therapy, today relying on lifestyle interventions and the amelioration of NAFLD-related complications [7]. The stage of fibrosis is the main marker, used to monitor the progression of the diseases, assign treatment, and predict a long-term outcome [8]. Current methods rely on invasive procedures (like biopsies) and non-invasive methods (blood-markers measurements and image-based). Invasive procedures are expensive and associated with further complications and represent only a small fraction of the liver, which can provide misleading results [9]. Blood-based tests rely on measurements of NAFLD-related markers and general indicators of liver and organism performance. Among others, the most popular criteria are the NAFLD fibrosis score (NFS), AST-to-platelet ratio index (APRI), and enhanced liver fibrosis test (ELF) [10]. Image-based methods (mainly ultrasound, spectroscopy, and magnetic resonance imaging (MRI)) aim to visualize and quantify changes in liver elasticity and hepatic fat content [11]. However, although MRI tools are very indicative, they are expensive and not available for wide application. Modern diagnostic strategies include also “omics” investigation, where lipidomic, proteomic, microRNA, and gut microbiome profiles are used to monitor liver status and disease progression [12].
The current understanding of the NAFLD pathogenesis process relies on the theory of so-called “hits”, where the first “hit” is based on the excess number of FAs (fatty acids) and cholesterol in the liver and circulation. Normally, FAs are metabolized into triglycerides, but due to the impaired β-oxidation, lipotoxic species of saturated FAs and oxidized cholesterol are formed and accumulated in hepatocytes [13]. Subsequent hepatocytes damage leads to apoptosis, necrosis, and necroptosis, causing the release of damage-associated molecular patterns, wide metabolic changes, and formation of the pro-inflammatory environment (known as sterile injury) where activated Kupffer cells and pro-inflammatory (M1-like) macrophages are associated with secreted chemo attractants and stimulating cytokines [14]. However, we should notice that inflammatory response is also an important part of the tissue repair and healing processes; thus, immune cells can release similar mediators in the early stages of liver injury [15]. Thus, immune cells and cytokines can serve as NAFLD/NASH biomarkers; however, obtained information would represent rather system-level inflammation, and every biomarker (or their combination) should be evaluated and supported by other diagnostic tools [16].
In the second “hit”, adipose tissue plays the main role as a source of inflammation [17]. Overrepresented macrophages (such as CCR2+ and CD11c+, CD206) can be found in liver and adipose tissue and are accompanied by the release of a high amount of pro-inflammatory cytokines and chemokines in the circulation. This “hit” (often called the adipose tissue–liver axis), together with the progression of liver disease, stimulates the development of IR and powers local and systemic chronic inflammation [18].
In this review, we have analyzed recently identified mutations in mitochondrial DNA and SNPs in nuclear genes, linked with NAFLD. The causative role of mitophagy, mitochondria-delivered ROS, lipotoxic by-products of β-oxidation, and mtDNA in NAFLD development and progression is shown. Additionally, urea cycle metabolites are suggested as a new non-invasive biomarker to evaluate the efficiency of mitochondria performance and the NAFLD stage. The majority of identified SNPs are involved in lipid/glucose metabolism, inflammation, and carcinogenesis. In total, identified SNPs and mtDNA mutations can be used for early NAFLD prediction, diagnosis, and to monitor the progression to more severe forms of the disease (NASH, cirrhosis, and HCC).

2. Considering NAFLD/NASH as a Mitochondrial Disease

2.1. Mitochondrial Mutations at the Origins of Liver Inflammation

Chronic inflammation, fibrosis, and cell death are the main drivers of NAFLD progression [19]. Many studies have supported the view that hepatic steatosis (fat accumulation) can facilitate oxidative stress, inflammation, and activate fibrogenic machinery [20]. Others have focused on the abnormal hepatic FAO (fatty acid oxidation) where mitochondrial malfunctions have been involved [21]. Such mitochondria produce less ATPs and more ROS, and due to the incomplete FAO, also release toxic lipid intermediates that can cause further liver injury [22,23]. Simultaneously, the antioxidant biosynthesis system is not sufficient to neutralize surplus ROS; thus, OS also contributes to the hepatocytes’ stress pathways, inflammation, fibrogenesis, and further NAFLD progression to NASH [24].
Recent research has highlighted that the liver mtDNA of NAFLD patients has a higher mutation rate in comparison to healthy people [25]. The expression analysis of the identified mtDNA mutations and polymorphic sites suggests that the OXPHOS (oxidative phosphorylation) is their primary target, leading to phenotype manifestation. Interestingly, identified mutations were common for the entire body and not somatic, while in patients with advanced fibrosis, damaging somatic mutations have been found in the mtCYB gene [25]. The similarity of liver and blood mtDNA mutations suggests that they can be inherited from the mother, not developed de novo. This assumption is supported by many cases of NAFLD development in early childhood when paediatric NAFLD was found to be associated not only with nutrition but also with MetS, T2DM, and the obesity of mothers [26]. In addition to the mtDNA mutations, inherited nuclear mutations may also be the primary cause of OS and higher mtDNA variability. Common POLG missense mutation p.Gln1236His can be a good example, because it is known to deplete the liver mtDNA [25].
Another set of evidence was found in Turkey NASH patients, where the disease progression was associated with nucleotide variations in the D-loop region. It was found that the Mt16318C→A variant was associated with NASH, while the Mt16129AA genotype was associated with more advanced stages of fibrosis. Similarly, other genotypes have been associated with different stages of the disease and other symptoms: the Mt16249 CC—advanced steatosis and lobular inflammation; the Mt16296 TT—hypothyroidism; the Mt16163 GG and Mt16294 TT—MetS; and the Mt16256 TT+CT genotypes—T2DM [27].
Accumulation of mutations in the mtCYB gene was found to be closely related to the NAFLD severity [28]. The mitochondrial cytochrome b is a fundamental part of Complex III, responsible for the electron transfer from coenzyme Q to cytochrome c, with further proton gradient generation and ATP synthesis. A complex III deficiency can cause different clinical manifestation; depending on the affected tissues, it can be multisystem disorders such as deafness, muscle weakness, cataract, epilepsy, cardiomyopathy, mitochondrial myopathy, and others [29]. NASH patients have a higher mtCYB mutations rate, leading to progressive chronic liver degeneration. This process is accompanied by increased release of the oncogenic metabolites (such as 2-hydroxyglutarate), DNA-damaging ROS, lipid peroxyl radicals, and global changes in the liver transcriptome [28].
In summary, mitochondrial mutations play an important role in NAFLD and NASH development and are associated with severe adipose tissue inflammation. Mitochondria are responsible for energy production via β-oxidation of FAs and are also the most important producer of cellular ROS. ROS and other toxic by-products of lipid peroxidation are damaging respiratory chain proteins and mtDNA. Accumulation of mtDNA and surplus ROS production are important hallmarks of NAFLD progression to NASH, cirrhosis, and, finally, liver failure.

2.2. Disrupted Mitophagy and NAFLD Progression

Mitophagy is the specialised form of autophagy, targeting the degradation of damaged or malfunctional mitochondria to sustain energy homeostasis. The process of mitochondria turnover via mitophagy is part of many normal physiological events, such as development, cell differentiation, and response to stresses and damage. There are several distinguished stages of mitophagy: initiation, membrane nucleation, phagophore formation and expansion, fusion with the lysosome, and, on the final stage, degradation [30]. In general, mitophagy can be classified as PINK1-dependent and Parkin-in/dependent pathways, while both pathways rely on LC3 (light chain, microtubule-associated protein). LC3 proteins (I and II) are crucial for mitophagy; however, the LC3 deletion can be compensated to restore the normal auto/mitophagy [31].
In the PINK1/Parkin pathway, PINK1 senses mitochondrial potential, and, without a specific trigger, is imported to and degraded in the mitochondria. When the mitochondrial potential is out of normal range, PINK1 stabilizes on the OMM (outer mitochondrial membrane) and recruits E3 ubiquitin ligase Parkin. Further, Parkin ubiquitinates several OMM proteins (mitofusins MFN1 and MFN2, VDAC (voltage-dependent anion channel) and others), and initiates phagophore formation and engulfment. Except for Parkin, other E3 ubiquitin ligases can be involved in PINK1-dependent mitophagy (such as SIAH1 and ARIH1). Receptor-mediated mitophagy (Parkin-independent) pathways can be facilitated by OMM (BCL2L13, BNIP3, FKBP8, NIX, and FUNDC1) and IMM (cardiolipin and PHB2) proteins, and some other receptors (p62, OPTN, AMBRA1, NBR1, TAXIBP1, and NDP52) [32].
Several studies have demonstrated a decreased efficiency of mitophagy in the context of aging and an unbalanced diet with NAFLD. Fat-induced liver damage is associated with inhibited mitophagy and following the mitochondria-mediated death of hepatocytes [33]. However, this also suggests several potential mitochondrial targets that can be used as a treatment to reduce inflammation via activated mitophagy, or as a prevention therapy to enhance mitophagy and protect against surplus lipid accumulation in the liver [34,35].
Mitochondria use FAO as the main energy source to fuel the cells. Excess substrates for the β-oxidations overload the mitochondrial respiratory chain and favor higher ROS production, which leads to a further dysregulation in mitochondrial functions, structure, transcription, and replication [36].
There are several hypotheses explaining the role of mitophagy in the NAFLD/NASH progression. Firstly, hyperlipidemia and hyperinsulinemia are suppressing mitophagy, which leads to the accumulation of lipogenic mitochondria [37]. This hypothesis is supported by the known effect of DM-related drugs, such as liraglutide, which is known to stimulate mitophagy and reduce inflammation and OS production [35]. Thus, effective and timely replacement of lipogenic mitochondria with functional fat-oxidizing mitochondria via the mitophagy process is crucial to prevent NAFLD progression. Additionally, it is known that NAFLD patients have lower acidity of the hepatic lysosomes, which cannot effectively recycle cellular components and sustain normal metabolic functions. As a result, it leads to an increased accumulation of damaged proteins and lipotoxic by-products and gradually increases hepatic TG content [38].
In total, ineffective β-oxidation and increased lipogenesis lead to lipids accumulation in the hepatocytes, ROS generation, and hepatocyte injury, promoting NAFLD progression to hepatic inflammation and fibrosis. This suggests that the stimulation of mitophagy may be a promising therapeutic strategy to limit ROS production and the subsequent death of hepatocytes.

2.3. Non-Invasive Biomarkers to Evaluate the Efficiency of Hepatic Mitochondria

Metabolic abnormalities can be detected in peripheral cells and used as biomarkers to estimate the NAFLD/NASH stage and liver status. The levels of OS and pro-inflammatory cytokines are the main markers to monitor NAFLD severity and progression [39]. Additionally, there are several metabolic pathways completely or partly localized to the mitochondria, such as citric acid and urea cycles. However, under NAFLD conditions, those pathways and their metabolites are compromised and thus can also can provide some insights into NAFLD pathogenesis and new targets for medical intervention.
Several mitochondria-related metabolites have been suggested as NAFLD markers: l-ornithine l-aspartate, citrulline/ornithine ratio, glutamine, glutathione, arginine, and glutamate [40]; and CPS-1 (carbamoyl phosphate synthase 1), OTC (Ornithine transcarbamoylase), FGF-21 (Fibroblast growth factor-21), and CK-18 (cytokeratin 18) [40,41]. As it was shown, dysfunctional mitochondria have affected CPS and OTC enzymes, leading to a compromised urea cycle and hyperammonemia (Figure 1). Additionally, hyperammonemia was associated with altered expression of the TLR-pathway, inflammation, increased OS, and hepatocyte apoptosis [42]. The mechanism is likely liver-specific, thus suggesting ammonia as a promising target for NAFLD treatment [43]. FGF-21 is a liver secreted hormone, hepatokine, regulating simple sugar intake in adipocytes and associated with T2DM and NAFLD. FGF-21 has a pleiotropic effect on metabolic homeostasis, which results in increased energy expenditure, fat utilization, and lipid excretion. On the cellular level, FGF-21 enhances mitochondrial oxidative capacity and induces several key metabolic genes via the AMPK/SIRT1 pathway [44].
Crucial metabolic pathways, such as the TCA cycle, β-oxidation of FAs, and several steps of the urea cycle are intersecting in mitochondria. The NAFLD progression to more severe forms is most likely based on the direct (from surplus FAs β-oxidation) and indirect (increased OS damage) injury. In this model, accumulated fat and related toxic by-products cause electron transfer chain malfunction, leading to reduced FAD and NAD production, which causes further defects in FAs oxidation and TCA cycle [45]. The particular importance of the urea cycle for the NAFLD progression can be explained by the exclusive location of the rate-limiting steps for ornithine and citrulline metabolism in liver mitochondria. An impaired urea cycle leads to reduced urea synthesis and increases ammonia concentration (hyperammonemia), which is associated with hepatic encephalopathy, inflammation, hepatocyte apoptosis, and high mortality [43]. Additionally, it is known that NASH patients with fibrosis have a much higher level of glutamate, which is also involved in both TCA and urea cycles. Thus, such a connection between TCA and urea cycles suggests that damage to the urea cycle can affect the TCA cycle and provide further cellular damage [46].
Those data suggested that pathological changes in mitochondria efficiency can be measured based on the urea cycle metabolites and thus serve as a non-invasive biomarker to monitor NAFLD severity and progression. Further analysis of urea-cycle-related metabolites may provide a better understanding of the involvement of hepatocyte mitochondria in the molecular mechanisms of NAFLD pathogenesis.

3. The Role of OS and Inflammation in NAFLD

NAFLD has become the primary chronic liver disease in both developing and developed countries, mostly due to sedentary lifestyle, lack of exercise, overnutrition, and poor feeding behavior, which are closely related to IR, T2DM, MetS, obesity, and other complications [47]. NASH, a more severe inflammatory form of the disease, in addition to the hepatocyte fat accumulation, requires other aetiological factors, such as mitochondrial oxidative injury. As a by-product of the OXPHOS process, mitochondria generate ROS, which is normally balanced by the scavenging antioxidant system, preventing cell damage [48]. However, an unbalanced ROS-antioxidant system leads to further disruption of mitochondrial functions, impaired mtDNA replication, and transcription, initiating a vicious circle [49]. A high-fat diet and overnutrition are the main triggers of the circle. Thus, excess substrates for the β-oxidations overload the mitochondrial respiratory chain and favors higher ROS production [36]. The current NASH development model suggests that fat-laden hepatocytes have shifted the redox balance with significant ROS overproduction by mitochondria. This model was proven on the experimental animals and humans, where hepatic mitochondrial capacity was challenged with high doses of available lipids [50].

3.1. OS Activates the NLRP3 Inflammasome and Triggers Chronic Inflammation State

The liver is responsible for the detoxification of harmful substances and endotoxins consumed with diet and created during different metabolic processes. Normally, antioxidants counterbalance ROS generated during detoxification reactions. However, under NAFLD conditions, the antioxidant pool is depleted, while ROS production is greatly increased. As a result, ROS causes damage to DNA, lipids, and proteins, forming oxidative damage products [51]. Following lipotoxic liver injury, ER stress and DNA damage promote further NAFLD inflammation and fibrotic progression [52].

3.1.1. The NLRP3 Inflammasome: An OS Sensor and Inflammation Driver

The NLRP3 inflammasome has a sensor (NLRP3), an adaptor (ASC), and an effector (caspase 1). NLRP3 is a three-domain protein, with an N-terminal pyrin domain (PYD), a central NACHT domain, and a C-terminal leucine-rich repeat domain (LRR domain). The self-regulation of NLRP3 relies on the ATPses activity of the NACHT domain, which can be autoinhibited by the LRR domain. An adaptor has two protein–protein interaction domains: N-terminal PYD and a C-terminal CARD (caspase recruitment domain). Caspase 1 has an N-terminal CARD domain and a C-terminal catalytic domain (large and small subunits) [53]. Stimulated NLRP3 can oligomerize via NACHT domain-mediated homotypic interactions with further recruitment of ASC through PYD–PYD interactions. Multiple ASC filaments combine into a speck structure, where caspase 1 is recruited through CARD–CARD interactions and self-activation [54].
Involvement of the NLRP3 inflammasome in an inflammatory process is a two-step process: priming and activation. Priming can be initiated through the PRRs (pattern recognition receptors such as TLRs (Toll-like receptors)), cytokines (TNFα, IL-1β), or NOD2 (nucleotide-binding oligomerization domain-containing protein 2), which activate NF-kB and up-regulate the expression of inflammasome components (caspase 1, NLRP3, and pro-IL-1β). Additionally, priming is stabilizing NLRP3 through PTMs (post-translational modifications): sumoylation, phosphorylation, and ubiquitylation [55].
Inflammasome activation via sensing of cellular stresses is not a fully elucidated process. However, it is known that NLRP3 can be activated by many unrelated stimuli, such as viral, bacterial or fungal infection, but also by internal sterile inflammation and environmental irritants. Among the recognized triggers, we can mention mitochondrial DNA and ROS, ion flux (K+, Cl, Ca2+), free fatty acids (FAs), and others [56]. While inflammasome description is far beyond the topic of this review, we wish to redirect interested readers to some more focused recent reviews [57,58].
Malfunctional mitochondria are one of the key NLRP3 activators. During cellular stress, ROS production by mitochondria is greatly increased. Mitophagy, as the main remover of damaged and retired mitochondria, is closely connected to the NLRP3 activation. Inhibited mitophagy and stimulated ROS production can significantly enhance NLRP3 activation [59]. FAs are known activators of NLRP3 inflammasome. AMPK, known to suppress inflammation by limiting ROS production and autophagy activation, thus inhibits NLRP3 activation. FAs, however, suppress AMPK and promote NLRP3 activation [60].
Activated NLRP3 inflammasome release the pro-inflammatory cytokines IL-1β and IL-18 and initiates pyroptosis, an inflammatory form of lytic programmed cell death. The key player of pyroptosis is gasdermin D (GSDMD), which is activated by caspase 1, binds to the plasma membrane and makes pores, thus, killing cells from within. GSDMD also can bind cardiolipin, presented in bacterial membranes, thus, demonstrating a bactericidal activity. Additionally, GSDMD can facilitate IL-1β and IL-18 release, thus, promoting both chronic and acute inflammatory conditions and leading to the emergence of inflammaging [61].

3.1.2. Chronic Inflammation

TLR signalling pathway is one of the key mechanisms involved in low-grade chronic inflammation state in obese, diabetic and NAFLD/NASH individuals. FAs and their derivatives enhance TLR4 activity in both liver and macrophages, and, together with oxLDLs, stimulate the NLRP3 inflammasome activation in atherosclerotic plaques. Further, generated ROS and NF-kB enhance the expression of pro-inflammatory genes. In addition to FAs, many derivatives (such as cholesterol crystals and ceramides) can prime and activate NLRP3 [62]. Prolonged TLR activity is leading to the chronic inflammation state and triggering the development of metabolic syndrome (MetS), which also fuels the inflammation pool, making this vicious cycle complete [63].
Endoplasmic reticulum (ER) stress is another pathway, linking inflammation, hepatocyte death and NAFLD progression. Severe ER stress can activate NLRP3 inflammasome and induce the inflammatory process, thus initiating and aggravating chronic diseases [64]. Chronically unresolved ER stress activates the so-called unfolded protein response (UPR), which uses chaperons to regain functional and properly folded proteins. UPR triggers a set of signalling pathways, leading to JNK phosphorylation and apoptosis, but also induction of NF-kD with the production of pro-inflammatory cytokines and following insulin resistance [65]. The UPR’s signalling mechanism is based on IRE1 (inositol-requiring enzyme 1) and transcription factor ATF6 (activating transcription factor 6). The IRE1 activates the NLRP3 inflammasome in the ASC-independent pathway and causes caspase-2-driven mitochondrial damage [66].
ER stress in combination with oxLDL up-regulates CD36 in macrophages, enhancing foam cell formation and thus promoting atherosclerosis progression and plaque necrosis [67]. In mitochondria, CD36 and oxLDL mediate the metabolic switch from oxidative phosphorylation to superoxide production and NF-kB activation. Thus, the CD36/oxLDL signalling pathway links FAs dysregulation, mitochondria ROS production, and chronic inflammation [68].
In total, the NLRP3 inflammasome itself, its upstream activators, and its downstream effectors are the main consequences of the OS and can serve as potential new targets against chronic inflammatory diseases such as NAFLD and related co-morbidities (atherosclerosis, MetS, IR, DM, and others).

4. Genetic Determinants of the NAFLD Pathogenesis

NAFLD pathogenesis includes environmental, behavioral, and genetic factors. Environmental and behavioral (such as nutrition, physical activities, feeding behavior, and pollutant levels in the environment), genetic (single nucleotide polymorphisms (SNPs) and mutations), and epigenetic (DNA methylation, expressed miRNAs, and histone modifications) factors interact and modulate individual risk of NAFLD development and the severity of progression [69]. It has been known for decades that some NAFLD susceptibility caused by genetic and epigenetic factors can be inherited [70]. Recent GWAS (genome-wide association studies) helped to define particular genes linked with the risk of NAFLD development [71] and NAFLD prevalence in different ethnic groups [72].
Candidate gene studies involve a detailed investigation of a small sample size, where a gene with known function is compared in NAFLD cases and healthy controls. GWAS requires huge sampling but is able to provide genetic association. During the past decades’ many genes, SNPs and epigenetic factors have been linked to NAFLD [69,73]. We wish to redirect interested readers to these reviews; here, we will further discuss recently identified mutations and SNPs (Table 1).
In general, identified SNPs and genes, associated with NAFLD/NASH, can be assigned to three groups: lipids/glucose metabolism-related, immune/inflammatory response, and other genes. Further, we will discuss several genes with well-studied functions and roles in different diseases.

4.1. SNPs in Genes Related to the Lipid/Glucose Metabolism

PNPLA3 (the Patatin-like phospholipase domain containing 3) gene is involved in the LD (lipid droplets) metabolism and VLDL (very-low-density lipoprotein) secretion; this gene’s SNP site was linked with NAFLD and studied by many researchers around the globe. The role of polymorphism in the PNPLA3 was shown to be the most crucial factor influencing the ethical differences in hepatic fat content [135]. In particular, the rs738409 polymorphism site (I148M substitution) was studied in several ethnic groups and found to be related to a higher risk of NAFLD development, elevated liver enzymes, and fibrosis [76,82,124,136]. Interestingly, the mitochondrial genome also plays a crucial role in NAFLD/NASH development and progression. As it was shown, the presence of haplogroup L has a protective role against NASH development [137]. There were several molecular mechanisms suggested, explaining the effect of I148M PNPLA3 substitution. The first one implies the involvement of PNPLA3 in the regulation of lipophagy, a specialized form of lipid droplets autophagy. PNPLA3 interacts with LC3-II, the central protein in autophagosome biogenesis, while in M148-PNPLA3 the level of lipophagy was decreased, which leads to slower LDs turnover and higher accumulation of intracellular lipids [138]. Another pathway links inflammation to the NAFLD progression, due to the higher inflammatory infiltration and liver damage found in NAFLD patients with PNPLA3 I148M mutation [139]. It was shown on HepG2 cells culture that NF-kB, the most crucial inflammation-regulating transcription factor, regulates PNPLA3 expression. Thus, the I148 PNPLA3 protein participates in palmitic-acid-induced inflammatory response through the ER stress pathway and increased TNF-α expression [140].
PON1 is the well-known member of the paraoxonase (PON) family, associated with HDL and acting like antioxidant-inhibiting LDL oxidation, thus having antiatherogenic effects [141]. The PON enzymes play an important role in lipid and glucose homeostasis and aging and are associated with different metabolic disorders: DM, NAFLD, CVD, neurodegenerative disease, and cancer [142,143]. Recently, PON1 genetic polymorphism and activity were linked to coronary heart disease in aged patients with confirmed DM; thus, PON1 can be considered as an additional diagnostic factor to evaluate cardiovascular risk [144]. Rs854560 polymorphism (L55M substitution) and low serum PON1 concentration were identified in NAFLD patients, suggesting their potential application in NAFLD early prediction and non-invasive diagnostics [74].
Perilipins (PLINs) proteins are the most abundant lipid droplet proteins, responsible for lipid storage. PLIN2 gene is constitutively expressed and correlates with lipid droplet density and TG content. Interestingly, plin2-/- mice have much lower TG content and are protected against fatty liver disease [145]. Recently identified among American NASH patients, the rs35568725 polymorphism site (Ser251Pro substitution) was suggested as a risk factor for NASH due to the effect on LD phenotype [75].
Recently, apolipoprotein was suggested as a diagnostic and therapeutic target of NASH. It was noticed that NASH patients have a tight correlation between abnormal apolipoprotein, increased liver fat content, and VLDL plasma concentration [146]. Interestingly, several polymorphism sites (rs10750097, rs1263173, rs17120035, and rs662799) of the APOA5 gene have been associated with NAFLD in the Chinese Han population [90]. Additionally, the relation of the APOC3 and APOE genes’ polymorphism sites to NAFLD was identified for other ethnic and age groups. The most probable mechanism is based on the role of the apolipoproteins in the accumulation of TG, which leads to lipotoxic liver injury [147].
The TM6SF2 gene (transmembrane 6 superfamily member 2) regulates VLDL metabolism via the reduction of apolipoproteins secretion. Polymorphism sites influence TM6SF2 protein’s stability and turnover, causing impaired ER-to-Golgi trafficking of VLDL particles and their accumulation in ER. TM6SF2 variants have been found in several ethnic groups and linked to lipid metabolism diseases [148]. Similarly, a variant of the FNDC5 gene, generating the soluble protein irisin, has altered protein stability, which is also influenced VLDL metabolism via apolipoprotein B gene regulation. An unstable FNDC5 variant resulted in increased steatosis, insulin resistance, decreased autophagic flux, and hepatocyte death [149].
PPARγ (peroxisome proliferator-activated receptor γ) is an important transcription factor, regulating many genes responsible for different aspects of cellular metabolism, differentiation, development, and tumorigenesis [150,151]. Four polymorphism sites (rs9817428, rs1175543, rs13433696, and rs2920502) of the PPARγ gene have been identified as NAFLD-related [99]. Since PPARγ expression level is increased in NAFLD, obese, and T2DM patients, it was suggested that PPARγ acts as a pro-steatosis factor via the de novo lipogenesis and activation of lipogenic genes [152,153].
Adiponectin is a protein hormone secreted from adipose tissue and regulating glucose levels and FAs breakdown. Impaired adiponectin regulation/activity was connected to several metabolic diseases (T2DM, atherosclerosis, NAFLD, obesity, and MetS). Adiponectin can act synergistically with leptin, the other hormone, regulating energy balance and modulating insulin resistance [154]. Thus, it is not surprising that NAFLD-related polymorphism sites also have been found in insulin-related genes, such as IRS2 (insulin receptor substrate 2) and IGF1 (insulin-like growth factor 1). In addition to the direct and genetic interaction between glucose and lipid metabolism-regulating genes, IRS2 also mediates the signalling of IL-4 and interacts with SOCS1 (suppressor of cytokine signaling 1), thus connecting the energy metabolism gene pool with the inflammatory/immune response genes [155].
In summary, defined dysregulation in lipid metabolism provides a complex influence on an organism and, in combination with IR and other factors, significantly increases the risk of a life-threatening co-morbidity such as atherosclerosis [156].

4.2. SNPs in Genes Related to the Immune/Inflammatory Response

There were several SNPs sites identified in genes associated with the immune functions and inflammatory response, primarily cytokines: IL-6, IL-6R, IL-27, IL-17A, IL-17RA, and TNF-α. IL-6 is the best-studied pleiotropic pro-inflammatory cytokine, associated with chronic inflammation and the development of many diseases: Alzheimer’s, Crohn’s, anaemia, rheumatoid arthritis, inflammatory bowel disease, cancer, multiple sclerosis, aging, and others [157,158]. The IL-6 receptor is the main interaction partner for IL-6, which activates several signalling pathological pathways (Ras/MAPK, PI3K–PKB/Akt, and JAK/STAT3) and regulates levels of VEGF and CD4+ T cells to execute its biological functions [159]. It is also known that, in the absence of inflammation, up to 30% of IL-6 can be produced by adipose tissue [160], which makes IL-6 the main chronic inflammation factor for diseases linked with the accumulation of surplus fat. In addition to the pro-inflammatory role, IL-6 acts as an anti-inflammatory myokine, produced from muscles upon contraction [161]. As an anti-inflammatory myokine, IL-6 inhibits IL-1 and TNF-α and activates IL-10 and IL-1ra. TNF-α is another cytokine with a defined SNP site (rs1800629), responsible for immune cells’ regulation and implicated in many human diseases and disorders (cancer, psoriasis, cognitive deficits, inflammatory bowel disease, Alzheimer’s, depression, and others) [162,163]. In addition to the role of the pro-inflammatory response mediator, IL-17A and its receptor IL-17RA have many immune regulatory functions, associated with allergic responses and the production of many other cytokines (IL-6, IL-1β, TNF-α, and β), chemokines, and prostaglandins [164]. IL-17 was linked to several immune/autoimmune diseases (lupus, asthma, rheumatoid arthritis, psoriasis, multiple sclerosis, and others) [165].
In total, cytokines have connected into a close co-regulation circuit, creating a self-promoting chronic inflammation micro-environment suitable for the development of many diseases, including obesity, liver diseases, and several types of cancer [166].

4.3. SNPs in Other Genes

Among other genes that were found to be associated with NAFLD, we wish to discuss genes associated with tumorigenesis and inflammation. The polymorphism site rs2303861 was identified in the CD82 (KAI1) gene [123], responsible for the downregulation of tumor progression of human cancers, immunity, inflammation, and cognitive function [167,168,169,170]. As it was shown on the mouse model of rheumatoid arthritis (chronic autoinflammatory joint disease), the level of CD82 is increased in RASF (rheumatoid arthritis synovial fibroblasts), where it plays an important role in cell adhesion and motility. RASFs are active cells, migrating and promoting joint inflammation and destruction in non-affected areas [171].
Telomerase reverse transcriptase (TERT) has NAFLD-related substitution C228T [119]. Telomerases are responsible for the regulation of telomeres length, so that senescent cells can potentially become immortal and turn into cancerous cells [172]. In the case of NAFLD, C228T TERT mutations have been suggested as a non-invasive diagnostic biomarker for the disease progression to HCC [173,174]. While the results are very promising, they have been obtained on a small group of patients, so wider studies required before this method can be introduced to the clinical practice.
Sam50, an important mitochondria outer membrane protein, encoded by the SAMM50 gene and involved in the regulation of mitophagy, mitochondrial morphology and removal of ROS [175]. Several SNPs were linked with NAFLD; however, the exact mechanism, leading to hepatic lipids accumulation and NAFLD progression was not known. SAMM50-knockdown cells have lower levels of FAO, ETC activity; however, those effects can be reversed by overexpression of PPARα, which is known to enhance FAO [125]. While these results provide direct mechanistic evidence for the involvement of SAMM50 in lipid metabolism, wider research on different ethnic groups required to prove the causative role of SAMM50 polymorphisms in NAFLD susceptibility.
Interesting results, obtained by Rausch et al., 2018 [101] have suggested several new associations between NAFLD and obesity, DM and IR. However, the most important point of this GWAS study is the absence of PNPLA3 gene polymorphism, which was associated with NAFLD in several other cross-ethnic studies. This can be explained by a specific cohort (Hispanic boys, up to 18 years of age). However, both aspects of the newly identified SNPs (ethnic and age specificity) required further investigation.
Vitamin D deficiency was linked to NAFLD progression; thus vitamin D supplementation was suggested as an effective NAFLD treatment [176]. Polymorphisms of Vitamin D metabolism-related and signalling genes were associated with diabetes 1 and 2 types and BMI [177,178,179]. Recently, polymorphism site rs1544410 of the Vitamin D receptor was linked to advanced liver fibrosis in Japanese NAFLD patients [130]. Interestingly, the involvement of the liver–gut microbiome axis was suggested as a NAFLD progression factor, acting through nutrient uptake from the diet and bile acid circulation [180].
Another interesting SNP site was identified in NPY (Neuropeptide Y), which is a six-amino-acid peptide neurotransmitter, expressed by chromaffin and noradrenergic cells. It was suggested that high NPY expression in the hypothalamus is related to the development of T2DM, IR, and obesity [181]. Probably, the rs16147 variant has altered interaction properties with transcription factor and other regulatory elements. In the case of obese NAFLD patients, this polymorphism site was connected with a lower percentage of steatohepatitis and lobular inflammation [132]. The molecular mechanism of NPY activity relies on the up-regulation of the SREBP2/HMGCR pathway, where SREBP2 is a crucial transcription factor, regulating cholesterol homeostasis, and HMGCR is the rate-limiting enzyme in cholesterol synthesis [182].
In total, we can conclude that many genes have polymorphism sites linked to NAFLD and, potentially, can be used as diagnostic biomarkers for early disease diagnostics, monitoring its progression to NASH and HCC. However, more wide population analysis and studies on different ethnic and age groups are required for successful clinical application.

4.4. Functional Association between NAFLD and PNPLA3 Risk Allele

In the last decade, our understanding of the NAFLD pathogenesis has significantly expanded. While tens of SNPs were identified as a NAFLD-development risk factor, only some of them have been studied in detail and assigned to a particular mechanism. Thus, PNPLA3, TM6SF2, GCKR, MBOAT7, and HSD17B13 are the most frequent genes, and their association with NAFLD development was shown for different ethnic groups [183].
However, the primary role in the NAFLD development among non-obese patients was suggested for the PNPLA3 gene, even further elaborated to the PASH (PNPLA3-associated steatohepatitis) concept [184]. It is necessary to note that the presence of a mutant gene variant is not causing the disease itself. This was shown in Pnpla3148M knock-in mice, which had a normal level of hepatic fat on a standard diet. However, under a high-sucrose diet challenge, the level of hepatic fat was increased two- to three-fold, and PNPLA3 protein quantity on hepatic LDs was 40-fold higher, with no difference in hepatic Pnpla3 mRNA quantity [185]. The exact molecular mechanism of the couriers of the PNPLA3 risk allele p.148M was further elucidated. Firstly, it was found that the 148M variant can avoid ubiquitylation and proteasomal degradation, which causes accumulation of the mutant protein on LDs [186], where it prevents the TG mobilization by decreasing hydrolysis/transacylation of PUFAs from PUFA-containing diacylglycerols [187]. Interestingly, such detention of the TGs in the liver has a positive effect on the cardiovascular system, because carriers of the 148M PNPLA3 gene variant have a lower risk of cardiovascular diseases [188].
The mutant variant of the PNPLA3 gene has a system-level influence on lipid metabolism, causing fat accumulation in hepatocytes and, under certain circumstances, NAFLD development. The PNPLA3 I148M mutant protein variant is the main common genetic factor, described for several ethnic groups and with well-studied molecular mechanism of action. Such deep and detailed characterization is the first prerequisite for the development of an effective personalized NAFLD treatment. The PNPLA3 gene variant is widely accepted as the main NAFLD-associated risk factor; however, future research should also consider the role of other genetic risk factors, the majority of which now lack detailed characterization.

5. A Bi-Directional NAFLD ↔ DM Relationship

The original connection between liver disease and DM was established in 1889 by Bernhard Naunyn [189] and reviewed in [190]. Since then, many studies have confirmed the close association between NAFLD and DM, MetS, and risk of cardiovascular diseases [191,192,193]. However, the main question of whether NAFLD is a cause or a consequence of DM is still unanswered [194]. Focusing on the different types of DM, we can notice that linkage between NAFLD and DM1T is not so pronounced and depends greatly on the diagnostic strategy (ultrasound, magnetic resonance imaging, biopsy, or transient elastography) with a pooled prevalence of 19.3–22% in adults [195]. Similar results were obtained for the Brazilian population, where NAFLD was diagnosed among DM1T patients with hepatic ultrasound and transient elastography and resulted in 12.6% and 16.8% prevalence, respectively [196]. In contrast, the risk of DM2T and MetS among NAFLD patients was increased two-fold [191]. Similar results were obtained in another study, where the role of abnormal glucose tolerance was examined as a factor, predicting NASH severity in children and adolescents with and without NAFLD. As a result, diabetic and prediabetes conditions were associated with a 2.2-fold increased risk of NASH [197].
Recent research has defined that IR increases circulating ALT (one of the main NAFLD markers). Thus, increased insulin level (due to IR) promotes de novo lipogenesis in the liver and further hepatic IR, with forced stimulation of hepatic gluconeogenesis, leading to the liver’s elevated output of TG and glucose to the circulation. Eventually, those would diminish β-cell function and cause DM2T [198]. In total, IR results in NAFLD, which increases the risk of DM2T, suggesting IR as the main factor connecting NAFLD and DM [199].
The proposed role of IR was supported also by genetic factors, when SNPs in several genes known to define genetic susceptibility to NAFLD were also linked with IR: PNPLA3, TM6SF2, GCKR, MBOAT7, and HSD17B13 (Table 1) [200]. Recently, SNPs in those genes were suggested as the main nutritional sensors, when an individual’s genetic variations impact nutrients utilization and metabolic processes and define the risk of disease development [201]. Among those, PNPLA3 and GCKR SNPs were shown to be the main variants, interacting with insulin, IR, levels of TG, and glucose to increase NAFLD risk in non-diabetic individuals [202]. An even more confident study on the German population suggests that NAFLD cases could be eliminated if the PNPLA3 mutant variant was absent [203].
The mutant variant of the MBOAT7 gene, known to localize on the ER–mitochondria membrane border, where biosynthesis of LD and fat occurs, is also regulated by insulin. The strong reduction of MBOAT7 expression occurs during IR and obesity, leading to altered mitochondrial dynamics and morphology, higher OS, and switches from oxidative phosphorylation toward anaerobic glycolysis [204,205].
In contrast, a recent study on the Japanese NAFLD patients found that the proportions of PNPLA3 and KCNQ1 variants did not differ much among the NAFLD non-diabetic and NAFLD diabetic groups. Similarly, the PNPLA3 genotype does not influence the prevalence of diabetes and incidence of new-onset diabetes [206]. Another study, conducted on Chinese NAFLD patients, found that the PNPLA3 GG genotype escalates liver steatosis but reduces the risk of DM2T in patients with obesity or IR [207].
In total, we can conclude that NAFLD and DM are closely associated; however, it is not possible to prove causality between them. It was suggested that IR is the main factor linking NAFLD development and increased DM2T risk. Environmental and dietary factors can affect some mutant gene variants and promote NAFLD and DM2T development; nevertheless, some research papers contradict this conclusion at least for Japanese and Chinese NAFLD patients. Further research on different ethnic groups is required to set up a solid connection between NAFLD, increased risk of DM development, and SNPs in related genes.

6. Therapy

The molecular mechanisms of NAFLD development and progression are not completely understood, and there is no approved medical treatment for NAFLD [208]. However, current knowledge allows the provision of effective management of NAFLD as a complex metabolic disorder, often associated with IR, obesity, and DM. There are several main targets in NAFLD treatments: (1) lifestyle modification, which includes changes in feeding behavior, healthy diet, and exercise, oriented around weight loss; (2) control and management of the cardiovascular risks (the leading cause of mortality among NAFLD patients); and (3) prevention of the NAFLD progression to more severe forms and development of associated complications [6]. Ideal treatment should combine glucose and lipid-lowering drugs, metabolism modulators, antioxidants, and anti-inflammatory and anti-fibrotic agents [209]. A single substance (natural or synthetic) cannot provide such complex activities; thus, several combined approaches and drugs should be used.
Several recent reviews have covered many aspects of NAFLD treatments, including plant-based compounds [210], DM-targeted drugs, lifestyle modifications, surgeries, and pharmacological approaches [211,212,213]. For those reasons, we will focus on the recent publications discussing NLRP3 inflammasome-oriented treatments (Table 2).
Further, we wish to point to two natural compounds, naringenin and apigenin, with wide therapeutic activities. Naringenin is a flavonoid, widely presented in many fruits and grasses. In addition to its known anti-cancer and antimicrobial properties [222], recent research has defined also liver-specific anti-inflammatory activity [221]. Interestingly, the discovered effect was based on the inhibition of the NLRP3 activation; later, this decreased expression and secretion of IL-1β pro-inflammatory cytokine and lipid deposition in hepatocytes [221]. As a promising NAFLD treatment, naringenin was studied in several model systems and cell lines, proving its high therapeutic potential [223]. Furthermore, it was reported that naringenin has effective anti-diabetic properties, investigated in many in vitro and in vivo studies [224]. Similarly, apigenin is related to the naringenin flavone compound, known historically as a coloring agent. However, now apigenin is known for its wide pharmacological activities on several signalling pathways (NF-κB, JAK/STAT, PI3K/AKT/mTOR, MAPK/ERK, and others); thus, it is an effective suppressor of chronic inflammation-mediated diseases [225] and several types of cancer [226]. Similarly to naringenin, apigenin prevents NLRP3 inflammasome activation and reduces the release of IL-1β and IL-18 cytokines, but also has a direct effect on the mitochondria (discussed earlier ammonia detoxification pathway) manifested via reduced urea acid production [219].
In total, presented data suggest several effective synthetic and natural compounds which can be used for the treatment of NAFLD and related morbidities. Synthetic compounds seem to have more targeted and profound activities; however, their safety (possible cancerogenic and toxic properties, effective doses) requires detailed investigation. Natural compounds, in general, are safe, but, on the other hand, have wider and less specific activities. Further clinical studies are required to define effective therapeutic drugs and their combinations for NAFLD treatment and prevention.

7. Conclusions

We can conclude that variability in many nuclear and mitochondrial genes was linked to NAFLD development and progression. This knowledge should be used for the future creation of a comprehensive list of risk factors used for individual NAFLD prediction and, ideally, personalised treatments. NAFLD is triggered by an unbalanced diet, sedentary lifestyle, and absence of physical activity, but it also can be caused by genetic susceptibility because of the inherited mutations. It was shown that hepatic mitochondria have impaired mitophagy and are responsible for exceeding ROS production, the release of lipotoxic by-products of FAs β-oxidation and mtDNA, involved in chronic inflammation, NAFLD progress to NASH, cirrhosis, HCC, and hepatocyte death. Recent results suggest that the efficiency of mitochondria can be examined in a non-invasive way via measurements of urea cycle metabolites and used for NAFLD diagnosis. However, presented data are ethnic-specific and cannot be used as a universal guide. Additionally, some age-dependent factors and co-morbidities can affect NAFLD diagnosis. To overcome this limitation, more cross-ethnic studies with the involvement of various age groups and detailed clinical examination should be conducted in future.

Author Contributions

S.A.D., E.E.B. and A.N.O. conceptualized the manuscript; S.A.D. wrote the manuscript text; M.S.B., T.V.P., L.V.N., A.V.S. and A.N.O. reviewed the text; E.E.B. and M.S.B. contributed to the methodology; T.V.P., L.V.N. and A.V.S. contributed to the formal analysis; A.N.O. obtained funding and supervised. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Russian Science Foundation (Grant # 18-15-00254).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funder had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

APOapolipoprotein
APRIAST-to-platelet ratio index
BMIbody mass index
CK-18cytokeratin 18
CPS-1carbamoyl phosphate synthase 1
DMdiabetes milieus
ELFenhanced liver fibrosis test
FAsfatty acids
FAOfatty acids oxidation
FGF-21fibroblast growth factor-21
FNDC5fibronectin type III domain-containing protein 5
GWASgenome-wide association studies
HCChepatocellular carcinoma
IMMinner mitochondrial membrane
IGF1insulin-like growth factor 1
ILinterleukin
IRinsulin resistance
IRS2insulin receptor substrate 2
LC3microtubule-associated proteins, light chain
LDlipid droplet
MetSmetabolic syndrome
MRImagnetic resonance imaging
NAFLDnon-alcoholic fatty liver disease
NASHnon-alcoholic steatohepatitis
NFSNAFLD fibrosis score
NPYNeuropeptide Y
OMMouter mitochondrial membrane
OSoxidative stress
OTCornithine transcarbamoylase
OXPHOSoxidative phosphorylation
PLINsPerilipins
PNPLA3Patatin-like Phospholipase Domain Containing 3
PONparaoxonase
PPARγperoxisome proliferator-activated receptor γ
PUFApolyunsaturated fatty acid
RASFsrheumatoid arthritis synovial fibroblasts
ROSreactive oxygen species
SNPsingle-nucleotide polymorphism
SOCS1suppressor of cytokine signaling 1
TGtriglyceride
TERTtelomerase reverse transcriptase
TM6SF2transmembrane 6 superfamily member 2
TNFtumor necrosis factor
VDACvoltage-dependent anion channel
VEGFvascular endothelial growth factor
VLDLvery-low-density lipoprotein

References

  1. Parthasarathy, G.; Revelo, X.; Malhi, H. Pathogenesis of Nonalcoholic Steatohepatitis: An Overview. Hepatol. Commun. 2020, 4, 478–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Dooghaie Moghadam, A.; Eslami, P.; Razavi-Khorasani, N.; Moazzami, B.; Makhdoomi Sharabiani, K.; Farokhi, E.; Mansour-Ghanaei, A.; Zahedi-Tajrishi, F.; Mehrvar, A.; Aghajanpoor Pasha, M.; et al. Recurrence of Fatty Liver Disease Following Liver Transplantation for NAFLD-Related Cirrhosis: Current Status and Challenges. Casp. J. Intern. Med. 2020, 11. [Google Scholar] [CrossRef]
  3. Nahon, P.; Allaire, M.; Nault, J.-C.; Paradis, V. Characterizing the Mechanism behind the Progression of NAFLD to Hepatocellular Carcinoma. Hepatic Oncol. 2020, 7, HEP36. [Google Scholar] [CrossRef]
  4. NIH NAFLD & NASH. 2021. Available online: https://www.niddk.nih.gov/health-information/liver-disease/nafld-nash/all-content (accessed on 3 April 2021).
  5. Estes, C.; Anstee, Q.M.; Arias-Loste, M.T.; Bantel, H.; Bellentani, S.; Caballeria, J.; Colombo, M.; Craxi, A.; Crespo, J.; Day, C.P.; et al. Modeling NAFLD Disease Burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the Period 2016–2030. J. Hepatol. 2018, 69, 896–904. [Google Scholar] [CrossRef]
  6. Ramanan, S.P.; Mohamed, M.W.F.; Aung, S.S.; Sange, I.; Hamid, P. Treatment of Fatty Liver Disease: The Present and the Future. Cureus 2021, 13, e12713. [Google Scholar] [CrossRef]
  7. Schulz, M.; Tacke, F. Identifying High-Risk NASH Patients: What We Know so Far. HMER 2020, 12, 125–138. [Google Scholar] [CrossRef]
  8. Campos-Murguía, A.; Ruiz-Margáin, A.; González-Regueiro, J.A.; Macías-Rodríguez, R.U. Clinical Assessment and Management of Liver Fibrosis in Non-Alcoholic Fatty Liver Disease. WJG 2020, 26, 5919–5943. [Google Scholar] [CrossRef]
  9. Davison, B.A.; Harrison, S.A.; Cotter, G.; Alkhouri, N.; Sanyal, A.; Edwards, C.; Colca, J.R.; Iwashita, J.; Koch, G.G.; Dittrich, H.C. Suboptimal Reliability of Liver Biopsy Evaluation Has Implications for Randomized Clinical Trials. J. Hepatol. 2020, 73, 1322–1332. [Google Scholar] [CrossRef] [PubMed]
  10. Han, M.A.T. Noninvasive Tests (NITs) for Hepatic Fibrosis in Fatty Liver Syndrome. Life 2020, 10, 198. [Google Scholar] [CrossRef] [PubMed]
  11. Ajmera, V.; Loomba, R. Imaging Biomarkers of NAFLD, NASH, and Fibrosis. Mol. Metab. 2021, 101167. [Google Scholar] [CrossRef] [PubMed]
  12. Loomba, R.; Adams, L.A. Advances in Non-Invasive Assessment of Hepatic Fibrosis. Gut 2020, 69, 1343–1352. [Google Scholar] [CrossRef] [PubMed]
  13. Tilg, H.; Adolph, T.E.; Moschen, A.R. Multiple Parallel Hits Hypothesis in Nonalcoholic Fatty Liver Disease: Revisited After a Decade. Hepatology 2021, 73, 833–842. [Google Scholar] [CrossRef] [PubMed]
  14. Parola, M.; Pinzani, M. Liver Fibrosis: Pathophysiology, Pathogenetic Targets and Clinical Issues. Mol. Asp. Med. 2019, 65, 37–55. [Google Scholar] [CrossRef] [PubMed]
  15. Hossain, M.; Kubes, P. Innate Immune Cells Orchestrate the Repair of Sterile Injury in the Liver and Beyond. Eur. J. Immunol. 2019, 49, 831–841. [Google Scholar] [CrossRef] [Green Version]
  16. Lambrecht, J.; Tacke, F. Controversies and Opportunities in the Use of Inflammatory Markers for Diagnosis or Risk Prediction in Fatty Liver Disease. Front. Immunol. 2021, 11, 634409. [Google Scholar] [CrossRef] [PubMed]
  17. Cordeiro, A.; Costa, R.; Andrade, N.; Silva, C.; Canabrava, N.; Pena, M.J.; Rodrigues, I.; Andrade, S.; Ramalho, A. Does Adipose Tissue Inflammation Drive the Development of Non-Alcoholic Fatty Liver Disease in Obesity? Clin. Res. Hepatol. Gastroenterol. 2020, 44, 394–402. [Google Scholar] [CrossRef]
  18. Lefere, S.; Tacke, F. Macrophages in Obesity and Non-Alcoholic Fatty Liver Disease: Crosstalk with Metabolism. JHEP Rep. 2019, 1, 30–43. [Google Scholar] [CrossRef] [Green Version]
  19. Schuppan, D.; Surabattula, R.; Wang, X.Y. Determinants of Fibrosis Progression and Regression in NASH. J. Hepatol. 2018, 68, 238–250. [Google Scholar] [CrossRef]
  20. Karsdal, M.A.; Detlefsen, S.; Daniels, S.J.; Nielsen, M.J.; Krag, A.; Schuppan, D. Is the Total Amount as Important as Localization and Type of Collagen in Liver Fibrosis Attributable to Steatohepatitis? Hepatology 2020, 71, 346–351. [Google Scholar] [CrossRef]
  21. Mansouri, A.; Gattolliat, C.-H.; Asselah, T. Mitochondrial Dysfunction and Signaling in Chronic Liver Diseases. Gastroenterology 2018, 155, 629–647. [Google Scholar] [CrossRef] [Green Version]
  22. Liangpunsakul, S.; Chalasani, N. Lipid Mediators of Liver Injury in Nonalcoholic Fatty Liver Disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 316, G75–G81. [Google Scholar] [CrossRef] [PubMed]
  23. Mendez-Sanchez, N.; Cruz-Ramon, V.; Ramirez-Perez, O.; Hwang, J.; Barranco-Fragoso, B.; Cordova-Gallardo, J. New Aspects of Lipotoxicity in Nonalcoholic Steatohepatitis. Int. J. Mol. Sci. 2018, 19, 2034. [Google Scholar] [CrossRef] [Green Version]
  24. Arroyave-Ospina, J.C.; Wu, Z.; Geng, Y.; Moshage, H. Role of Oxidative Stress in the Pathogenesis of Non-Alcoholic Fatty Liver Disease: Implications for Prevention and Therapy. Antioxidants 2021, 10, 174. [Google Scholar] [CrossRef] [PubMed]
  25. Sookoian, S.; Flichman, D.; Scian, R.; Rohr, C.; Dopazo, H.; Gianotti, T.F.; Martino, J.S.; Castaño, G.O.; Pirola, C.J. Mitochondrial Genome Architecture in Non-Alcoholic Fatty Liver Disease: Mitochondrial Genome Mutations and NAFLD. J. Pathol. 2016, 240, 437–449. [Google Scholar] [CrossRef] [PubMed]
  26. Mandala, A.; Janssen, R.C.; Palle, S.; Short, K.R.; Friedman, J.E. Pediatric Non-Alcoholic Fatty Liver Disease: Nutritional Origins and Potential Molecular Mechanisms. Nutrients 2020, 12, 3166. [Google Scholar] [CrossRef] [PubMed]
  27. Hasturk, B.; Yilmaz, Y.; Eren, F. Potential Clinical Variants Detected in Mitochondrial DNA D-Loop Hypervariable Region I of Patients with Non-Alcoholic Steatohepatitis. Hormones 2019, 18, 463–475. [Google Scholar] [CrossRef]
  28. Pirola, C.J.; Garaycoechea, M.; Flichman, D.; Castaño, G.O.; Sookoian, S. Liver Mitochondrial DNA Damage and Genetic Variability of Cytochrome b—A Key Component of the Respirasome—Drive the Severity of Fatty Liver Disease. J. Intern. Med. 2021, 289, 84–96. [Google Scholar] [CrossRef] [PubMed]
  29. Paiva Coelho, M.; Martins, E.; Vilarinho, L. Diagnosis, Management, and Follow-up of Mitochondrial Disorders in Childhood: A Personalized Medicine in the New Era of Genome Sequence. Eur. J. Pediatr. 2019, 178, 21–32. [Google Scholar] [CrossRef]
  30. Vazquez-Calvo, C.; Suhm, T.; Büttner, S.; Ott, M. The Basic Machineries for Mitochondrial Protein Quality Control. Mitochondrion 2020, 50, 121–131. [Google Scholar] [CrossRef]
  31. Suzuki, H.; Tabata, K.; Morita, E.; Kawasaki, M.; Kato, R.; Dobson, R.C.J.; Yoshimori, T.; Wakatsuki, S. Structural Basis of the Autophagy-Related LC3/Atg13 LIR Complex: Recognition and Interaction Mechanism. Structure 2014, 22, 47–58. [Google Scholar] [CrossRef] [Green Version]
  32. Lou, G.; Palikaras, K.; Lautrup, S.; Scheibye-Knudsen, M.; Tavernarakis, N.; Fang, E.F. Mitophagy and Neuroprotection. Trends Mol. Med. 2020, 26, 8–20. [Google Scholar] [CrossRef]
  33. Gao, A.; Jiang, J.; Xie, F.; Chen, L. Bnip3 in Mitophagy: Novel Insights and Potential Therapeutic Target for Diseases of Secondary Mitochondrial Dysfunction. Clin. Chim. Acta 2020, 506, 72–83. [Google Scholar] [CrossRef] [PubMed]
  34. Li, X.; Shi, Z.; Zhu, Y.; Shen, T.; Wang, H.; Shui, G.; Loor, J.J.; Fang, Z.; Chen, M.; Wang, X.; et al. Cyanidin-3- O -glucoside Improves Non-alcoholic Fatty Liver Disease by Promoting PINK1-mediated Mitophagy in Mice. Br. J. Pharm. 2020, 177, 3591–3607. [Google Scholar] [CrossRef] [PubMed]
  35. Yu, X.; Hao, M.; Liu, Y.; Ma, X.; Lin, W.; Xu, Q.; Zhou, H.; Shao, N.; Kuang, H. Liraglutide Ameliorates Non-Alcoholic Steatohepatitis by Inhibiting NLRP3 Inflammasome and Pyroptosis Activation via Mitophagy. Eur. J. Pharmacol. 2019, 864, 172715. [Google Scholar] [CrossRef]
  36. Grattagliano, I.; Montezinho, L.P.; Oliveira, P.J.; Frühbeck, G.; Gómez-Ambrosi, J.; Montecucco, F.; Carbone, F.; Wieckowski, M.R.; Wang, D.Q.-H.; Portincasa, P. Targeting Mitochondria to Oppose the Progression of Nonalcoholic Fatty Liver Disease. Biochem. Pharmacol. 2019, 160, 34–45. [Google Scholar] [CrossRef]
  37. Zhou, T.; Chang, L.; Luo, Y.; Zhou, Y.; Zhang, J. Mst1 Inhibition Attenuates Non-Alcoholic Fatty Liver Disease via Reversing Parkin-Related Mitophagy. Redox Biol. 2019, 21, 101120. [Google Scholar] [CrossRef] [PubMed]
  38. Zeng, J.; Shirihai, O.S.; Grinstaff, M.W. Degradable Nanoparticles Restore Lysosomal PH and Autophagic Flux in Lipotoxic Pancreatic Beta Cells. Adv. Healthc. Mater. 2019, 8, 1801511. [Google Scholar] [CrossRef] [PubMed]
  39. Tincopa, M.A. Diagnostic and Interventional Circulating Biomarkers in Nonalcoholic Steatohepatitis. Endocrinol. Diab. Metab. 2020, 3. [Google Scholar] [CrossRef]
  40. Canbay, A.; Sowa, J.-P. L-Ornithine l-Aspartate (LOLA) as a Novel Approach for Therapy of Non-Alcoholic Fatty Liver Disease. Drugs 2019, 79, 39–44. [Google Scholar] [CrossRef] [Green Version]
  41. He, L.; Deng, L.; Zhang, Q.; Guo, J.; Zhou, J.; Song, W.; Yuan, F. Diagnostic Value of CK-18, FGF-21, and Related Biomarker Panel in Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Biomed. Res. Int. 2017, 2017, 1–12. [Google Scholar] [CrossRef]
  42. Yan, J.; Shen, S.; He, Y.; Li, Z. TLR5 Silencing Reduced Hyperammonaemia-Induced Liver Injury by Inhibiting Oxidative Stress and Inflammation Responses via Inactivating NF-ΚB and MAPK Signals. Chem. Biol. Interact. 2019, 299, 102–110. [Google Scholar] [CrossRef] [PubMed]
  43. Thomsen, K.L.; De Chiara, F.; Rombouts, K.; Vilstrup, H.; Andreola, F.; Mookerjee, R.P.; Jalan, R. Ammonia: A Novel Target for the Treatment of Non-Alcoholic Steatohepatitis. Med. Hypotheses 2018, 113, 91–97. [Google Scholar] [CrossRef] [PubMed]
  44. BonDurant, L.D.; Potthoff, M.J. Fibroblast Growth Factor 21: A Versatile Regulator of Metabolic Homeostasis. Annu. Rev. Nutr. 2018, 38, 173–196. [Google Scholar] [CrossRef] [PubMed]
  45. De Chiara, F.; Heebøll, S.; Marrone, G.; Montoliu, C.; Hamilton-Dutoit, S.; Ferrandez, A.; Andreola, F.; Rombouts, K.; Grønbæk, H.; Felipo, V.; et al. Urea Cycle Dysregulation in Non-Alcoholic Fatty Liver Disease. J. Hepatol. 2018, 69, 905–915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ajaz, S.; McPhail, M.J.; Gnudi, L.; Trovato, F.M.; Mujib, S.; Napoli, S.; Carey, I.; Agarwal, K. Mitochondrial Dysfunction as a Mechanistic Biomarker in Patients with Non-Alcoholic Fatty Liver Disease (NAFLD). Mitochondrion 2021, 57, 119–130. [Google Scholar] [CrossRef] [PubMed]
  47. Younossi, Z.M. Non-Alcoholic Fatty Liver Disease—A Global Public Health Perspective. J. Hepatol. 2019, 70, 531–544. [Google Scholar] [CrossRef] [Green Version]
  48. Prieto, I.; Monsalve, M. ROS Homeostasis, a Key Determinant in Liver Ischemic-Preconditioning. Redox Biol. 2017, 12, 1020–1025. [Google Scholar] [CrossRef] [Green Version]
  49. Lee, J.; Park, J.-S.; Roh, Y.S. Molecular Insights into the Role of Mitochondria in Non-Alcoholic Fatty Liver Disease. Arch. Pharm. Res. 2019, 42, 935–946. [Google Scholar] [CrossRef] [PubMed]
  50. Farzanegi, P.; Dana, A.; Ebrahimpoor, Z.; Asadi, M.; Azarbayjani, M.A. Mechanisms of Beneficial Effects of Exercise Training on Non-Alcoholic Fatty Liver Disease (NAFLD): Roles of Oxidative Stress and Inflammation. Eur. J. Sport Sci. 2019, 19, 994–1003. [Google Scholar] [CrossRef] [PubMed]
  51. Ore, A.; Akinloye, O. Oxidative Stress and Antioxidant Biomarkers in Clinical and Experimental Models of Non-Alcoholic Fatty Liver Disease. Medicina 2019, 55, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Delli Bovi, A.P.; Marciano, F.; Mandato, C.; Siano, M.A.; Savoia, M.; Vajro, P. Oxidative Stress in Non-Alcoholic Fatty Liver Disease. An Updated Mini Review. Front. Med. 2021, 8, 595371. [Google Scholar] [CrossRef]
  53. Hafner-Bratkovič, I.; Sušjan, P.; Lainšček, D.; Tapia-Abellán, A.; Cerović, K.; Kadunc, L.; Angosto-Bazarra, D.; Pelegrin, P.; Jerala, R. NLRP3 Lacking the Leucine-Rich Repeat Domain Can Be Fully Activated via the Canonical Inflammasome Pathway. Nat. Commun. 2018, 9, 5182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Lechtenberg, B.C.; Mace, P.D.; Riedl, S.J. Structural Mechanisms in NLR Inflammasome Signaling. Curr. Opin. Struct. Biol. 2014, 29, 17–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Mangan, M.S.J.; Olhava, E.J.; Roush, W.R.; Seidel, H.M.; Glick, G.D.; Latz, E. Targeting the NLRP3 Inflammasome in Inflammatory Diseases. Nat. Rev. Drug Discov. 2018, 17, 588–606. [Google Scholar] [CrossRef]
  56. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef] [Green Version]
  57. Deets, K.A.; Vance, R.E. Inflammasomes and Adaptive Immune Responses. Nat. Immunol. 2021, 22, 412–422. [Google Scholar] [CrossRef]
  58. Seok, J.K.; Kang, H.C.; Cho, Y.-Y.; Lee, H.S.; Lee, J.Y. Therapeutic Regulation of the NLRP3 Inflammasome in Chronic Inflammatory Diseases. Arch. Pharm. Res. 2021, 44, 16–35. [Google Scholar] [CrossRef]
  59. Yabal, M.; Calleja, D.J.; Simpson, D.S.; Lawlor, K.E. Stressing out the Mitochondria: Mechanistic Insights into NLRP3 Inflammasome Activation. J. Leukoc. Biol. 2019, 105, 377–399. [Google Scholar] [CrossRef]
  60. Ye, Z.; Wang, S.; Zhang, C.; Zhao, Y. Coordinated Modulation of Energy Metabolism and Inflammation by Branched-Chain Amino Acids and Fatty Acids. Front. Endocrinol. 2020, 11, 617. [Google Scholar] [CrossRef]
  61. Meyers, A.K.; Zhu, X. The NLRP3 Inflammasome: Metabolic Regulation and Contribution to Inflammaging. Cells 2020, 9, 1808. [Google Scholar] [CrossRef] [PubMed]
  62. Monserrat-Mesquida, M.; Quetglas-Llabrés, M.; Capó, X.; Bouzas, C.; Mateos, D.; Pons, A.; Tur, J.A.; Sureda, A. Metabolic Syndrome Is Associated with Oxidative Stress and Proinflammatory State. Antioxidants 2020, 9, 236. [Google Scholar] [CrossRef] [Green Version]
  63. Wong, S.K.; Chin, K.-Y.; Ima-Nirwana, S. Toll-like Receptor as a Molecular Link between Metabolic Syndrome and Inflammation: A Review. Curr. Drug Targets 2019, 20, 1264–1280. [Google Scholar] [CrossRef] [PubMed]
  64. Li, W.; Cao, T.; Luo, C.; Cai, J.; Zhou, X.; Xiao, X.; Liu, S. Crosstalk between ER Stress, NLRP3 Inflammasome, and Inflammation. Appl. Microbiol. Biotechnol. 2020, 104, 6129–6140. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, L.; Chen, R.; Wang, H.; Liang, F. Mechanisms Linking Inflammation to Insulin Resistance. Int. J. Endocrinol. 2015, 2015, 1–9. [Google Scholar] [CrossRef] [PubMed]
  66. Bronner, D.N.; Abuaita, B.H.; Chen, X.; Fitzgerald, K.A.; Nuñez, G.; He, Y.; Yin, X.-M.; O’Riordan, M.X.D. Endoplasmic Reticulum Stress Activates the Inflammasome via NLRP3- and Caspase-2-Driven Mitochondrial Damage. Immunity 2015, 43, 451–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Puig, N.; Estruch, M.; Jin, L.; Sanchez-Quesada, J.L.; Benitez, S. The Role of Distinctive Sphingolipids in the Inflammatory and Apoptotic Effects of Electronegative LDL on Monocytes. Biomolecules 2019, 9, 300. [Google Scholar] [CrossRef] [Green Version]
  68. Chen, Y.; Yang, M.; Huang, W.; Chen, W.; Zhao, Y.; Schulte, M.L.; Volberding, P.; Gerbec, Z.; Zimmermann, M.T.; Zeighami, A.; et al. Mitochondrial Metabolic Reprogramming by CD36 Signaling Drives Macrophage Inflammatory Responses. Circ. Res. 2019, 125, 1087–1102. [Google Scholar] [CrossRef]
  69. Jonas, W.; Schürmann, A. Genetic and Epigenetic Factors Determining NAFLD Risk. Mol. Metab. 2020, in press, 101111. [Google Scholar] [CrossRef]
  70. Struben, V.M.D.; Hespenheide, E.E.; Caldwell, S.H. Nonalcoholic Steatohepatitis and Cryptogenic Cirrhosis within Kindreds. Am. J. Med. 2000, 108, 9–13. [Google Scholar] [CrossRef]
  71. Di Costanzo, A.; Belardinilli, F.; Bailetti, D.; Sponziello, M.; D’Erasmo, L.; Polimeni, L.; Baratta, F.; Pastori, D.; Ceci, F.; Montali, A.; et al. Evaluation of Polygenic Determinants of Non-Alcoholic Fatty Liver Disease (NAFLD) By a Candidate Genes Resequencing Strategy. Sci. Rep. 2018, 8, 3702. [Google Scholar] [CrossRef] [PubMed]
  72. Han, M.A.T.; Yu, Q.; Tafesh, Z.; Pyrsopoulos, N. Diversity in NAFLD: A Review of Manifestations of Nonalcoholic Fatty Liver Disease in Different Ethnicities Globally. J. Clin. Transl. Hepatol. 2020, 1–10. [Google Scholar] [CrossRef]
  73. Choudhary, N.S.; Duseja, A. Genetic and Epigenetic Disease Modifiers: Non-Alcoholic Fatty Liver Disease (NAFLD) and Alcoholic Liver Disease (ALD). Transl. Gastroenterol. Hepatol. 2021, 6, 2. [Google Scholar] [CrossRef]
  74. Milaciu, M.V.; Vesa, Ș.C.; Bocșan, I.C.; Ciumărnean, L.; Sâmpelean, D.; Negrean, V.; Pop, R.M.; Matei, D.M.; Pașca, S.; Răchișan, A.L.; et al. Paraoxonase-1 Serum Concentration and PON1 Gene Polymorphisms: Relationship with Non-Alcoholic Fatty Liver Disease. J. Clin. Med. 2019, 8, 2200. [Google Scholar] [CrossRef] [Green Version]
  75. Faulkner, C.S.; White, C.M.; Shah, V.H.; Jophlin, L.L. A Single Nucleotide Polymorphism of PLIN2 Is Associated with Nonalcoholic Steatohepatitis and Causes Phenotypic Changes in Hepatocyte Lipid Droplets: A Pilot Study. Biochim. Et Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2020, 1865, 158637. [Google Scholar] [CrossRef]
  76. Delik, A.; Akkız, H.; Dinçer, S. The Effect of PNPLA3 Polymorphism as Gain in Function Mutation in the Pathogenesis of Non-Alcoholic Fatty Liver Disease. Indian J. Gastroenterol. 2020, 39, 84–91. [Google Scholar] [CrossRef] [PubMed]
  77. Xu, M.; Li, Y.; Zhang, S.; Wang, X.; Shen, J.; Zhang, S. Interaction of TM6SF2 E167K and PNPLA3 I148M Variants in NAFLD in Northeast China. Ann. Hepatol. 2019, 18, 456–460. [Google Scholar] [CrossRef]
  78. Lisboa, Q.C.; Nardelli, M.J.; Pereira, P.D.A.; Miranda, D.M.; Ribeiro, S.N.; Costa, R.S.N.; Versiani, C.A.; Vidigal, P.V.T.; Ferrari, T.C.D.A.; Couto, C.A. PNPLA3 and TM6SF2 Polymorphisms in Brazilian Patients with Nonalcoholic Fatty Liver Disease. World J. Hepatol. 2020, 12, 792–806. [Google Scholar] [CrossRef] [PubMed]
  79. Chatterjee, A.; Basu, A.; Das, K.; Chowdhury, A.; Basu, P. Exome-Wide Scan Identifies Significant Association of Rs4788084 in IL27 Promoter with Increase in Hepatic Fat Content among Indians. Gene 2021, 775, 145431. [Google Scholar] [CrossRef]
  80. Zhu, J.; Xu, D.; Yang, R.; Liu, M.; Liu, Y. The Triglyceride Glucose Index (TyG) and CDKAL1 Gene Rs10946398 SNP Are Associated with NAFLD in Chinese Adults. Minerva Endocrinol. 2020. [Google Scholar] [CrossRef]
  81. Li, Y.; Liu, S.; Gao, Y.; Ma, H.; Zhan, S.; Yang, Y.; Xin, Y.; Xuan, S. Association of TM6SF2 Rs58542926 Gene Polymorphism with the Risk of Non-Alcoholic Fatty Liver Disease and Colorectal Adenoma in Chinese Han Population. BMC Biochem. 2019, 20, 3. [Google Scholar] [CrossRef] [PubMed]
  82. Chen, L.-Z.; Ding, H.-Y.; Liu, S.-S.; Liu, Q.; Jiang, X.-J.; Xin, Y.-N.; Xuan, S.-Y. Combining I148M and E167K Variants to Improve Risk Prediction for Nonalcoholic Fatty Liver Disease in Qingdao Han Population, China. Lipids Health Dis. 2019, 18, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Ma, Y.; Belyaeva, O.V.; Brown, P.M.; Fujita, K.; Valles, K.; Karki, S.; de Boer, Y.S.; Koh, C.; Chen, Y.; Du, X.; et al. 17-Beta Hydroxysteroid Dehydrogenase 13 Is a Hepatic Retinol Dehydrogenase Associated With Histological Features of Nonalcoholic Fatty Liver Disease. Hepatology 2019, 69, 1504–1519. [Google Scholar] [CrossRef]
  84. Adant, I.; Declercq, M.; Bird, M.; Bauters, M.; Boeckx, N.; Devriendt, K.; Cassiman, D.; Witters, P. Two Cases of Non-Alcoholic Fatty Liver Disease Caused by Biallelic ABHD5 Mutations. J. Hepatol. 2020, 72, 1030–1032. [Google Scholar] [CrossRef] [PubMed]
  85. Youssefian, L.; Vahidnezhad, H.; Saeidian, A.H.; Pajouhanfar, S.; Sotoudeh, S.; Mansouri, P.; Amirkashani, D.; Zeinali, S.; Levine, M.A.; Peris, K.; et al. Inherited Non-Alcoholic Fatty Liver Disease and Dyslipidemia Due to Monoallelic ABHD5 Mutations. J. Hepatol. 2019, 71, 366–370. [Google Scholar] [CrossRef]
  86. Aller, R.; Izaola, O.; Primo, D.; de Luis, D. Cholesteryl Ester Transfer Protein Variant (RS1800777) with Liver Histology in Non-Alcoholic Fatty Liver Disease Patients. Ann. Nutr. Metab. 2018, 73, 265–270. [Google Scholar] [CrossRef]
  87. Liu, J.; Xing, J.; Wang, B.; Wei, C.; Yang, R.; Zhu, Y.; Qiu, H. Correlation Between Adiponectin Gene Rs1501299 Polymorphism and Nonalcoholic Fatty Liver Disease Susceptibility: A Systematic Review and Meta-Analysis. Med. Sci. Monit. 2019, 25, 1078–1086. [Google Scholar] [CrossRef]
  88. Liu, M.; Liu, S.; Shang, M.; Liu, X.; Wang, Y.; Li, Q.; Mambiya, M.; Yang, L.; Zhang, Q.; Zhang, K.; et al. Association between ADIPOQ G276T and C11377G Polymorphisms and the Risk of Non-alcoholic Fatty Liver Disease: An Updated Meta-analysis. Mol. Genet. Genom. Med. 2019, 7, e624. [Google Scholar] [CrossRef]
  89. Pan, X.; Zheng, M.; Zou, T.; Liu, W.; Gu, X.; Zhang, X.; Cheng, X. The LEPR K109R and Q223R Might Contribute to the Risk of NAFLD: A Meta-Analysis. Curr. Mol. Med. 2018, 18. [Google Scholar] [CrossRef] [PubMed]
  90. Xu, Y.; Lu, L.-L.; Liu, S.-S.; Du, S.-X.; Zhu, H.-L.; Dong, Q.-J.; Xin, Y.-N. Apolipoprotein A5 Gene Polymorphisms Are Associated with Non-Alcoholic Fatty Liver Disease. Hepatobiliary Pancreat. Dis. Int. 2018, 17, 214–219. [Google Scholar] [CrossRef]
  91. Xu, Q.-Y.; Li, H.; Cao, H.-X.; Pan, Q.; Fan, J.-G. APOC3 Rs2070667 Associates with Serum Triglyceride Profile and Hepatic Inflammation in Nonalcoholic Fatty Liver Disease. Biomed. Res. Int. 2020, 2020, 1–9. [Google Scholar] [CrossRef]
  92. Jain, V.; Kumar, A.; Ahmad, N.; Jana, M.; Kalaivani, M.; Kumar, B.; Shastri, S.; Jain, O.; Kabra, M. Genetic Polymorphisms Associated with Obesity and Non-Alcoholic Fatty Liver Disease in Asian Indian Adolescents. J. Pediatric Endocrinol. Metab. 2019, 32, 749–758. [Google Scholar] [CrossRef]
  93. Jamialahmadi, O.; Mancina, R.M.; Ciociola, E.; Tavaglione, F.; Luukkonen, P.K.; Baselli, G.; Malvestiti, F.; Thuillier, D.; Raverdy, V.; Männistö, V.; et al. Exome-Wide Association Study on Alanine Aminotransferase Identifies Sequence Variants in the GPAM and APOE Associated With Fatty Liver Disease. Gastroenterology 2021, S0016508520355621. [Google Scholar] [CrossRef]
  94. Gravito-Soares, M.; Gravito-Soares, E.; Gomes, D.; Tome, L. Lysosomal Acid Lipase: Can It Be a New Non-Invasive Serum Biomarker of Cryptogenic Liver Fibrosis and Cirrhosis? Ann. Hepatol. 2019, 18, 78–88. [Google Scholar] [CrossRef] [PubMed]
  95. Metwally, M.; Bayoumi, A.; Romero-Gomez, M.; Thabet, K.; John, M.; Adams, L.A.; Huo, X.; Aller, R.; García-Monzón, C.; Teresa Arias-Loste, M.; et al. A Polymorphism in the Irisin-Encoding Gene (FNDC5) Associates with Hepatic Steatosis by Differential MiRNA Binding to the 3′UTR. J. Hepatol. 2019, 70, 494–500. [Google Scholar] [CrossRef] [PubMed]
  96. Gao, F.; Zheng, K.I.; Zhu, P.-W.; Li, Y.-Y.; Ma, H.-L.; Li, G.; Tang, L.-J.; Rios, R.S.; Liu, W.-Y.; Pan, X.-Y.; et al. FNDC5 Polymorphism Influences the Association between Sarcopenia and Liver Fibrosis in Adults with Biopsy-Proven Non-Alcoholic Fatty Liver Disease. Br. J. Nutr. 2020, 1–12. [Google Scholar] [CrossRef]
  97. Cai, W.; Weng, D.; Yan, P.; Lin, Y.; Dong, Z.; Mailamuguli Yao, H. Genetic Polymorphisms Associated with Nonalcoholic Fatty Liver Disease in Uyghur Population: A Case-Control Study and Meta-Analysis. Lipids Health Dis. 2019, 18, 14. [Google Scholar] [CrossRef] [Green Version]
  98. Hasan, E.M.; Abd Al Aziz, R.A.; Sabry, D.; Darweesh, S.K.; Badary, H.A.; Elsharkawy, A.; Abouelkhair, M.M.; Yosry, A. Genetic Variants in Nicotinamide-N-Methyltransferase (NNMT) Gene Are Related to the Stage of Non-Alcoholic Fatty Liver Disease Diagnosed by Controlled Attenuation Parameter (CAP)-FibroScan. J. Gastrointest. Liver Dis. 2018, 27, 265–272. [Google Scholar] [CrossRef]
  99. Zhu, P.; Lu, H.; Jing, Y.; Zhou, H.; Ding, Y.; Wang, J.; Guo, D.; Guo, Z.; Dong, C. Interaction Between AGTR1 and PPARγ Gene Polymorphisms on the Risk of Nonalcoholic Fatty Liver Disease. Genet. Test. Mol. Biomark. 2019, 23, 166–175. [Google Scholar] [CrossRef]
  100. Musso, G.; Saba, F.; Cassader, M.; Paschetta, E.; De Michieli, F.; Pinach, S.; Framarin, L.; Berrutti, M.; Leone, N.; Parente, R.; et al. Angiotensin II Type 1 Receptor Rs5186 Gene Variant Predicts Incident NAFLD and Associated Hypertension: Role of Dietary Fat-Induced Pro-Inflammatory Cell Activation. Am. J. Gastroenterol. 2019, 114, 607–619. [Google Scholar] [CrossRef]
  101. Rausch, J.C.; Lavine, J.E.; Chalasani, N.; Guo, X.; Kwon, S.; Schwimmer, J.B.; Molleston, J.P.; Loomba, R.; Brunt, E.M.; Chen, Y.-D.I.; et al. Genetic Variants Associated With Obesity and Insulin Resistance in Hispanic Boys With Nonalcoholic Fatty Liver Disease. J. Pediatric Gastroenterol. Nutr. 2018, 66, 789–796. [Google Scholar] [CrossRef]
  102. Bale, G.; Vishnubhotla, R.V.; Mitnala, S.; Sharma, M.; Padaki, R.N.; Pawar, S.C.; Duvvur, R.N. Whole-Exome Sequencing Identifies a Variant in Phosphatidylethanolamine N-Methyltransferase Gene to Be Associated With Lean-Nonalcoholic Fatty Liver Disease. J. Clin. Exp. Hepatol. 2019, 9, 561–568. [Google Scholar] [CrossRef] [PubMed]
  103. Ji, F.; Liu, Y.; Hao, J.-G.; Wang, L.-P.; Dai, M.-J.; Shen, G.-F.; Yan, X.-B. KLB Gene Polymorphism Is Associated with Obesity and Non-Alcoholic Fatty Liver Disease in the Han Chinese. Aging 2019, 11, 7847–7858. [Google Scholar] [CrossRef]
  104. Dongiovanni, P.; Crudele, A.; Panera, N.; Romito, I.; Meroni, M.; De Stefanis, C.; Palma, A.; Comparcola, D.; Fracanzani, A.L.; Miele, L.; et al. β-Klotho Gene Variation Is Associated with Liver Damage in Children with NAFLD. J. Hepatol. 2020, 72, 411–419. [Google Scholar] [CrossRef] [Green Version]
  105. Yan, X.; Jin, W.; Zhang, J.; Wang, M.; Liu, S.; Xin, Y. Association of TCF7L2 Rs7903146 Gene Polymorphism with the Risk of NAFLD and CAD in the Chinese Han Population. J. Clin. Transl. Hepatol. 2020, 8, 1–6. [Google Scholar] [CrossRef] [PubMed]
  106. Bhatt, S.P.; Misra, A.; Pandey, R.M. Rs7903146 (C/T) Polymorphism of Transcription Factor 7 like 2 (TCF7L-2) Gene Is Independently Associated with Non-Alcoholic Fatty Liver Disease in Asian Indians. Diabetes Metab. Syndr. Clin. Res. Rev. 2020, 14, 175–180. [Google Scholar] [CrossRef]
  107. Perez-Diaz-del-Campo, N.; Abete, I.; Cantero, I.; Marin-Alejandre, B.A.; Monreal, J.I.; Elorz, M.; Herrero, J.I.; Benito-Boillos, A.; Riezu-Boj, J.I.; Milagro, F.I.; et al. Association of the SH2B1 Rs7359397 Gene Polymorphism with Steatosis Severity in Subjects with Obesity and Non-Alcoholic Fatty Liver Disease. Nutrients 2020, 12, 1260. [Google Scholar] [CrossRef] [PubMed]
  108. Sabzikarian, M.; Mahmoudi, T.; Tabaeian, S.P.; Rezamand, G.; Asadi, A.; Farahani, H.; Nobakht, H.; Dabiri, R.; Mansour-Ghanaei, F.; Derakhshan, F.; et al. The Common Variant of Rs6214 in Insulin like Growth Factor 1 (IGF1) Gene: A Potential Protective Factor for Non-Alcoholic Fatty Liver Disease. Arch. Physiol. Biochem. 2020, 1–6. [Google Scholar] [CrossRef]
  109. Dabiri, R.; Mahmoudi, T.; Sabzikarian, M.; Asadi, A.; Farahani, H.; Nobakht, H.; Maleki, I.; Mansour-Ghanaei, F.; Derakshshan, F.; Zali, M.R. A 3′-Untranslated Region Variant (Rs2289046) of Insulin Receptor Substrate 2 Gene Is Associated with Susceptibility to Nonalcoholic Fatty Liver Disease. Acta Gastroenterol. Belg. 2020, 83, 271–276. [Google Scholar]
  110. Teo, K.; Abeysekera, K.W.M.; Adams, L.; Aigner, E.; Anstee, Q.M.; Banales, J.M.; Banerjee, R.; Basu, P.; Berg, T.; Bhatnagar, P.; et al. Rs641738C>T near MBOAT7 Is Associated with Liver Fat, ALT and Fibrosis in NAFLD: A Meta-Analysis. J. Hepatol. 2021, 74, 20–30. [Google Scholar] [CrossRef] [PubMed]
  111. Thangapandi, V.R.; Knittelfelder, O.; Brosch, M.; Patsenker, E.; Vvedenskaya, O.; Buch, S.; Hinz, S.; Hendricks, A.; Nati, M.; Herrmann, A.; et al. Loss of Hepatic Mboat7 Leads to Liver Fibrosis. Gut 2021, 70, 940–950. [Google Scholar] [CrossRef]
  112. Topchieva, L.V.; Kurbatova, I.V.; Dudanova, O.P.; Sokolovskaya, A.A.; Shipovskaya, A.A. IL6R Gene Polymorphic Variant Rs2228145(C >A) as a Marker of Genetic Liability to Nonalcoholic Steatohepatitis in the Russian Population of Karelia. Bull. Exp. Biol. Med. 2018, 165, 64–68. [Google Scholar] [CrossRef]
  113. Damavandi, N.; Zeinali, S. Association of Xenobiotic-Metabolizing Enzymes (GSTM1 and GSTT 1), and pro-Inflammatory Cytokines (TNF-α and IL-6) Genetic Polymorphisms with Non-Alcoholic Fatty Liver Disease. Mol. Biol. Rep. 2021, 48, 1225–1231. [Google Scholar] [CrossRef] [PubMed]
  114. Crudele, A.; Dato, S.; Re, O.L.; Maugeri, A.; Sanna, P.; Giallongo, S.; Oben, J.; Panera, N.; De Rango, F.; Mosca, A.; et al. Pediatric Non-Alcoholic Fatty Liver Disease Is Affected by Genetic Variants Involved in Lifespan/Healthspan. J. Pediatric Gastroenterol. Nutr. 2021. Publish Ahead of Print. [Google Scholar] [CrossRef] [PubMed]
  115. Kempinska-Podhorodecka, A.; Wunsch, E.; Milkiewicz, P.; Stachowska, E.; Milkiewicz, M. The Association between SOCS1−1656G>A Polymorphism, Insulin Resistance and Obesity in Nonalcoholic Fatty Liver Disease (NAFLD) Patients. J. Clin. Med. 2019, 8, 1912. [Google Scholar] [CrossRef] [Green Version]
  116. Wang, J.-Z.; Zhang, Y.-H.; Bai, J.; Liu, Y.-W.; Du, W.-T. PIN1, a Perspective on Genetic Biomarker for Nonalcoholic Fatty Liver Disease (NAFLD). Metab. Open 2019, 3, 100014. [Google Scholar] [CrossRef] [PubMed]
  117. Akbulut, U.E.; Emeksiz, H.C.; Citli, S.; Cebi, A.H.; Korkmaz, H.A.A.; Baki, G. IL-17A, MCP-1, CCR-2, and ABCA1 Polymorphisms in Children with Non-Alcoholic Fatty Liver Disease. J. De Pediatr. 2019, 95, 350–357. [Google Scholar] [CrossRef] [PubMed]
  118. Namjou, B.; Lingren, T.; Huang, T.; Parameswaran, S.; Cobb, B.L.; Stanaway, I.B.; Connolly, J.J.; Mentch, F.D.; Benoit, B.; Niu, X.; et al. GWAS and Enrichment Analyses of Non-Alcoholic Fatty Liver Disease Identify New Trait-Associated Genes and Pathways across EMERGE Network. BMC Med. 2019, 17, 135. [Google Scholar] [CrossRef]
  119. Akuta, N.; Kawamura, Y.; Kobayashi, M.; Arase, Y.; Saitoh, S.; Fujiyama, S.; Sezaki, H.; Hosaka, T.; Kobayashi, M.; Suzuki, Y.; et al. TERT Promoter Mutation in Serum Cell-Free DNA Is a Diagnostic Marker of Primary Hepatocellular Carcinoma in Patients with Nonalcoholic Fatty Liver Disease. Oncology 2021, 99, 114–123. [Google Scholar] [CrossRef]
  120. Liu, Q.; Liu, S.-S.; Zhao, Z.-Z.; Zhao, B.-T.; Du, S.-X.; Jin, W.-W.; Xin, Y.-N. TRIB1 Rs17321515 Gene Polymorphism Increases the Risk of Coronary Heart Disease in General Population and Non-Alcoholic Fatty Liver Disease Patients in Chinese Han Population. Lipids Health Dis. 2019, 18, 165. [Google Scholar] [CrossRef] [Green Version]
  121. Liu, Q.; Xue, F.; Meng, J.; Liu, S.-S.; Chen, L.-Z.; Gao, H.; Geng, N.; Jin, W.-W.; Xin, Y.-N.; Xuan, S.-Y. TRIB1 Rs17321515 and Rs2954029 Gene Polymorphisms Increase the Risk of Non-Alcoholic Fatty Liver Disease in Chinese Han Population. Lipids Health Dis. 2019, 18, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Nimer, N.; Choucair, I.; Wang, Z.; Nemet, I.; Li, L.; Gukasyan, J.; Weeks, T.L.; Alkhouri, N.; Zein, N.; Tang, W.H.W.; et al. Bile Acids Profile, Histopathological Indices and Genetic Variants for Non-Alcoholic Fatty Liver Disease Progression. Metabolism 2021, 116, 154457. [Google Scholar] [CrossRef]
  123. Habibzadeh, P.; Honarvar, B.; Silawi, M.; Bahramjahan, S.; Kazemi, A.; Faghihi, M.A.; Lankarani, K. Association between Rs2303861 Polymorphism in CD82 Gene and Non-Alcoholic Fatty Liver Disease: A Preliminary Case-Control Study. Croat. Med. J. 2019, 60, 361–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Chung, G.E.; Lee, Y.; Yim, J.Y.; Choe, E.K.; Kwak, M.-S.; Yang, J.I.; Park, B.; Lee, J.-E.; Kim, J.A.; Kim, J.S. Genetic Polymorphisms of PNPLA3 and SAMM50 Are Associated with Nonalcoholic Fatty Liver Disease in a Korean Population. Gut Liver 2018, 12, 316–323. [Google Scholar] [CrossRef] [PubMed]
  125. Li, Z.; Shen, W.; Wu, G.; Qin, C.; Zhang, Y.; Wang, Y.; Song, G.; Xiao, C.; Zhang, X.; Deng, G.; et al. The Role of SAMM50 in Non-alcoholic Fatty Liver Disease: From Genetics to Mechanisms. FEBS Open Bio 2021. [Google Scholar] [CrossRef]
  126. Nakajima, S.; Tanaka, H.; Sawada, K.; Hayashi, H.; Hasebe, T.; Abe, M.; Hasebe, C.; Fujiya, M.; Okumura, T. Polymorphism of Receptor-Type Tyrosine-Protein Phosphatase Delta Gene in the Development of Non-Alcoholic Fatty Liver Disease: PTPRD Genes in the Development of NAFLD. J. Gastroenterol. Hepatol. 2018, 33, 283–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Brady, G.F.; Kwan, R.; Ulintz, P.J.; Nguyen, P.; Bassirian, S.; Basrur, V.; Nesvizhskii, A.I.; Loomba, R.; Omary, M.B. Nuclear Lamina Genetic Variants, Including a Truncated LAP2, in Twins and Siblings with Nonalcoholic Fatty Liver Disease. Hepatology 2018, 67, 1710–1725. [Google Scholar] [CrossRef] [Green Version]
  128. Seko, Y.; Yamaguchi, K.; Mizuno, N.; Okuda, K.; Takemura, M.; Taketani, H.; Hara, T.; Umemura, A.; Nishikawa, T.; Moriguchi, M.; et al. Combination of PNPLA3 and TLL1 Polymorphism Can Predict Advanced Fibrosis in Japanese Patients with Nonalcoholic Fatty Liver Disease. J. Gastroenterol. 2018, 53, 438–448. [Google Scholar] [CrossRef]
  129. Chung, G.E.; Shin, E.; Kwak, M.-S.; In Yang, J.; Lee, J.-E.; Choe, E.K.; Yim, J.Y. The Association of Genetic Polymorphisms with Nonalcoholic Fatty Liver Disease in a Longitudinal Study. BMC Gastroenterol. 2020, 20, 344. [Google Scholar] [CrossRef] [PubMed]
  130. Arai, T.; Atsukawa, M.; Tsubota, A.; Koeda, M.; Yoshida, Y.; Okubo, T.; Nakagawa, A.; Itokawa, N.; Kondo, C.; Nakatsuka, K.; et al. Association of Vitamin D Levels and Vitamin D-Related Gene Polymorphisms with Liver Fibrosis in Patients with Biopsy-Proven Nonalcoholic Fatty Liver Disease. Dig. Liver Dis. 2019, 51, 1036–1042. [Google Scholar] [CrossRef] [PubMed]
  131. Hao, X.; Ma, C.; Xiang, T.; Ou, L.; Zeng, Q. Associations Among Methylene Tetrahydrofolate Reductase Rs1801133 C677T Gene Variant, Food Groups, and Non-Alcoholic Fatty Liver Disease Risk in the Chinese Population. Front. Genet. 2021, 12, 568398. [Google Scholar] [CrossRef]
  132. Aller, R.; López-Gomez, J.J.; Izaola, O.; Primo, D.; de Luis, D. Role of Neuropeptide Y Gene Variant (Rs161477) in Liver Histology in Obese Patients with Non-Alcoholic Fatty Liver Disease. Endocrinol. Diabetes Y Nutr. 2019, 66, 217–222. [Google Scholar] [CrossRef]
  133. Hao, X.; Zeng, Q. The Association and Interaction of Aldehyde Dehydrogenase 2 Polymorphisms with Food Group Intake and Probability of Having Non-Alcoholic Fatty Liver Disease. Diabetes Metab. Syndr. Obes. Targets Ther. 2020, 13, 5049–5057. [Google Scholar] [CrossRef]
  134. Dongiovanni, P.; Meroni, M.; Baselli, G.; Mancina, R.M.; Ruscica, M.; Longo, M.; Rametta, R.; Cespiati, A.; Pelusi, S.; Ferri, N.; et al. PCSK7 Gene Variation Bridges Atherogenic Dyslipidemia with Hepatic Inflammation in NAFLD Patients. J. Lipid Res. 2019, 60, 1144–1153. [Google Scholar] [CrossRef]
  135. Romeo, S.; Kozlitina, J.; Xing, C.; Pertsemlidis, A.; Cox, D.; Pennacchio, L.A.; Boerwinkle, E.; Cohen, J.C.; Hobbs, H.H. Genetic Variation in PNPLA3 Confers Susceptibility to Nonalcoholic Fatty Liver Disease. Nat. Genet. 2008, 40, 1461–1465. [Google Scholar] [CrossRef] [Green Version]
  136. Mazo, D.F.; Malta, F.M.; Stefano, J.T.; Salles, A.P.M.; Gomes-Gouvea, M.S.; Nastri, A.C.S.; Almeida, J.R.; Pinho, J.R.R.; Carrilho, F.J.; Oliveira, C.P. Validation of PNPLA3 Polymorphisms as Risk Factor for NAFLD and Liver Fibrosis in an Admixed Population. Ann. Hepatol. 2019, 18, 466–471. [Google Scholar] [CrossRef] [PubMed]
  137. Mehta, R.; Jeiran, K.; Koenig, A.B.; Otgonsuren, M.; Goodman, Z.; Baranova, A.; Younossi, Z. The Role of Mitochondrial Genomics in Patients with Non-Alcoholic Steatohepatitis (NASH). BMC Med. Genet. 2016, 17, 63. [Google Scholar] [CrossRef] [Green Version]
  138. Negoita, F.; Blomdahl, J.; Wasserstrom, S.; Winberg, M.E.; Osmark, P.; Larsson, S.; Stenkula, K.G.; Ekstedt, M.; Kechagias, S.; Holm, C.; et al. PNPLA3 Variant M148 Causes Resistance to Starvation-mediated Lipid Droplet Autophagy in Human Hepatocytes. J. Cell Biochem. 2019, 120, 343–356. [Google Scholar] [CrossRef] [Green Version]
  139. Carpino, G.; Pastori, D.; Baratta, F.; Overi, D.; Labbadia, G.; Polimeni, L.; Di Costanzo, A.; Pannitteri, G.; Carnevale, R.; Del Ben, M.; et al. PNPLA3 Variant and Portal/Periportal Histological Pattern in Patients with Biopsy-Proven Non-Alcoholic Fatty Liver Disease: A Possible Role for Oxidative Stress. Sci. Rep. 2017, 7, 15756. [Google Scholar] [CrossRef] [PubMed]
  140. Yuan, S.; Liu, H.; Yuan, D.; Xu, J.; Chen, Y.; Xu, X.; Xu, F.; Liang, H. PNPLA3 I148M Mediates the Regulatory Effect of NF-kB on Inflammation in PA-treated HepG2 Cells. J. Cell Mol. Med. 2020, 24, 1541–1552. [Google Scholar] [CrossRef] [PubMed]
  141. Aviram, M.; Rosenblat, M.; Bisgaier, C.L.; Newton, R.S.; Primo-Parmo, S.L.; La Du, B.N. Paraoxonase Inhibits High-Density Lipoprotein Oxidation and Preserves Its Functions. A Possible Peroxidative Role for Paraoxonase. J. Clin. Investig. 1998, 101, 1581–1590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Levy, D.; Reichert, C.O.; Bydlowski, S.P. Paraoxonases Activities and Polymorphisms in Elderly and Old-Age Diseases: An Overview. Antioxidants 2019, 8, 118. [Google Scholar] [CrossRef] [Green Version]
  143. Meneses, M.J.; Silvestre, R.; Sousa-Lima, I.; Macedo, M.P. Paraoxonase-1 as a Regulator of Glucose and Lipid Homeostasis: Impact on the Onset and Progression of Metabolic Disorders. Int. J. Mol. Sci. 2019, 20, 4049. [Google Scholar] [CrossRef] [Green Version]
  144. Wysocka, A.; Cybulski, M.; Wysokiński, A.P.; Berbeć, H.; Stążka, J.; Zapolski, T. Paraoxonase 1 Activity, Polymorphism and Atherosclerosis Risk Factors in Patients Undergoing Coronary Artery Surgery. J. Clin. Med. 2019, 8, 441. [Google Scholar] [CrossRef] [Green Version]
  145. Tsai, T.-H.; Chen, E.; Li, L.; Saha, P.; Lee, H.-J.; Huang, L.-S.; Shelness, G.S.; Chan, L.; Chang, B.H.-J. The Constitutive Lipid Droplet Protein PLIN2 Regulates Autophagy in Liver. Autophagy 2017, 13, 1130–1144. [Google Scholar] [CrossRef] [Green Version]
  146. Zhang, Y.; He, H.; Zeng, Y.-P.; Yang, L.-D.; Jia, D.; An, Z.-M.; Jia, W.-G. Lipoprotein A, Combined with Alanine Aminotransferase and Aspartate Aminotransferase, Contributes to Predicting the Occurrence of NASH: A Cross-Sectional Study. Lipids Health Dis. 2020, 19, 134. [Google Scholar] [CrossRef]
  147. Nemes, K.; Åberg, F. Interpreting Lipoproteins in Nonalcoholic Fatty Liver Disease. Curr. Opin. Lipidol. 2017, 28, 355–360. [Google Scholar] [CrossRef] [PubMed]
  148. Zhang, X.; Liu, S.; Dong, Q.; Xin, Y.; Xuan, S. The Genetics of Clinical Liver Diseases: Insight into the TM6SF2 E167K Variant. J. Clin. Transl. Hepatol. 2018, 6, 1–6. [Google Scholar] [CrossRef] [PubMed]
  149. Canivet, C.M.; Bonnafous, S.; Rousseau, D.; Leclere, P.S.; Lacas-Gervais, S.; Patouraux, S.; Sans, A.; Luci, C.; Bailly-Maitre, B.; Iannelli, A.; et al. Hepatic FNDC5 Is a Potential Local Protective Factor against Non-Alcoholic Fatty Liver. Biochim. Et Biophys. Acta (BBA) Mol. Basis Dis. 2020, 165705. [Google Scholar] [CrossRef] [PubMed]
  150. Matheson, J.; Le Foll, B. Therapeutic Potential of Peroxisome Proliferator-Activated Receptor (PPAR) Agonists in Substance Use Disorders: A Synthesis of Preclinical and Human Evidence. Cells 2020, 9, 1196. [Google Scholar] [CrossRef]
  151. Augimeri, G.; Giordano, C.; Gelsomino, L.; Plastina, P.; Barone, I.; Catalano, S.; Andò, S.; Bonofiglio, D. The Role of PPARγ Ligands in Breast Cancer: From Basic Research to Clinical Studies. Cancers 2020, 12, 2623. [Google Scholar] [CrossRef]
  152. Majid, M.; Masood, A.; Kadla, S.A.; Hameed, I.; Ganai, B.A. Association of Pro12Ala Polymorphism of Peroxisome Proliferator-Activated Receptor Gamma 2 (PPARγ2) Gene with Type 2 Diabetes Mellitus in Ethnic Kashmiri Population. Biochem. Genet. 2017, 55, 10–21. [Google Scholar] [CrossRef] [PubMed]
  153. Murakami-Nishida, S.; Matsumura, T.; Senokuchi, T.; Ishii, N.; Kinoshita, H.; Yamada, S.; Morita, Y.; Nishida, S.; Motoshima, H.; Kondo, T.; et al. Pioglitazone Suppresses Macrophage Proliferation in Apolipoprotein-E Deficient Mice by Activating PPARγ. Atherosclerosis 2019, 286, 30–39. [Google Scholar] [CrossRef] [PubMed]
  154. Boutari, C.; Mantzoros, C.S. Adiponectin and Leptin in the Diagnosis and Therapy of NAFLD. Metabolism 2020, 103, 154028. [Google Scholar] [CrossRef] [PubMed]
  155. McCormick, S.M.; Gowda, N.; Fang, J.X.; Heller, N.M. Suppressor of Cytokine Signaling (SOCS)1 Regulates Interleukin-4 (IL-4)-Activated Insulin Receptor Substrate (IRS)-2 Tyrosine Phosphorylation in Monocytes and Macrophages via the Proteasome. J. Biol. Chem. 2016, 291, 20574–20587. [Google Scholar] [CrossRef] [Green Version]
  156. Stols-Gonçalves, D.; Hovingh, G.K.; Nieuwdorp, M.; Holleboom, A.G. NAFLD and Atherosclerosis: Two Sides of the Same Dysmetabolic Coin? Trends Endocrinol. Metab. 2019, 30, 891–902. [Google Scholar] [CrossRef]
  157. Childs, B.G.; Gluscevic, M.; Baker, D.J.; Laberge, R.-M.; Marquess, D.; Dananberg, J.; van Deursen, J.M. Senescent Cells: An Emerging Target for Diseases of Ageing. Nat. Rev. Drug Discov. 2017, 16, 718–735. [Google Scholar] [CrossRef] [Green Version]
  158. Kaur, S.; Bansal, Y.; Kumar, R.; Bansal, G. A Panoramic Review of IL-6: Structure, Pathophysiological Roles and Inhibitors. Bioorganic Med. Chem. 2020, 28, 115327. [Google Scholar] [CrossRef] [PubMed]
  159. Rose-John, S. Interleukin-6 Signalling in Health and Disease. F1000Research 2020, 9, 1013. [Google Scholar] [CrossRef] [PubMed]
  160. Fäldt, J.; Wernstedt, I.; Fitzgerald, S.M.; Wallenius, K.; Bergström, G.; Jansson, J.-O. Reduced Exercise Endurance in Interleukin-6-Deficient Mice. Endocrinology 2004, 145, 2680–2686. [Google Scholar] [CrossRef] [Green Version]
  161. Domin, R.; Dadej, D.; Pytka, M.; Zybek-Kocik, A.; Ruchała, M.; Guzik, P. Effect of Various Exercise Regimens on Selected Exercise-Induced Cytokines in Healthy People. Int. J. Environ. Res. Public Halth 2021, 18, 1261. [Google Scholar] [CrossRef] [PubMed]
  162. Holbrook, J.; Lara-Reyna, S.; Jarosz-Griffiths, H.; McDermott, M.F. Tumour Necrosis Factor Signalling in Health and Disease. F1000Research 2019, 8, 111. [Google Scholar] [CrossRef]
  163. Jung, M.K.; Lee, J.S.; Kwak, J.-E.; Shin, E.-C. Tumor Necrosis Factor and Regulatory T Cells. Yonsei Med. J. 2019, 60, 126. [Google Scholar] [CrossRef] [PubMed]
  164. Ramirez-Carrozzi, V.; Ota, N.; Sambandam, A.; Wong, K.; Hackney, J.; Martinez-Martin, N.; Ouyang, W.; Pappu, R. Cutting Edge: IL-17B Uses IL-17RA and IL-17RB to Induce Type 2 Inflammation from Human Lymphocytes. J. Immunol. 2019, 202, 1935–1941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Brembilla, N.C.; Senra, L.; Boehncke, W.-H. The IL-17 Family of Cytokines in Psoriasis: IL-17A and Beyond. Front. Immunol. 2018, 9, 1682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Kern, L.; Mittenbühler, M.; Vesting, A.; Ostermann, A.; Wunderlich, C.; Wunderlich, F. Obesity-Induced TNFα and IL-6 Signaling: The Missing Link between Obesity and Inflammation—Driven Liver and Colorectal Cancers. Cancers 2018, 11, 24. [Google Scholar] [CrossRef] [Green Version]
  167. Florin, L.; Lang, T. Tetraspanin Assemblies in Virus Infection. Front. Immunol. 2018, 9, 1140. [Google Scholar] [CrossRef]
  168. Prabhu, V.V.; Devaraj, S.N. KAI1/CD82, Metastasis Suppressor Gene as a Therapeutic Target for Non-Small-Cell Lung Carcinoma. J. Env. Pathol. Toxicol. Oncol. 2017, 36, 269–275. [Google Scholar] [CrossRef]
  169. Yeung, L.; Hickey, M.J.; Wright, M.D. The Many and Varied Roles of Tetraspanins in Immune Cell Recruitment and Migration. Front. Immunol. 2018, 9, 1644. [Google Scholar] [CrossRef] [Green Version]
  170. Zhao, Y.; Kiss, T.; DelFavero, J.; Li, L.; Li, X.; Zheng, L.; Wang, J.; Jiang, C.; Shi, J.; Ungvari, Z.; et al. CD82-TRPM7-Numb Signaling Mediates Age-Related Cognitive Impairment. GeroScience 2020, 42, 595–611. [Google Scholar] [CrossRef]
  171. Neumann, E.; Schwarz, M.C.; Hasseli, R.; Hülser, M.-L.; Classen, S.; Sauerbier, M.; Rehart, S.; Mueller-Ladner, U. Tetraspanin CD82 Affects Migration, Attachment and Invasion of Rheumatoid Arthritis Synovial Fibroblasts. Ann. Rheum. Dis. 2018, 77, 1619–1626. [Google Scholar] [CrossRef]
  172. Lu, A.T.; Xue, L.; Salfati, E.L.; Chen, B.H.; Ferrucci, L.; Levy, D.; Joehanes, R.; Murabito, J.M.; Kiel, D.P.; Tsai, P.-C.; et al. GWAS of Epigenetic Aging Rates in Blood Reveals a Critical Role for TERT. Nat. Commun. 2018, 9, 387. [Google Scholar] [CrossRef] [Green Version]
  173. Akuta, N.; Suzuki, F.; Kobayashi, M.; Fujiyama, S.; Kawamura, Y.; Sezaki, H.; Hosaka, T.; Kobayashi, M.; Saitoh, S.; Arase, Y.; et al. Detection of TERT Promoter Mutation in Serum Cell-free DNA Using Wild-type Blocking PCR Combined with Sanger Sequencing in Hepatocellular Carcinoma. J. Med. Virol. 2020, 92, 3604–3608. [Google Scholar] [CrossRef] [PubMed]
  174. Trung, N.T.; Hoan, N.X.; Trung, P.Q.; Binh, M.T.; Van Tong, H.; Toan, N.L.; Bang, M.H.; Song, L.H. Clinical Significance of Combined Circulating TERT Promoter Mutations and MiR-122 Expression for Screening HBV-Related Hepatocellular Carcinoma. Sci Rep. 2020, 10, 8181. [Google Scholar] [CrossRef]
  175. Liu, S.; Gao, Y.; Zhang, C.; Li, H.; Pan, S.; Wang, X.; Du, S.; Deng, Z.; Wang, L.; Song, Z.; et al. SAMM50 Affects Mitochondrial Morphology through the Association of Drp1 in Mammalian Cells. FEBS Lett. 2016, 590, 1313–1323. [Google Scholar] [CrossRef]
  176. Ebrahimpour-Koujan, S.; Sohrabpour, A.A.; Foroughi, F.; Alvandi, E.; Esmaillzadeh, A. Effects of Vitamin D Supplementation on Liver Fibrogenic Factors in Non-Alcoholic Fatty Liver Patients with Steatohepatitis: Study Protocol for a Randomized Clinical Trial. Trials 2019, 20, 153. [Google Scholar] [CrossRef] [PubMed]
  177. Bakos, B.; Szili, B.; Szabó, B.; Horváth, P.; Kirschner, G.; Kósa, J.P.; Toldy, E.; Lakatos, P.; Tabák, Á.G.; Takács, I. Genetic Variants of VDR and CYP2R1 Affect BMI Independently of Serum Vitamin D Concentrations. BMC Med. Genet. 2020, 21, 129. [Google Scholar] [CrossRef]
  178. Hu, Z.; Tao, S.; Liu, H.; Pan, G.; Li, B.; Zhang, Z. The Association between Polymorphisms of Vitamin D Metabolic-Related Genes and Vitamin D 3 Supplementation in Type 2 Diabetic Patients. J. Diabetes Res. 2019, 2019, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Nam, H.; Rhie, Y.; Lee, K. Vitamin D Level and Gene Polymorphisms in Korean Children with Type 1 Diabetes. Pediatr. Diabetes 2019. [Google Scholar] [CrossRef]
  180. Zhang, Z.; Thorne, J.L.; Moore, J.B. Vitamin D and Nonalcoholic Fatty Liver Disease. Curr. Opin. Clin. Nutr. Metab. Care 2019, 22, 449–458. [Google Scholar] [CrossRef]
  181. Pacheco, D.; Izaola, O.; Primo, D.; de Luis, D. Allele a of the Rs16147 Variant of Neuropeptide Y Predicts Early Metabolic Improvements after Bariatric Surgery with Biliopancreatic Diversion in Morbid Obese Subjects. Clin. Nutr. Open Sci. 2021, 36, 26–34. [Google Scholar] [CrossRef]
  182. Chen, F.; Zhou, Y.; Yang, K.; Shen, M.; Wang, Y. NPY Stimulates Cholesterol Synthesis Acutely by Activating the SREBP2-HMGCR Pathway through the Y1 and Y5 Receptors in Murine Hepatocytes. Life Sci. 2020, 262, 118478. [Google Scholar] [CrossRef] [PubMed]
  183. Kozlitina, J. Genetic Risk Factors and Disease Modifiers of Nonalcoholic Steatohepatitis. Gastroenterol. Clin. N. Am. 2020, 49, 25–44. [Google Scholar] [CrossRef]
  184. Krawczyk, M.; Portincasa, P.; Lammert, F. PNPLA3-Associated Steatohepatitis: Toward a Gene-Based Classification of Fatty Liver Disease. Semin. Liver Dis. 2013, 33, 369–379. [Google Scholar] [CrossRef] [Green Version]
  185. Smagris, E.; BasuRay, S.; Li, J.; Huang, Y.; Lai, K.V.; Gromada, J.; Cohen, J.C.; Hobbs, H.H. Pnpla3I148M Knockin Mice Accumulate PNPLA3 on Lipid Droplets and Develop Hepatic Steatosis. Hepatology 2015, 61, 108–118. [Google Scholar] [CrossRef] [Green Version]
  186. BasuRay, S.; Smagris, E.; Cohen, J.C.; Hobbs, H.H. The PNPLA3 Variant Associated with Fatty Liver Disease (I148M) Accumulates on Lipid Droplets by Evading Ubiquitylation. Hepatology 2017, 66, 1111–1124. [Google Scholar] [CrossRef] [Green Version]
  187. Luukkonen, P.K.; Nick, A.; Hölttä-Vuori, M.; Thiele, C.; Isokuortti, E.; Lallukka-Brück, S.; Zhou, Y.; Hakkarainen, A.; Lundbom, N.; Peltonen, M.; et al. Human PNPLA3-I148M Variant Increases Hepatic Retention of Polyunsaturated Fatty Acids. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [PubMed]
  188. Simons, N.; Isaacs, A.; Koek, G.H.; Kuč, S.; Schaper, N.C.; Brouwers, M.C.G.J. PNPLA3, TM6SF2, and MBOAT7 Genotypes and Coronary Artery Disease. Gastroenterology 2017, 152, 912–913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Naunyn, B. Der Diabetes Melitus; Alfred Hölder: Wien, Austria, 1898. [Google Scholar]
  190. Göke, B. Die Leber im Zentrum des Kohlenhydratstoffwechsels. Diabetologe 2016, 12, 460. [Google Scholar] [CrossRef] [Green Version]
  191. Ballestri, S.; Zona, S.; Targher, G.; Romagnoli, D.; Baldelli, E.; Nascimbeni, F.; Roverato, A.; Guaraldi, G.; Lonardo, A. Nonalcoholic Fatty Liver Disease Is Associated with an Almost Twofold Increased Risk of Incident Type 2 Diabetes and Metabolic Syndrome. Evidence from a Systematic Review and Meta-Analysis: NAFLD Foreruns Metabolic Syndrome. J. Gastroenterol. Hepatol. 2016, 31, 936–944. [Google Scholar] [CrossRef] [PubMed]
  192. Marušić, M.; Paić, M.; Knobloch, M.; Liberati Pršo, A.-M. NAFLD, Insulin Resistance, and Diabetes Mellitus Type 2. Can. J. Gastroenterol. Hepatol. 2021, 2021, 1–9. [Google Scholar] [CrossRef]
  193. Caussy, C.; Aubin, A.; Loomba, R. The Relationship Between Type 2 Diabetes, NAFLD, and Cardiovascular Risk. Curr. Diab. Rep. 2021, 21, 15. [Google Scholar] [CrossRef]
  194. Lonardo, A.; Nascimbeni, F.; Mantovani, A.; Targher, G. Hypertension, Diabetes, Atherosclerosis and NASH: Cause or Consequence? J. Hepatol. 2018, 68, 335–352. [Google Scholar] [CrossRef]
  195. De Vries, M.; Westerink, J.; Kaasjager, K.H.A.H.; de Valk, H.W. Prevalence of Nonalcoholic Fatty Liver Disease (NAFLD) in Patients With Type 1 Diabetes Mellitus: A Systematic Review and Meta-Analysis. J. Clin. Endocrinol. Metab. 2020, 105, 3842–3853. [Google Scholar] [CrossRef]
  196. Barros, B.S.V.; Monteiro, F.C.; Terra, C.; Gomes, M.B. Prevalence of Non-Alcoholic Fatty Liver Disease and Its Associated Factors in Individuals with Type 1 Diabetes: A Cross-Sectional Study in a Tertiary Care Center in Brazil. Diabetol. Metab. Syndr. 2021, 13, 33. [Google Scholar] [CrossRef]
  197. Nobili, V.; Mantovani, A.; Cianfarani, S.; Alisi, A.; Mosca, A.; Sartorelli, M.R.; Maffeis, C.; Loomba, R.; Byrne, C.D.; Targher, G. Prevalence of Prediabetes and Diabetes in Children and Adolescents with Biopsy-Proven Non-Alcoholic Fatty Liver Disease. J. Hepatol. 2019, 71, 802–810. [Google Scholar] [CrossRef] [PubMed]
  198. Taylor, R.; Al-Mrabeh, A.; Zhyzhneuskaya, S.; Peters, C.; Barnes, A.C.; Aribisala, B.S.; Hollingsworth, K.G.; Mathers, J.C.; Sattar, N.; Lean, M.E.J. Remission of Human Type 2 Diabetes Requires Decrease in Liver and Pancreas Fat Content but Is Dependent upon Capacity for β Cell Recovery. Cell Metab. 2018, 28, 547–556.e3. [Google Scholar] [CrossRef] [Green Version]
  199. De Silva, N.M.G.; Borges, M.C.; Hingorani, A.; Engmann, J.; Shah, T.; Zhang, X.; Luan, J.; Langenberg, C.; Wong, A.; Kuh, D.; et al. Liver Function and Risk of Type 2 Diabetes: Bidirectional Mendelian Randomization Study. Diabetes 2019. [Google Scholar] [CrossRef] [Green Version]
  200. Fujii, H.; Kawada, N.; Japan Study Group of NAFLD (JSG-NAFLD). The Role of Insulin Resistance and Diabetes in Nonalcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2020, 21, 3863. [Google Scholar] [CrossRef] [PubMed]
  201. Meroni, M.; Longo, M.; Rustichelli, A.; Dongiovanni, P. Nutrition and Genetics in NAFLD: The Perfect Binomium. Int. J. Mol. Sci. 2020, 21, 2986. [Google Scholar] [CrossRef] [Green Version]
  202. Barata, L.; Feitosa, M.F.; Bielak, L.F.; Halligan, B.; Baldridge, A.S.; Guo, X.; Yerges-Armstrong, L.M.; Smith, A.V.; Yao, J.; Palmer, N.D.; et al. Insulin Resistance Exacerbates Genetic Predisposition to Nonalcoholic Fatty Liver Disease in Individuals Without Diabetes. Hepatol. Commun. 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. NAFLD Clinical Study Group (NAFLD CSG); Krawczyk, M.; Bantel, H.; Rau, M.; Schattenberg, J.M.; Grünhage, F.; Pathil, A.; Demir, M.; Kluwe, J.; Boettler, T.; et al. Could Inherited Predisposition Drive Non-Obese Fatty Liver Disease? Results from German Tertiary Referral Centers. J. Hum. Genet. 2018, 63, 621–626. [Google Scholar] [CrossRef]
  204. Mancina, R.M.; Dongiovanni, P.; Petta, S.; Pingitore, P.; Meroni, M.; Rametta, R.; Borén, J.; Montalcini, T.; Pujia, A.; Wiklund, O.; et al. The MBOAT7-TMC4 Variant Rs641738 Increases Risk of Nonalcoholic Fatty Liver Disease in Individuals of European Descent. Gastroenterology 2016, 150, 1219–1230.e6. [Google Scholar] [CrossRef] [Green Version]
  205. Longo, M.; Meroni, M.; Erconi, V.; Carli, F.; Macchi, C.; Ronchi, D.; Sabatini, S.; Paolini, E.; De Caro, E.R.; Alisi, A.; et al. TM6SF2/PNPLA3/MBOAT7 Loss-of-Function Genetic Variants Impact on NAFLD Development and Progression Both in Patients and in in Vitro Models. Dig. Liver Dis. 2021, 53, S27–S28. [Google Scholar] [CrossRef]
  206. Kogiso, T.; Sagawa, T.; Kodama, K.; Taniai, M.; Hashimoto, E.; Tokushige, K. Development and Course of Diabetes According to Genetic Factors and Diabetes Treatment among Patients with Nonalcoholic Fatty Liver Disease. Nutrition 2021, 83, 111080. [Google Scholar] [CrossRef]
  207. Xia, M.-F.; Lin, H.-D.; Chen, L.-Y.; Wu, L.; Ma, H.; Li, Q.; Aleteng, Q.; Hu, Y.; He, W.-Y.; Gao, J.; et al. The PNPLA3 Rs738409 C>G Variant Interacts with Changes in Body Weight over Time to Aggravate Liver Steatosis, but Reduces the Risk of Incident Type 2 Diabetes. Diabetologia 2019, 62, 644–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. 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, Vol. XX, No. X, 2017. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef] [PubMed]
  209. Petroni, M.L.; Brodosi, L.; Bugianesi, E.; Marchesini, G. Management of Non-Alcoholic Fatty Liver Disease. BMJ 2021, 372, m4747. [Google Scholar] [CrossRef]
  210. Li, H.-Y.; Gan, R.-Y.; Shang, A.; Mao, Q.-Q.; Sun, Q.-C.; Wu, D.-T.; Geng, F.; He, X.-Q.; Li, H.-B. Plant-Based Foods and Their Bioactive Compounds on Fatty Liver Disease: Effects, Mechanisms, and Clinical Application. Oxidative Med. Cell. Longev. 2021, 2021, 1–23. [Google Scholar] [CrossRef]
  211. Mantovani, A.; Dalbeni, A. Treatments for NAFLD: State of Art. Int. J. Mol. Sci. 2021, 22, 2350. [Google Scholar] [CrossRef]
  212. Attia, S.L.; Softic, S.; Mouzaki, M. Evolving Role for Pharmacotherapy in NAFLD/NASH. Clin. Transl. Sci. 2021, 14, 11–19. [Google Scholar] [CrossRef]
  213. Lian, J.; Fu, J. Efficacy of Various Hypoglycemic Agents in the Treatment of Patients with Nonalcoholic Liver Disease With or Without Diabetes: A Network Meta-Analysis. Front. Endocrinol. 2021, 12, 649018. [Google Scholar] [CrossRef] [PubMed]
  214. Liu, J.; Wang, T.; He, K.; Xu, M.; Gong, J.-P. Cardiolipin Inhibitor Ameliorates the Non-Alcoholic Steatohepatitis through Suppressing NLRP3 Inflammasome Activation. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 8158–8167. [Google Scholar] [CrossRef]
  215. Shi, H.; Zhang, Y.; Xing, J.; Liu, L.; Qiao, F.; Li, J.; Chen, Y. Baicalin Attenuates Hepatic Injury in Non-Alcoholic Steatohepatitis Cell Model by Suppressing Inflammasome-Dependent GSDMD-Mediated Cell Pyroptosis. Int. Immunopharmacol. 2020, 81, 106195. [Google Scholar] [CrossRef]
  216. Chen, H.-W.; Yen, C.-C.; Kuo, L.-L.; Lo, C.-W.; Huang, C.-S.; Chen, C.-C.; Lii, C.-K. Benzyl Isothiocyanate Ameliorates High-Fat/Cholesterol/Cholic Acid Diet-Induced Nonalcoholic Steatohepatitis through Inhibiting Cholesterol Crystal-Activated NLRP3 Inflammasome in Kupffer Cells. Toxicol. Appl. Pharmacol. 2020, 393, 114941. [Google Scholar] [CrossRef] [PubMed]
  217. Hwangbo, H.; Kim, M.Y.; Ji, S.Y.; Kim, S.Y.; Lee, H.; Kim, G.-Y.; Park, C.; Keum, Y.-S.; Hong, S.H.; Cheong, J.; et al. Auranofin Attenuates Non-Alcoholic Fatty Liver Disease by Suppressing Lipid Accumulation and NLRP3 Inflammasome-Mediated Hepatic Inflammation In Vivo and In Vitro. Antioxidants 2020, 9, 1040. [Google Scholar] [CrossRef] [PubMed]
  218. Yang, G.; Jang, J.H.; Kim, S.W.; Han, S.-H.; Ma, K.-H.; Jang, J.-K.; Kang, H.C.; Cho, Y.-Y.; Lee, H.S.; Lee, J.Y. Sweroside Prevents Non-Alcoholic Steatohepatitis by Suppressing Activation of the NLRP3 Inflammasome. Int. J. Mol. Sci. 2020, 21, 2790. [Google Scholar] [CrossRef] [Green Version]
  219. Lv, Y.; Gao, X.; Luo, Y.; Fan, W.; Shen, T.; Ding, C.; Yao, M.; Song, S.; Yan, L. Apigenin Ameliorates HFD-Induced NAFLD through Regulation of the XO/NLRP3 Pathways. J. Nutr. Biochem. 2019, 71, 110–121. [Google Scholar] [CrossRef]
  220. Flores-Costa, R.; Duran-Güell, M.; Casulleras, M.; López-Vicario, C.; Alcaraz-Quiles, J.; Diaz, A.; Lozano, J.J.; Titos, E.; Hall, K.; Sarno, R.; et al. Stimulation of Soluble Guanylate Cyclase Exerts Antiinflammatory Actions in the Liver through a VASP/NF-ΚB/NLRP3 Inflammasome Circuit. Proc. Natl. Acad. Sci. USA 2020, 117, 28263–28274. [Google Scholar] [CrossRef] [PubMed]
  221. Wang, Q.; Ou, Y.; Hu, G.; Wen, C.; Yue, S.; Chen, C.; Xu, L.; Xie, J.; Dai, H.; Xiao, H.; et al. Naringenin Attenuates Non-alcoholic Fatty Liver Disease by Down-regulating the NLRP3/NF-κB Pathway in Mice. Br. J. Pharm. 2020, 177, 1806–1821. [Google Scholar] [CrossRef]
  222. Patel, K.; Singh, G.K.; Patel, D.K. A Review on Pharmacological and Analytical Aspects of Naringenin. Chin. J. Integr. Med. 2018, 24, 551–560. [Google Scholar] [CrossRef]
  223. Zobeiri, M.; Belwal, T.; Parvizi, F.; Naseri, R.; Farzaei, M.H.; Nabavi, S.F.; Sureda, A.; Nabavi, S.M. Naringenin and Its Nano-Formulations for Fatty Liver: Cellular Modes of Action and Clinical Perspective. Curr. Pharm. Biotechnol. 2018, 19, 196–205. [Google Scholar] [CrossRef] [PubMed]
  224. Den Hartogh, D.J.; Tsiani, E. Antidiabetic Properties of Naringenin: A Citrus Fruit Polyphenol. Biomolecules 2019, 9, 99. [Google Scholar] [CrossRef] [Green Version]
  225. Jantan, I.; Haque, M.d.A.; Arshad, L.; Harikrishnan, H.; Septama, A.W.; Mohamed-Hussein, Z.-A. Dietary Polyphenols Suppress Chronic Inflammation by Modulation of Multiple Inflammation-Associated Cell Signaling Pathways. J. Nutr. Biochem. 2021, 108634. [Google Scholar] [CrossRef] [PubMed]
  226. Ahmed, S.A.; Parama, D.; Daimari, E.; Girisa, S.; Banik, K.; Harsha, C.; Dutta, U.; Kunnumakkara, A.B. Rationalizing the Therapeutic Potential of Apigenin against Cancer. Life Sci. 2021, 267, 118814. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Involvement of the liver mitochondria in ammonia detoxification. Under NAFLD/NASH conditions, the expression and activities of CSP1 and OTC enzymes are reduced (magenta arrows) and the efficiency of the urea cycle is diminished, which leads to hyperammonemia (green arrows) and activation of pro-fibrotic and pro-inflammatory factors favoring the disease progression.
Figure 1. Involvement of the liver mitochondria in ammonia detoxification. Under NAFLD/NASH conditions, the expression and activities of CSP1 and OTC enzymes are reduced (magenta arrows) and the efficiency of the urea cycle is diminished, which leads to hyperammonemia (green arrows) and activation of pro-fibrotic and pro-inflammatory factors favoring the disease progression.
Ijms 22 04459 g001
Table 1. List of Single-nucleotide polymorphism sites linked to NAFLD/NASH.
Table 1. List of Single-nucleotide polymorphism sites linked to NAFLD/NASH.
GeneNamePolymorphismSubstitutionAssociation with NAFLDReferencePatients Notes
Lipids/free fatty acids/glucose metabolism
PON1Paraoxonase-1rs854560L55MNAFLD development risk factor[74]Romanian NAFLD patients
PLIN2Perilipin-2rs35568725S251PNAFLD development risk factor[75]American NASH patients
PNPLA3Patatin-like phospholipase domain-containing protein 3rs738409444C>GI148MNAFLD development risk factor[76]Turkish NAFLD patients
[77] Chinese NAFLD patients
Associated with NAFLD and NASH susceptibility and progression[78] Brazilian NAFLD patients
rs2281135 Associated with hepatocyte ballooning, lobular and portal inflammation, and NASH[79] Indian NAFLD patients
CDKAL1Cdk5 regulatory associated protein 1-like 1rs10946398CAssociated with the high triglyceride glucose index and NAFLD development[80] Northern Chinese NAFLD patients
TM6SF2Transmembrane 6 Superfamily Member 2rs58542926CT/TT
E167K
High risk of NAFLD and CRA[81,82]Chinese Han NAFLD and CRA patients
[77] Chinese NAFLD patients
[78] Brazilian NAFLD patients
HSD17B1317β-Hydroxysteroid dehydrogenase type 13rs6834314G/GIncreased steatosis but decreased inflammation, ballooning, Mallory–Denk bodies[83]Caucasians NAFLD patients
rs62305723P260S
ABHD5Abhydrolase Domain-Containing Protein 5 c.193A>GT65ANAFLD development risk factor[84]Two Romanian origin sisters, two and five years old, normal BMI
40G>T; G
14Ter
[85]Seven families of Italian origin
CETPCholesteryl ester transfer protein rs1800777G/AAssociated with the presence of steatosis and lobulillar inflammation[86]Caucasian NAFLD patients
ADIPOQGBP-28; regulating glucose levels and fatty acid breakdownrs1501299G/TIncreased NAFLD susceptibility[87]Meta-analysis
rs11377C/GA risk factor for NAFLD development[88] Meta-analysis
LEPRLeptin receptor Q223RRisk factor for NAFLD in Chinese population[89] Meta-analysis
K109RRisk factor for NAFLD in Southeast Asian population
APOA5Apolipoprotein A5rs10750097(G/G)Associated with NAFLD[90]Chinese Han NAFLD patients
rs1263173(A/A)
rs17120035(T/T)
rs662799(G/G)
APOC3Apolipoprotein C3rs2070667AAssociated with high-grade lobular inflammation in NAFLD patients[91] Chinese Han NAFLD patients
455T>Cassociated with NAFLD[92] Asian Indian adolescents with overweight/obesity
APOEApolipoprotein Ers429358CAssociated with steatosis and liver damage[93] UK biobank samples
GPAM/
GPAT1
Glycerol-3-phosphate acyltransferasers2792751T
LALLysosomal acid lipaseE8SJM-C.894G>ALAL activity associated with cryptogenic fibrosis and cirrhosis; not associated with cryptogenic liver steatosis[94]Portugal patients with abnormal liver enzymes
FNDC5Irisin, the cleaved extracellular fragment of the Fibronectin type III domain-containing protein 5 rs3480GG allele is associated with more severe steatosis in NAFLD Through a microRNA-mediated mechanism controlling FNDC5 mRNA stability[95]Caucasian NAFLD patients
AAAssociated with severe fibrosis in NAFLD patients with sarcopenia[96] Chinese NAFLD patients
GCKRGlucokinase regulatory proteinrs1260326Ty associated with NAFLD among Asian, liver biopsy, adult, and paediatric groups[97]Asian NAFLD patients
rs780094TSignificantly increased in NAFLD cases
NNMTNicotinamide-N-Methyltransferase rs694539AASignificantly correlated with the steatosis degree, NAFLD, and NASH risk factor[98]Egyptian NAFLD patients
PPARγPeroxisome proliferator-activated receptor-γrs9817428CNAFLD susceptibility[99]Chinese NAFLD patients
rs1175543G
rs13433696G
rs2920502C
ATGR1Angiotensin II type 1 receptorrs1492100T
rs5186A1166CA1166C variant affects liver disease, insulin resistance, and endothelial dysfunction in NAFLD[100] Non-diabetic Italian NAFLD patients
SPATS2LSpermatogenesis Associated Serine Rich 2 Likers295120A/CAssociated with obesity/adiposity in NAFLD paediatric patients[101]Hispanic NAFLD paediatric patients
rs99521T/G
rs295120A/C
SEMA6ASemaphorin 6Ars2303752T/C
CAMK1DCalcium/calmodulin-dependent protein kinase IDrs17583338T/C
GAS2Growth Arrest Specific 2rs11026723A/G
NCKAP5NCK Associated Protein 5rs12619898G/A
rs17397163G/A
rs11687204C/T
rs17397380A/C
Unknown rs8005339A/G
RFX8Regulatory Factor X 8rs10865041G/TAssociated with IR in NAFLD paediatric patients
FAM19A1TAFA Chemokine Like Family Member 1rs9846667A/G
WBSCR17Williams–Beuren Syndrome Chromosomal Region 17 Proteinrs11773571C/T
DZANK1Double Zinc Ribbon and Ankyrin Repeat Domains 1rs4361192C/T
LINC00851Long Intergenic Non-Protein Coding RNA 851rs2295067A/G
Unknown rs8046133G/A
OPCMLOpioid Binding Protein/cell Adhesion Molecule Likers3923850A/GDM susceptibility in NAFLD pediatric patients
rs11727927G/A
Unknown rs11644684C/A
PEMTPhosphatidylethanolamine N-methyltransferasers7946TRisk of NAFLD development[102]Lean-NAFLD Indian patients
KLBKlotho beta rs7674434GAssociated with obesity and hepatic inflammation[103] Obese/non-obese NAFLD/non-NAFLD Chinese patients
rs12152703T
rs17618244G>AAssociated with NAFLD severity in paediatric patients[104] Italian NAFLD paediatric patients
TCF7L2Transcription factor 7-like 2rs7903146CT + TTA protective factor against the development of NAFLD[105]Chinese NAFLD and CAD patients
C/TAssociated with NAFLD in Asian Indians[106] Non-diabetic Asian Indian NAFLD patients
SH2B1SH2B adapter protein 1rs7359397TAssociated with a higher risk of developing a severe stage of NAFLD[107] Spanish overweight/obese patients with NAFLD
IGF1Insulin-like growth factor 1rs6214AA and AGProtective effects for NAFLD susceptibility[108] Iranian NAFLD patients
IRS2Insulin receptor substrate 2rs2289046GG + AGA marker of decreased NAFLD susceptibility[109]Iranian NAFLD patients
MBOAT7Lysophosphatidylinositol acyltransferase 1 (LPIAT1)rs641738C>TAssociated with higher liver fat, NAFLD presence, and severity risk factor[110]Caucasian adults, a meta-analysis
NAFLD/NASH development risk factor[111]Caucasian NAFLD patients
Immune/inflammatory response
IL6RInterleukin 6 Receptorrs2228145C>AAssociated with NAFLD development in Russian population of Karelia[112]Karelian NASH patients
TNF-αTumor Necrosis Factor αrs1800629AG/AAAssociated with NAFLD in the Iranian population[113]Iranian NAFLD patients
IL-6Interleukin 6rs1800795 CG/CC
GNAFLD-associated hyperglycemia in children[114]Italian NAFLD paediatric patients
ANRILP15 Antisense RNA rs1556516G
IL27Interleukin 27rs4788048 Associated with hepatocyte ballooning, lobular and portal inflammation and NASH[79] Indian NAFLD patients
SOCS1Suppressor of cytokine signaling 1rs2433301656G>AAssociated with obese NAFLD patients[115] Polish NAFLD patients
PIN1Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1rs2233678GAssociated with a high NAFLD risk[116]Chinese patient case-study
rs2287839C
IL-17AInterleukin-17Ars2275913197G/AAssociate with NAFLD development in obese Turkish children[117] Obese Turkish children with NAFLD
IL17RAInterleukin 17 Receptor Ars5748926THigh NAFLD activity score, a promising biomarker[118] eMERGE Network data
Other genes
TERTTelomerase Reverse Transcriptase C228TCatalytic subunit of telomerase; risk factor of NAFLD to HCC promotion[119]NAFLD Japanese patients
TRIB1Tribbles-1rs17321515AAssociated with the risk of NAFLD in the Chinese Han population[120,121] Chinese Han NAFLD patients
rs2954029A
rs2954021AMarker of transition from simple hepatic steatosis into NASH[122] American NAFLD patients
CD82 (KAI1)Metastasis suppressor, a membrane glycoproteinrs2303861A/GAssociated with the risk of NAFLD in the Iranian population[123]Iranian NAFLD patients
SAMM50Sorting and assembly machinery component 50 homologrs2143571GAssociated with the presence and severity of NAFLD in a Korean population[124]Korean NAFLD patients
rs3761472A
rs2073080T
rs738491TT + CTRisk and severity of NAFLD in Chinese Han population[125] Chinese Han NAFLD patients
rs2073082AG + GG
PTPRDProtein tyrosine phosphatase receptor type Drs35929428 GA; R995CRisk factor for NAFLD development, hepatic lipid accumulation, and fibrosis[126]Japanese NAFLD patients
TMPO/LAP2Lamina-associated polypeptide-2 InsA;T99fsAssociated with NAFLD; increased lipid droplet accumulation[127]Twin-based study
TLL1Tolloid-like 1rs17047200AT/TTadvanced risk of fibrosis[128]Japanese NAFLD patients
KCL1Kinesin light chain 1rs4906353TAssociation with a high risk of NAFLD development [129]Korean NAFLD patients
VDRVitamin D receptorrs1544410CCAssociated with advanced fibrosis in NAFLD patients[130] Japanese NAFLD patients
MTHFRMethylene tetrahydrofolate reductasers1801133C677TAssociation with a high risk of NAFLD development[131] Chinese NAFLD patients
NPYNeuropeptide Yrs16147AA lower percentage of steatohepatitis and lobular inflammation in obese NAFLD patients[132] Spanish NAFLD patients
ALDH2Aldehyde dehydrogenase 2rs671GA and AAAssociated with increased probability of NAFLD among Chinese subjects[133] Chinese NAFLD patients
PCSK7Proprotein convertase subtilisin/kexin type 7rs236918CAssociated with higher triglycerides, aminotransferases, and hepatic inflammation[134]Cross-sectional Liver Biopsy Cohort
Table 2. NLRP3 inflammasome-targeting NAFLD treatments.
Table 2. NLRP3 inflammasome-targeting NAFLD treatments.
TreatmentFuntion/TargetLine/MutantEffectReference
Cardiolipin inhibitor shRNA-CLS1NLRP3 inflammasomeC57BL/6shRNA-CLS1 treatment significantly reduced the levels of IL-1β and IL-18; NLRP3, ASC, and Caspase-1 [214]
Baicalin, a flavone glycosideNLRP3 and GSDMDHuman HepG2 cellsDown-regulates NLRP3, Caspase1, ASC, GSDMD, IL-1β, and IL-18[215]
Benzyl
isothiocyanate
NLRP3 inflammasomeC57BL/6Suppressed lipid accumulation, macrophage infiltration, fibrosis, crown-like structure formation, p20 caspase 1, and p17 IL-1β expression[216]
AuranofinNLRP3 InflammasomeC57BL/6 Decreases the body weight, epididymal fat weight, levels of AST, glucose, triglyceride, cholesterol, and LDL-c; suppressed the expressions of IL-1β, IL-18, caspase-1, NLRP3, NADPH oxidase 4, and PPARγ[217]
SwerosideNLRP3 InflammasomeC57BL/6Inhibits NLRP3 inflammasome activation by decreasing IL-1β and caspase-1 production; reduces serum AST and ALT levels, hepatic immune cell infiltration, hepatic triglyceride accumulation, and liver fibrosis[218]
ApigenininflammationC57BL/6Apigenin reverses activation of the NLRP3 inflammasome, reduces inflammatory cytokines IL-1β and IL-18 released, inhibits xanthine oxidase activity, and reduces uric acid and ROS[219]
Soluble guanylate cyclase stimulator praliciguat (PRL)VASP/
NF-κB/NLRP3 inflammasome
C57BL/6The PRL anti-inflammatory effect was associated with suppression of hepatic levels of IL-1β, NLPR3, ASC, and c-caspase-1. Mechanistically, PRL induces the protein kinase G (PKG)-mediated phosphorylation of the VASP, thus reducing NF-κB activity and Il1b and Nlrp3 gene transcription[220]
Naringenin; NLRP3 inhibitor MCC950NLRP3, inflammationNLRP3−/− HepG2 cells, primary hepatocytes, and Kuffer cells; C57BL/6Naringenin inhibits activation of the NLRP3/NF-κB pathway, lipid deposition, and IL-1β expression[221]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dabravolski, S.A.; Bezsonov, E.E.; Baig, M.S.; Popkova, T.V.; Nedosugova, L.V.; Starodubova, A.V.; Orekhov, A.N. Mitochondrial Mutations and Genetic Factors Determining NAFLD Risk. Int. J. Mol. Sci. 2021, 22, 4459. https://doi.org/10.3390/ijms22094459

AMA Style

Dabravolski SA, Bezsonov EE, Baig MS, Popkova TV, Nedosugova LV, Starodubova AV, Orekhov AN. Mitochondrial Mutations and Genetic Factors Determining NAFLD Risk. International Journal of Molecular Sciences. 2021; 22(9):4459. https://doi.org/10.3390/ijms22094459

Chicago/Turabian Style

Dabravolski, Siarhei A., Evgeny E. Bezsonov, Mirza S. Baig, Tatyana V. Popkova, Ludmila V. Nedosugova, Antonina V. Starodubova, and Alexander N. Orekhov. 2021. "Mitochondrial Mutations and Genetic Factors Determining NAFLD Risk" International Journal of Molecular Sciences 22, no. 9: 4459. https://doi.org/10.3390/ijms22094459

APA Style

Dabravolski, S. A., Bezsonov, E. E., Baig, M. S., Popkova, T. V., Nedosugova, L. V., Starodubova, A. V., & Orekhov, A. N. (2021). Mitochondrial Mutations and Genetic Factors Determining NAFLD Risk. International Journal of Molecular Sciences, 22(9), 4459. https://doi.org/10.3390/ijms22094459

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