IMR Press / RCM / Volume 22 / Issue 3 / DOI: 10.31083/j.rcm2203082
Open Access Review
Pathophysiological mechanisms and clinical evidence of relationship between Nonalcoholic fatty liver disease (NAFLD) and cardiovascular disease
Show Less
1 Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
2 Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
3 Division of Cardiology, A.O.R.N. “Sant’Anna & San Sebastiano”, I-81100 Caserta, Italy
4 Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80138 Naples, Italy
*Correspondence: ferdinandocarlo.sasso@unicampania.it (Ferdinando Carlo Sasso)
These authors contributed equally.
Academic Editor: Peter A. McCullough
Rev. Cardiovasc. Med. 2021, 22(3), 755–768; https://doi.org/10.31083/j.rcm2203082
Submitted: 6 August 2021 | Revised: 6 September 2021 | Accepted: 9 September 2021 | Published: 24 September 2021
(This article belongs to the Special Issue Featured Papers in Cardiovascular Medicine 2021)
Copyright: © 2021 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license (https://creativecommons.org/licenses/by/4.0/).
Abstract

Evidence suggests a close connection between Nonalcoholic Fatty Liver Disease (NAFLD) and increased cardiovascular (CV) risk. Several cross-sectional studies report that NAFLD is related to preclinical atherosclerotic damage, and to coronary, cerebral and peripheral vascular events. Similar results have been showed by prospective studies and also by meta-analyzes on observational studies. The pathophysiological mechanisms of NAFLD are related to insulin resistance, which causes a dysfunction in adipokine production, especially adiponectin, from adipose tissue. A proinflammatory state and an increase in oxidative stress, due to increased reacting oxygen species (ROS) formation with consequent oxidation of free fatty acids and increased de novo lipogenesis with accumulation of triglycerides, are observed. These mechanisms may have an impact on atherosclerotic plaque formation and progression, and they can lead to increased cardiovascular risk in subjects with NAFLD. This review extensively discusses and comments current and developing NAFLD therapies and their possible impact on cardiovascular outcome.

Keywords
NAFLD
Cardiovascular disease
Pathophysiology
Type 2 diabetes
Cardiovascular risk
1. Introduction

Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver dysfunction. It affects about 25% of the population and is associated with several metabolic risk factors such as hypertension, diabetes, obesity and dyslipidemia [1, 2]. Evidence suggests a close link between Nonalcoholic Fatty Liver Disease (NAFLD) and increased cardiovascular (CV) risk. Several cross-sectional studies report hepatic steatosis as an independent risk factor for both an increased carotid intima-media thickness and diastolic cardiac dysfunction [3, 4]. Moreover, a clinical investigation conducted on a cohort of more than 2000 subjects with type 2 diabetes showed that a condition of hepatic steatosis, even after correction for the main cardio-metabolic risk factors, is associated with a higher prevalence of coronary, cerebral and peripheral vascular disease [5].

Prospective studies confirm the association between NAFLD and CV risk. A large longitudinal study with a two-year follow-up demonstrated that the presence of NAFLD at baseline assessment is an independent risk factor for the carotid disease [6]. Similarly, in the Valpolicella study conducted on diabetic subjects, after correction for cardiometabolic risk factors, NAFLD appeared as an independent risk factor for the incidence of cardio-cerebrovascular events [7].

Furthermore, a recent meta-analysis showed that subjects with NAFLD, compared to the control group without steatosis, had a 1.6 times higher risk of developing CV events, and this risk is higher for non-fatal events [8]. The same meta-analysis demonstrates that the presence of “severe NAFLD” is associated with an approximately 2.5 times higher risk of developing fatal and non-fatal CV events.

In a cohort of patients with a histological diagnosis of NAFLD, carotid intima-media thickness increased significantly and progressively along with the stage of hepatic fibrosis [9]. Furthermore, the severity of hepatic fibrosis assessed through liver biopsy has been independently associated with the worsening of both systolic and diastolic cardiac dysfunction [10].

A large retrospective study conducted on subjects with no history of CV disease undergoing to serial Computed Tomography (CT) to evaluate the Coronary Artery Calcium (CAC) score, showed that the risk of progression of CAC increased progressively from subjects without steatosis to those with steatosis and advanced fibrosis [11].

The aim of this review is to provide a comprehensive overview on the pathophysiological mechanisms underlying the development of NAFLD and its link between with CV risk. Finally, the therapeutic strategies for NAFLD treatment and their impact on CV outcome will be discussed.

2. NAFLD diagnosis

NAFLD is characterized by a large spectrum of disorders, ranging from simple steatosis to non-alcoholic steatohepatitis, which both in turn may progress into overt cirrhosis and hepatocellular carcinoma [12, 13, 14]. NAFLD diagnosis can be performed only in absence of other causes of chronic liver disease such as alcohol consumption (<30 g/day for men and <20 g/day for women), congenital or autoimmune diseases, viruses, and iatrogenic injury [15]. Liver biopsy is the gold standard for NAFLD diagnosis, by detecting a liver fat accumulation in >5% of hepatocytes and other hallmarks and may also distinguish from other causes of liver injury (e.g., non-alcoholic fatty liver and steatohepatitis) [16]. The distinction between NAFLD and non-alcoholic steatohepatitis (NASH) is possible only by histological analysis. It detects the signs of inflammation and the degree of fibrosis. The histological findings most commonly associated with this condition are the hepatocyte ballooning, the perisinusoidal fibrosis and the Mallory bodies [17].

Recently, new non-invasive diagnostic tools, such as ultrasonography, CT, and magnetic resonance imaging (MRI), have emerged, showing a greater safety profile and availability, and sharing a good sensitivity and specificity compared to gold standard [15, 18]. Ultrasonography has been recommended as the first-line diagnostic tool to detect NAFLD, though its sensitivity decreases when less than 30% of hepatocytes are involved as well as in obese patients [15, 19]. Controlled attenuation parameter (CAP), available on the equipment “Fibroscan ®”, represents a new and accurate technique to detect and quantify hepatic steatosis. Moreover, transient elastography, by Fibroscan is also able to detect liver stiffness, which is a reliable measure of liver fibrosis [20, 21, 22]. This equipment has two M and XL probes capable of supplying ultrasounds and an elastic wave to measure the stiffness of the liver tissue and steatosis. XL probe allows to study obese patients who, due to their thick panniculus adipose, can hinder the propagation of the elastic wave [23, 24]. Several clinical and imaging scores have been validated to assess liver steatosis. The fatty liver index is calculated through serum triglycerides levels, waist circumference, and γ-glutamyl-transpeptidase [25]. This score showed good performances in ruling in (60) or ruling out (<30) patients with steatosis, though it fails to stratify patients with a score ranging from 30 to 59, which remains undetermined. The ultrasound fatty liver index is based, instead, on ultrasonography parameters, suggesting hepatic steatosis when a score >2 is present [26]. The NAFLD Fat score evaluates several parameters (metabolic syndrome, type 2 diabetes mellitus (T2DM), fasting serum levels of insulin, aspartate aminotransferase (AST)/alanine aminotransferase (ALT) ratio to predict NAFLD and liver fat content [27]. In patients with known NAFLD, estimation of hepatic fibrosis degree can be accurately performed by using NAFLD fibrosis score (NFS), calculated through age, altered glucose metabolism, body mass index (BMI), AST/ALT ratio, albumin levels and platelet count [28]. The NFS is suggestive of hepatic fibrosis (equivalent to F3-F4 on liver biopsy) when >0.675. Two other scores, the Fibrosis-4 score and the AST to Platelet ratio index, have been developed to screen cirrhosis, though both validated in a cohort of subjects affected by hepatitis C virus [29, 30].

3. NAFLD main pathophysiological mechanism

The primary NAFLD hallmark is fat accumulation (mostly triglycerides) in the liver. However, how this accumulation leads to a liver dysfunction is compound and incompletely understood [31, 32, 33, 34]. Of note, though triglycerides represent the major part of intrahepatic lipid in NAFLD patients, new evidence suggests that changes in liver lipid composition (including ceramides, diacylglycerol, triglyceride/diacylglycerol ratio, phospholipids and free cholesterol) may play a significant role [35, 36, 37]. Particularly, alterations of free cholesterol metabolism seem involved in the development and progression of NAFLD and consequently associated with a higher CV risk. It has been observed that alteration of microbiota could alter the absorption and elimination of free cholesterol, thus determining an increase of its serum levels. Moreover, some authors found that mitochondrial abnormalities and altered activation of acyl coenzyme A cholesterol acyl transferase, SREBP-2 maturation, altered hydroxymethylglutaryl coenzyme A reductase (HMG-CoA-R) expression and decreased phosphorylation of HMG-CoA-R, induced by genic factors, could affect synthesis and transport of free cholesterol [31, 37, 38].

Adipose tissue is also involved in hormone secretion (e.g., adipokines) [39]. Obesity represents an important cause of adipose tissue dysfunction, which plays a relevant role in the metabolic disorders’ progression, particularly in insulin resistance and NAFLD [40, 41]. As a result of adipose tissue dysfunction, there is a reduction of adiponectin, an insulin-sensitizing adipokine. Adiponectin plays a crucial role in increasing free fatty acids (FFA) oxidation and decreasing FFA influx to the liver, gluconeogenesis, and de novo lipogenesis, thus enhancing a protective hepatic role [42, 43, 44]. In addition, it also plays a hepatic anti-inflammatory and antifibrotic role, by reducing proinflammatory cytokines and the activation and proliferation of hepatic stellate cells [45, 46, 47, 48, 49]. Impaired insulin action, through stimulation of lipogenic enzymes via sterol receptor-binding protein 1c (SREBP-1c), results in an increase in circulating FFA, an increased storage into the liver and increased de novo lipogenesis [50, 51, 52, 53]. Furthermore, it seems that insulin resistance is also involved in the triglyceride synthesis increase through the Kennedy pathway and in fatty acids β-oxidation decrease, though this latter is still controversial [54]. Skeletal muscle insulin resistance may also be implicated in steatosis development, by redistributing postprandial glucose from peripheral glycogen storage to hepatic de novo lipogenesis [55, 56]. Lipogenesis and triglycerides accumulation may also be affected by lipogenic action of dietary fructose. In addition, fructose causes liver damage due to its metabolism. It is highly energy dependent and activates a cascade of impaired protein synthesis, mitochondrial dysfunction and oxidative stress [57, 58, 59]. Moreover, the increased lipid burden is also responsible for increased mitochondria activity (β-oxidation of free fatty acid, adenosine triphosphate (ATP) production and reactive oxygen species (ROS) generation) and mass [60]. Over time, mitochondria may become exhausted, leading to uncoupling, with an increase in oxidative stress due to increased ROS formation and impaired hepatic insulin resistance, thus favouring NASH development [61, 62, 63]. Mitochondrial ROS production plays a leading role in propagating hepatocyte damage through lipid peroxidation products and (tumor necrosis factor-α) TNF-α production, both of which booster of mitochondrial injury, permeability, and uncoupling [61, 64].

Genetic factors associated with CV events in NAFLD are still undefined. Single-nucleotide polymorphisms (SNPs) may have a role on NAFLD-related cardiovascular disease (CVD) risk. The two most frequently SNPs in NAFLD are the patatin-like phospholipase domain-containing protein 3 (PNPLA3) and the transmembrane 6 superfamily member 2 (TM6SF2). PNPLA3 modulates the lipid droplet profile and appears to be related to tryglicerides (TG) metabolism [65]. It is shown that carriers of this mutation could increase atherosclerosis, paradoxically lowering serum TG levels [66, 67]. In a Danish cohort study, using Mendelian randomization to test a genetic variant in the gene encoding for PNPLA3, a genetically elevated hepatic fat content was not found associated to an increased risk of ischemic heart disease [68]. TM6SF2 modulates TG and cholesterol secretion in the liver through the excretion of very low-density lipoprotein (VLDL) [69]. The carriers of this mutation could raise the risk of NAFLD development because of increased TG and lipid retention in the liver, but they may exhibit some degree of cardio protection resulting in reduced serum levels of TG, low-density lipoprotein (LDL) and total cholesterol [70]. The glucokinase regulator (GCKR) is another susceptibility gene for NAFLD, which encodes liver-specific glucokinase regulatory protein (GKRP) and is involved in both hepatic de novo lipogenesis and NAFLD development [52, 71]. A recent study shown that common variants of the GCKR gene weakly could be associated with CVD risk (Odds Ratio per risk allele: 1.02, 95% confidence interval (CI): 1.0–1.04). However, the increased CVD risk might also be due to changes (increased levels) in VLDL [72].

Recent findings have proposed dysbiosis as a contributing mechanism in NAFLD development and progression. In fact, alteration in intestinal composition seems to increase intestinal tissue permeability, due to increased ethanol production. Therefore, gut-derived pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide, can reach the liver through blood flow, stimulate proinflammatory pathways and drive to liver inflammation and fibrosis [73, 74, 75, 76]. Moreover, gut microbiota increases the choline metabolism, which results in an increase in triglycerides content in the liver, due to the lack of VLDL secretion. In addition, it seems that dysbiosis is also associated to endothelial lipoprotein lipase inhibition, due to the decreased secretion of fasting-induced adipocyte factors and angiopoietin-like 4, with a consequent lack of hydrolyzation in the liver of triglycerides from VLDL particles, and increased gluconeogenesis and lipogenesis due to increased substrate [73].

Even though NAFLD is strongly associated with obesity, a substantial proportion of lean subjects without increased waist circumference can also develop NAFLD. Visceral obesity as opposed to general obesity, genetic predisposition, unhealthy dietary pattern consisting of high cholesterol and fructose intake may be associated with “lean” NAFLD [77].

4. NAFLD and CV risk: a phatological link

Recently, numerous epidemiologic studies have documented that NAFLD is associated to increased CV morbidity and mortality both in general population and in patients with diabetes mellitus (DM), a population already burdened by chronic vascular and extravascular complications [78, 79, 80, 81, 82, 83, 84, 85].

Paik et al. [86] proved CVD as one of the major causes of death among NAFLD patients. NAFLD and CVD share several common cardiometabolic risk factors such as genetics, systemic inflammation, endothelial dysfunction, hepatic insulin resistance, adipose tissue dysfunction, oxidative stress and lipid metabolism [87, 88]. Moreover, evidence shows that NAFLD is closely interrelated with several metabolic conditions that expose subjects to an increased risk of CV disease [89, 90, 91, 92]. The association between NAFLD and CV events is also underlined by recent European guidelines, which recommend CV screening in all subjects with NAFLD [93]. Many markers of subclinical atherosclerosis, such as increased carotid intima-media thickness, reduced flow-mediated vasodilation, arterial stiffness and increased coronary artery calcification, are reported in patients with NAFLD [94]. In a cohort of 755 patients, Moon et al. [95] reported a severe and independent association between NAFLD and carotid artery inflammation, which may be the expression of plaque vulnerability, as assessed by positron emission tomography with 18 F-fluorodeoxyglucose. Patients with NAFLD develop hyperglycemia, hyperinsulinemia, hyperlipidemia, and vascular endothelial cell damage due to insulin resistance, which can also cause smooth muscle cell (SMC) proliferation. Thus, insulin resistance counts for the development of both NAFLD and atherosclerosis [96]. In a meta-analysis of almost 300,000 individuals, subjects with NAFLD had a >2-fold increased risk of DM compared to the controls (hazard ratio [HR]: 2.22; 95% CI: 1.84 to 2.60) with more advanced fibrosis scores conferring higher risk (HR: 4. 27; 95% CI: 3.54 to 5.94) [97]. NAFLD represents an additional risk conferred to patients with DM, as the prevalence of CVD in patients with DM and NAFLD is higher compared to the risk in patients with DM without NAFLD [98]. In NAFLD, systemic inflammation is characterized by increased levels of TNF-α, interleukin (IL)-6, monocyte chemotactic protein-1, and C-reactive protein, which are associated to endothelial dysfunction. They alter vascular tone and play a crucial role in vascular plaque formation and liver injury [99, 100]. Moreover, steatosis hepatocytes release extracellular vesicles, which stimulate endothelial inflammation [101]. The vesicles from hepatocytes with steatosis were characterized by altered micro-ribonucleic acid (miRNA) expression, thereby leading to vascular inflammation via micro-RNA-1 (miR-1) release and activation of nuclear factor kappalight-chain-enhancer of activated B cells (NF-κB). It is reasonable that steatotic hepatocyte-derived extracellular vesicles induce vascular inflammation and facilitate atherogenesis through miR-1 delivery, NF-κB activation and reduced Krüppel-like factor 4 [102]. In addition, increased levels of matrix metalloproteinases have also been found in NAFLD and could play a critical role in the atheromasic process through unstable atherosclerotic plaques [103]. Increased levels of asymmetric dimethylarginine, an endogenous nitric oxide synthase antagonist, are frequent in subjects with NAFLD [101]. Hyperhomocysteinemia is typical of NAFLD: it is mainly due to changes in methionine metabolism and is associated with increased intrahepatic vascular resistance compromising nitric oxide formation and causing oxidative stress, which increases platelet activation [104]. NAFLD modifies serum lipid profiles, leading to abnormally elevated levels of TG, VLDL, and LDL, as well as abnormally reduced levels of high-density lipoprotein (HDL) [105]. Subjects with obesity and/or type 2 DM have hypersecretion of VLDL due to high plasma levels of free fatty acids and high hepatic fat content [35]. Elevated serum concentrations of VLDL and LDL have been linked to hepatic lobular inflammation, regardless of steatosis [106]. Investigations of lipoprotein subclasses reveal that subjects with NASH have significantly smaller LDL peak diameter and particle size, higher LDL particle concentration, higher low-density lipoprotein subfraction IVb (LDL-IVb) levels, and reduced HDL2b levels [107]. Probably, this altered composition of serum lipoproteins contributes to the increased CVD risk. Adiponectin produced in the liver could activate vascular endothelial nitric oxide synthase and gradually reduce atherosclerotic lesions and its levels are reduced in patients with NAFLD [108, 109, 110]. Instead, leptin, also produced by the liver, is closely related to insulin resistance and is higher in subjects with NAFLD, related to the severity of the disease [111]. Visfatin, an adipocytokine produced by visceral adipose tissue activity, correlates to several risk factors for AS including endothelial dysfunction, inflammation, vascular endothelial proliferation, and atherosclerotic plaque formation. Visfatin probably contributes to the atherosclerotic process in NAFLD, since its increase is related to the stage of NAFLD and in foamy macrophages in AS plaques [112]. The intestinal microbiota has been observed as a risk factor contributing to NAFLD development [113]. Gut microbiota-derived lipopolysaccharide levels are increased in subjects with NAFLD and exhibit endotoxic effects. When endotoxin plasma concentrations increase in the portal vein, systemic endotoxemia and mild chronic inflammation are observed [114]. Gut microorganisms could contribute to AS through indirect pathways in patients with NAFLD [115].

All these pathophysiological mechanisms can result in remodeling of left ventricle, increased ventricular mass and diastolic dysfunction. In NAFLD, portal pressure is increased as a result of changes in sinusoidal morphology, reduced sinusoidal flow, and increased intrahepatic resistance, mainly to the presence of fibrosis in more advanced stages of disease [110]. NAFLD leads to an increase in body surface area, which further increases left ventricular filling pressure, cardiac output, and volume overload [116, 117]. Recently, the connection between NAFLD and increased risk of arrhythmias, mainly permanent atrial fibrillation (AF), has achieved extensive scientific interest [90]. Mantovani et al. [7] found that NAFLD doubles the risk of prevalent AF independently of other AF risk factors. In addition, several studies also shown that NAFLD is associated with an increased risk of incident AF, especially among subjects with established T2DM. Park et al. [118] observed that individuals with NAFLD and advanced fibrosis had a greater risk of prevalent AF even after adjusting for multiple CVD risk factors. The presence and severity of NAFLD is associated with a higher risk for corrected QT (QTc) interval prolongation resulting in an increased risk of both ventricular tachyarrhythmias and sudden cardiac death [90]. In a hospital-based cohort of 751 elderly patients with T2DM, NAFLD shows a three-fold greater risk of cardiac conduction defects (right bundle branch block and left anterior hemiblock, persistent first-degree atrioventricular block), so it would result in a close association between NAFLD and cardiac arrhythmias [119]. However, further research is still needed to better explain the increased risk of cardiac death observed in patients with NAFLD. Furthermore, Mantovani et al. [120] observed that ultrasound-detected NAFLD is associated with an approximately 3.5-fold higher risk of both aortic valve sclerosis and mitral annular calcification in a population of hospitalized T2DM subjects with no history of known liver disease, heart failure, moderate-to-severe heart valve disease.

Moreover, an association of excessive peri-organ adipose tissue, namely intrahepatic, epicardial/pericardial, perivascular, intramuscular, peripancreatic and perirenal fat, with cardiometabolic and CVD risk factors was observed [121].

NAFLD, especially in advanced stage of liver fibrosis and severity of disease, increases the risk of coronary atherosclerosis, cardiomyopathy and arrhythmias, which clinically result in increased CVD morbidity and mortality. Further studies are still needed to better establish whether NAFLD independently contributes to the risk of developing adverse CVD outcomes and other cardiac/arrhythmic events. These mechanisms are summarized in Fig. 1.

Fig. 1.

NAFLD and CV risk: a pathophysiological link. NAFLD, Non-alcoholic fatty liver disease; TNF-α, Tumor necrosis factor; IL-6, Interleukin; CRP, C-reactive protein; TG, Triglycerides; VLDL, very-low-density lipoprotein; LDL, low-density-lipoprotein.

5. NAFLD treatment and impact on cardiovascular outcomes

Non-diabetic patients with NAFLD show a reduction in endothelium-mediated vasodilation of the brachial artery, which correlates with the extent of histologically documented liver damage, irrespective of age, sex, body mass index, and the presence of insulin resistance [122]. The implication of this association is that the treatment of NAFLD can have CV benefits and impact on CV outcomes. NAFLD is a common condition, particularly in patients who often consult the cardiologist, such as obese, diabetic and patients with metabolic syndrome. The close association with classic risk factors but especially the high prevalence of CV diseases in patients with NAFLD, mean that this disease cannot be considered the exclusive domain of the hepatologist. The possible clinical scenarios that may present to the cardiologist could be: (1) evaluation of a patient in primary prevention (based on available evidence and because it can be considered a “modifiable factor, NAFLD should be researched for optimal and complete cardiovascular risk estimation”); (2) patient with recent CV event (in these subjects the suspicion of NAFLD is less likely because, especially in the acute phase, it is possible to have an increase in transaminases related to the recent coronary event). Hence the need to treat the CV risk factors that cause NAFLD and to treat NAFLD as a CV risk factor.

If this condition of several comorbidities is observed in elderly patients, they become fragile subjects with different phenotypes. Different frailty phenotypes are differentially associated with adverse events that can worsen the prognosis [123, 124].

Currently, NAFLD has no approved drug therapy, and the objective of treatment is to prevent or reduce the liver injury. In this scenario, the target is to reduce NAFLD related risk factors and hypertriglyceridemia, diabetes, high cholesterol levels and obesity [125].

5.1 Change in lifestyle

The first suggested steps are lifestyle and diet changes. It is well known that a Mediterranean diet and balanced physical activity are essential to prevent the progression of NAFLD and incidence of CVD. Through a reduction of body weight, BMI, systolic blood pressure, ALT/AST, fasting glucose, total cholesterol and low-density lipoprotein cholesterol (LDL-C) and fatty liver index and liver stiffness many authors observe a risk reduction of cirrhosis and CVD [126, 127, 128, 129, 130].

The European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD), European Association for the Study of Obesity (EASO) and American Association for the Study of Liver Diseases (AASLD) practice guidelines for the management of NAFLD recommend that, in overweight/obese NAFLD patients, a 5–10% weight loss is the key goal of lifestyle interventions [17, 131]. Exercise has both direct effects on lipid metabolism and glycemic control, with an increase in VLDL clearance, promoting reduction of liver fat storage [132].

At the same time, these interventions have a direct and indirect effect on CV outcome. In fact, whereas these interventions and lifestyle modifications have shown benefit in patients with NAFLD, reducing liver involvement and consequently its CV effects by indirect impact, the direct role of lifestyle in the prevention of CV disease has been extensively established [133].

5.2 Drugs

In addition to Mediterranean diet and lifestyle changes, some drugs are being evaluated for the treatment of NAFLD, and mainly those who act on CV risk factors [134].

5.2.1 Probiotics

Some authors affirm that intestinal permeability, increased in a condition of hepatic disease, results in the release of bacterial products (e.g., endotoxins) in mesenteric circulation, and these products could promote the progression of liver damage [135]. Randomized clinical trials showed that by decreasing intestinal permeability and endotoxemia, probiotics could be considered significant treatment options [136, 137]. Compared to placebo they offered reduction of fatty liver index, ALT/AST, total cholesterol, homeostatic model assessment of insulin resistance (HOMA-IR), (ALT: weighted mean difference (WMD) –23.71, 95% CI: –33.46–13.95, P < 0.00001; AST: WMD –19.77, 95% CI: –32.55–7.00, P = 0.002; total cholesterol: WMD –0.28, 95% CI: –0.55–0.01, P = 0.04; HOMA-IR: WMD –0.46, 95% CI: –0.73–0.19, P = 0.0008) [138, 139, 140]. However, larger studies are needed to better clarify these effects and to evaluate efficacy on CV outcomes.

5.2.2 Vitamin E

Vitamin E is a well-known anti-oxidant agent and has been proposed in this setting, although there is a lack of clinical evidence. In the PIVENS trial, after 96 weeks of treatment, Vitamin E reduces AST/ALT levels, hepatic steatosis and lobular inflammation (P = 0.001, P = 0.005 and P = 0.02 respectively) [141]. Also, in this case, no effect was demonstrated on liver stiffness or on CV risk factors.

5.2.3 Omega-3 fatty acid

Omega-3 fatty acid, by reducing lipogenesis, seems to determine a triglyceride serum and hepatic fat content reduction and consequently a less production of VLDL. Therefore, this drug has been proposed as a theoretically effective drug to reduce CV risk in patients with NAFLD, diabetes or hypertriglyceridemia. Results from REDUCE-IT trial have shown a beneficial effect of icosapent ethyl 4 g/d in addition to statin vs placebo, both on the primary composite end point (major adverse cardiovascular events) (hazard ratio [HR], 0.69 [95% CI, 0.59–0.80]; P = 0.000001), and on secondary CV endpoints. However, similar effects were not achieved in two recent trials, STRENGTH (NCT02104817) and EFFECT I trial (NCT02354976), respectively on the achievement of CV outcome risk reduction and on the reduction of liver fat storage compared to the control group [142, 143, 144]. Gastrointestinal effects, such as abdominal pain and flatulence, were the main reported adverse events, connected to omega-3 fatty acid assumption [142].

5.2.4 Statins

Statins, due to the inhibition of 3-hydroxy-3methyl-glutaryl-coenzyme A reductase (HMG-CoA reductase), determines an LDL cholesterol levels reduction, thus leading to a reduction of CV risk [145]. Also for what concerns the use of statins, it seems that these drugs have a beneficial effect on the progression of NAFLD and on the reduction of the related CV risk, despite their potential effect on CK and AST/ALT elevation [146]. However, the use of different endpoints in the different studies does not allow to confirm the efficacy of these drugs on the evolution of NAFLD. Statin and ezetimibe are well known as safe and in a population of subjects not at high CV risk, AST/ALT and LDL cholesterol were significantly reduced at the end of the follow-up [145]. In a recent work conducted on a similar population, a multidrug approach with atorvastatin and Vitamin E and C, allowed a risk reduction of disease progression after a follow up of 3.6 years [147]. Some authors have shown the same effects also in populations with NAFLD and diabetes and hypertension [148, 149]. A sub analysis of the Greek Atorvastatin and Coronary Heart Disease Evaluation (GREACE) study evaluated 437 patients with NAFLD (227 of whom were treated with a statin and 210 were not). The authors observed that NAFLD patients who received statins had lower incidence of CV morbidity, without significant liver-related adverse events [150]. Several studies showed that statin use in individuals with NAFLD, is associated with a considerable improvement of steatosis, inflammation, and liver fibrosis [151, 152]. An observational study of 11,593,409 Korean subjects, showed that the statins were associated with a reduced risk of NAFLD (adjusted odds ratio (OR): 0.66, 95% CI: 0.65–0.67), as well as with a reduced risk of significant liver fibrosis [152]. Interestingly, in a recent systematic review and metanalysis it was described that in the interventional studies, ALT, AST and gamma-glutamyltransferase (GGT) were reduced after statin treatment on average between 25 and 35%, while observational studies showed an effect null, suggesting the hepatic safety of statins in patients with NAFLD [153]. Moreover, genetics may play a role in the pathogenesis of NAFLD/NASH and its treatment with statins [154].

5.2.5 Ezetimibe

Ezetimibe, a lipid-lowering agent acting by blocking the mediator of cholesterol absorption in the intestine - namely, the Niemann-Pick C1-Like 1 (NPC1L1) protein, showed a beneficial effect on NAFLD. In fact, in a meta-analysis that considered 273 NAFLD patients with and without T2DM, it showed to significantly reduce serum liver biomarkers and hepatic steatosis [155]. It is well-known that the combination of statin and ezetimibe and their effect on LDL-C values is crucial to reduce CV risk. The use of statins, ezetimibe, or their combination strategy is a mainstay in the therapy of patients at CV risk, and their use should be even more strongly recommended in NAFLD patients in light of the liver effects [133]. Moreover, a post-hoc analysis on 14,819 patients enrolled in the IMPROVE-IT study showed that NFS identifies patients at highest risk of recurrent CV events and most likely to benefit from dual lipid-lowering therapy (ezetimibe/simvastatin) [156]. Finally, Ezetimibe is generally well tolerated and also the addition with statins does not increase the risk of side effects [133].

5.2.6 PCSK9 inhibitors

Inhibitors of proprotein convertase subtilisin/kexin type 9 (PCSK9-i), a new class of lipid lowering treatment, promote the activity of LDL receptors, by inhibiting their degradation. Data on PCSK9-i are poor, however, a recent observational study conducted on patients with familial hypercholesterolemia has shown as also these drugs, through their mechanism, particularly in a subgroup with low TG/HDL levels, could determine a reduction of steatosis biomarkers, triglyceride-glucose index (TyG) and hepatic steatosis index (HSI) [157]. Some authors have suggested how these drugs could be an optimal solution to prevent NAFLD and reduce CV risk, however new trials are needed to better define this hypothesis [158].

5.2.7 Anti-hyperglycemic drug

In diabetic patients with NAFLD, insulin-sensitizing drug therapy is still the preferred choice, and many patients with T2DM are already treated with these drugs. Both metformin and thiazolidinediones have been shown to be useful not only in reducing steatosis, but also in reducing inflammation and fibrosis during treatment [159, 160, 161, 162]. Nevertheless, despite beneficial effects of metformin on liver enzymes and HbA1c levels, a recent metanalysis of randomized controlled trials that involving studies with patients with biopsy-proven NAFLD, showed a small beneficial effect on liver steatosis or inflammation and no effects on the NASH resolution and liver fibrosis [163]. It should be considered that metformin, which has numerous protective extra metabolic effects, can reduce the risk for CV events and death in T2DM patients who are overweight or obese, but the EASL-EASO-EASD and AASLD practice guidelines for the management of NAFLD, to date, do not support the use of metformin for the treatment of NAFLD [17, 131, 164, 165, 166]. On the other hand, the same guidelines recommend the use of pioglitazone in patients with NASH, in fact, pioglitazone has shown to improve advanced fibrosis in NASH patients, regardless of T2DM, and in absence of significant side effects [17, 163, 167]. However, mechanisms related to this beneficial effect on the liver are still unknown.

Recently, new antidiabetic drugs have demonstrated a great efficacy on CV outcomes [168, 169]. Moreover, some trials have evaluated if these treatments could have a beneficial effect also on NAFLD progression. In a recent study, both liraglutide and sitagliptin separately, in a population of subjects with diabetes and NAFLD, have shown efficacy on the reduction of intrahepatic lipid and visceral adipose tissue, evaluated through MRI after 24 weeks of treatment (15.4% ± 5.6% to 12.5% ± 6.4%, p < 0.001 for liraglutide and 15.5% ± 5.6% to 11.7% ± 5.0%, P = 0.001; 153.4 ± 31.5 to 139.8 ± 27.3, P = 0.027 for sitagliptin) [170]. Glucagon-like peptide-1 receptor agonists (GLP-1 RA) and Dipeptidyl Peptidase 4 inhibitors (DPP4i) seem to determine an increase of adiponectin levels, a reduction of IL-6 levels, and a modulation of CD36 pathway, thus resulting in a reduction of liver fat content [170, 171]. Results on the efficacy of DPP4 inhibitors are controversial. As elsewhere demonstrated, it seems that sitagliptin has no positive effect on NAFLD progression compared to placebo [172]. However, as suggested by the authors, the small sample size could explain these not significant results and thus is challenging to generalize these results [173]. Other studies have confirmed the protective role of liraglutide, on CV system and on liver damage, although it has not demonstrated a significant reduction in visceral adipose tissue [174, 175, 176]. Semaglutide, in patients with NASH have demonstrated a resolution of the liver impairment, even if with no significant fibrosis eradication, as previously emerged from the LEAN trial (P < 0.001 for semaglutide 0.4 mg vs. placebo) [177]. As discussed by the authors, these differences could be due to the short period of follow up [178]. These drugs are generally well tolerated, and mild-moderate gastrointestinal effects are recognized as the main reported adverse events [170].

Effects of SGLT2 inhibitors on NAFLD and type 2 diabetes mellitus have been evaluated in some few studies. The sodium-glucose co-transporter 2 inhibitors (SGLT2i) through an inhibition of SGLT2 receptors, induce urinary glucose excretion and consequently a reduction of serum glucose levels and an increase of insulin sensitivity. The documented adverse events are generally few and mainly related to urinary tract infections, related to the mechanism of action. Some animal models suggest that these effects could determine a reduction of liver lipogenesis and a down regulation of transcription factors that play a key role in fatty acid synthesis [179]. Dapagliflozin has shown efficacy on the reduction of liver damage indexes and on the liver disease progression, but also a reduction of visceral fat as compared to the control group (decrease in CAP from 314 ± 61 to 290 ± 73 dB/m (P = 0.0424)) [180, 181]. Similar results have been obtained by empagliflozin in other two trials, in which, at the end of the follow up, in the experimental group there is a significant reduction of liver fat evaluated by MRI (–22% (–36, –7; P = 0.009) and 16.2% to 11.3%; P < 0.0001). Authors suggest that empagliflozin could be useful in an early phase of disease to prevent the progression of liver steatosis in subjects with DM [182, 183]. Similar results have been observed also for canagliflozin, by measuring liver fat by MRI [184].

5.2.8 Specific NAFLD drugs

Several specific NAFLD drugs are in development and others are being studied in phase 3 trials [185]. Obeticholic acid (OCA), which is an analogue of chenodeoxycholic acid, is a synthetic ligand agonist of the farnesoid X receptor (FXR) [186]. This class of agent showed to improve insulin resistance, modulate glucose and lipid metabolism, and have direct anti-inflammatory effects in animal NASH models. While showing efficacy on hepatic outcome, this drug appears to increase LDL-C levels, exposing patients to increased CV risk [186, 187]. On the other hand, tight glycemic control improves the CV outcome even in non-diabetic subjects [188, 189]. Moreover, it has recently been documented that a reduction of multiple risk factors significantly improves the risk of fatal and non-fatal major adverse CV events [190].

IONIS-DGAT2Rx is a 20-O-methoxyethyl chimeric antisense oligonucleotide inhibitor that mediates degradation of Diacylglycerol-O acyltransferase 2 (DGAT2) mRNA. DGAT2 is one of two enzyme isoforms involved in the final step of triglyceride synthesis. It has been shown to be effective in improving liver parameters, but its CV safety needs to be better elucidated, as some CV events (i.e., cardiac arrest, ischemic cerebral infarction, deep-vein thrombosis) have been reported [191].

NAFLD should become part of the cardiologist’s background because of its high prevalence, close association with the main CV risk factors and relationship with coronary disease that seem to indicate an autonomous role of NAFLD on the atherosclerosis progression.

In recent years some evidence from clinical trials have shown that bariatric surgery, although not considered the gold standard of treatment, could determine a beneficial effect in patients with NAFLD and liver fibrosis. Notably, it has been shown that patients observed before and after surgery, present a reduction of both steatosis and liver fibrosis, evaluated through liver biopsy. Moreover, in these patients it has been reported an improvement of biochemical parameters (e.g., AST/ALT) [192]. Some authors suggest that the beneficial effect could also improve the CV profile, however there is still no clear evidence on this point and future studies are required to better define this association [193, 194].

6. Conclusions

The recent literature has extensively documented patients with NAFLD are exposed to an increased risk of progression of liver disease, and a higher risk of extrahepatic complications, particularly CV disease. This risk seems to be conditioned by several factors among which the most relevant is the ability of NAFLD to interfere with cardiometabolic risk factors.

New drugs, evaluated in phase 3 trials for the treatment of NAFLD, can also impact metabolic risk factors such as glycemic control and lipid profile. Therefore, the possible protective effect of these drugs will be evaluated not only on liver damage but also on CV risk.

In conclusion, NAFLD seems to be the epiphenomenon of a systemic disease that can also affect the CV system, requiring a multidisciplinary diagnostic and therapeutic assessment.

Author contributions

FCS, RG, ACa, EV, ACe, PC designed the research study. RG, ACa, EV, ACe, FCS performed the research. ACe, LR provided help and advice on the table and figures; TS, RM, CS, EM, FG analyzed the literature data. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Acknowledgment

Raffaele Galiero was supported by the Programma VALERE, University of Campania “Luigi Vanvitelli”.

Funding

This research received no external funding.

Conflict of interest

The authors declare no conflict of interest.

References
[1]
Ye Q, Zou B, Yeo YH, Li J, Huang DQ, Wu Y, et al. Global prevalence, incidence, and outcomes of non-obese or lean non-alcoholic fatty liver disease: a systematic review and meta-analysis. The Lancet Gastroenterology & Hepatology. 2020; 5: 739–752.
[2]
Younossi ZM, Koenig AB, 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.
[3]
Fracanzani AL, Burdick L, Raselli S, Pedotti P, Grigore L, Santorelli G, et al. Carotid Artery Intima-media Thickness in Nonalcoholic Fatty Liver Disease. The American Journal of Medicine. 2008; 121: 72–78.
[4]
Bonapace S, Perseghin G, Molon G, Canali G, Bertolini L, Zoppini G, et al. Nonalcoholic fatty liver disease is associated with left ventricular diastolic dysfunction in patients with type 2 diabetes. Diabetes Care. 2012; 35: 389–395.
[5]
Targher G, Bertolini L, Padovani R, Rodella S, Tessari R, Zenari L, et al. Prevalence of nonalcoholic fatty liver disease and its association with cardiovascular disease among type 2 diabetic patients. Diabetes Care. 2007; 30: 1212–1218.
[6]
Pais R, Giral P, Khan J, Rosenbaum D, Housset C, Poynard T, et al. Fatty liver is an independent predictor of early carotid atherosclerosis. Journal of Hepatology. 2016; 65: 95–102.
[7]
Mantovani A, Dauriz M, Sandri D, Bonapace S, Zoppini G, Tilg H, et al. Association between non‐alcoholic fatty liver disease and risk of atrial fibrillation in adult individuals: an updated meta‐analysis. Liver International. 2019; 39: 758–769.
[8]
Targher G, Byrne CD, Lonardo A, Zoppini G, Barbui C. Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: a meta-analysis. Journal of Hepatology. 2016; 65: 589–600.
[9]
Targher G, Bertolini L, Padovani R, Rodella S, Zoppini G, Zenari L, et al. Relations between carotid artery wall thickness and liver histology in subjects with nonalcoholic fatty liver disease. Diabetes Care. 2006; 29: 1325–1330.
[10]
Petta S, Argano C, Colomba D, Cammà C, Di Marco V, Cabibi D, et al. Epicardial fat, cardiac geometry and cardiac function in patients with non-alcoholic fatty liver disease: Association with the severity of liver disease. Journal of Hepatology. 2015; 62: 928–933.
[11]
Sinn DH, Kang D, Chang Y, Ryu S, Gu S, Kim H, et al. Non-alcoholic fatty liver disease and progression of coronary artery calcium score: a retrospective cohort study. Gut. 2017; 66: 323–329.
[12]
Acierno C, Caturano A, Pafundi PC, Nevola R, Adinolfi LE, Sasso FC. Nonalcoholic fatty liver disease and type 2 diabetes: pathophysiological mechanisms shared between the two faces of the same coin. Exploration of Medicine. 2020; 1: 287–306.
[13]
Ascione A, Fontanella L, Imparato M, Rinaldi L, De Luca M. Mortality from cirrhosis and hepatocellular carcinoma in Western Europe over the last 40 years. Liver International. 2017; 37: 1193–1201.
[14]
Valente G, Rinaldi L, Sgambato M, Piai G. Conversion from twice-daily to once-daily tacrolimus in stable liver transplant patients: effectiveness in a real-world setting. Transplantation Proceedings. 2013; 45: 1273–1275.
[15]
European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD), European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. Diabetologia. 2016; 59: 1121–1140.
[16]
Masarone M, Rosato V, Aglitti A, Bucci T, Caruso R, Salvatore T, et al. Liver biopsy in type 2 diabetes mellitus: Steatohepatitis represents the sole feature of liver damage. PLoS ONE. 2017; 12: e0178473.
[17]
European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD), European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. Journal of Hepatology. 2016; 64: 1388–1402.
[18]
Rinaldi L, Pafundi PC, Galiero R, Caturano A, Morone MV, Silvestri C, et al. Mechanisms of Non-Alcoholic Fatty Liver Disease in the Metabolic Syndrome. A Narrative Review. Antioxidants. 2021; 10: 270.
[19]
Wieckowska A, McCullough AJ, Feldstein AE. Noninvasive diagnosis and monitoring of nonalcoholic steatohepatitis: present and future. Hepatology. 2007; 46: 582–589.
[20]
Petta S, Wong VW, Cammà C, Hiriart J, Wong GL, Marra F, et al. Improved noninvasive prediction of liver fibrosis by liver stiffness measurement in patients with nonalcoholic fatty liver disease accounting for controlled attenuation parameter values. Hepatology. 2017; 65: 1145–1155.
[21]
Lombardi R, Airaghi L, Targher G, Serviddio G, Maffi G, Mantovani A, et al. Liver fibrosis by FibroScan® independently of established cardiovascular risk parameters associates with macrovascular and microvascular complications in patients with type 2 diabetes. Liver International. 2020; 40: 347–354.
[22]
Lombardi R, Petta S, Pisano G, Dongiovanni P, Rinaldi L, Adinolfi LE, et al. FibroScan Identifies Patients with Nonalcoholic Fatty Liver Disease and Cardiovascular Damage. Clinical Gastroenterology and Hepatology. 2020; 18: 517–519.
[23]
Petroff D, Blank V, Newsome PN, Shalimar, Voican CS, Thiele M, et al. Assessment of hepatic steatosis by controlled attenuation parameter using the M and XL probes: an individual patient data meta-analysis. The Lancet Gastroenterology & Hepatology. 2021; 6: 185–198.
[24]
Rinaldi L, Valente G, Piai G. Serial Liver Stiffness Measurements and Monitoring of Liver-Transplanted Patients in a Real-Life Clinical Practice. Hepatitis Monthly. 2016; 16: e41162.
[25]
Leung C, Rivera L, Furness JB, Angus PW. The role of the gut microbiota in NAFLD. Nature Reviews Gastroenterology & Hepatology. 2016; 13: 412–425.
[26]
Mehal WZ. The Gordian Knot of dysbiosis, obesity and NAFLD. Nature Reviews Gastroenterology & Hepatology. 2013; 10: 637–644.
[27]
Sharpton SR, Ajmera V, Loomba R. Emerging Role of the Gut Microbiome in Nonalcoholic Fatty Liver Disease: from Composition to Function. Clinical Gastroenterology and Hepatology. 2019; 17: 296–306.
[28]
Bedogni G, Bellentani S, Miglioli L, Masutti F, Passalacqua M, Castiglione A, et al. The Fatty Liver Index: a simple and accurate predictor of hepatic steatosis in the general population. BMC Gastroenterology. 2006; 6: 33.
[29]
Ballestri S, Lonardo A, Romagnoli D, Carulli L, Losi L, Day CP, et al. Ultrasonographic fatty liver indicator, a novel score which rules out NASH and is correlated with metabolic parameters in NAFLD. Liver International. 2012; 32: 1242–1252.
[30]
Kotronen A, Peltonen M, Hakkarainen A, Sevastianova K, Bergholm R, Johansson LM, et al. Prediction of Non-Alcoholic Fatty Liver Disease and Liver Fat Using Metabolic and Genetic Factors. Gastroenterology. 2009; 137: 865–872.
[31]
Marjot T, Moolla A, Cobbold JF, Hodson L, Tomlinson JW. Nonalcoholic Fatty Liver Disease in Adults: Current Concepts in Etiology, Outcomes, and Management. Endocrine Reviews. 2020; 41: 66–117.
[32]
Sasso FC, Pafundi PC, Caturano A, Galiero R, Vetrano E, Nevola R, et al. Impact of direct acting antivirals (DAAs) on cardiovascular events in HCV cohort with pre-diabetes. Nutrition, Metabolism and Cardiovascular Diseases. 2021; 31: 2345–2353.
[33]
Adinolfi LE, Petta S, Fracanzani AL, Coppola C, Narciso V, Nevola R, et al. Impact of hepatitis C virus clearance by direct-acting antiviral treatment on the incidence of major cardiovascular events: a prospective multicentre study. Atherosclerosis. 2020; 296: 40–47.
[34]
Adinolfi LE, Petta S, Fracanzani AL, Nevola R, Coppola C, Narciso V, et al. Reduced incidence of type 2 diabetes in patients with chronic hepatitis C virus infection cleared by direct‐acting antiviral therapy: a prospective study. Diabetes, Obesity and Metabolism. 2020; 22: 2408–2416.
[35]
Cusi K. Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: pathophysiology and clinical implications. Gastroenterology. 2012; 142: 711–725.e6.
[36]
Petersen MC, Shulman GI. Roles of Diacylglycerols and Ceramides in Hepatic Insulin Resistance. Trends in Pharmacological Sciences. 2017; 38: 649–665.
[37]
Puri P, Baillie RA, Wiest MM, Mirshahi F, Choudhury J, Cheung O, et al. A lipidomic analysis of nonalcoholic fatty liver disease. Hepatology. 2007; 46: 1081–1090.
[38]
Min H, Kapoor A, Fuchs M, Mirshahi F, Zhou H, Maher J, et al. Increased hepatic synthesis and dysregulation of cholesterol metabolism is associated with the severity of nonalcoholic fatty liver disease. Cell Metabolism. 2012; 15: 665–674.
[39]
Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. The Journal of Clinical Endocrinology and Metabolism. 2004; 89: 2548–2556.
[40]
Jung UJ, Choi M. Obesity and its metabolic complications: the role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. International Journal of Molecular Sciences. 2014; 15: 6184–6223.
[41]
Wang Y, Rimm EB, Stampfer MJ, Willett WC, Hu FB. Comparison of abdominal adiposity and overall obesity in predicting risk of type 2 diabetes among men. The American Journal of Clinical Nutrition. 2005; 81: 555–563.
[42]
Buechler C, Wanninger J, Neumeier M. Adiponectin, a key adipokine in obesity related liver diseases. World Journal of Gastroenterology. 2011; 17: 2801–2811.
[43]
Yoon MJ, Lee GY, Chung J, Ahn YH, Hong SH, Kim JB. Adiponectin increases fatty acid oxidation in skeletal muscle cells by sequential activation of AMP-activated protein kinase, p38 mitogen-activated protein kinase, and peroxisome proliferator-activated receptor alpha. Diabetes. 2006; 55: 2562–2570.
[44]
Liu Q, Yuan B, Lo KA, Patterson HC, Sun Y, Lodish HF. Adiponectin regulates expression of hepatic genes critical for glucose and lipid metabolism. Proceedings of the National Academy of Sciences of the United States of America. 2012; 109: 14568–14573.
[45]
Matsumoto H, Tamura S, Kamada Y, Kiso S, Fukushima J, Wada A, et al. Adiponectin deficiency exacerbates lipopolysaccharide/D-galactosamine-induced liver injury in mice. World Journal of Gastroenterology. 2006; 12: 3352–3358.
[46]
Ouchi N, Walsh K. Adiponectin as an anti-inflammatory factor. Clinica Chimica Acta. 2007; 380: 24–30.
[47]
Ding X, Saxena NK, Lin S, Xu A, Srinivasan S, Anania FA. The Roles of Leptin and Adiponectin. The American Journal of Pathology. 2005; 166: 1655–1669.
[48]
Adachi M, Brenner DA. High molecular weight adiponectin inhibits proliferation of hepatic stellate cells via activation of adenosine monophosphate-activated protein kinase. Hepatology. 2008; 47: 677–685.
[49]
Kumar P, Raeman R, Chopyk DM, Smith T, Verma K, Liu Y, et al. Adiponectin inhibits hepatic stellate cell activation by targeting the PTEN/AKT pathway. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease. 2018; 1864: 3537–3545.
[50]
Boden G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes. 1997; 46: 3–10.
[51]
Suganami T, Nishida J, Ogawa Y. A paracrine loop between adipocytes and macrophages aggravates inflammatory changes: role of free fatty acids and tumor necrosis factor alpha. Arteriosclerosis, Thrombosis, and Vascular Biology. 2005; 25: 2062–2068.
[52]
Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD, Parks EJ. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. The Journal of Clinical Investigation. 2005; 115: 1343–1351.
[53]
Shimomura I, Bashmakov Y, Horton JD. Increased levels of nuclear SREBP-1c associated with fatty livers in two mouse models of diabetes mellitus. The Journal of Biological Chemistry. 1999; 274: 30028–30032.
[54]
Watt MJ, Miotto PM, De Nardo W, Montgomery MK. The Liver as an Endocrine Organ—Linking NAFLD and Insulin Resistance. Endocrine Reviews. 2019; 40: 1367–1393.
[55]
Flannery C, Dufour S, Rabøl R, Shulman GI, Petersen KF. Skeletal muscle insulin resistance promotes increased hepatic de novo lipogenesis, hyperlipidemia, and hepatic steatosis in the elderly. Diabetes. 2012; 61: 2711–2717.
[56]
DeFronzo RA, Tripathy D. Skeletal Muscle Insulin Resistance is the Primary Defect in Type 2 Diabetes. Diabetes Care. 2009; 32: S157–S163.
[57]
Mäenpää PH, Raivio KO, Kekomäki MP. Liver adenine nucleotides: fructose-induced depletion and its effect on protein synthesis. Science. 1968; 161: 1253–1254.
[58]
Lanaspa MA, Sanchez-Lozada LG, Choi Y, Cicerchi C, Kanbay M, Roncal-Jimenez CA, et al. Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: potential role in fructose-dependent and -independent fatty liver. The Journal of Biological Chemistry. 2012; 287: 40732–40744.
[59]
Choi Y, Shin H, Choi HS, Park J, Jo I, Oh E, et al. Uric acid induces fat accumulation via generation of endoplasmic reticulum stress and SREBP-1c activation in hepatocytes. Laboratory Investigation. 2014; 94: 1114–1125.
[60]
Murphy MP. How mitochondria produce reactive oxygen species. The Biochemical Journal. 2009; 417: 1–13.
[61]
Koliaki C, Szendroedi J, Kaul K, Jelenik T, Nowotny P, Jankowiak F, et al. Adaptation of hepatic mitochondrial function in humans with non-alcoholic fatty liver is lost in steatohepatitis. Cell Metabolism. 2015; 21: 739–746.
[62]
Serviddio G, Bellanti F, Tamborra R, Rollo T, Capitanio N, Romano AD, et al. Uncoupling protein-2 (UCP2) induces mitochondrial proton leak and increases susceptibility of non-alcoholic steatohepatitis (NASH) liver to ischaemia-reperfusion injury. Gut. 2008; 57: 957–965.
[63]
Cortez-Pinto H, Chatham J, Chacko VP, Arnold C, Rashid A, Diehl AM. Alterations in liver ATP homeostasis in human nonalcoholic steatohepatitis: a pilot study. The Journal of the American Medical Association. 1999; 282: 1659–1664.
[64]
Leclercq IA, Farrell GC, Field J, Bell DR, Gonzalez FJ, Robertson GR. CYP2E1 and CYP4a as microsomal catalysts of lipid peroxides in murine nonalcoholic steatohepatitis. The Journal of Clinical Investigation. 2000; 105: 1067–1075.
[65]
Smagris E, BasuRay S, Li J, Huang Y, Lai KV, Gromada J, et al. Pnpla3i148M knockin mice accumulate PNPLA3 on lipid droplets and develop hepatic steatosis. Hepatology. 2015; 61: 108–118.
[66]
Petta S, Valenti L, Marchesini G, Di Marco V, Licata A, Cammà C, et al. PNPLA3 GG genotype and carotid atherosclerosis in patients with non-alcoholic fatty liver disease. PLoS ONE. 2013; 8: e74089.
[67]
Palmer CNA, Maglio C, Pirazzi C, Burza MA, Adiels M, Burch L, et al. Paradoxical lower serum triglyceride levels and higher type 2 diabetes mellitus susceptibility in obese individuals with the PNPLA3 148M variant. PLoS ONE. 2012; 7: e39362.
[68]
Lauridsen BK, Stender S, Kristensen TS, Kofoed KF, Køber L, Nordestgaard BG, et al. Liver fat content, non-alcoholic fatty liver disease, and ischaemic heart disease: Mendelian randomization and meta-analysis of 279 013 individuals. European Heart Journal. 2018; 39: 385–393.
[69]
Kozlitina J, Smagris E, Stender S, Nordestgaard BG, Zhou HH, Tybjærg-Hansen A, et al. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nature Genetics. 2014; 46: 352–356.
[70]
Dongiovanni P, Petta S, Maglio C, Fracanzani AL, Pipitone R, Mozzi E, et al. Transmembrane 6 superfamily member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease. Hepatology. 2015; 61: 506–514.
[71]
Brouwers MCGJ, Jacobs C, Bast A, Stehouwer CDA, Schaper NC. Modulation of Glucokinase Regulatory Protein: a Double-Edged Sword? Trends in Molecular Medicine. 2015; 21: 583–594.
[72]
Brouwers MCGJ, Simons N, Stehouwer CDA, Isaacs A. Non-alcoholic fatty liver disease and cardiovascular disease: assessing the evidence for causality. Diabetologia. 2020; 63: 253–260.
[73]
Hu H, Lin A, Kong M, Yao X, Yin M, Xia H, et al. Intestinal microbiome and NAFLD: molecular insights and therapeutic perspectives. Journal of Gastroenterology. 2020; 55: 142–158.
[74]
Angulo P, Hui JM, Marchesini G, Bugianesi E, George J, Farrell GC, et al. The NAFLD fibrosis score: a noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology. 2007; 45: 846–854.
[75]
McPherson S, Hardy T, Dufour J, Petta S, Romero-Gomez M, Allison M, et al. Age as a Confounding Factor for the Accurate Non-Invasive Diagnosis of Advanced NAFLD Fibrosis. The American Journal of Gastroenterology. 2017; 112: 740–751.
[76]
Lin Z, Xin Y, Dong Q, Wang Q, Jiang X, Zhan S, et al. Performance of the aspartate aminotransferase-to-platelet ratio index for the staging of hepatitis C-related fibrosis: an updated meta-analysis. Hepatology. 2011; 53: 726–736.
[77]
Chrysavgis L, Ztriva E, Protopapas A, Tziomalos K, Cholongitas E. Nonalcoholic fatty liver disease in lean subjects: Prognosis, outcomes and management. World Journal of Gastroenterology. 2020; 26: 6514–6528.
[78]
Williams KH, Shackel NA, Gorrell MD, McLennan SV, Twigg SM. Diabetes and nonalcoholic Fatty liver disease: a pathogenic duo. Endocrine Reviews. 2013; 34: 84–129.
[79]
Targher G, Byrne CD. Clinical Review: Nonalcoholic fatty liver disease: a novel cardiometabolic risk factor for type 2 diabetes and its complications. The Journal of Clinical Endocrinology and Metabolism. 2013; 98: 483–495.
[80]
Peters SAE, Huxley RR, Sattar N, Woodward M. Sex Differences in the Excess Risk of Cardiovascular Diseases Associated with Type 2 Diabetes: Potential Explanations and Clinical Implications. Current Cardiovascular Risk Reports. 2015; 9: 36.
[81]
Ballestri S, Lonardo A, Bonapace S, Byrne CD, Loria P, Targher G. Risk of cardiovascular, cardiac and arrhythmic complications in patients with non-alcoholic fatty liver disease. World Journal of Gastroenterology. 2014; 20: 1724–1745.
[82]
Targher G, Byrne CD, Tilg H. NAFLD and increased risk of cardiovascular disease: clinical associations, pathophysiological mechanisms and pharmacological implications. Gut. 2020; 69: 1691–1705.
[83]
Mantovani A, Scorletti E, Mosca A, Alisi A, Byrne CD, Targher G. Complications, morbidity and mortality of nonalcoholic fatty liver disease. Metabolism. 2020; 111: 154170.
[84]
Torella D, Iaconetti C, Tarallo R, Marino F, Giurato G, Veneziano C, et al. miRNA Regulation of the Hyperproliferative Phenotype of Vascular Smooth Muscle Cells in Diabetes. Diabetes. 2018; 67: 2554–2568.
[85]
Sasso FC, Salvatore T, Tranchino G, Cozzolino D, Caruso AA, Persico M, et al. Cochlear dysfunction in type 2 diabetes: a complication independent of neuropathy and acute hyperglycemia. Metabolism: Clinical and Experimental. 1999; 48: 1346–1350.
[86]
Paik JM, Henry L, De Avila L, Younossi E, Racila A, Younossi ZM. Mortality Related to Nonalcoholic Fatty Liver Disease is Increasing in the United States. Hepatology Communications. 2019; 3: 1459–1471.
[87]
Ismaiel A., Dumitraşcu D.L. Cardiovascular Risk in Fatty Liver Disease: The Liver-Heart Axis-Literature Review. Frontiers in Medicine. 2019; 6: 202.
[88]
Sasso FC, Pafundi PC, Gelso A, Bono V, Costagliola C, Marfella R, et al. NO BLIND Study Group. Telemedicine for screening diabetic retinopathy: The NO BLIND Italian multicenter study. Diabetes/Metabolism Research and Reviews. 2019; 35: e3113.
[89]
Adams LA, Anstee QM, Tilg H, Targher G. Non-alcoholic fatty liver disease and its relationship with cardiovascular disease and other extrahepatic diseases. Gut. 2017; 66: 1138–1153.
[90]
Anstee QM, Mantovani A, Tilg H, Targher G. Risk of cardiomyopathy and cardiac arrhythmias in patients with nonalcoholic fatty liver disease. Nature Reviews Gastroenterology & Hepatology. 2018; 15: 425–439.
[91]
Zhou Y, Zhou X, Wu S, Fan D, Van Poucke S, Chen Y, et al. Nonalcoholic fatty liver disease contributes to subclinical atherosclerosis: a systematic review and meta-analysis. Hepatology Communications. 2018; 2: 376–392.
[92]
Bonci E, Chiesa C, Versacci P, Anania C, Silvestri L, Pacifico L. Association of Nonalcoholic Fatty Liver Disease with Subclinical Cardiovascular Changes: a Systematic Review and Meta-Analysis. BioMed Research International. 2015; 2015: 213737.
[93]
Wu S, Wu F, Ding Y, Hou J, Bi J, Zhang Z. Association of non-alcoholic fatty liver disease with major adverse cardiovascular events: a systematic review and meta-analysis. Scientific Reports. 2016; 6: 33386.
[94]
Oni ET, Agatston AS, Blaha MJ, Fialkow J, Cury R, Sposito A, et al. A systematic review: burden and severity of subclinical cardiovascular disease among those with nonalcoholic fatty liver; should we care? Atherosclerosis. 2013; 230: 258–267.
[95]
Moon SH, Noh TS, Cho YS, Hong SP, Hyun SH, Choi JY, et al. Association between nonalcoholic fatty liver disease and carotid artery inflammation evaluated by 18F-fluorodeoxyglucose positron emission tomography. Angiology. 2015; 66: 472–480.
[96]
Severova MM, Saginova EA, Galliamov MG, Ermakov NV, Rodina AV, Fomin VV. Clinicopathogenetic characteristics of cardiorenal syndrome in non-alcoholic fatty liver disease. Terapevticheskii arkhiv. 2012; 84: 15–20. (In Russian)
[97]
Mantovani A, Byrne CD, Bonora E, Targher G. Nonalcoholic Fatty Liver Disease and Risk of Incident Type 2 Diabetes: a Meta-analysis. Diabetes Care. 2018; 41: 372–382.
[98]
Targher G, Bertolini L, Padovani R, Poli F, Scala L, Tessari R, et al. Increased prevalence of cardiovascular disease in Type 2 diabetic patients with non-alcoholic fatty liver disease. Diabetic Medicine. 2006; 23: 403–409.
[99]
Kaneto H, Katakami N, Matsuhisa M, Matsuoka T. Role of reactive oxygen species in the progression of type 2 diabetes and atherosclerosis. Mediators of Inflammation. 2010; 2010: 453892.
[100]
Francque SM, van der Graaff D, Kwanten WJ. Non-alcoholic fatty liver disease and cardiovascular risk: Pathophysiological mechanisms and implications. Journal of Hepatology. 2016; 65: 425–443.
[101]
Jiang F, Chen Q, Wang W, Ling Y, Yan Y, Xia P. Hepatocyte-derived extracellular vesicles promote endothelial inflammation and atherogenesis via microRNA-1. Journal of Hepatology. 2020; 72: 156–166.
[102]
Sunbul M, Kivrak T, Durmus E, Akin H, Aydin Y, Ergelen R, et al. Nonalcoholic Steatohepatitis Score is an Independent Predictor of Right Ventricular Dysfunction in Patients with Nonalcoholic Fatty Liver Disease. Cardiovascular Therapeutics. 2015; 33: 294–299.
[103]
Ando W, Yokomori H, Tsutsui N, Yamanouchi E, Suzuki Y, Oda M, et al. Serum matrix metalloproteinase-1 level represents disease activity as opposed to fibrosis in patients with histologically proven nonalcoholic steatohepatitis. Clinical and Molecular Hepatology. 2018; 24: 61–76.
[104]
Pacana T, Cazanave S, Verdianelli A, Patel V, Min H, Mirshahi F, et al. Dysregulated Hepatic Methionine Metabolism Drives Homocysteine Elevation in Diet-Induced Nonalcoholic Fatty Liver Disease. PLoS ONE. 2015; 10: e0136822.
[105]
Siddiqui MS, Fuchs M, Idowu MO, Luketic VA, Boyett S, Sargeant C, et al. Severity of nonalcoholic fatty liver disease and progression to cirrhosis are associated with atherogenic lipoprotein profile. Clinical Gastroenterology and Hepatology. 2015; 13: 1000–1008.e3.
[106]
Männistö VT, Simonen M, Soininen P, Tiainen M, Kangas AJ, Kaminska D, et al. Lipoprotein subclass metabolism in nonalcoholic steatohepatitis. Journal of Lipid Research. 2014; 55: 2676–2684.
[107]
Corey KE, Misdraji J, Gelrud L, Zheng H, Chung RT, Krauss RM. Nonalcoholic steatohepatitis is associated with an atherogenic lipoprotein subfraction profile. Lipids in Health and Disease. 2014; 13: 100.
[108]
Wang X, Pu H, Ma C, Jiang T, Wei Q, Zhang C, et al. Adiponectin abates atherosclerosis by reducing oxidative stress. Medical Science Monitor. 2014; 20: 1792–1800.
[109]
Sasso FC, Pafundi PC, Marfella R, Calabrò P, Piscione F, Furbatto F, et al. Adiponectin and insulin resistance are related to restenosis and overall new PCI in subjects with normal glucose tolerance: the prospective AIRE Study. Cardiovascular Diabetology. 2019; 18: 24.
[110]
Haukeland JW, Damås JK, Konopski Z, Løberg EM, Haaland T, Goverud I, et al. Systemic inflammation in nonalcoholic fatty liver disease is characterized by elevated levels of CCL2. Journal of Hepatology. 2006; 44: 1167–1174.
[111]
Fitzpatrick E, Mitry RR, Quaglia A, Hussain MJ, DeBruyne R, Dhawan A. Serum levels of CK18 M30 and leptin are useful predictors of steatohepatitis and fibrosis in paediatric NAFLD. Journal of Pediatric Gastroenterology and Nutrition. 2010; 51: 500–506.
[112]
Xu X, Lu L, Dong Q, Li X, Zhang N, Xin Y, et al. Research advances in the relationship between nonalcoholic fatty liver disease and atherosclerosis. Lipids in Health and Disease. 2015; 14: 158.
[113]
Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature. 2012; 482: 179–185.
[114]
Rao R. Endotoxemia and gut barrier dysfunction in alcoholic liver disease. Hepatology. 2009; 50: 638–644.
[115]
Lonardo A, Sookoian S, Pirola CJ, Targher G. Non-alcoholic fatty liver disease and risk of cardiovascular disease. Metabolism. 2016; 65: 1136–1150.
[116]
Francque S, Laleman W, Verbeke L, Van Steenkiste C, Casteleyn C, Kwanten W, et al. Increased intrahepatic resistance in severe steatosis: endothelial dysfunction, vasoconstrictor overproduction and altered microvascular architecture. Laboratory Investigation. 2012; 92: 1428–1439.
[117]
VanWagner LB, Wilcox JE, Colangelo LA, Lloyd-Jones DM, Carr JJ, Lima JA, et al. Association of nonalcoholic fatty liver disease with subclinical myocardial remodeling and dysfunction: a population-based study. Hepatology. 2015; 62: 773–783.
[118]
Park HE, Lee H, Choi S, Kim HS, Chung GE. The risk of atrial fibrillation in patients with non-alcoholic fatty liver disease and a high hepatic fibrosis index. Scientific Reports. 2020; 10: 5023.
[119]
Mantovani A, Rigolon R, Pichiri I, Bonapace S, Morani G, Zoppini G, et al. Nonalcoholic fatty liver disease is associated with an increased risk of heart block in hospitalized patients with type 2 diabetes mellitus. PLoS ONE. 2017; 12: e0185459.
[120]
Mantovani A, Pernigo M, Bergamini C, Bonapace S, Lipari P, Valbusa F, et al. Heart valve calcification in patients with type 2 diabetes and nonalcoholic fatty liver disease. Metabolism: Clinical and Experimental. 2015; 64: 879–887.
[121]
Katsiki N, G. Athyros V, P. Mikhailidis D. Abnormal Peri-Organ or Intra-organ Fat (APIFat) Deposition: an Underestimated Predictor of Vascular Risk? Current Vascular Pharmacology. 2016; 14: 432–441.
[122]
Davignon J, Ganz P. Role of endothelial dysfunction in atherosclerosis. Circulation. 2004; 109: III27–III32.
[123]
Marcucci M, Franchi C, Nobili A, Mannucci PM, Ardoino I. Defining Aging Phenotypes and Related Outcomes: Clues to Recognize Frailty in Hospitalized Older Patients. The journals of gerontology. Series A, Biological sciences and medical sciences 2017; 72: 395–402.
[124]
Lenti MV, Pasina L, Cococcia S, Cortesi L, Miceli E, Caccia Dominioni C, et al. Mortality rate and risk factors for gastrointestinal bleeding in elderly patients. European Journal of Internal Medicine. 2019; 61: 54–61.
[125]
Lonardo A, Ballestri S, Guaraldi G, Nascimbeni F, Romagnoli D, Zona S, et al. Fatty liver is associated with an increased risk of diabetes and cardiovascular disease - Evidence from three different disease models: NAFLD, HCV and HIV. World Journal of Gastroenterology. 2016; 22: 9674–9693.
[126]
Abenavoli L, Greco M, Milic N, Accattato F, Foti D, Gulletta E, et al. Effect of Mediterranean Diet and Antioxidant Formulation in Non-Alcoholic Fatty Liver Disease: a Randomized Study. Nutrients. 2017; 9: 870.
[127]
Huber Y, Pfirrmann D, Gebhardt I, Labenz C, Gehrke N, Straub BK, et al. Improvement of non-invasive markers of NAFLD from an individualised, web-based exercise program. Alimentary Pharmacology & Therapeutics. 2019; 50: 930–939.
[128]
Zhang H, He J, Pan L, Ma Z, Han C, Chen C, et al. Effects of Moderate and Vigorous Exercise on Nonalcoholic Fatty Liver Disease: a Randomized Clinical Trial. JAMA Internal Medicine. 2015; 176: 1074–1082.
[129]
Di Francia R, Rinaldi L, Troisi A, Di Benedetto F, Berretta M. Effect of anti-oxidant agents in patients with hepatocellular diseases. European Review for Medical and Pharmacological Sciences. 2016; 19: 3993–3995.
[130]
Di Francia R, Rinaldi L, Cillo M, Varriale E, Facchini G, D’Aniello C, et al. Antioxidant diet and genotyping as tools for the prevention of liver disease. European Review for Medical and Pharmacological Sciences. 2016; 20: 5155–5163.
[131]
Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. 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.
[132]
Romero-Gómez M, Zelber-Sagi S, Trenell M. Treatment of NAFLD with diet, physical activity and exercise. Journal of Hepatology. 2017; 67: 829–846.
[133]
Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, et al. ESC Scientific Document Group. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. European Heart Journal. 2020; 41: 111–188.
[134]
Sheka AC, Adeyi O, Thompson J, Hameed B, Crawford PA, Ikramuddin S. Nonalcoholic Steatohepatitis. The Journal of the American Medical Association. 2020; 323: 1175.
[135]
Loguercio C, Federico A, Tuccillo C, Terracciano F, D’Auria MV, De Simone C, et al. Beneficial effects of a probiotic VSL#3 on parameters of liver dysfunction in chronic liver diseases. Journal of Clinical Gastroenterology. 2005; 39: 540–543.
[136]
Malasanos TH, Stacpoole PW. Biological effects of omega-3 fatty acids in diabetes mellitus. Diabetes Care. 1991; 14: 1160–1179.
[137]
Malaguarnera M, Gargante MP, Malaguarnera G, Salmeri M, Mastrojeni S, Rampello L, et al. Bifidobacterium combined with fructo-oligosaccharide versus lactulose in the treatment of patients with hepatic encephalopathy. European Journal of Gastroenterology & Hepatology. 2010; 22: 199–206.
[138]
Ma Y, Li L, Yu C, Shen Z, Chen L, Li Y. Effects of probiotics on nonalcoholic fatty liver disease: a meta-analysis. World Journal of Gastroenterology. 2013; 19: 6911–6918.
[139]
Gao X, Zhu Y, Wen Y, Liu G, Wan C. Efficacy of probiotics in non-alcoholic fatty liver disease in adult and children: a meta-analysis of randomized controlled trials. Hepatology Research. 2016; 46: 1226–1233.
[140]
Kobyliak N, Abenavoli L, Mykhalchyshyn G, Kononenko L, Boccuto L, Kyriienko D, et al. A Multi-strain Probiotic Reduces the Fatty Liver Index, Cytokines and Aminotransferase levels in NAFLD Patients: Evidence from a Randomized Clinical Trial. Journal of Gastrointestinal and Liver Diseases. 2018; 27: 41–49.
[141]
Sanyal AJ, Chalasani N, Kowdley KV, McCullough A, Diehl AM, Bass NM, et al. Pioglitazone, Vitamin E, or Placebo for Nonalcoholic Steatohepatitis. New England Journal of Medicine. 2010; 362: 1675–1685.
[142]
Oscarsson J, Önnerhag K, Risérus U, Sundén M, Johansson L, Jansson P, et al. Effects of free omega-3 carboxylic acids and fenofibrate on liver fat content in patients with hypertriglyceridemia and non-alcoholic fatty liver disease: a double-blind, randomized, placebo-controlled study. Journal of Clinical Lipidology. 2018; 12: 1390–1403.e4.
[143]
Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, et al. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk. The Journal of the American Medical Association. 2020; 324: 2268.
[144]
Bhatt DL, Miller M, Brinton EA, Jacobson TA, Steg PG, Ketchum SB, et al. REDUCE-it USA. Circulation. 2020; 141: 367–375.
[145]
Abel T, Fehér J, Dinya E, Eldin MG, Kovács A. Safety and efficacy of combined ezetimibe/simvastatin treatment and simvastatin monotherapy in patients with non-alcoholic fatty liver disease. Medical Science Monitor. 2009; 15: MS6–M11.
[146]
Pasternak RC, Smith SC, Bairey-Merz CN, Grundy SM, Cleeman JI, Lenfant C. ACC/AHA/NHLBI Clinical Advisory on the Use and Safety of Statins. Circulation. 2002; 106: 1024–1028.
[147]
Foster T, Budoff MJ, Saab S, Ahmadi N, Gordon C, Guerci AD. Atorvastatin and antioxidants for the treatment of nonalcoholic fatty liver disease: the St Francis Heart Study randomized clinical trial. The American Journal of Gastroenterology. 2011; 106: 71–77.
[148]
Mitsiou E, Boutari C, Kotsis V, Georgianou E, Doumas M, Karagiannis A, et al. Effect of Low (5 mg) vs. High (20–40 mg) Rosuvastatin Dose on 24h Arterial Stiffness, Central Haemodynamics, and Non-Alcoholic Fatty Liver Disease in Patients with Optimally Controlled Arterial Hypertension. Current Vascular Pharmacology. 2018; 16: 393–400.
[149]
Doumas M, Imprialos K, Dimakopoulou A, Stavropoulos K, Binas A, Athyros VG. The Role of Statins in the Management of Nonalcoholic Fatty Liver Disease. Current Pharmaceutical Design. 2018; 24: 4587–4592.
[150]
Athyros VG, Tziomalos K, Gossios TD, Griva T, Anagnostis P, Kargiotis K, et al. Safety and efficacy of long-term statin treatment for cardiovascular events in patients with coronary heart disease and abnormal liver tests in the Greek Atorvastatin and Coronary Heart Disease Evaluation (GREACE) Study: a post-hoc analysis. Lancet. 2010; 376: 1916–1922.
[151]
Dongiovanni P, Petta S, Mannisto V, Mancina RM, Pipitone R, Karja V, et al. Statin use and non-alcoholic steatohepatitis in at risk individuals. Journal of Hepatology. 2015; 63: 705–712.
[152]
Lee JI, Lee HW, Lee KS, Lee HS, Park J. Effects of Statin Use on the Development and Progression of Nonalcoholic Fatty Liver Disease: a Nationwide Nested Case-Control Study. American Journal of Gastroenterology. 2021; 116: 116–124.
[153]
Pastori D, Pani A, Di Rocco A, Menichelli D, Gazzaniga G, Farcomeni A, et al. Statin liver safety in non-alcoholic fatty liver disease: A systematic review and metanalysis. British Journal of Clinical Pharmacology. 2021. (in press)
[154]
Sfikas G, Psallas M, Koumaras C, Imprialos K, Perdikakis E, Doumas M, et al. Prevalence, diagnosis and treatment with 3 different statins of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis in military personnel. Do genetics play a role? Current Vascular Pharmacology. 2020. (in press)
[155]
Nakade Y, Murotani K, Inoue T, Kobayashi Y, Yamamoto T, Ishii N, et al. Ezetimibe for the treatment of non-alcoholic fatty liver disease: a meta-analysis. Hepatology Research. 2017; 47: 1417–1428.
[156]
Simon TG, Corey KE, Cannon CP, Blazing M, Park J, O’Donoghue ML, et al. The nonalcoholic fatty liver disease (NAFLD) fibrosis score, cardiovascular risk stratification and a strategy for secondary prevention with ezetimibe. International Journal of Cardiology. 2018; 270: 245–252.
[157]
Scicali R, Di Pino A, Urbano F, Ferrara V, Marchisello S, Di Mauro S, et al. Analysis of steatosis biomarkers and inflammatory profile after adding on PCSK9 inhibitor treatment in familial hypercholesterolemia subjects with nonalcoholic fatty liver disease: a single lipid center real-world experience. Nutrition, Metabolism and Cardiovascular Diseases. 2021; 31: 869–879.
[158]
Theocharidou E, Papademetriou M, Reklou A, Sachinidis A, Boutari C, Giouleme O. The Role of PCSK9 in the Pathogenesis of Non-alcoholic Fatty Liver Disease and the Effect of PCSK9 Inhibitors. Current Pharmaceutical Design. 2018; 24: 3654–3657.
[159]
Bugianesi E, Gentilcore E, Manini R, Natale S, Vanni E, Villanova N, et al. A randomized controlled trial of metformin versus vitamin E or prescriptive diet in nonalcoholic fatty liver disease. The American Journal of Gastroenterology. 2005; 100: 1082–1090.
[160]
Belfort R, Harrison SA, Brown K, Darland C, Finch J, Hardies J, et al. A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis. The New England Journal of Medicine. 2006; 355: 2297–2307.
[161]
Neuschwander-Tetri BA, Brunt EM, Wehmeier KR, Oliver D, Bacon BR. Improved nonalcoholic steatohepatitis after 48 weeks of treatment with the PPAR-gamma ligand rosiglitazone. Hepatology. 2003; 38: 1008–1017.
[162]
Promrat K, Lutchman G, Uwaifo GI, Freedman RJ, Soza A, Heller T, et al. A pilot study of pioglitazone treatment for nonalcoholic steatohepatitis. Hepatology. 2004; 39: 188–196.
[163]
Mantovani A, Byrne CD, Scorletti E, Mantzoros CS, Targher G. Efficacy and safety of anti-hyperglycaemic drugs in patients with non-alcoholic fatty liver disease with or without diabetes: an updated systematic review of randomized controlled trials. Diabetes & Metabolism. 2020; 46: 427–441.
[164]
Morgillo F, Fasano M, Della Corte CM, Sasso FC, Papaccio F, Viscardi G, et al. Results of the safety run-in part of the METAL (METformin in Advanced Lung cancer) study: a multicentre, open-label phase i-II study of metformin with erlotinib in second-line therapy of patients with stage IV non-small-cell lung cancer. ESMO Open. 2017; 2: e000132.
[165]
Salvatore T, Pafundi PC, Morgillo F, Di Liello R, Galiero R, Nevola R, et al. Metformin: an old drug against old age and associated morbidities. Diabetes Research and Clinical Practice. 2020; 160: 108025.
[166]
Cosentino F, Grant PJ, Aboyans V, Bailey CJ, Ceriello A, Delgado V, et al. 2019 ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD. European Heart Journal. 2020; 41: 255–323.
[167]
Musso G, Cassader M, Paschetta E, Gambino R. Thiazolidinediones and Advanced Liver Fibrosis in Nonalcoholic Steatohepatitis. JAMA Internal Medicine. 2017; 177: 633.
[168]
Tsapas A, Karagiannis T, Avgerinos I, Matthews DR, Bekiari E. Comparative Effectiveness of Glucose-Lowering Drugs for Type 2 Diabetes. Annals of Internal Medicine. 2021; 174: 141.
[169]
Palmiero G, Cesaro A, Vetrano E, Pafundi PC, Galiero R, Caturano A, et al. Impact of SGLT2 Inhibitors on Heart Failure: From Pathophysiology to Clinical Effects. International Journal of Molecular Sciences. 2021; 22: 5863.
[170]
Yan J, Yao B, Kuang H, Yang X, Huang Q, Hong T, et al. Liraglutide, Sitagliptin, and Insulin Glargine Added to Metformin: the Effect on Body Weight and Intrahepatic Lipid in Patients with Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease. Hepatology. 2019; 69: 2414–2426.
[171]
Hosui A, Tatsumi T, Hikita H, Saito Y, Hiramatsu N, Tsujii M, et al. Signal transducer and activator of transcription 5 plays a crucial role in hepatic lipid metabolism through regulation of CD36 expression. Hepatology Research. 2017; 47: 813–825.
[172]
Cui J, Philo L, Nguyen P, Hofflich H, Hernandez C, Bettencourt R, et al. Sitagliptin vs. placebo for non-alcoholic fatty liver disease: a randomized controlled trial. Journal of Hepatology. 2016; 65: 369–376.
[173]
Fukuhara T, Hyogo H, Ochi H, Fujino H, Kan H, Naeshiro N, et al. Efficacy and safety of sitagliptin for the treatment of nonalcoholic fatty liver disease with type 2 diabetes mellitus. Hepato-Gastroenterology. 2014; 61: 323–328.
[174]
Khoo J, Hsiang JC, Taneja R, Koo S, Soon G, Kam CJ, et al. Randomized trial comparing effects of weight loss by liraglutide with lifestyle modification in non‐alcoholic fatty liver disease. Liver International. 2019; 39: 941–949.
[175]
Bizino MB, Jazet IM, de Heer P, van Eyk HJ, Dekkers IA, Rensen PCN, et al. Placebo-controlled randomised trial with liraglutide on magnetic resonance endpoints in individuals with type 2 diabetes: a pre-specified secondary study on ectopic fat accumulation. Diabetologia. 2020; 63: 65–74.
[176]
Petit J, Cercueil J, Loffroy R, Denimal D, Bouillet B, Fourmont C, et al. Effect of Liraglutide Therapy on Liver Fat Content in Patients with Inadequately Controlled Type 2 Diabetes: the Lira-NAFLD Study. The Journal of Clinical Endocrinology and Metabolism. 2017; 102: 407–415.
[177]
Newsome PN, Buchholtz K, Cusi K, Linder M, Okanoue T, Ratziu V, et al. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. New England Journal of Medicine. 2021; 384: 1113–1124.
[178]
Armstrong MJ, Gaunt P, Aithal GP, Barton D, Hull D, Parker R, et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet. 2016; 387: 679–690.
[179]
Honda Y, Imajo K, Kato T, Kessoku T, Ogawa Y, Tomeno W, et al. The Selective SGLT2 Inhibitor Ipragliflozin Has a Therapeutic Effect on Nonalcoholic Steatohepatitis in Mice. PLoS ONE. 2016; 11: e0146337.
[180]
Aso Y, Kato K, Sakurai S, Kishi H, Shimizu M, Jojima T, et al. Impact of dapagliflozin, an SGLT2 inhibitor, on serum levels of soluble dipeptidyl peptidase-4 in patients with type 2 diabetes and non-alcoholic fatty liver disease. International Journal of Clinical Practice. 2019; 73: e13335.
[181]
Shimizu M, Suzuki K, Kato K, Jojima T, Iijima T, Murohisa T, et al. Evaluation of the effects of dapagliflozin, a sodium-glucose co-transporter-2 inhibitor, on hepatic steatosis and fibrosis using transient elastography in patients with type 2 diabetes and non-alcoholic fatty liver disease. Diabetes, Obesity and Metabolism. 2019; 21: 285–292.
[182]
Kuchay MS, Krishan S, Mishra SK, Farooqui KJ, Singh MK, Wasir JS, et al. Effect of Empagliflozin on Liver Fat in Patients with Type 2 Diabetes and Nonalcoholic Fatty Liver Disease: a Randomized Controlled Trial (E-LIFT Trial). Diabetes Care. 2018; 41: 1801–1808.
[183]
Kahl S, Gancheva S, Straßburger K, Herder C, Machann J, Katsuyama H, et al. Empagliflozin Effectively Lowers Liver Fat Content in well-Controlled Type 2 Diabetes: a Randomized, Double-Blind, Phase 4, Placebo-Controlled Trial. Diabetes Care. 2020; 43: 298–305.
[184]
Inoue M, Hayashi A, Taguchi T, Arai R, Sasaki S, Takano K, et al. Effects of canagliflozin on body composition and hepatic fat content in type 2 diabetes patients with non‐alcoholic fatty liver disease. Journal of Diabetes Investigation. 2019; 10: 1004–1011.
[185]
Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nature Medicine. 2018; 24: 908–922.
[186]
Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM, Goodman Z, et al. REGENERATE Study Investigators. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2019; 394: 2184–2196.
[187]
Siddiqui MS, Van Natta ML, Connelly MA, Vuppalanchi R, Neuschwander-Tetri BA, Tonascia J, et al. Impact of obeticholic acid on the lipoprotein profile in patients with non-alcoholic steatohepatitis. Journal of Hepatology. 2020; 72: 25–33.
[188]
Marfella R, Sasso FC, Cacciapuoti F, Portoghese M, Rizzo MR, Siniscalchi M, et al. Tight glycemic control may increase regenerative potential of myocardium during acute infarction. The Journal of Clinical Endocrinology and Metabolism. 2012; 97: 933–942.
[189]
Sasso FC, Rinaldi L, Lascar N, Marrone A, Pafundi PC, Adinolfi LE, et al. Role of Tight Glycemic Control during Acute Coronary Syndrome on CV Outcome in Type 2 Diabetes. Journal of Diabetes Research. 2018; 2018: 3106056.
[190]
Sasso FC, Pafundi PC, Simeon V, De Nicola L, Chiodini P, Galiero R, et al. Efficacy and durability of multifactorial intervention on mortality and MACEs: a randomized clinical trial in type-2 diabetic kidney disease. Cardiovascular Diabetology. 2021; 20: 145.
[191]
Loomba R, Morgan E, Watts L, Xia S, Hannan LA, Geary RS, et al. Novel antisense inhibition of diacylglycerol O-acyltransferase 2 for treatment of non-alcoholic fatty liver disease: a multicentre, double-blind, randomised, placebo-controlled phase 2 trial. The Lancet Gastroenterology & Hepatology. 2020; 5: 829–838.
[192]
Fakhry TK, Mhaskar R, Schwitalla T, Muradova E, Gonzalvo JP, Murr MM. Bariatric surgery improves nonalcoholic fatty liver disease: a contemporary systematic review and meta-analysis. Surgery for Obesity and Related Diseases. 2019; 15: 502–511.
[193]
Ammar W, Basset HA, AL Faramawy A, Hegazy T, Sharaf Y. Bariatric surgery and cardiovascular outcome. The Egyptian Heart Journal. 2020; 72: 67.
[194]
Albaugh VL, Kindel TL, Nissen SE, Aminian A. Cardiovascular Risk Reduction Following Metabolic and Bariatric Surgery. Surgical Clinics of North America. 2021; 101: 269–294.
Share
Back to top