Next Article in Journal
The Role of Nutritional Support in Cured/Chronic Patients
Previous Article in Journal
Characteristic-Grouped Adiposity Indicators for Identifying Metabolic Syndrome in Adolescents: Develop and Valid Risk Screening Tools Using Dual Population
Previous Article in Special Issue
Ethyl Acetate Fraction of Amomum xanthioides Ameliorates Nonalcoholic Fatty Liver Disease in a High-Fat Diet Mouse Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pediatric Non-Alcoholic Fatty Liver Disease: Nutritional Origins and Potential Molecular Mechanisms

1
Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
2
Department of Pediatrics, Section of Gastroenterology, Hepatology & Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
3
Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
4
Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
*
Author to whom correspondence should be addressed.
Nutrients 2020, 12(10), 3166; https://doi.org/10.3390/nu12103166
Submission received: 10 September 2020 / Revised: 12 October 2020 / Accepted: 13 October 2020 / Published: 16 October 2020
(This article belongs to the Special Issue Nutrition and Metabolic (Non-alcoholic) Fatty Liver Disease (NAFLD))

Abstract

:
Non-alcoholic fatty liver disease (NAFLD) is the number one chronic liver disease worldwide and is estimated to affect nearly 40% of obese youth and up to 10% of the general pediatric population without any obvious signs or symptoms. Although the early stages of NAFLD are reversible with diet and lifestyle modifications, detecting such stages is hindered by a lack of non-invasive methods of risk assessment and diagnosis. This absence of non-invasive means of diagnosis is directly related to the scarcity of long-term prospective studies of pediatric NAFLD in children and adolescents. In the majority of pediatric NAFLD cases, the mechanisms driving the origin and rapid progression of NAFLD remain unknown. The progression from NAFLD to non-alcoholic steatohepatitis (NASH) in youth is associated with unique histological features and possible immune processes and metabolic pathways that may reflect different mechanisms compared with adults. Recent data suggest that circulating microRNAs (miRNAs) are important new biomarkers underlying pathways of liver injury. Several factors may contribute to pediatric NAFLD development, including high-sugar diets, in utero exposures via epigenetic alterations, changes in the neonatal microbiome, and altered immune system development and mitochondrial function. This review focuses on the unique aspects of pediatric NAFLD and how nutritional exposures impact the immune system, mitochondria, and liver/gastrointestinal metabolic health. These factors highlight the need for answers to how NAFLD develops in children and for early stage-specific interventions.

1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is a generic term that describes a spectrum of diseases including non-alcoholic fatty liver (NAFL), non-alcoholic steatohepatitis (NASH), fibrosis, and NAFLD-cirrhosis [1]. The global epidemic of NAFLD is increasing exponentially owing to the growing prevalence of obesity and type 2 diabetes (T2D) in children and adults along with the aging population [2]. Recent estimates indicate that the global prevalence of NAFLD is 25%, with the highest prevalence in the Middle East and South America and the lowest in Africa [3]. NAFLD is the most common liver disorder and currently is the second most common cause of liver transplantation [4]. NAFLD is estimated to affect 34% of obese children aged 2–19 years and 10% of the general pediatric population [5]. Pediatric NAFLD is associated with extrahepatic complications such as early atherosclerosis and cardiac dysfunction [6,7] and abnormal renal function [8]. Recently, NAFLD has been renamed by some as metabolic (dysfunction) associated fatty liver disease (MAFLD) as the majority of patients with fatty liver have metabolic dysfunction in the form of T2D, dyslipidemia, and increased insulin resistance [9]. Pediatric NAFLD has a complex pathophysiology and is different from adult NAFLD with multiple inputs, including perinatal events. Understanding these differences may lead to new biomarkers and opportunities for novel therapeutics.

2. Clinical Pathophysiology

Despite advances in understanding NAFLD in adults, major gaps remain in defining pathways and mechanisms unique to NAFLD pathology in children. The pathophysiology of pediatric NAFLD is multi-factorial and includes complex interactions among hormonal, nutritional, genetic, and environmental factors [10] that may begin in utero [11]. Initially, NAFLD involves hepatic steatosis, which comprises lipid accumulation arising from excessive influx of fatty acids from endogenous fat depots, excess consumption of dietary fat, and hepatic de novo lipogenesis (DNL). NASH is characterized by inflammation, oxidative stress, mitochondrial dysfunction, and fibrosis [12].
Guidelines for diagnosing NAFLD in children were updated in 2017 by the North American Society of Pediatric Gastroenterology, Hepatology and Nutrition (NASPGHAN) [13]. That expert group emphasized that obese children should be prioritized for screening because of their higher likelihood to have NAFLD. They also recognized that an unmet challenge is to identify reliable and minimally invasive biomarkers for the disease. The only currently NAPSGHAN-recommended blood test for screening for pediatric NAFLD is alanine aminotransferase (ALT). The American Academy of Pediatrics endorsed the NASPGHAN recommendation to measure ALT beginning at ages 9–11 years for all obese children, and for overweight children with additional risk factors such as insulin resistance, diabetes, dyslipidemia, sleep apnea, central adiposity, or a family history of NAFLD [14]. NAFLD is likely present in obese children who have ALT values that are 2-fold higher than the sex-specific normal range. An advantage of ALT is that it is inexpensive, and the measurement has been standardized among laboratories. However, normal ranges reported among locations are variable, which complicates interpretation, and, more importantly, ALT values do not reliably differentiate NAFLD severity, or distinguish uncomplicated NAFLD from NASH in children or adults [13,15,16]. Several other potential circulating biomarkers have been proposed [17,18,19,20,21,22,23,24] but few have been tested in children with biopsy-proven NAFLD, or tracked over the course of treatment or disease progression [13,16,25].
Imaging tools have been used for screening, but most approaches have recognized limitations [13,24]. Standard B-mode ultrasound lacks specificity and sensitivity for steatosis and is not recommended [13]. Computed tomography and magnetic resonance (MR) modalities are better than ultrasound; however, they carry concerns about radiation (the former), expense and availability of instrumentation (the latter), and the need for sedation in some children (both) [13]. Newer imaging tools that are gaining acceptance and application include MR elastography, ultrasound-based shear wave elastography using acoustic radiation force impulse (ARFI) techniques, and ultrasound-based vibration-controlled transient elastography (FibroScan) [26,27]. MR elastography has not yet been shown to be as useful in children as it has in adults and requires an MRI machine and a specific surface coil, so it is likely to be implemented only in medical centers with specialty clinics. ARFI was shown to be useful in determining liver fibrosis in pediatric patients with chronic liver disease [28] and showed high correlation with aspartate aminotransferase (AST)/ALT ratios and detecting NAFLD in childhood obesity [29]. The FibroScan has been validated for measuring liver steatosis in adults and children and is FDA approved for clinical and research applications [24,30]. There are small, medium, and extra-large probes that can be selected to accommodate the size of the patient, which obviously spans a large potential range from younger children to adolescents and young adults. Like MR-based approaches, it is not yet widely available in pediatric clinics [31]. Thus, imaging techniques have promise but additional tools are needed for comprehensive liver health profiling.
The current gold standard for confirmation of NAFLD is histological examination of liver tissue obtained by biopsy, as it rules out other causes of liver dysfunction [13]. Histological differences have been demonstrated in pediatric versus adult NAFLD, with children being more likely to display portal inflammation and fibrosis and less ballooning and peri-sinusoidal fibrosis than adults [25]. Whether those distinctions result in different trajectories of disease progression or responses to treatment is not yet known. It should be noted that, in a separate clinical guideline for pediatric NAFLD released by the American Association for the Study of Liver Diseases in 2018, there were no recommended blood or imaging tests for screening for NAFLD in obese children because of the paucity of evidence [32]. Recommendations for clinical work up and liver biopsy, however, were similar to those made by NASPGHAN [26].
Bile acids are commonly studied as biomarkers and therapeutic targets for NAFLD. Bile acids are synthesized from cholesterol in the liver and are the major components of bile. Altered bile acid composition and metabolism have been reported during the progression of NAFLD [33]. Currently, little evidence exists linking the development of cholestasis with NAFLD/NASH. Metabolomic analysis revealed significantly increased serum levels of glycochenodeoxycholate, glycocholate, and taurocholate in patients with NAFLD compared with healthy controls [34]. Research in this field is complicated by the complexity of the liver-bile-intestinal axis and is therefore more focused on pharmacotherapies like the FXR-agonist, obeticholic acid, and peroxisome proliferator-activated receptor (PPAR) agonists such as saroglitazar rather than on bile acids as clinical biomarkers. Free fatty acids and their metabolites, which contribute to liver injury via increased oxidative stress, are typically elevated in children and adults with obesity and NAFLD [35,36,37,38]. As with bile acids, free fatty acids are mostly used as therapeutic targets rather than clinical biomarkers.
Genetic factors are associated with NAFLD susceptibility and progression. A variant in the patatin-like phospholipase domain-containing protein 3 gene (PNPLA3) is associated with increased liver fat, fibrosis, and risk for carcinoma, with a higher prevalence of the at-risk allele in Hispanic youth [39,40]. A variant in the glucokinase regulatory protein (GCKR) gene was associated with an increased rate of DNL in obese adolescents [41], and a minor allele in the transmembrane 6 superfamily 2 human gene (TM6SF2) was associated with higher fibrosis and NAFLD Activity Score in children [42]. Lysosomal acid lipase (LAL) deficiency is observed in two recessive genetic disorders involving increased lysosomal cholesterol ester storage. LAL activity was shown to be significantly reduced in children [43] and adults [44,45] with NAFLD, suggesting a possible role of LAL reduction in the progression of NAFLD [46]. It is not yet known whether the presence of these polymorphisms modify the response to lifestyle or pharmacological interventions designed to slow or reverse the development and progression of NAFLD. However, Van Name et al. [47] demonstrated that a small group (n = 17) of obese children who completed a 12-week diet with low n-6/n-3 fatty acids had favorable changes in lipids, liver fat, and insulin sensitivity and these changes were the same or slightly better in patients with the PNPLA3 “at risk” genotype.

3. Role of Nutrients in Pediatric NAFLD

Animal and human evidence supports the adverse effects of high sugar intake, particularly fructose, on obesity and pediatric NAFLD risk, including in utero exposure [48,49,50,51]. Fructose consumption acutely stimulates hepatic DNL in adolescents and adults [41,52,53]. In mice chronically consuming a high-fat diet (HFD), the addition of dietary sugars promoted triglyceride production [54,55]. Extra glucose promoted lipid synthesis through activation of the transcription factor carbohydrate-responsive element-binding protein (Chrebp), while extra fructose activated both Chrebp and sterol regulatory element-binding protein 1 (Srebp1) [54]. Dietary glucose and fructose exerted different effects on mitochondrial protein acetylation and malonyl CoA, resulting in a greater reduction in fatty acid oxidation and greater lipid synthesis in response to the extra fructose diet compared with the extra glucose diet [55].
It is perhaps not surprising that a reduction of dietary sugars results in several improvements in liver health. In one study, obese adolescents who were habitually high consumers of dietary sugar were placed on a prescribed diet for nine days that limited added sugar and fructose to 10% and 4% of energy intake, respectively [56,57]. In response to this short-term intervention, liver fat, DNL, and fasting insulin decreased, accompanied by a small (1.1 kg) amount of weight loss [56,57]. Subsequent interventions lasting eight weeks that were designed to be more sustainable in pediatric clinical settings yielded consistent results [58,59]. Schwimmer et al. [58] enrolled boys 11 to 16 years old with NAFLD and at least 10% hepatic fat content. They were randomly assigned to either a usual diet (control) or a low-sugar diet with a goal of less than 3% of daily energy from added sugars (intervention). For intervention participants, meals for their entire family were provided and existing foods with excess sugar were removed from their home. In response to the low-sugar diet, hepatic fat content was reduced by about 8% versus only 1% change in the control group. There was also greater reduction in plasma liver enzyme activities in the low-sugar group, but no significant changes were observed in fasting insulin or triglycerides. In a similar manner, Goss et al. [59] compared the effect of moderate reductions in dietary carbohydrate versus dietary fat in boys and girls with obesity and NAFLD. All meals were provided for the first two weeks and then families were given instructions on how to follow dietary guidelines for the remaining six weeks. Following the intervention, hepatic lipid was reduced in the low-carbohydrate group by 6% but only 1% in the low-fat group. The low-carbohydrate group also maintained fasting insulin sensitivity (HOMA-IR), whereas the low-fat group had an increase in HOMA-IR. Each of these studies demonstrate that dietary sugar reduction can lead to improvement in liver steatosis and potentially other health outcomes in obese adolescents.
Studies in humans have shown that individuals with NAFLD have low omega-3 polyunsaturated fatty acid (n-3 PUFA) intake and high n-6/n-3 PUFA intake ratio [60,61]. Owing to their effect on hepatic lipid metabolism and inflammation, n-3 PUFA as a nutrient supplementation has been recommended for improving NAFLD [62,63]. A randomized control study by Nobili et al. [64] reported that docosahexaenoic acid taken orally for 6 months reduced liver fat content and improved insulin sensitivity in children with NAFLD. Furthermore, Janczyk et al. [65] reported that although n-3 PUFA supplementation for 6 months did not improve steatosis as determined by ultrasound and ALT levels, it improved AST and gamma-glutamyl transpeptidase levels in children with NAFLD compared with placebo. Another study found that n-3 PUFA supplementation for 12 months had beneficial effects on steatosis and ALT levels in children with NAFLD and obesity [66]. Overall, nutrient intervention with modified PUFA levels appears to be safe and efficacious for the treatment of NAFLD in children. However, the real challenge for clinicians, behaviorists, and public health officials is to transfer these effects from small, highly controlled feeding studies to strategies that can be broadly implemented, with affordable, palatable, and sustainable diet options for families.

4. Mitochondrial Dysfunction and Oxidative Stress in the Progression of Pediatric NAFLD

Altered mitochondrial dynamics and function has been implicated in the pathophysiology of NAFLD, insulin resistance, T2D, and cardiovascular diseases in adults [67,68,69]. Examination of mitochondrial ultrastructure in NASH patients by electron microscopy identified the structural abnormalities characterized by the presence of megamitochondria with crystalline inclusions [70], swollen, rounded mitochondria, and loss of cristae [71]. Likewise, an in-depth investigation of mitochondrial ultrastructure in liver biopsy specimens from children aged 2–14 years with previously clinicopathologically-diagnosed NASH revealed numerous mitochondrial abnormalities such as adult NASH patients [72].
One emerging concept about NAFLD progression is that the liver makes several initial adaptations to nutrient overload and oxidative stress but eventually becomes overwhelmed and shows signs of decompensation. Mitochondria are intimately involved in this process. For example, Kolkaki et al. [73] performed detailed functional analyses of mitochondria acquired from liver samples from adults classified as normal-weight healthy controls, obese with or without liver steatosis, or obese with NASH. They found that mitochondrial respiration increased in the obese steatotic group compared with normal-weight and obese groups without steatosis. The NASH group had respiration rates similar to the normal-weight controls, despite ~30% higher mitochondrial mass, attributable in part to increased mitochondrial leak. Additionally, the NASH group had increased markers of oxidative stress, lipid peroxidation, and inflammation. These data support the concept that liver mitochondrial oxidative capacity in healthy people with obesity expands to accommodate increased carbon flux but eventually oxidative stress during NASH development causes mitochondrial failure. Further support for this idea was provided in a murine study, which demonstrated that elevated fatty acid availability caused the liver to increase its oxidative capacity, presumably to handle the excess lipid, but with the cost of increased oxidative stress and inflammation as carbon flux through anaplerotic reactions in mitochondria was increased [74]. The same authors reported that, in human liver samples, the calculated rate of oxidative flux was positively correlated with the severity of histopathology [74]. Additional human studies by the same group confirmed that oxidative flux through liver mitochondria was increased in people with liver steatosis compared with people without steatosis, and it was accompanied by increased anaplerosis, leading to increased gluconeogenesis and oxidative stress [38,75]. In rodents, the generation of mitochondria-derived damage-associated molecular patterns (mito-DAMPs) in the liver triggered inflammation and fibrosis [76]. That study also showed that mito-DAMPs were increased in humans with NASH and fibrosis. Thus, the liver responds to excess lipid or carbohydrate input through enhanced mitochondrial oxidation or anaplerotic carbon flux, but under chronic nutrient overload, oxidative stress leads to mitochondrial failure. In some animal studies, inherently low mitochondrial capacity predisposes the liver to oxidative stress, insulin resistance, steatosis, and fibrosis [77,78], but whether this is also the case in humans has not been demonstrated.
Oxidative stress plays a key role in the pathogenesis of NAFLD and progression to NASH (reviewed recently in [79]). Nobili et al. [80] found that oxidative stress is highly prevalent in pediatric NAFLD. Reductions in oxidative stress parameters have been observed in clinical trials in children with NAFLD after treatment with vitamin E plus hydroxytyrosol [81], calorie restriction with lycopene-rich tomato juice supplementation [82], and n-3 PUFA supplementation [83]. Furthermore, lipid oxidation products (LPOs) such as 4-hydroxynonenal and malondialdehyde are produced as a result of the interaction between reactive oxygen species (ROS) and PUFAs derived from membrane phospholipids [84]. The resulting LPOs generate mutagenic and carcinogenic exocyclic etheno-DNA adducts leading to hepatic carcinogenesis. LPO-mediated etheno-DNA adducts have been positively correlated with NAFLD in adults [85] and NASH in children [86]. Hence, oxidative stress along with ROS, LPOs, and etheno-DNA adducts are important mechanisms in the onset of NAFLD and progression to hepatocellular carcinoma.

5. Gut Microbial Dysbiosis in Pediatric NAFLD

Substantial evidence now indicates that an imbalance in the gut microbiota, i.e., microbial dysbiosis, plays a critical role in the development of NAFLD through altered energy homeostasis, exacerbated inflammation, and impaired choline and bile acid metabolism [87,88]; these alterations are beyond the scope of this review. In an analysis of the gut microbiome in children, NAFLD and its severity were associated with a greater abundance of genes encoding inflammatory bacterial products [89]. One pathway implicated in NAFLD pathogenesis is bacterial translocation, or the passage of intestinal bacteria into circulation [90,91,92]. In non-human primates (NHP), short-term fructose consumption increased serum levels of lipopolysaccharide (LPS) [93], an active component of the outer membrane of intestinal bacteria. Subsequently, elevated LPS promoted NASH independent of weight gain [93]. These relationships have been explored in humans as well. A handful of dietary analyses document inverse relations of healthy diets (e.g., Mediterranean diet) [94,95] and positive relations of sugar (fructose in particular [96,97]), fat [98,99], and total energy intake [100] with serum endotoxin. Higher serum endotoxin has been linked to NAFLD risk and progression in children [101,102,103] and adults [104,105,106,107]. Genes involved in LPS biosynthesis were enriched in the gut microbiome of children with NASH [89]. In rodents fed an HFD, translocation of gut microbial products into the blood may even precede obesity [108]. Short-chain fatty acids (SCFAs) butyrate, acetate, and propionate, produced by the fermentation of carbohydrates, can act in liver and adipose tissue to decrease expression of PPARG which in turn increases fatty acid oxidation [109]. However, children with NASH ferment dietary carbohydrates that augment oxidative stress through production of endogenous alcohol [110]. These SCFAs also have the potential to dampen immunity and reduce gut permeability [111], but their roles in pediatric NAFLD are complex and not well understood.
The maintenance of homeostasis and tolerance to environmental exposures controlled by immune mechanisms may be determined by microbial-host interactions occurring during a narrow time frame contained within the earliest days of life [112,113]. Studies in neonates have underscored the importance of microbial pioneers on chronic inflammatory disease development in later life [114,115]. Animal models suggest that gut microbiome dysbiosis in early life profoundly alters the development of the innate and adaptive immune systems [116]. Disruption of this early colonization process has been linked with childhood immunological diseases and contributes to an increased risk of metabolic diseases [117,118,119]. Lemas et al. [120] found that infants born to obese mothers have an altered gut microbiota including reduced Gammaproteobacteria, an aerobic LPS-producing microbe with an essential role in immune cell function. Germ-free mice colonized with stool from 2-week-old infants born to mothers with obesity showed elevated markers of inflammation and endoplasmic reticulum stress in their livers, and these mice progressed rapidly to NAFLD when challenged with a Western-style diet (WSD) [121]. Soderborg et al. also found that these colonized mice had increased periportal inflammation, which correlated with an increased modified pediatric NAFLD histological score [121] and increased collagen disposition around the portal triad [122] compared with mice colonized with stool from 2-week-old infants from normal-weight mothers. In the liver, LPS infiltration leads to pro-inflammatory cytokine production and liver Kupffer cell (KC) activation, which can lead to the activation of hepatic stellate cells and fibrosis [123]. These results support the concept that early maternal microbial dysbiosis contributes to the detrimental effects on offspring immune function that can drive hepatic inflammation. More research is needed, however, on the how changes in serum endotoxins, SCFAs, and microbial dysbiosis affect the mechanisms for pediatric NAFLD.

6. Micro-RNAs in Fatty Liver Disease: Predictors and Prognosticators

MicroRNAs (miRNAs) are small (18–25 nucleotides) non-coding RNAs that regulate gene expression at the post-transcriptional level. The binding of miRNAs to specific messenger RNAs (mRNAs) inhibits translation, leading to reduced protein abundance of the target genes [124,125]. Each miRNA species has the potential to bind complementary sequences in multiple mRNAs and could influence several pathways. miRNAs can act both locally (intracellularly) and remotely through their secretion into the circulation and uptake into distant tissues. Blood-borne miRNAs are found within exosomes and bound to vesicle-free proteins and lipid particles, making them remarkably resistant to degradation [126,127]. The abundances of many miRNAs are altered in the liver and/or circulation in humans with NAFLD or animal/cell culture models of the disease, and several serum miRNAs have been examined as potential biomarkers [124,128,129,130]. We focus here on a few miRNAs with evidence to support their regulatory roles in the lipogenic, gluconeogenic, or β-oxidation pathways at the nexus of NAFLD development and progression (Figure 1). Importantly, most of the available evidence about the involvement of miRNAs in NAFLD comes from studies of adults or animals. We hypothesize that many of the same miRNAs operate similarly in pediatric NAFLD, but this remains to be determined.
miRNA-122 is the most extensively studied miRNA relevant to NAFLD and has the most promise as a biomarker because it is highly abundant in the liver and circulation [131,132]. It is produced almost exclusively by the liver, and its serum content increases with the presence and severity of NAFLD in adults [17,126,133] and animal models of NAFLD [127,134,135]. When considered separately or in combination with other biomarkers, miR-122 performed as well or better than ALT for predicting NAFLD severity [17,133]. The only published study of serum miR-122 content in pediatric NAFLD found that it was elevated in one cohort of children with the disease, but unaffected in another cohort, with no clear explanation for that discrepancy [136]. In adult human liver, miR-122 content is progressively reduced across the range from healthy to simple steatosis to NASH [127,137,138,139]. Initially, a healthy liver responds to rising levels of fatty acids by increasing miR-122 production, which then represses lipogenic enzymes and transcription factors such as SREBP1, fatty acid synthase (FASN), HMG-CoA reductase (HMGCR), and two enzymes required for triglyceride synthesis, 1-acyl-sn-glycerol-3-phosphase acyltransferase alpha 1 (AGPAT1) and diacylglycerol-acyltransferase 1 (DGAT1) [127,137]. This results in reduced hepatic lipid production and increased fat oxidation in normal liver [127,137]. However, longer-term nutrient overload hepatic miR-122 export accelerates and the loss of its inhibitory actions may contribute to exacerbated lipid production as NAFLD progresses to NASH and hepatic carcinoma [127,135]. The reason for excess miR-122 export in NAFLD is not yet established, although there is evidence that nutrient stress results in the upregulation of a protein that stabilizes miR-122 and directs it toward cellular export [140]. Strategies to maintain miR-122 abundance within the liver are currently being explored for hepatocellular carcinoma treatment [141,142].
miR-192 is also highly expressed in liver, has high serum content, and has promise as a biomarker for NAFLD in adults [17,126,133,143]. Since miR-192 is also abundantly produced in the gut, a change in serum miR-192 concentration may not always be the result of a change in liver health [132]. Nevertheless, serum miR-192 is increased in patients with NAFLD by up to six-fold compared with healthy comparison groups [17,126,133,143]. In animals and liver cell cultures, hepatocyte content of miR-192 declined in response to HFD or fatty acid exposure, resulting in the upregulation of one of its lipogenic targets, stearoyl-CoA desaturase (SCD1) [135,144,145]. Additionally, rats fed an HFD had increased liver miR-192 export, which activated M1 macrophages and resulted in increased liver inflammation [143]. In humans with NAFLD, serum miR-192 content was positively correlated with inflammation activity score and disease stage [143].
Other miRNAs are putative regulators of liver lipogenesis, gluconeogenesis, and fat oxidation. miR-155 represses SREBP1 and FASN in hepatocytes by acting through the liver X receptor (LXR), leading to lower triglyceride production [146]. Adults with NAFLD had lower serum and liver miR-155 content than healthy controls, while liver transcripts for SREBP1 and FASN were higher [146]. mIR-155 generation in adipose tissue is also involved in activating macrophages via the suppressor of cytokine signaling 1 (SOCS1) [129], leading to liver inflammation and insulin resistance in hepatocytes and adipocytes by targeting PPARG [129]. miR-30a promotes a shift from β-oxidation to DNL through the sirtuin 1 (SIRT1)/AMP-activated protein kinase (AMPK) pathway, and miR-30b/30c is an important co-regulator of that action [147,148]. miR-34a is increased in serum and liver from patients with NAFLD [137,149,150,151], while miR-30b/-30c is reduced in liver and serum [148,151]. miR-29a is involved in lipid oxidation through its action on the mitochondrial fat transporter, CD-36, leading to mitochondrial damage in mouse models [130]. In adults with NAFLD, serum miR-29a is increased and may be a biomarker for fibrosis and cirrhosis [130]. miR-27a and -27b regulate gluconeogenesis in mice by suppressing glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), two key enzymes responsible for hepatic gluconeogenesis [152]. This action is through forkhead box O1 (FOXO1). With chronic over nutrition, rodents display reduced liver content of miR-27a/27b while the serum level is increased [152,153]. Serum miR-27a/27b is also increased in adults with NAFLD or T2D, and children with obesity [133,152,154]. Lastly, miR-26a regulates multiple members of the hepatic gluconeogenic, β-oxidation, and insulin signaling pathways in mice [155]. In obese adults and obese mice, liver miR-26a content is reduced and restoring liver miR-26a rescues some of the adverse obesity-associated phenotype [155]. Recent work showed that the reduced liver miR-26a in obese mice also leads to an endoplasmic reticulum stress response, which can also be restored with miR-26a replacement [156]. Thus, an emerging picture is that oversupply of the liver results in increased content of several miRNAs, like those described above. This initial response helps suppress lipogenic and gluconeogenic pathways. However, with chronic overnutrition, several miRNAs are reduced in the liver, allowing the pathways they modulate to proceed unchecked. The loss of specific mRNAs in the liver is thought to be due to higher export rates to the circulation, which is why serum miRNA measurements have been tested as potential biomarkers of liver health in patients with NAFLD.
Other lines of evidence support that miRNAs regulate metabolic function early in life. For example, during pregnancy, the fetus is exposed to miRNAs from the placenta and umbilical cord and the profile of several miRNAs in those tissues are altered in mothers with diabetes [157,158,159]. Hyperglycemia increased the placental expression of miR-130b, which in turn suppressed PPARG coactivator 1 alpha (PPARGC1A) [159], a master regulator of oxidative phenotype. As a result, the abundance of mitochondrial transcription factor A (TFAM) and mitochondrial DNA content was reduced in trophoblasts [159]. How this impacts the future health and development of offspring is yet to be determined. Another way in which infants are exposed to miRNAs is through mother’s milk [160,161]. miRNAs in milk are packaged in exosomes so they survive digestion in the infant gut [160]. A question to be resolved is whether the health of the mother alters the abundance of specific miRNAs in her milk and how that might affect the development of her offspring and impact their future risk for metabolic diseases.

7. Maternal Over-Nutrition and Developmental Programming of NAFLD

Extensive animal and human evidence supports that maternal nutrition and obesity play key roles in the development of metabolic syndrome, insulin resistance, and NAFLD in offspring [11,162], summarized in Figure 2. Studies in humans demonstrated that increased intrahepatocellular lipid accumulation in neonates born to obese mothers correlated highly with maternal body mass index (BMI) [163,164]. Longitudinal, cross-sectional studies demonstrated that high or low birth weight is associated with greater odds of severe steatosis and fibrosis in adolescents with biopsy-confirmed NAFLD, independent of childhood BMI [165]. A variety of dietary approaches in animal models demonstrated a strong association between maternal obesity and the exacerbated risk of developing NAFLD in offspring [166,167,168,169]. Studies using NHP demonstrated that fetal offspring of HFD-fed dams show early signs of NAFLD; they had elevated liver triglycerides, markers of oxidative stress, and expression of gluconeogenic genes, independent of maternal obesity [170]. Interestingly, changing the maternal HFD to a low-fat diet during a subsequent pregnancy improved fetal hepatic triglyceride levels and gluconeogenic gene expression [170], suggesting that the maternal diet is a major risk factor for early NAFLD.
Experimental evidence from rodent models of maternal obesity strongly suggests impaired mitochondrial function as well as excess ROS generation in oocytes and zygotes [171] and in offspring at weaning [172] from diet-induced obese dams. Offspring exhibited peripheral insulin resistance as well as reduced liver mitochondrial DNA (mtDNA) and gene abundance of Ppargc1a [172], a key transcription factor in mitochondrial biogenesis [173]. Furthermore, oocytes and blastocytes from obese dams displayed reduced mtDNA, mitochondrial membrane potential, enhanced autophagy, and reduced mitochondrial biogenesis [174], which is linked to lipotoxic insult in liver. Interestingly, treating these obese mice with salubrinol, an endoplasmic reticulum (ER) stress inhibitor, before ovulation restored mtDNA content in association with enhanced mitochondrial replication factors, TFAM and DRP1 [174]. Likewise, Bruce et al. [168] observed that feeding HFD before or during pregnancy and lactation reduced hepatic mitochondrial electron transport chain activity and upregulated lipogenesis, driving the development of a NASH-like phenotype in offspring. In NHP, juvenile 1-year-old offspring from obese, insulin-resistant dams have a pattern of hepatic gene expression and cytokine and lipogenic responses, i.e., upregulation of SREBP1, SREBP2, FASN, ACC1, LPIN1, and DGAT1 gene expression, consistent with the early stages of NAFLD, despite weaning to a normal diet at 7 months of age [175]. Furthermore, Alfaradhi et al. [176] demonstrated in mice that peripheral insulin resistance preceded by oxidative stress in offspring of obese dams triggers the expression of hepatic genes including Pparg to promote DNL in liver. In an elegant study, Ashino et al. [177] also observed the association between maternal insulin resistance and development of NAFLD in the mouse offspring of obese dams independent of post-weaning diet.
In addition to the miRNAs discussed above, NAFLD is associated with changes in DNA methylation in offspring from humans and animal models. One study in humans aimed at identifying transcriptional changes associated with offspring metabolic reprogramming identified changes in DNA methylation in genomic regions of liver transcription factors and monocyte differentiation [178]. Wankhade et al. [179] identified that a combination of maternal HFD and post-weaning methionine- and choline-deficient diet in mice altered the DNA methylation of liver transcription factors including Ephb2 and Vwf, while maternal HFD alone changed the methylation of Hnf4a, Ppargc1b, and Fgf21. In mice with liver-specific IRS-1 deletion, parental insulin resistance was associated with epigenetically reprogrammed members of the TGF-β family, including neuronal regeneration-related protein (Nrep) and growth differentiation factor 15 (Gdf15), which modulate the expression of several genes involved in hepatic lipid metabolism [180]. In line with these data, in utero exposure to maternal HFD in NHP increased fetal liver H3K14 acetylation concomitant with reduced SIRT1 expression [181]. This reinforces the importance of parental nutrition/insulin resistance during pregnancy and lactation with epigenetic outcomes in offspring, but evidence in humans for a causal mechanism linking maternal obesity with offspring insulin resistance and pediatric NAFLD remains to be explored.

8. Programmed/Trained Inflammation in the Pathogenesis of Pediatric NAFLD

A number of innate immune cells are involved in hepatic inflammation, particularly KCs, in children with NAFLD [182,183], summarized in Figure 3. In pediatric NASH, numerous activated macrophages are found in the spaces between damaged hepatocytes [184]. However, how portal inflammation develops in childhood NASH and differs from adult NASH is unknown. The liver harbors a large population of macrophages characterized by two distinct heterogeneous subsets: resident (KCs) and infiltrating macrophages [185]. Strong experimental evidence from both in vitro and in vivo studies indicates that KCs activate hepatic stellate cells to transdifferentiate into myofibroblasts, a major collagen-producing cell type in fibrosis, in an NF-κB-dependent manner in mice [186]. Stienstra et al. [187] identified that the depletion of KCs by clodronate liposomes in HFD-fed mice significantly reduced liver triglyceride levels with the concomitant upregulation of genes involved in fatty acid oxidation, including Ppara concomitant with the downregulation of Il1b. Mouralidarane et al. [169] showed that while exposure to maternal obesity followed by an obesogenic diet increased the number of KCs in mouse offspring liver, their phagocytic function was impaired and ROS production enhanced. Another study showed that the phagocytic function of KCs is significantly impaired in rats with diet-induced NASH and in patients with NAFLD regardless of KC numbers [188]. Impaired KC phagocytosis causes hyper-production of inflammatory mediators and activates hepatic inflammation leading to liver injury in NAFLD. In children with NASH, Lotowska et al. [184] found activated KCs in close proximity to transformed hepatic stellate cells and intensive fibrosis.
Portal infiltration of macrophages is an early event in human NAFLD, occurring at the stage of steatosis before inflammation or fibrosis develops, and predicting progressive disease [189]. KCs and recruited circulating monocytes from the bone marrow (bone-marrow-derived macrophages (BMDMs)) both contribute to NAFLD pathology [190,191]. Studies in humans and in murine experimental models have shown that the hepatic recruitment of myeloid cells is regulated by chemokine receptor 2 (CCR2) and plays a critical role in obesity-induced inflammation by stimulating natural killer T cell apoptosis and releasing pro-inflammatory cytokines [192,193,194]. Recruited macrophages are highly plastic and acquire a distinct polarization state depending on the local microenvironment [195]. Mice exposed to maternal WSD have increased pro-inflammatory macrophage activation and in adulthood have increased recruited macrophages in the liver and accelerated hepatic fibrosis [196]. In another murine model, maternal HFD has been shown to restrict the expansion and renewal of fetal hematopoietic stem cells by altering the transcriptional output of genes regulating metabolism, stress response, proliferation, and other functions [197]. These data corroborate the premise that hepatic macrophages can be programmed by early diet exposure to retain a pro-inflammatory phenotype. Altogether, these data indicate the involvement of immune dysfunction in the pathogenesis of the development of NAFLD.

9. Conclusions

The pathophysiology of pediatric NAFLD is complex, multifactorial, and encompasses many unknown interactions between multiple cells types in the liver leading to NAFLD and inflammatory damage of hepatocytes. Cross-sectional studies have shown that the entire spectrum of NAFLD may occur during childhood, from hepatic steatosis to NASH to advanced fibrosis [198,199]. Despite advances in understanding adult liver pathophysiology, our understanding of pediatric NAFLD is incomplete for several reasons. First, most studies have focused on the endpoint of the disease and progressive factors have not been examined, leaving major gaps in our understanding of the many roles that make pediatric NAFLD unique from adult disease. Second, the majority of studies have examined processes in adult humans and animal models, and we know very little about critical physiological vulnerability during the perinatal period or critical transition periods such as puberty, which accelerate insulin resistance and disease patterns. Third, the lack of a suitable pediatric animal model is a major challenge for the field. Lastly, very few studies have been performed matching non-invasive discovery tools with cellular pathways for disease in individual pediatric NAFLD patients, one third of whom may advance very rapidly to NASH and cirrhosis.
Maternal obesity is associated with increased risk for childhood obesity and NAFLD in both humans and animal models. Offspring from dams fed a WSD during pregnancy are programmed for inflammation, even if the offspring are switched to a normal diet at weaning [175,196]. However, the mechanisms by which maternal WSD exposure imparts lasting effects on the development and progression of liver disease in offspring are unknown. Both antenatal and postnatal factors have a profound impact on the developing infant gut microbiome and on immune cell development [200]. An understanding of how microbial-derived factors shape macrophage metabolism and polarization toward inflammation in the gut and other tissues is needed. Gut microbes and inflammation may play an active role in promoting the progression, if not the initiation, of NAFLD in children and adults, in part through epigenetic rewiring in liver and immune cells [201]. The release of macrophage-derived mediators contributes to inflammation and fibrogenesis, suggesting that cross-talk between macrophages, stellate cells, and endothelial cells plays a key role in the progression of NAFLD to NASH. DNA methylation, covalent modification of histones, and the expression of non-coding RNAs are the epigenetic phenomena that affect inflammatory processes in the context of NAFLD [202] and need to be understood further. Remodeling these pathways during pregnancy or lactation holds the promise for altered developmental programming in the next generation. Halting and reversing pediatric NAFLD/NASH will require identification of new biomarkers for individualized therapy. Understanding how miRNAs control metabolic and inflammatory pathways in liver and their cross-talk with other extracellular tissues, particularly in pediatric NAFLD, has received relatively little attention. Advancing our understanding of early-life contributors to pediatric NAFLD via maternal and childhood diet and obesity, the gut microbiota, and metabolic and inflammatory pathways is a critical and unmet need and should motivate future studies.

Author Contributions

A.M. and J.E.F. drafted the manuscript. A.M., R.C.J., S.P., K.R.S., and J.E.F. contributed to writing the manuscript and critical revision. J.E.F. had primary responsibility for final content. All authors have read and approved the final manuscript.

Funding

Support from the National Institutes of Health grants R24-DK090964 and R56-DK114711 to J.E.F.

Conflicts of Interest

J.E.F. is a consultant to the scientific advisory board of Janssen Pharmaceuticals. All other authors report no conflict of interest.

References

  1. Bedossa, P. Pathology of non-alcoholic fatty liver disease. Liver Int. 2017, 37 (Suppl. S1), 85–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Younossi, Z.M. Non-alcoholic fatty liver disease—A global public health perspective. J. Hepatol. 2019, 70, 531–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef] [Green Version]
  4. Fazel, Y.; Koenig, A.B.; Sayiner, M.; Goodman, Z.D.; Younossi, Z.M. Epidemiology and natural history of non-alcoholic fatty liver disease. Metabolism 2016, 65, 1017–1025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Anderson, E.L.; Howe, L.D.; Jones, H.E.; Higgins, J.P.; Lawlor, D.A.; Fraser, A. The prevalence of non-alcoholic fatty liver disease in children and adolescents: A systematic review and meta-analysis. PLoS ONE 2015, 10, e0140908. [Google Scholar] [CrossRef] [Green Version]
  6. Di Sessa, A.; Umano, G.R.; Miraglia Del Giudice, E.; Santoro, N. From the liver to the heart: Cardiac dysfunction in obese children with non-alcoholic fatty liver disease. World J. Hepatol. 2017, 9, 69–73. [Google Scholar] [CrossRef] [PubMed]
  7. Pacifico, L.; Perla, F.M.; Roggini, M.; Andreoli, G.; D’Avanzo, M.; Chiesa, C. A systematic review of NAFLD-associated extrahepatic disorders in youths. J. Clin. Med. 2019, 8, 868. [Google Scholar] [CrossRef] [Green Version]
  8. Yodoshi, T.; Arce-Clachar, A.C.; Sun, Q.; Fei, L.; Bramlage, K.; Xanthakos, S.A.; Flores, F.; Mouzaki, M. Glomerular hyperfiltration is associated with liver disease severity in children with nonalcoholic fatty liver disease. J. Pediatr. 2020, 222, 127–133. [Google Scholar] [CrossRef]
  9. Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wai-Sun Wong, V.; Dufour, J.F.; Schattenberg, J.M.; et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J. Hepatol. 2020, 73, 202–209. [Google Scholar] [CrossRef]
  10. Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef]
  11. Wesolowski, S.R.; El Kasmi, K.C.; Jonscher, K.R.; Friedman, J.E. Developmental origins of NAFLD: A womb with a clue. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 81–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Dowman, J.K.; Tomlinson, J.W.; Newsome, P.N. Pathogenesis of non-alcoholic fatty liver disease. QJM 2010, 103, 71–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Vos, M.B.; Abrams, S.H.; Barlow, S.E.; Caprio, S.; Daniels, S.R.; Kohli, R.; Mouzaki, M.; Sathya, P.; Schwimmer, J.B.; Sundaram, S.S.; et al. NASPGHAN clinical practice guideline for the diagnosis and treatment of nonalcoholic fatty liver disease in children: Recommendations from the Expert Committee on NAFLD (ECON) and the North American Society of Pediatric Gastroenterology, Hepatology and Nutrition (NASPGHAN). J. Pediatr. Gastroenterol. Nutr. 2017, 64, 319–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. AAP Endorses New Guidelines on Non-Alcoholic Fatty Liver Disease. Available online: https://www.aappublications.org/news/2016/12/02/FattyLiver120216 (accessed on 10 September 2020).
  15. Maximos, M.; Bril, F.; Portillo Sanchez, P.; Lomonaco, R.; Orsak, B.; Biernacki, D.; Suman, A.; Weber, M.; Cusi, K. The role of liver fat and insulin resistance as determinants of plasma aminotransferase elevation in nonalcoholic fatty liver disease. Hepatology 2015, 61, 153–160. [Google Scholar] [CrossRef]
  16. Alkhouri, N.; Alisi, A.; Okwu, V.; Matloob, A.; Ferrari, F.; Crudele, A.; De Vito, R.; Lopez, R.; Feldstein, A.E.; Nobili, V. Circulating soluble Fas and Fas ligand levels are elevated in children with nonalcoholic steatohepatitis. Dig. Dis. Sci. 2015, 60, 2353–2359. [Google Scholar] [CrossRef]
  17. Becker, P.P.; Rau, M.; Schmitt, J.; Malsch, C.; Hammer, C.; Bantel, H.; Mullhaupt, B.; Geier, A. Performance of serum microRNAs -122, -192 and -21 as biomarkers in patients with non-alcoholic steatohepatitis. PLoS ONE 2015, 10, e0142661. [Google Scholar] [CrossRef] [PubMed]
  18. Alkhouri, N.; Feldstein, A.E. Noninvasive diagnosis of nonalcoholic fatty liver disease: Are we there yet? Metabolism 2016, 65, 1087–1095. [Google Scholar] [CrossRef] [Green Version]
  19. Decaris, M.L.; Li, K.W.; Emson, C.L.; Gatmaitan, M.; Liu, S.; Wang, Y.; Nyangau, E.; Colangelo, M.; Angel, T.E.; Beysen, C.; et al. Identifying nonalcoholic fatty liver disease patients with active fibrosis by measuring extracellular matrix remodeling rates in tissue and blood. Hepatology 2017, 65, 78–88. [Google Scholar] [CrossRef]
  20. Feng, R.; Luo, C.; Li, C.; Du, S.; Okekunle, A.P.; Li, Y.; Chen, Y.; Zi, T.; Niu, Y. Free fatty acids profile among lean, overweight and obese non-alcoholic fatty liver disease patients: A case—Control study. Lipids Health Dis. 2017, 16, 165. [Google Scholar] [CrossRef] [Green Version]
  21. 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, 9729107. [Google Scholar] [CrossRef] [PubMed]
  22. Isokuortti, E.; Zhou, Y.; Peltonen, M.; Bugianesi, E.; Clement, K.; Bonnefont-Rousselot, D.; Lacorte, J.M.; Gastaldelli, A.; Schuppan, D.; Schattenberg, J.M.; et al. Use of HOMA-IR to diagnose non-alcoholic fatty liver disease: A population-based and inter-laboratory study. Diabetologia 2017, 60, 1873–1882. [Google Scholar] [CrossRef] [PubMed]
  23. Peter, A.; Kovarova, M.; Staiger, H.; Machann, J.; Schick, F.; Königsrainer, A.; Königsrainer, I.; Schleicher, E.; Fritsche, A.; Haring, H.U.; et al. The hepatokines fetuin-A and fetuin-B are upregulated in the state of hepatic steatosis and may differently impact on glucose homeostasis in humans. Am. J. Physiol. Endocrinol. Metab. 2018, 314, E266–E273. [Google Scholar] [CrossRef] [PubMed]
  24. Yoneda, M.; Imajo, K.; Takahashi, H.; Ogawa, Y.; Eguchi, Y.; Sumida, Y.; Yoneda, M.; Kawanaka, M.; Saito, S.; Tokushige, K.; et al. Clinical strategy of diagnosing and following patients with nonalcoholic fatty liver disease based on invasive and noninvasive methods. J. Gastroenterol. 2018, 53, 181–196. [Google Scholar] [CrossRef] [Green Version]
  25. Crespo, M.; Lappe, S.; Feldstein, A.E.; Alkhouri, N. Similarities and differences between pediatric and adult nonalcoholic fatty liver disease. Metabolism 2016, 65, 1161–1171. [Google Scholar] [CrossRef]
  26. Nobili, V.; Alisi, A.; Valenti, L.; Miele, L.; Feldstein, A.E.; Alkhouri, N. NAFLD in children: New genes, new diagnostic modalities and new drugs. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 517–530. [Google Scholar] [CrossRef]
  27. Mansoor, S.; Collyer, E.; Alkhouri, N. A comprehensive review of noninvasive liver fibrosis tests in pediatric nonalcoholic Fatty liver disease. Curr. Gastroenterol. Rep. 2015, 17, 23. [Google Scholar] [CrossRef]
  28. Noruegas, M.J.; Matos, H.; Gonçalves, I.; Cipriano, M.A.; Sanches, C. Acoustic radiation force impulse-imaging in the assessment of liver fibrosis in children. Pediatr. Radiol. 2012, 42, 201–204. [Google Scholar] [CrossRef]
  29. Kamble, R.; Sodhi, K.S.; Thapa, B.R.; Saxena, A.K.; Bhatia, A.; Dayal, D.; Khandelwal, N. Liver acoustic radiation force impulse (ARFI) in childhood obesity: Comparison and correlation with biochemical markers. J. Ultrasound 2017, 20, 33–42. [Google Scholar] [CrossRef] [Green Version]
  30. Ferraioli, G.; Calcaterra, V.; Lissandrin, R.; Guazzotti, M.; Maiocchi, L.; Tinelli, C.; De Silvestri, A.; Regalbuto, C.; Pelizzo, G.; Larizza, D.; et al. Noninvasive assessment of liver steatosis in children: The clinical value of controlled attenuation parameter. BMC Gastroenterol. 2017, 17, 61. [Google Scholar] [CrossRef]
  31. Tapper, E.B.; Loomba, R. Noninvasive imaging biomarker assessment of liver fibrosis by elastography in NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 274–282. [Google Scholar] [CrossRef]
  32. Chalasani, N.; Younossi, Z.; Lavine, J.E.; Charlton, M.; Cusi, K.; Rinella, M.; Harrison, S.A.; Brunt, E.M.; Sanyal, A.J. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018, 67, 328–357. [Google Scholar] [CrossRef] [PubMed]
  33. Yu, Q.; Jiang, Z.; Zhang, L. Bile acid regulation: A novel therapeutic strategy in non-alcoholic fatty liver disease. Pharmacol. Ther. 2018, 190, 81–90. [Google Scholar] [CrossRef] [PubMed]
  34. Kalhan, S.C.; Guo, L.; Edmison, J.; Dasarathy, S.; McCullough, A.J.; Hanson, R.W.; Milburn, M. Plasma metabolomic profile in nonalcoholic fatty liver disease. Metabolism 2011, 60, 404–413. [Google Scholar] [CrossRef] [Green Version]
  35. Burgert, T.S.; Taksali, S.E.; Dziura, J.; Goodman, T.R.; Yeckel, C.W.; Papademetris, X.; Constable, R.T.; Weiss, R.; Tamborlane, W.V.; Savoye, M.; et al. Alanine aminotransferase levels and fatty liver in childhood obesity: Associations with insulin resistance, adiponectin, and visceral fat. J. Clin. Endocrinol. Metab. 2006, 91, 4287–4294. [Google Scholar] [CrossRef] [PubMed]
  36. Cheng, S.; Wiklund, P.; Autio, R.; Borra, R.; Ojanen, X.; Xu, L.; Törmäkangas, T.; Alen, M. Adipose tissue dysfunction and altered systemic amino acid metabolism are associated with non-alcoholic fatty liver disease. PLoS ONE 2015, 10, e0138889. [Google Scholar] [CrossRef]
  37. Sabin, M.A.; De Hora, M.; Holly, J.M.; Hunt, L.P.; Ford, A.L.; Williams, S.R.; Baker, J.S.; Retallick, C.J.; Crowne, E.C.; Shield, J.P. Fasting nonesterified fatty acid profiles in childhood and their relationship with adiposity, insulin sensitivity, and lipid levels. Pediatrics 2007, 120, e1426–e1433. [Google Scholar] [CrossRef]
  38. Fletcher, J.A.; Deja, S.; Satapati, S.; Fu, X.; Burgess, S.C.; Browning, J.D. Impaired ketogenesis and increased acetyl-CoA oxidation promote hyperglycemia in human fatty liver. JCI Insight 2019, 5, e127737. [Google Scholar] [CrossRef]
  39. Goran, M.I.; Walker, R.; Le, K.A.; Mahurkar, S.; Vikman, S.; Davis, J.N.; Spruijt-Metz, D.; Weigensberg, M.J.; Allayee, H. Effects of PNPLA3 on liver fat and metabolic profile in Hispanic children and adolescents. Diabetes 2010, 59, 3127–3130. [Google Scholar] [CrossRef] [Green Version]
  40. Santoro, N.; Feldstein, A.E.; Enoksson, E.; Pierpont, B.; Kursawe, R.; Kim, G.; Caprio, S. The association between hepatic fat content and liver injury in obese children and adolescents: Effects of ethnicity, insulin resistance, and common gene variants. Diabetes Care 2013, 36, 1353–1360. [Google Scholar] [CrossRef] [Green Version]
  41. Santoro, N.; Caprio, S.; Pierpont, B.; Van Name, M.; Savoye, M.; Parks, E.J. Hepatic de novo lipogenesis in obese youth is modulated by a common variant in the GCKR gene. J. Clin. Endocrinol. Metab. 2015, 100, E1125–E1132. [Google Scholar] [CrossRef] [Green Version]
  42. Goffredo, M.; Caprio, S.; Feldstein, A.E.; D’Adamo, E.; Shaw, M.M.; Pierpont, B.; Savoye, M.; Zhao, H.; Bale, A.E.; Santoro, N. Role of TM6SF2 rs58542926 in the pathogenesis of nonalcoholic pediatric fatty liver disease: A multiethnic study. Hepatology 2016, 63, 117–125. [Google Scholar] [CrossRef] [Green Version]
  43. Selvakumar, P.K.; Kabbany, M.N.; Lopez, R.; Tozzi, G.; Alisi, A.; Alkhouri, N.; Nobili, V. Reduced lysosomal acid lipase activity-A potential role in the pathogenesis of non alcoholic fatty liver disease in pediatric patients. Dig. Liver Dis. 2016, 48, 909–913. [Google Scholar] [CrossRef]
  44. Tovoli, F.; Napoli, L.; Negrini, G.; D’Addato, S.; Tozzi, G.; D’Amico, J.; Piscaglia, F.; Bolondi, L. A relative deficiency of lysosomal acid lypase activity characterizes non-alcoholic fatty liver disease. Int. J. Mol. Sci. 2017, 18, 1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Baratta, F.; Pastori, D.; Tozzi, G.; D’Erasmo, L.; Di Costanzo, A.; Arca, M.; Ettorre, E.; Ginanni Corradini, S.; Violi, F.; Angelico, F.; et al. Lysosomal acid lipase activity and liver fibrosis in the clinical continuum of non-alcoholic fatty liver disease. Liver Int. 2019, 39, 2301–2308. [Google Scholar] [CrossRef] [PubMed]
  46. Baratta, F.; Pastori, D.; Ferro, D.; Carluccio, G.; Tozzi, G.; Angelico, F.; Violi, F.; Del Ben, M. Reduced lysosomal acid lipase activity: A new marker of liver disease severity across the clinical continuum of non-alcoholic fatty liver disease? World J. Gastroenterol. 2019, 25, 4172–4180. [Google Scholar] [CrossRef]
  47. Van Name, M.A.; Savoye, M.; Chick, J.M.; Galuppo, B.T.; Feldstein, A.E.; Pierpont, B.; Johnson, C.; Shabanova, V.; Ekong, U.; Valentino, P.L.; et al. A low ω-6 to ω-3 PUFA ratio (n-6:n-3 PUFA) diet to treat fatty liver disease in obese youth. J. Nutr. 2020, 150, 2314–2321. [Google Scholar] [CrossRef] [PubMed]
  48. Starling, A.P.; Sauder, K.A.; Kaar, J.L.; Shapiro, A.L.; Siega-Riz, A.M.; Dabelea, D. Maternal dietary patterns during pregnancy are associated with newborn body composition. J. Nutr. 2017, 147, 1334–1339. [Google Scholar] [CrossRef] [Green Version]
  49. Goran, M.I.; Dumke, K.; Bouret, S.G.; Kayser, B.; Walker, R.W.; Blumberg, B. The obesogenic effect of high fructose exposure during early development. Nat. Rev. Endocrinol. 2013, 9, 494–500. [Google Scholar] [CrossRef] [Green Version]
  50. Sloboda, D.M.; Li, M.; Patel, R.; Clayton, Z.E.; Yap, C.; Vickers, M.H. Early life exposure to fructose and offspring phenotype: Implications for long term metabolic homeostasis. J. Obes. 2014, 2014, 203474. [Google Scholar] [CrossRef]
  51. Lee, W.C.; Wu, K.L.H.; Leu, S.; Tain, Y.L. Translational insights on developmental origins of metabolic syndrome: Focus on fructose consumption. Biomed. J. 2018, 41, 96–101. [Google Scholar] [CrossRef]
  52. Hudgins, L.C.; Parker, T.S.; Levine, D.M.; Hellerstein, M.K. A dual sugar challenge test for lipogenic sensitivity to dietary fructose. J. Clin. Endocrinol. Metab. 2011, 96, 861–868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Beysen, C.; Ruddy, M.; Stoch, A.; Mixson, L.; Rosko, K.; Riiff, T.; Turner, S.M.; Hellerstein, M.K.; Murphy, E.J. Dose-dependent quantitative effects of acute fructose administration on hepatic de novo lipogenesis in healthy humans. Am. J. Physiol. Endocrinol. Metab. 2018, 315, E126–E132. [Google Scholar] [CrossRef] [PubMed]
  54. Softic, S.; Gupta, M.K.; Wang, G.X.; Fujisaka, S.; O’Neill, B.T.; Rao, T.N.; Willoughby, J.; Harbison, C.; Fitzgerald, K.; Ilkayeva, O.; et al. Divergent effects of glucose and fructose on hepatic lipogenesis and insulin signaling. J. Clin. Investig. 2017, 127, 4059–4074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Softic, S.; Meyer, J.G.; Wang, G.X.; Gupta, M.K.; Batista, T.M.; Lauritzen, H.P.M.M.; Fujisaka, S.; Serra, D.; Herrero, L.; Willoughby, J.; et al. Dietary sugars alter hepatic fatty acid oxidation via transcriptional and post-translational modifications of mitochondrial proteins. Cell Metab. 2019, 30, 735–753.e734. [Google Scholar] [CrossRef] [PubMed]
  56. Lustig, R.H.; Mulligan, K.; Noworolski, S.M.; Tai, V.W.; Wen, M.J.; Erkin-Cakmak, A.; Gugliucci, A.; Schwarz, J.M. Isocaloric fructose restriction and metabolic improvement in children with obesity and metabolic syndrome. Obesity 2016, 24, 453–460. [Google Scholar] [CrossRef] [PubMed]
  57. Schwarz, J.M.; Noworolski, S.M.; Erkin-Cakmak, A.; Korn, N.J.; Wen, M.J.; Tai, V.W.; Jones, G.M.; Palii, S.P.; Velasco-Alin, M.; Pan, K.; et al. Effects of dietary fructose restriction on liver fat, de novo lipogenesis, and insulin kinetics in children with obesity. Gastroenterology 2017, 153, 743–752. [Google Scholar] [CrossRef] [Green Version]
  58. Schwimmer, J.B.; Ugalde-Nicalo, P.; Welsh, J.A.; Angeles, J.E.; Cordero, M.; Harlow, K.E.; Alazraki, A.; Durelle, J.; Knight-Scott, J.; Newton, K.P.; et al. Effect of a low free sugar diet vs usual diet on nonalcoholic fatty liver disease in adolescent boys: A randomized clinical trial. JAMA 2019, 321, 256–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Goss, A.M.; Dowla, S.; Pendergrass, M.; Ashraf, A.; Bolding, M.; Morrison, S.; Amerson, A.; Soleymani, T.; Gower, B. Effects of a carbohydrate-restricted diet on hepatic lipid content in adolescents with non-alcoholic fatty liver disease: A pilot, randomized trial. Pediatr. Obes. 2020, 15, e12630. [Google Scholar] [CrossRef]
  60. Musso, G.; Gambino, R.; De Michieli, F.; Cassader, M.; Rizzetto, M.; Durazzo, M.; Fagà, E.; Silli, B.; Pagano, G. Dietary habits and their relations to insulin resistance and postprandial lipemia in nonalcoholic steatohepatitis. Hepatology 2003, 37, 909–916. [Google Scholar] [CrossRef]
  61. Araya, J.; Rodrigo, R.; Videla, L.A.; Thielemann, L.; Orellana, M.; Pettinelli, P.; Poniachik, J. Increase in long-chain polyunsaturated fatty acid n − 6/n − 3 ratio in relation to hepatic steatosis in patients with non-alcoholic fatty liver disease. Clin. Sci. 2004, 106, 635–643. [Google Scholar] [CrossRef] [Green Version]
  62. Jump, D.B.; Lytle, K.A.; Depner, C.M.; Tripathy, S. Omega-3 polyunsaturated fatty acids as a treatment strategy for nonalcoholic fatty liver disease. Pharmacol. Ther. 2018, 181, 108–125. [Google Scholar] [CrossRef]
  63. Jump, D.B. N-3 polyunsaturated fatty acid regulation of hepatic gene transcription. Curr. Opin. Lipidol. 2008, 19, 242–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Nobili, V.; Bedogni, G.; Alisi, A.; Pietrobattista, A.; Risé, P.; Galli, C.; Agostoni, C. Docosahexaenoic acid supplementation decreases liver fat content in children with non-alcoholic fatty liver disease: Double-blind randomised controlled clinical trial. Arch. Dis. Child 2011, 96, 350–353. [Google Scholar] [CrossRef]
  65. Janczyk, W.; Lebensztejn, D.; Wierzbicka-Rucińska, A.; Mazur, A.; Neuhoff-Murawska, J.; Matusik, P.; Socha, P. Omega-3 fatty acids therapy in children with nonalcoholic fatty liver disease: A randomized controlled trial. J. Pediatr. 2015, 166, 1358–1363.e1351–1358–e1353. [Google Scholar] [CrossRef] [PubMed]
  66. Boyraz, M.; Pirgon, Ö.; Dündar, B.; Çekmez, F.; Hatipoğlu, N. Long-term treatment with n-3 polyunsaturated fatty acids as a monotherapy in children with nonalcoholic fatty liver disease. J. Clin. Res. Pediatr. Endocrinol. 2015, 7, 121–127. [Google Scholar] [CrossRef]
  67. Lowell, B.B.; Shulman, G.I. Mitochondrial dysfunction and type 2 diabetes. Science 2005, 307, 384–387. [Google Scholar] [CrossRef] [Green Version]
  68. Mitchell, T.; Darley-Usmar, V. Metabolic syndrome and mitochondrial dysfunction: Insights from preclinical studies with a mitochondrially targeted antioxidant. Free Radic. Biol. Med. 2012, 52, 838–840. [Google Scholar] [CrossRef] [Green Version]
  69. Jha, S.K.; Jha, N.K.; Kumar, D.; Ambasta, R.K.; Kumar, P. Linking mitochondrial dysfunction, metabolic syndrome and stress signaling in neurodegeneration. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1132–1146. [Google Scholar] [CrossRef]
  70. Caldwell, S.H.; Swerdlow, R.H.; Khan, E.M.; Iezzoni, J.C.; Hespenheide, E.E.; Parks, J.K.; Parker, W.D., Jr. Mitochondrial abnormalities in non-alcoholic steatohepatitis. J. Hepatol. 1999, 31, 430–434. [Google Scholar] [CrossRef]
  71. Sanyal, A.J.; Campbell-Sargent, C.; Mirshahi, F.; Rizzo, W.B.; Contos, M.J.; Sterling, R.K.; Luketic, V.A.; Shiffman, M.L.; Clore, J.N. Nonalcoholic steatohepatitis: Association of insulin resistance and mitochondrial abnormalities. Gastroenterology 2001, 120, 1183–1192. [Google Scholar] [CrossRef]
  72. Lotowska, J.M.; Sobaniec-Lotowska, M.E.; Bockowska, S.B.; Lebensztejn, D.M. Pediatric non-alcoholic steatohepatitis: The first report on the ultrastructure of hepatocyte mitochondria. World J. Gastroenterol. 2014, 20, 4335–4340. [Google Scholar] [CrossRef]
  73. Koliaki, C.; Szendroedi, J.; Kaul, K.; Jelenik, T.; Nowotny, P.; Jankowiak, F.; Herder, C.; Carstensen, M.; Krausch, M.; Knoefel, W.T.; et al. Adaptation of hepatic mitochondrial function in humans with non-alcoholic fatty liver is lost in steatohepatitis. Cell Metab. 2015, 21, 739–746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Satapati, S.; Kucejova, B.; Duarte, J.A.; Fletcher, J.A.; Reynolds, L.; Sunny, N.E.; He, T.; Nair, L.A.; Livingston, K.A.; Fu, X.; et al. Mitochondrial metabolism mediates oxidative stress and inflammation in fatty liver. J. Clin. Investig. 2015, 125, 4447–4462. [Google Scholar] [CrossRef] [Green Version]
  75. Sunny, N.E.; Parks, E.J.; Browning, J.D.; Burgess, S.C. Excessive hepatic mitochondrial TCA cycle and gluconeogenesis in humans with nonalcoholic fatty liver disease. Cell Metab. 2011, 14, 804–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. An, P.; Wei, L.L.; Zhao, S.; Sverdlov, D.Y.; Vaid, K.A.; Miyamoto, M.; Kuramitsu, K.; Lai, M.; Popov, Y.V. Hepatocyte mitochondria-derived danger signals directly activate hepatic stellate cells and drive progression of liver fibrosis. Nat. Commun. 2020, 11, 2362. [Google Scholar] [CrossRef] [PubMed]
  77. Thyfault, J.P.; Rector, R.S.; Uptergrove, G.M.; Borengasser, S.J.; Morris, E.M.; Wei, Y.; Laye, M.J.; Burant, C.F.; Qi, N.R.; Ridenhour, S.E.; et al. Rats selectively bred for low aerobic capacity have reduced hepatic mitochondrial oxidative capacity and susceptibility to hepatic steatosis and injury. J. Physiol. 2009, 587, 1805–1816. [Google Scholar] [CrossRef] [PubMed]
  78. Rector, R.S.; Thyfault, J.P.; Uptergrove, G.M.; Morris, E.M.; Naples, S.P.; Borengasser, S.J.; Mikus, C.R.; Laye, M.J.; Laughlin, M.H.; Booth, F.W.; et al. Mitochondrial dysfunction precedes insulin resistance and hepatic steatosis and contributes to the natural history of non-alcoholic fatty liver disease in an obese rodent model. J. Hepatol. 2010, 52, 727–736. [Google Scholar] [CrossRef] [Green Version]
  79. Ferro, D.; Baratta, F.; Pastori, D.; Cocomello, N.; Colantoni, A.; Angelico, F.; Del Ben, M. New insights into the pathogenesis of non-alcoholic fatty liver disease: Gut-derived lipopolysaccharides and oxidative stress. Nutrients 2020, 12, 2762. [Google Scholar] [CrossRef]
  80. Nobili, V.; Parola, M.; Alisi, A.; Marra, F.; Piemonte, F.; Mombello, C.; Sutti, S.; Povero, D.; Maina, V.; Novo, E.; et al. Oxidative stress parameters in paediatric non-alcoholic fatty liver disease. Int. J. Mol. Med. 2010, 26, 471–476. [Google Scholar] [CrossRef] [Green Version]
  81. Nobili, V.; Alisi, A.; Mosca, A.; Crudele, A.; Zaffina, S.; Denaro, M.; Smeriglio, A.; Trombetta, D. The antioxidant effects of hydroxytyrosol and vitamin E on pediatric nonalcoholic fatty liver disease, in a clinical trial: A new treatment? Antioxid. Redox Signal. 2019, 31, 127–133. [Google Scholar] [CrossRef]
  82. Negri, R.; Trinchese, G.; Carbone, F.; Caprio, M.G.; Stanzione, G.; di Scala, C.; Micillo, T.; Perna, F.; Tarotto, L.; Gelzo, M.; et al. Randomised clinical trial: Calorie restriction regimen with tomato juice supplementation ameliorates oxidative stress and preserves a proper immune surveillance modulating mitochondrial bioenergetics of T-lymphocytes in obese children affected by non-alcoholic fatty liver disease (NAFLD). J. Clin. Med. 2020, 9, 141. [Google Scholar] [CrossRef] [Green Version]
  83. Spahis, S.; Alvarez, F.; Ahmed, N.; Dubois, J.; Jalbout, R.; Paganelli, M.; Grzywacz, K.; Delvin, E.; Peretti, N.; Levy, E. Non-alcoholic fatty liver disease severity and metabolic complications in obese children: Impact of omega-3 fatty acids. J. Nutr. Biochem. 2018, 58, 28–36. [Google Scholar] [CrossRef] [PubMed]
  84. Linhart, K.; Bartsch, H.; Seitz, H.K. The role of reactive oxygen species (ROS) and cytochrome P-450 2E1 in the generation of carcinogenic etheno-DNA adducts. Redox Biol. 2014, 3, 56–62. [Google Scholar] [CrossRef] [Green Version]
  85. Linhart, K.B.; Glassen, K.; Peccerella, T.; Waldherr, R.; Linhart, H.; Bartsch, H.; Seitz, H.K. The generation of carcinogenic etheno-DNA adducts in the liver of patients with nonalcoholic fatty liver disease. Hepatobiliary Surg. Nutr. 2015, 4, 117–123. [Google Scholar] [CrossRef]
  86. Teufel, U.; Peccerella, T.; Engelmann, G.; Bruckner, T.; Flechtenmacher, C.; Millonig, G.; Stickel, F.; Hoffmann, G.F.; Schirmacher, P.; Mueller, S.; et al. Detection of carcinogenic etheno-DNA adducts in children and adolescents with non-alcoholic steatohepatitis (NASH). Hepatobiliary Surg. Nutr. 2015, 4, 426–435. [Google Scholar] [CrossRef]
  87. Grabherr, F.; Grander, C.; Effenberger, M.; Adolph, T.E.; Tilg, H. Gut dysfunction and non-alcoholic fatty liver disease. Front. Endocrinol. 2019, 10, 611. [Google Scholar] [CrossRef] [Green Version]
  88. Boursier, J.; Diehl, A.M. Nonalcoholic fatty liver disease and the gut microbiome. Clin. Liver Dis. 2016, 20, 263–275. [Google Scholar] [CrossRef] [PubMed]
  89. Schwimmer, J.B.; Johnson, J.S.; Angeles, J.E.; Behling, C.; Belt, P.H.; Borecki, I.; Bross, C.; Durelle, J.; Goyal, N.P.; Hamilton, G.; et al. Microbiome signatures associated with steatohepatitis and moderate to severe fibrosis in children with nonalcoholic fatty liver disease. Gastroenterology 2019, 157, 1109–1122. [Google Scholar] [CrossRef] [Green Version]
  90. Fukunishi, S.; Nishio, H.; Fukuda, A.; Takeshita, A.; Hanafusa, T.; Higuchi, K.; Suzuki, K. Development of fibrosis in nonalcoholic steatosis through combination of a synthetic diet rich in disaccharide and low-dose lipopolysaccharides in the livers of Zucker (fa/fa) rats. J. Clin. Biochem. Nutr. 2009, 45, 322–328. [Google Scholar] [CrossRef]
  91. Imajo, K.; Fujita, K.; Yoneda, M.; Nozaki, Y.; Ogawa, Y.; Shinohara, Y.; Kato, S.; Mawatari, H.; Shibata, W.; Kitani, H.; et al. Hyperresponsivity to low-dose endotoxin during progression to nonalcoholic steatohepatitis is regulated by leptin-mediated signaling. Cell Metab. 2012, 16, 44–54. [Google Scholar] [CrossRef] [Green Version]
  92. Ilan, Y. Leaky gut and the liver: A role for bacterial translocation in nonalcoholic steatohepatitis. World J. Gastroenterol. 2012, 18, 2609–2618. [Google Scholar] [CrossRef] [PubMed]
  93. Kavanagh, K.; Wylie, A.T.; Tucker, K.L.; Hamp, T.J.; Gharaibeh, R.Z.; Fodor, A.A.; Cullen, J.M. Dietary fructose induces endotoxemia and hepatic injury in calorically controlled primates. Am. J. Clin. Nutr. 2013, 98, 349–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ahola, A.J.; Lassenius, M.I.; Forsblom, C.; Harjutsalo, V.; Lehto, M.; Groop, P.H. Dietary patterns reflecting healthy food choices are associated with lower serum LPS activity. Sci. Rep. 2017, 7, 6511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Pastori, D.; Carnevale, R.; Nocella, C.; Novo, M.; Santulli, M.; Cammisotto, V.; Menichelli, D.; Pignatelli, P.; Violi, F. Gut-derived serum lipopolysaccharide is associated with enhanced risk of major adverse cardiovascular events in atrial fibrillation: Effect of adherence to mediterranean diet. J. Am. Heart Assoc. 2017, 6. [Google Scholar] [CrossRef]
  96. Spruss, A.; Bergheim, I. Dietary fructose and intestinal barrier: Potential risk factor in the pathogenesis of nonalcoholic fatty liver disease. J. Nutr. Biochem. 2009, 20, 657–662. [Google Scholar] [CrossRef]
  97. Vos, M.B.; Lavine, J.E. Dietary fructose in nonalcoholic fatty liver disease. Hepatology 2013, 57, 2525–2531. [Google Scholar] [CrossRef]
  98. Erridge, C.; Attina, T.; Spickett, C.M.; Webb, D.J. A high-fat meal induces low-grade endotoxemia: Evidence of a novel mechanism of postprandial inflammation. Am. J. Clin. Nutr. 2007, 86, 1286–1292. [Google Scholar] [CrossRef]
  99. Deopurkar, R.; Ghanim, H.; Friedman, J.; Abuaysheh, S.; Sia, C.L.; Mohanty, P.; Viswanathan, P.; Chaudhuri, A.; Dandona, P. Differential effects of cream, glucose, and orange juice on inflammation, endotoxin, and the expression of Toll-like receptor-4 and suppressor of cytokine signaling-3. Diabetes Care 2010, 33, 991–997. [Google Scholar] [CrossRef] [Green Version]
  100. Amar, J.; Burcelin, R.; Ruidavets, J.B.; Cani, P.D.; Fauvel, J.; Alessi, M.C.; Chamontin, B.; Ferriéres, J. Energy intake is associated with endotoxemia in apparently healthy men. Am. J. Clin. Nutr. 2008, 87, 1219–1223. [Google Scholar] [CrossRef] [Green Version]
  101. Giorgio, V.; Miele, L.; Principessa, L.; Ferretti, F.; Villa, M.P.; Negro, V.; Grieco, A.; Alisi, A.; Nobili, V. Intestinal permeability is increased in children with non-alcoholic fatty liver disease, and correlates with liver disease severity. Dig. Liver Dis. 2014, 46, 556–560. [Google Scholar] [CrossRef]
  102. Nier, A.; Engstler, A.J.; Maier, I.B.; Bergheim, I. Markers of intestinal permeability are already altered in early stages of non-alcoholic fatty liver disease: Studies in children. PLoS ONE 2017, 12, e0183282. [Google Scholar] [CrossRef] [Green Version]
  103. Nier, A.; Brandt, A.; Conzelmann, I.B.; Özel, Y.; Bergheim, I. Non-alcoholic fatty liver disease in overweight children: Role of fructose intake and dietary pattern. Nutrients 2018, 10, 1329. [Google Scholar] [CrossRef] [Green Version]
  104. Bauer, T.M.; Schwacha, H.; Steinbrückner, B.; Brinkmann, F.E.; Ditzen, A.K.; Aponte, J.J.; Pelz, K.; Berger, D.; Kist, M.; Blum, H.E. Small intestinal bacterial overgrowth in human cirrhosis is associated with systemic endotoxemia. Am. J. Gastroenterol. 2002, 97, 2364–2370. [Google Scholar] [CrossRef]
  105. Bauer, T.M.; Steinbrückner, B.; Brinkmann, F.E.; Ditzen, A.K.; Schwacha, H.; Aponte, J.J.; Pelz, K.; Kist, M.; Blum, H.E. Small intestinal bacterial overgrowth in patients with cirrhosis: Prevalence and relation with spontaneous bacterial peritonitis. Am. J. Gastroenterol. 2001, 96, 2962–2967. [Google Scholar] [CrossRef] [PubMed]
  106. Harte, A.L.; da Silva, N.F.; Creely, S.J.; McGee, K.C.; Billyard, T.; Youssef-Elabd, E.M.; Tripathi, G.; Ashour, E.; Abdalla, M.S.; Sharada, H.M.; et al. Elevated endotoxin levels in non-alcoholic fatty liver disease. J. Inflamm. 2010, 7, 15. [Google Scholar] [CrossRef] [Green Version]
  107. Wong, V.W.; Wong, G.L.; Chan, H.Y.; Yeung, D.K.; Chan, R.S.; Chim, A.M.; Chan, C.K.; Tse, Y.K.; Woo, J.; Chu, W.C.; et al. Bacterial endotoxin and non-alcoholic fatty liver disease in the general population: A prospective cohort study. Aliment. Pharmacol. Ther. 2015, 42, 731–740. [Google Scholar] [CrossRef] [PubMed]
  108. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
  109. den Besten, G.; Bleeker, A.; Gerding, A.; van Eunen, K.; Havinga, R.; van Dijk, T.H.; Oosterveer, M.H.; Jonker, J.W.; Groen, A.K.; Reijngoud, D.J.; et al. Short-chain fatty acids protect against high-fat diet-induced obesity via a PPARγ-dependent switch from lipogenesis to fat oxidation. Diabetes 2015, 64, 2398–2408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Zhu, L.; Baker, S.S.; Gill, C.; Liu, W.; Alkhouri, R.; Baker, R.D.; Gill, S.R. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: A connection between endogenous alcohol and NASH. Hepatology 2013, 57, 601–609. [Google Scholar] [CrossRef]
  111. Albillos, A.; de Gottardi, A.; Rescigno, M. The gut-liver axis in liver disease: Pathophysiological basis for therapy. J. Hepatol. 2020, 72, 558–577. [Google Scholar] [CrossRef] [Green Version]
  112. Koch, M.A.; Reiner, G.L.; Lugo, K.A.; Kreuk, L.S.; Stanbery, A.G.; Ansaldo, E.; Seher, T.D.; Ludington, W.B.; Barton, G.M. Maternal IgG and IgA antibodies dampen mucosal T helper cell responses in early life. Cell 2016, 165, 827–841. [Google Scholar] [CrossRef] [Green Version]
  113. Zeng, M.Y.; Cisalpino, D.; Varadarajan, S.; Hellman, J.; Warren, H.S.; Cascalho, M.; Inohara, N.; Núñez, G. Gut microbiota-induced immunoglobulin G controls systemic infection by symbiotic bacteria and pathogens. Immunity 2016, 44, 647–658. [Google Scholar] [CrossRef] [Green Version]
  114. Arrieta, M.C.; Stiemsma, L.T.; Dimitriu, P.A.; Thorson, L.; Russell, S.; Yurist-Doutsch, S.; Kuzeljevic, B.; Gold, M.J.; Britton, H.M.; Lefebvre, D.L.; et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci. Transl. Med. 2015, 7, 307ra152. [Google Scholar] [CrossRef]
  115. Nogacka, A.M.; Salazar, N.; Arboleya, S.; Suárez, M.; Fernández, N.; Solís, G.; de Los Reyes-Gavilán, C.G.; Gueimonde, M. Early microbiota, antibiotics and health. Cell. Mol. Life Sci. 2018, 75, 83–91. [Google Scholar] [CrossRef]
  116. Mirpuri, J.; Raetz, M.; Sturge, C.R.; Wilhelm, C.L.; Benson, A.; Savani, R.C.; Hooper, L.V.; Yarovinsky, F. Proteobacteria-specific IgA regulates maturation of the intestinal microbiota. Gut Microbes 2014, 5, 28–39. [Google Scholar] [CrossRef] [Green Version]
  117. Al Nabhani, Z.; Eberl, G. Imprinting of the immune system by the microbiota early in life. Mucosal Immunol. 2020, 13, 183–189. [Google Scholar] [CrossRef] [Green Version]
  118. Vatanen, T.; Kostic, A.D.; d’Hennezel, E.; Siljander, H.; Franzosa, E.A.; Yassour, M.; Kolde, R.; Vlamakis, H.; Arthur, T.D.; Hämäläinen, A.M.; et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell 2016, 165, 842–853. [Google Scholar] [CrossRef] [Green Version]
  119. Houghteling, P.D.; Walker, W.A. Why is initial bacterial colonization of the intestine important to infants’ and children’s health? J. Pediatr. Gastroenterol. Nutr. 2015, 60, 294–307. [Google Scholar] [CrossRef] [Green Version]
  120. Lemas, D.J.; Young, B.E.; Baker, P.R., 2nd; Tomczik, A.C.; Soderborg, T.K.; Hernandez, T.L.; de la Houssaye, B.A.; Robertson, C.E.; Rudolph, M.C.; Ir, D.; et al. Alterations in human milk leptin and insulin are associated with early changes in the infant intestinal microbiome. Am. J. Clin. Nutr. 2016, 103, 1291–1300. [Google Scholar] [CrossRef]
  121. Soderborg, T.K.; Clark, S.E.; Mulligan, C.E.; Janssen, R.C.; Babcock, L.; Ir, D.; Young, B.E.; Krebs, N.F.; Lemas, D.J.; Johnson, L.K.; et al. The gut microbiota in infants of obese mothers increases inflammation and susceptibility to NAFLD. Nat. Commun. 2018, 9, 4462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Soderborg, T.K.; Friedman, J.E. Imbalance in gut microbes from babies born to obese mothers increases gut permeability and myeloid cell adaptations that provoke obesity and NAFLD. Microb. Cell 2018, 6, 102–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Jayakumar, S.; Loomba, R. Review article: Emerging role of the gut microbiome in the progression of nonalcoholic fatty liver disease and potential therapeutic implications. Aliment. Pharmacol. Ther. 2019, 50, 144–158. [Google Scholar] [CrossRef] [PubMed]
  124. Willeit, P.; Skroblin, P.; Kiechl, S.; Fernández-Hernando, C.; Mayr, M. Liver microRNAs: Potential mediators and biomarkers for metabolic and cardiovascular disease? Eur. Heart J. 2016, 37, 3260–3266. [Google Scholar] [CrossRef] [Green Version]
  125. Weiland, M.; Gao, X.H.; Zhou, L.; Mi, Q.S. Small RNAs have a large impact: Circulating microRNAs as biomarkers for human diseases. RNA Biol. 2012, 9, 850–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Pirola, C.J.; Fernández Gianotti, T.; Castaño, G.O.; Mallardi, P.; San Martino, J.; Mora Gonzalez Lopez Ledesma, M.; Flichman, D.; Mirshahi, F.; Sanyal, A.J.; Sookoian, S. Circulating microRNA signature in non-alcoholic fatty liver disease: From serum non-coding RNAs to liver histology and disease pathogenesis. Gut 2015, 64, 800–812. [Google Scholar] [CrossRef] [Green Version]
  127. Chai, C.; Rivkin, M.; Berkovits, L.; Simerzin, A.; Zorde-Khvalevsky, E.; Rosenberg, N.; Klein, S.; Yaish, D.; Durst, R.; Shpitzen, S.; et al. Metabolic circuit involving free fatty acids, microRNA 122, and triglyceride synthesis in liver and muscle tissues. Gastroenterology 2017, 153, 1404–1415. [Google Scholar] [CrossRef]
  128. Yu, J.; Peng, J.; Luan, Z.; Zheng, F.; Su, W. MicroRNAs as a novel tool in the diagnosis of liver lipid dysregulation and fatty liver disease. Molecules 2019, 24, 230. [Google Scholar] [CrossRef] [Green Version]
  129. Mori, M.A.; Ludwig, R.G.; Garcia-Martin, R.; Brandão, B.B.; Kahn, C.R. Extracellular miRNAs: From biomarkers to mediators of physiology and disease. Cell Metab. 2019, 30, 656–673. [Google Scholar] [CrossRef]
  130. Lin, H.Y.; Yang, Y.L.; Wang, P.W.; Wang, F.S.; Huang, Y.H. The emerging role of microRNAs in NAFLD: Highlight of microRNA-29a in modulating oxidative stress, inflammation, and beyond. Cells 2020, 9, 1041. [Google Scholar] [CrossRef]
  131. Chang, J.; Nicolas, E.; Marks, D.; Sander, C.; Lerro, A.; Buendia, M.A.; Xu, C.; Mason, W.S.; Moloshok, T.; Bort, R.; et al. miR-122, a mammalian liver-specific microRNA, is processed from hcr mRNA and may downregulate the high affinity cationic amino acid transporter CAT-1. RNA Biol. 2004, 1, 106–113. [Google Scholar] [CrossRef] [Green Version]
  132. Iguchi, T.; Niino, N.; Tamai, S.; Sakurai, K.; Mori, K. Comprehensive analysis of circulating microRNA specific to the liver, heart, and skeletal muscle of cynomolgus monkeys. Int. J. Toxicol. 2017, 36, 220–228. [Google Scholar] [CrossRef] [PubMed]
  133. Tan, Y.; Ge, G.; Pan, T.; Wen, D.; Gan, J. A pilot study of serum microRNAs panel as potential biomarkers for diagnosis of nonalcoholic fatty liver disease. PLoS ONE 2014, 9, e105192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Long, J.K.; Dai, W.; Zheng, Y.W.; Zhao, S.P. miR-122 promotes hepatic lipogenesis via inhibiting the LKB1/AMPK pathway by targeting Sirt1 in non-alcoholic fatty liver disease. Mol. Med. 2019, 25, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Povero, D.; Eguchi, A.; Li, H.; Johnson, C.D.; Papouchado, B.G.; Wree, A.; Messer, K.; Feldstein, A.E. Circulating extracellular vesicles with specific proteome and liver microRNAs are potential biomarkers for liver injury in experimental fatty liver disease. PLoS ONE 2014, 9, e113651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Brandt, S.; Roos, J.; Inzaghi, E.; Kotnik, P.; Kovac, J.; Battelino, T.; Cianfarani, S.; Nobili, V.; Colajacomo, M.; Kratzer, W.; et al. Circulating levels of miR-122 and nonalcoholic fatty liver disease in pre-pubertal obese children. Pediatr. Obes. 2018, 13, 175–182. [Google Scholar] [CrossRef]
  137. Cheung, O.; Puri, P.; Eicken, C.; Contos, M.J.; Mirshahi, F.; Maher, J.W.; Kellum, J.M.; Min, H.; Luketic, V.A.; Sanyal, A.J. Nonalcoholic steatohepatitis is associated with altered hepatic microRNA expression. Hepatology 2008, 48, 1810–1820. [Google Scholar] [CrossRef] [Green Version]
  138. Braza-Boïls, A.; Marí-Alexandre, J.; Molina, P.; Arnau, M.A.; Barceló-Molina, M.; Domingo, D.; Girbes, J.; Giner, J.; Martínez-Dolz, L.; Zorio, E. Deregulated hepatic microRNAs underlie the association between non-alcoholic fatty liver disease and coronary artery disease. Liver Int. 2016, 36, 1221–1229. [Google Scholar] [CrossRef]
  139. Miyaaki, H.; Ichikawa, T.; Kamo, Y.; Taura, N.; Honda, T.; Shibata, H.; Milazzo, M.; Fornari, F.; Gramantieri, L.; Bolondi, L.; et al. Significance of serum and hepatic microRNA-122 levels in patients with non-alcoholic fatty liver disease. Liver Int. 2014, 34, e302–e307. [Google Scholar] [CrossRef]
  140. Mukherjee, K.; Ghoshal, B.; Ghosh, S.; Chakrabarty, Y.; Shwetha, S.; Das, S.; Bhattacharyya, S.N. Reversible HuR-microRNA binding controls extracellular export of miR-122 and augments stress response. EMBO Rep. 2016, 17, 1184–1203. [Google Scholar] [CrossRef]
  141. Guo, H.; Xu, M.; Cao, Z.; Li, W.; Chen, L.; Xie, X.; Wang, W.; Liu, J. Ultrasound-assisted miR-122-loaded polymeric nanodroplets for hepatocellular carcinoma gene therapy. Mol. Pharm. 2020, 17, 541–553. [Google Scholar] [CrossRef]
  142. Ning, Q.; Liu, Y.F.; Ye, P.J.; Gao, P.; Li, Z.P.; Tang, S.Y.; He, D.X.; Tang, S.S.; Wei, H.; Yu, C.Y. Delivery of liver-specific miRNA-122 using a targeted macromolecular prodrug toward synergistic therapy for hepatocellular carcinoma. ACS Appl. Mater. Interfaces 2019, 11, 10578–10588. [Google Scholar] [CrossRef]
  143. Liu, X.L.; Pan, Q.; Cao, H.X.; Xin, F.Z.; Zhao, Z.H.; Yang, R.X.; Zeng, J.; Zhou, H.; Fan, J.G. Lipotoxic hepatocyte-derived exosomal microRNA 192-5p activates macrophages through rictor/Akt/forkhead box transcription factor O1 signaling in nonalcoholic fatty liver disease. Hepatology 2020, 72, 454–469. [Google Scholar] [CrossRef]
  144. Párrizas, M.; Brugnara, L.; Esteban, Y.; González-Franquesa, A.; Canivell, S.; Murillo, S.; Gordillo-Bastidas, E.; Cussó, R.; Cadefau, J.A.; García-Roves, P.M.; et al. Circulating miR-192 and miR-193b are markers of prediabetes and are modulated by an exercise intervention. J. Clin. Endocrinol. Metab. 2015, 100, E407–E415. [Google Scholar] [CrossRef] [Green Version]
  145. Liu, X.L.; Cao, H.X.; Wang, B.C.; Xin, F.Z.; Zhang, R.N.; Zhou, D.; Yang, R.X.; Zhao, Z.H.; Pan, Q.; Fan, J.G. miR-192-5p regulates lipid synthesis in non-alcoholic fatty liver disease through SCD-1. World J. Gastroenterol. 2017, 23, 8140–8151. [Google Scholar] [CrossRef]
  146. Wang, L.; Zhang, N.; Wang, Z.; Ai, D.M.; Cao, Z.Y.; Pan, H.P. Decreased miR-155 level in the peripheral blood of non-alcoholic fatty liver disease patients may serve as a biomarker and may influence LXR activity. Cell. Physiol. Biochem. 2016, 39, 2239–2248. [Google Scholar] [CrossRef] [Green Version]
  147. Li, X.; Lian, F.; Liu, C.; Hu, K.Q.; Wang, X.D. Isocaloric pair-fed high-carbohydrate diet induced more hepatic steatosis and inflammation than high-fat diet mediated by miR-34a/SIRT1 axis in mice. Sci. Rep. 2015, 5, 16774. [Google Scholar] [CrossRef] [PubMed]
  148. Latorre, J.; Ortega, F.J.; Liñares-Pose, L.; Moreno-Navarrete, J.M.; Lluch, A.; Comas, F.; Oliveras-Cañellas, N.; Ricart, W.; Höring, M.; Zhou, Y.; et al. Compounds that modulate AMPK activity and hepatic steatosis impact the biosynthesis of microRNAs required to maintain lipid homeostasis in hepatocytes. EBioMedicine 2020, 53, 102697. [Google Scholar] [CrossRef] [PubMed]
  149. Guo, Y.; Xiong, Y.; Sheng, Q.; Zhao, S.; Wattacheril, J.; Flynn, C.R. A micro-RNA expression signature for human NAFLD progression. J. Gastroenterol. 2016, 51, 1022–1030. [Google Scholar] [CrossRef] [Green Version]
  150. Cermelli, S.; Ruggieri, A.; Marrero, J.A.; Ioannou, G.N.; Beretta, L. Circulating microRNAs in patients with chronic hepatitis C and non-alcoholic fatty liver disease. PLoS ONE 2011, 6, e23937. [Google Scholar] [CrossRef] [Green Version]
  151. Zarrinpar, A.; Gupta, S.; Maurya, M.R.; Subramaniam, S.; Loomba, R. Serum microRNAs explain discordance of non-alcoholic fatty liver disease in monozygotic and dizygotic twins: A prospective study. Gut 2016, 65, 1546–1554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Wang, S.; Ai, H.; Liu, L.; Zhang, X.; Gao, F.; Zheng, L.; Yi, J.; Sun, L.; Yu, C.; Zhao, H.; et al. Micro-RNA-27a/b negatively regulates hepatic gluconeogenesis by targeting FOXO1. Am. J. Physiol. Endocrinol. Metab. 2019, 317, E911–E924. [Google Scholar] [CrossRef]
  153. Alisi, A.; Da Sacco, L.; Bruscalupi, G.; Piemonte, F.; Panera, N.; De Vito, R.; Leoni, S.; Bottazzo, G.F.; Masotti, A.; Nobili, V. Mirnome analysis reveals novel molecular determinants in the pathogenesis of diet-induced nonalcoholic fatty liver disease. Lab. Investig. 2011, 91, 283–293. [Google Scholar] [CrossRef]
  154. Can, U.; Buyukinan, M.; Yerlikaya, F.H. The investigation of circulating microRNAs associated with lipid metabolism in childhood obesity. Pediatr. Obes. 2016, 11, 228–234. [Google Scholar] [CrossRef] [PubMed]
  155. Fu, X.; Dong, B.; Tian, Y.; Lefebvre, P.; Meng, Z.; Wang, X.; Pattou, F.; Han, W.; Wang, X.; Lou, F.; et al. MicroRNA-26a regulates insulin sensitivity and metabolism of glucose and lipids. J. Clin. Investig. 2015, 125, 2497–2509. [Google Scholar] [CrossRef] [Green Version]
  156. Xu, H.; Tian, Y.; Tang, D.; Zou, S.; Liu, G.; Song, J.; Zhang, G.; Du, X.; Huang, W.; He, B.; et al. An endoplasmic reticulum stress-microRNA-26a feedback circuit in nonalcoholic fatty liver disease. Hepatology 2020. [Google Scholar] [CrossRef] [PubMed]
  157. Shah, K.B.; Chernausek, S.D.; Teague, A.M.; Bard, D.E.; Tryggestad, J.B. Maternal diabetes alters microRNA expression in fetal exosomes, human umbilical vein endothelial cells and placenta. Pediatr. Res. 2020. [Google Scholar] [CrossRef]
  158. Tryggestad, J.B.; Vishwanath, A.; Jiang, S.; Mallappa, A.; Teague, A.M.; Takahashi, Y.; Thompson, D.M.; Chernausek, S.D. Influence of gestational diabetes mellitus on human umbilical vein endothelial cell miRNA. Clin. Sci. 2016, 130, 1955–1967. [Google Scholar] [CrossRef] [Green Version]
  159. Jiang, S.; Teague, A.M.; Tryggestad, J.B.; Chernausek, S.D. Role of microRNA-130b in placental PGC-1alpha/TFAM mitochondrial biogenesis pathway. Biochem. Biophys. Res. Commun. 2017, 487, 607–612. [Google Scholar] [CrossRef] [PubMed]
  160. Liao, Y.; Du, X.; Li, J.; Lonnerdal, B. Human milk exosomes and their microRNAs survive digestion in vitro and are taken up by human intestinal cells. Mol. Nutr. Food Res. 2017, 61, 1700082. [Google Scholar] [CrossRef]
  161. Benmoussa, A.; Provost, P. Milk microRNAs in health and disease. Compr. Rev. Food Sci. F 2019, 18, 703–722. [Google Scholar] [CrossRef] [Green Version]
  162. Shankar, K.; Harrell, A.; Liu, X.; Gilchrist, J.M.; Ronis, M.J.; Badger, T.M. Maternal obesity at conception programs obesity in the offspring. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2008, 294, R528–R538. [Google Scholar] [CrossRef]
  163. Modi, N.; Murgasova, D.; Ruager-Martin, R.; Thomas, E.L.; Hyde, M.J.; Gale, C.; Santhakumaran, S.; Dore, C.J.; Alavi, A.; Bell, J.D. The influence of maternal body mass index on infant adiposity and hepatic lipid content. Pediatr. Res. 2011, 70, 287–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Brumbaugh, D.E.; Tearse, P.; Cree-Green, M.; Fenton, L.Z.; Brown, M.; Scherzinger, A.; Reynolds, R.; Alston, M.; Hoffman, C.; Pan, Z.; et al. Intrahepatic fat is increased in the neonatal offspring of obese women with gestational diabetes. J. Pediatr. 2013, 162, 930–936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Newton, K.P.; Feldman, H.S.; Chambers, C.D.; Wilson, L.; Behling, C.; Clark, J.M.; Molleston, J.P.; Chalasani, N.; Sanyal, A.J.; Fishbein, M.H.; et al. Low and high birth weights are risk factors for nonalcoholic fatty liver disease in children. J. Pediatr. 2017, 187, 141–146. [Google Scholar] [CrossRef] [PubMed]
  166. Gregorio, B.M.; Souza-Mello, V.; Carvalho, J.J.; Mandarim-de-Lacerda, C.A.; Aguila, M.B. Maternal high-fat intake predisposes nonalcoholic fatty liver disease in C57BL/6 offspring. Am. J. Obstet. Gynecol. 2010, 203, 495.e491–e498. [Google Scholar] [CrossRef] [PubMed]
  167. Oben, J.A.; Mouralidarane, A.; Samuelsson, A.M.; Matthews, P.J.; Morgan, M.L.; McKee, C.; Soeda, J.; Fernandez-Twinn, D.S.; Martin-Gronert, M.S.; Ozanne, S.E.; et al. Maternal obesity during pregnancy and lactation programs the development of offspring non-alcoholic fatty liver disease in mice. J. Hepatol. 2010, 52, 913–920. [Google Scholar] [CrossRef]
  168. Bruce, K.D.; Cagampang, F.R.; Argenton, M.; Zhang, J.; Ethirajan, P.L.; Burdge, G.C.; Bateman, A.C.; Clough, G.F.; Poston, L.; Hanson, M.A.; et al. Maternal high-fat feeding primes steatohepatitis in adult mice offspring, involving mitochondrial dysfunction and altered lipogenesis gene expression. Hepatology 2009, 50, 1796–1808. [Google Scholar] [CrossRef]
  169. Mouralidarane, A.; Soeda, J.; Visconti-Pugmire, C.; Samuelsson, A.M.; Pombo, J.; Maragkoudaki, X.; Butt, A.; Saraswati, R.; Novelli, M.; Fusai, G.; et al. Maternal obesity programs offspring nonalcoholic fatty liver disease by innate immune dysfunction in mice. Hepatology 2013, 58, 128–138. [Google Scholar] [CrossRef]
  170. McCurdy, C.E.; Bishop, J.M.; Williams, S.M.; Grayson, B.E.; Smith, M.S.; Friedman, J.E.; Grove, K.L. Maternal high-fat diet triggers lipotoxicity in the fetal livers of nonhuman primates. J. Clin. Investig. 2009, 119, 323–335. [Google Scholar] [CrossRef] [Green Version]
  171. Igosheva, N.; Abramov, A.Y.; Poston, L.; Eckert, J.J.; Fleming, T.P.; Duchen, M.R.; McConnell, J. Maternal diet-induced obesity alters mitochondrial activity and redox status in mouse oocytes and zygotes. PLoS ONE 2010, 5, e10074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Borengasser, S.J.; Lau, F.; Kang, P.; Blackburn, M.L.; Ronis, M.J.; Badger, T.M.; Shankar, K. Maternal obesity during gestation impairs fatty acid oxidation and mitochondrial SIRT3 expression in rat offspring at weaning. PLoS ONE 2011, 6, e24068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Burgueño, A.L.; Cabrerizo, R.; Gonzales Mansilla, N.; Sookoian, S.; Pirola, C.J. Maternal high-fat intake during pregnancy programs metabolic-syndrome-related phenotypes through liver mitochondrial DNA copy number and transcriptional activity of liver PPARGC1A. J. Nutr. Biochem. 2013, 24, 6–13. [Google Scholar] [CrossRef] [PubMed]
  174. Wu, L.L.; Russell, D.L.; Wong, S.L.; Chen, M.; Tsai, T.S.; St John, J.C.; Norman, R.J.; Febbraio, M.A.; Carroll, J.; Robker, R.L. Mitochondrial dysfunction in oocytes of obese mothers: Transmission to offspring and reversal by pharmacological endoplasmic reticulum stress inhibitors. Development 2015, 142, 681–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Thorn, S.R.; Baquero, K.C.; Newsom, S.A.; El Kasmi, K.C.; Bergman, B.C.; Shulman, G.I.; Grove, K.L.; Friedman, J.E. Early life exposure to maternal insulin resistance has persistent effects on hepatic NAFLD in juvenile nonhuman primates. Diabetes 2014, 63, 2702–2713. [Google Scholar] [CrossRef] [Green Version]
  176. Alfaradhi, M.Z.; Fernandez-Twinn, D.S.; Martin-Gronert, M.S.; Musial, B.; Fowden, A.; Ozanne, S.E. Oxidative stress and altered lipid homeostasis in the programming of offspring fatty liver by maternal obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2014, 307, R26–R34. [Google Scholar] [CrossRef]
  177. Ashino, N.G.; Saito, K.N.; Souza, F.D.; Nakutz, F.S.; Roman, E.A.; Velloso, L.A.; Torsoni, A.S.; Torsoni, M.A. Maternal high-fat feeding through pregnancy and lactation predisposes mouse offspring to molecular insulin resistance and fatty liver. J. Nutr. Biochem. 2012, 23, 341–348. [Google Scholar] [CrossRef]
  178. Sureshchandra, S.; Wilson, R.M.; Rais, M.; Marshall, N.E.; Purnell, J.Q.; Thornburg, K.L.; Messaoudi, I. Maternal pregravid obesity remodels the DNA methylation landscape of cord blood monocytes disrupting their inflammatory program. J. Immunol. 2017, 199, 2729–2744. [Google Scholar] [CrossRef] [Green Version]
  179. Wankhade, U.D.; Zhong, Y.; Kang, P.; Alfaro, M.; Chintapalli, S.V.; Thakali, K.M.; Shankar, K. Enhanced offspring predisposition to steatohepatitis with maternal high-fat diet is associated with epigenetic and microbiome alterations. PLoS ONE 2017, 12, e0175675. [Google Scholar] [CrossRef] [Green Version]
  180. De Jesus, D.F.; Orime, K.; Kaminska, D.; Kimura, T.; Basile, G.; Wang, C.H.; Haertle, L.; Riemens, R.; Brown, N.K.; Hu, J.; et al. Parental metabolic syndrome epigenetically reprograms offspring hepatic lipid metabolism in mice. J. Clin. Investig. 2020, 130, 2391–2407. [Google Scholar] [CrossRef] [Green Version]
  181. Suter, M.A.; Chen, A.; Burdine, M.S.; Choudhury, M.; Harris, R.A.; Lane, R.H.; Friedman, J.E.; Grove, K.L.; Tackett, A.J.; Aagaard, K.M. A maternal high-fat diet modulates fetal SIRT1 histone and protein deacetylase activity in nonhuman primates. FASEB J. 2012, 26, 5106–5114. [Google Scholar] [CrossRef] [Green Version]
  182. de Oliveira, F.L.; Panera, N.; De Stefanis, C.; Mosca, A.; D’Oria, V.; Crudele, A.; De Vito, R.; Nobili, V.; Alisi, A. The number of liver galectin-3 positive cells is dually correlated with NAFLD severity in children. Int. J. Mol. Sci. 2019, 20, 3460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Carpino, G.; Nobili, V.; Renzi, A.; De Stefanis, C.; Stronati, L.; Franchitto, A.; Alisi, A.; Onori, P.; De Vito, R.; Alpini, G.; et al. Macrophage activation in pediatric nonalcoholic fatty liver disease (NAFLD) correlates with hepatic progenitor cell response via Wnt3a pathway. PLoS ONE 2016, 11, e0157246. [Google Scholar] [CrossRef]
  184. Lotowska, J.M.; Sobaniec-Lotowska, M.E.; Lebensztejn, D.M. The role of Kupffer cells in the morphogenesis of nonalcoholic steatohepatitis - ultrastructural findings. The first report in pediatric patients. Scand. J. Gastroenterol. 2013, 48, 352–357. [Google Scholar] [CrossRef] [PubMed]
  185. Tacke, F.; Zimmermann, H.W. Macrophage heterogeneity in liver injury and fibrosis. J. Hepatol. 2014, 60, 1090–1096. [Google Scholar] [CrossRef] [Green Version]
  186. Pradere, J.P.; Kluwe, J.; De Minicis, S.; Jiao, J.J.; Gwak, G.Y.; Dapito, D.H.; Jang, M.K.; Guenther, N.D.; Mederacke, I.; Friedman, R.; et al. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology 2013, 58, 1461–1473. [Google Scholar] [CrossRef] [Green Version]
  187. Stienstra, R.; Saudale, F.; Duval, C.; Keshtkar, S.; Groener, J.E.; van Rooijen, N.; Staels, B.; Kersten, S.; Müller, M. Kupffer cells promote hepatic steatosis via interleukin-1beta-dependent suppression of peroxisome proliferator-activated receptor alpha activity. Hepatology 2010, 51, 511–522. [Google Scholar] [CrossRef] [PubMed]
  188. Asanuma, T.; Ono, M.; Kubota, K.; Hirose, A.; Hayashi, Y.; Saibara, T.; Inanami, O.; Ogawa, Y.; Enzan, H.; Onishi, S.; et al. Super paramagnetic iron oxide MRI shows defective Kupffer cell uptake function in non-alcoholic fatty liver disease. Gut 2010, 59, 258–266. [Google Scholar] [CrossRef] [Green Version]
  189. Gadd, V.L.; Skoien, R.; Powell, E.E.; Fagan, K.J.; Winterford, C.; Horsfall, L.; Irvine, K.; Clouston, A.D. The portal inflammatory infiltrate and ductular reaction in human nonalcoholic fatty liver disease. Hepatology 2014, 59, 1393–1405. [Google Scholar] [CrossRef]
  190. Krenkel, O.; Tacke, F. Liver macrophages in tissue homeostasis and disease. Nat. Rev. Immunol. 2017, 17, 306–321. [Google Scholar] [CrossRef]
  191. Kazankov, K.; Jørgensen, S.M.D.; Thomsen, K.L.; Møller, H.J.; Vilstrup, H.; George, J.; Schuppan, D.; Grønbaek, H. The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 145–159. [Google Scholar] [CrossRef]
  192. Deng, Z.B.; Liu, Y.; Liu, C.; Xiang, X.; Wang, J.; Cheng, Z.; Shah, S.V.; Zhang, S.; Zhang, L.; Zhuang, X.; et al. Immature myeloid cells induced by a high-fat diet contribute to liver inflammation. Hepatology 2009, 50, 1412–1420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Obstfeld, A.E.; Sugaru, E.; Thearle, M.; Francisco, A.M.; Gayet, C.; Ginsberg, H.N.; Ables, E.V.; Ferrante, A.W., Jr. C-C chemokine receptor 2 (CCR2) regulates the hepatic recruitment of myeloid cells that promote obesity-induced hepatic steatosis. Diabetes 2010, 59, 916–925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Parker, R.; Weston, C.J.; Miao, Z.; Corbett, C.; Armstrong, M.J.; Ertl, L.; Ebsworth, K.; Walters, M.J.; Baumart, T.; Newland, D.; et al. CC chemokine receptor 2 promotes recruitment of myeloid cells associated with insulin resistance in nonalcoholic fatty liver disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 314, G483–G493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Biswas, S.K.; Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef]
  196. Friedman, J.E.; Dobrinskikh, E.; Alfonso-Garcia, A.; Fast, A.; Janssen, R.C.; Soderborg, T.K.; Anderson, A.L.; Reisz, J.A.; D’Alessandro, A.; Frank, D.N.; et al. Pyrroloquinoline quinone prevents developmental programming of microbial dysbiosis and macrophage polarization to attenuate liver fibrosis in offspring of obese mice. Hepatol. Commun. 2018, 2, 313–328. [Google Scholar] [CrossRef] [Green Version]
  197. Kamimae-Lanning, A.N.; Krasnow, S.M.; Goloviznina, N.A.; Zhu, X.; Roth-Carter, Q.R.; Levasseur, P.R.; Jeng, S.; McWeeney, S.K.; Kurre, P.; Marks, D.L. Maternal high-fat diet and obesity compromise fetal hematopoiesis. Mol. Metab. 2015, 4, 25–38. [Google Scholar] [CrossRef]
  198. Carter-Kent, C.; Brunt, E.M.; Yerian, L.M.; Alkhouri, N.; Angulo, P.; Kohli, R.; Ling, S.C.; Xanthakos, S.A.; Whitington, P.F.; Charatcharoenwitthaya, P.; et al. Relations of steatosis type, grade, and zonality to histological features in pediatric nonalcoholic fatty liver disease. J. Pediatr. Gastroenterol. Nutr. 2011, 52, 190–197. [Google Scholar] [CrossRef]
  199. Alkhouri, N.; Sedki, E.; Alisi, A.; Lopez, R.; Pinzani, M.; Feldstein, A.E.; Nobili, V. Combined paediatric NAFLD fibrosis index and transient elastography to predict clinically significant fibrosis in children with fatty liver disease. Liver Int. 2013, 33, 79–85. [Google Scholar] [CrossRef]
  200. Nash, M.J.; Frank, D.N.; Friedman, J.E. Early microbes modify immune system development and metabolic homeostasis- the “Restaurant” hypothesis revisited. Front. Endocrinol. 2017, 8, 349. [Google Scholar] [CrossRef]
  201. Negi, S.; Das, D.K.; Pahari, S.; Nadeem, S.; Agrewala, J.N. Potential role of gut microbiota in induction and regulation of innate immune memory. Front. Immunol. 2019, 10, 2441. [Google Scholar] [CrossRef] [Green Version]
  202. Campisano, S.; La Colla, A.; Echarte, S.M.; Chisari, A.N. Interplay between early-life malnutrition, epigenetic modulation of the immune function and liver diseases. Nutr. Res. Rev. 2019, 32, 128–145. [Google Scholar] [CrossRef] [PubMed]
Figure 1. In normal liver, miR-122, -192, and -155 act as suppressors of lipid biosynthesis, while miR-27a/b suppresses gluconeogenesis. In the context of excess fatty acids and carbohydrates, as occurs during the development of non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH), each of those miRNAs are found in reduced abundance within the liver, presumably due to excess export. The loss of the inhibitory actions allows for a metabolic shift in favor of lipid and glucose production and away from lipid oxidation. The increase in miR-34a, acting through the sirtuin 1 (SIRT1)/AMP-activated protein kinase (AMPK) axis, also contributes to a reduction in lipid oxidation. miR-155 and -192 play additional roles in activating M1 macrophages, contributing to the inflammatory and insulin-resistant milieu in adipose tissue and the liver. Green arrows reflect stimulatory actions and blunted red arrow represent inhibitory actions. β-OX, beta oxidation; FFA, free fatty acids; High-carb, high carbohydrate; Inflam, inflammation; IR, insulin resistance; miR, microRNA; miRNA, microRNA; TCA, tricarboxylic acid; TG, triglycerides; VLDL, very-low density lipoproteins.
Figure 1. In normal liver, miR-122, -192, and -155 act as suppressors of lipid biosynthesis, while miR-27a/b suppresses gluconeogenesis. In the context of excess fatty acids and carbohydrates, as occurs during the development of non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH), each of those miRNAs are found in reduced abundance within the liver, presumably due to excess export. The loss of the inhibitory actions allows for a metabolic shift in favor of lipid and glucose production and away from lipid oxidation. The increase in miR-34a, acting through the sirtuin 1 (SIRT1)/AMP-activated protein kinase (AMPK) axis, also contributes to a reduction in lipid oxidation. miR-155 and -192 play additional roles in activating M1 macrophages, contributing to the inflammatory and insulin-resistant milieu in adipose tissue and the liver. Green arrows reflect stimulatory actions and blunted red arrow represent inhibitory actions. β-OX, beta oxidation; FFA, free fatty acids; High-carb, high carbohydrate; Inflam, inflammation; IR, insulin resistance; miR, microRNA; miRNA, microRNA; TCA, tricarboxylic acid; TG, triglycerides; VLDL, very-low density lipoproteins.
Nutrients 12 03166 g001
Figure 2. Maternal malnutrition imparts long-lasting detrimental effects on offspring liver. Placental transfer of excess circulating free fatty acids (FFA) and carbohydrates from obese mother induces mitochondrial dysfunction and endoplasmic reticulum (ER) stress. The resulting mitochondrial dysfunction predisposes the offspring liver to reduced lipid peroxidation by decreasing SIRT3 and PGC1α expression, oxidative stress, inflammation, insulin resistance (IR), and increased gluconeogenesis leading to the development of NAFLD. In utero exposure to an obesogenic environment also induces early microbial dysbiosis, leading to the reprogramming of offspring liver towards an inflammatory phenotype by epigenetic modifications. β-OX, beta oxidation; ETC, electron transport chain; ROS, reactive oxygen species; TCA, tricarboxylic acid.
Figure 2. Maternal malnutrition imparts long-lasting detrimental effects on offspring liver. Placental transfer of excess circulating free fatty acids (FFA) and carbohydrates from obese mother induces mitochondrial dysfunction and endoplasmic reticulum (ER) stress. The resulting mitochondrial dysfunction predisposes the offspring liver to reduced lipid peroxidation by decreasing SIRT3 and PGC1α expression, oxidative stress, inflammation, insulin resistance (IR), and increased gluconeogenesis leading to the development of NAFLD. In utero exposure to an obesogenic environment also induces early microbial dysbiosis, leading to the reprogramming of offspring liver towards an inflammatory phenotype by epigenetic modifications. β-OX, beta oxidation; ETC, electron transport chain; ROS, reactive oxygen species; TCA, tricarboxylic acid.
Nutrients 12 03166 g002
Figure 3. Bone-marrow-derived and liver resident macrophages in offspring from obese mothers have trained memories towards inflammatory phenotypes and secrete inflammatory and fibrogenic factors to induce hepatic stellate cell activation. Activated stellate cells undergo rapid proliferation to further worsen inflammation, ultimately leading to fibrosis, organ failure, and death. HCC, hepatocellular carcinoma.
Figure 3. Bone-marrow-derived and liver resident macrophages in offspring from obese mothers have trained memories towards inflammatory phenotypes and secrete inflammatory and fibrogenic factors to induce hepatic stellate cell activation. Activated stellate cells undergo rapid proliferation to further worsen inflammation, ultimately leading to fibrosis, organ failure, and death. HCC, hepatocellular carcinoma.
Nutrients 12 03166 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/nu12103166

AMA Style

Mandala A, Janssen RC, Palle S, Short KR, Friedman JE. Pediatric Non-Alcoholic Fatty Liver Disease: Nutritional Origins and Potential Molecular Mechanisms. Nutrients. 2020; 12(10):3166. https://doi.org/10.3390/nu12103166

Chicago/Turabian Style

Mandala, Ashok, Rachel C. Janssen, Sirish Palle, Kevin R. Short, and Jacob E. Friedman. 2020. "Pediatric Non-Alcoholic Fatty Liver Disease: Nutritional Origins and Potential Molecular Mechanisms" Nutrients 12, no. 10: 3166. https://doi.org/10.3390/nu12103166

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