Next Article in Journal
SARS-CoV-2 Renal Impairment in Critical Care: An Observational Study of 42 Cases (Kidney COVID)
Previous Article in Journal
Hepatitis C: A Pharmacological Therapeutic Update
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interplay between Heart Disease and Metabolic Steatosis: A Contemporary Perspective

by
Mohammad Said Ramadan
1,
Vincenzo Russo
2,3,
Gerardo Nigro
2,3,
Emanuele Durante-Mangoni
1,4,* and
Rosa Zampino
4,5
1
Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
2
Department of Translational Medical Sciences, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy
3
Cardiology Unit, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy
4
Infectious and Transplant Medicine Unit, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy
5
Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(8), 1569; https://doi.org/10.3390/jcm10081569
Submission received: 12 March 2021 / Revised: 26 March 2021 / Accepted: 3 April 2021 / Published: 8 April 2021
(This article belongs to the Section Endocrinology & Metabolism)

Abstract

:
The liver-heart axis is a growing field of interest owing to rising evidence of complex bidirectional interplay between the two organs. Recent data suggest non-alcoholic fatty liver disease (NAFLD) has a significant, independent association with a wide spectrum of structural and functional cardiac diseases, and seems to worsen cardiovascular disease (CVD) prognosis. Conversely, the effect of cardiac disease on NAFLD is not well studied and data are mostly limited to cardiogenic liver disease. We believe it is important to further investigate the heart-liver relationship because of the tremendous global health and economic burden the two diseases pose, and the impact of such investigations on clinical decision making and management guidelines for both diseases. In this review, we summarize the current knowledge on NAFLD diagnosis, its systemic manifestations, and associations with CVD. More specifically, we review the pathophysiological mechanisms that govern the interplay between NAFLD and CVD and evaluate the relationship between different CVD treatments and NAFLD progression.

1. Introduction

Non-Alcoholic Fatty Liver Disease (NAFLD) is currently thought to cause most cases of chronic liver disease (CLD) worldwide, accounting for as high as 75% of them in some studies [1]. NAFLD is currently recognized as the hepatic clinico-pathological manifestation of metabolic syndrome (MetS)—which also includes abdominal obesity, arterial hypertension, atherogenic dyslipidemia, and reduced insulin sensitivity, all of which were subsequently shown to be closely associated with NAFLD [2,3,4]. The prevalence of NAFLD is on the rise, with variable estimations ranging from 20% to 50% among adults in Western countries [5,6,7]. This rise should not be surprising, due to NAFLD association with MetS, and the current growing epidemics of MetS in the West [8]. Further strengthening the association with MetS, it was shown that NAFLD prevalence rises to 70–75% among individuals with type 2 diabetes (T2DM) and up to 95–99% in those with obesity [9,10]. Although NAFLD was initially identified as a fatty liver disease arising in the absence of significant alcohol intake, it has subsequently been increasingly correlated to a glucose and lipid metabolic derangement. Accordingly, an expert panel recently proposed to change nomenclature from NAFLD to ‘metabolic associated fatty liver disease’ (MAFLD) [11]. The relationship of NAFLD with visceral obesity and alterations in glucose (insulin resistance, diabetes mellitus) and lipid (hypertriglyceridemia, low HDL cholesterol, hypercholesterolemia) [12] metabolism implicates this disease in a large range of clinical conditions relevant not only to liver disease, but also cardiovascular disease (CVD), diabetes, and cancer, making NAFLD/MALFD an emerging topic in clinical medicine.

2. NAFLD Diagnosis: Current Approaches

NAFLD is a broad term comprising a wide spectrum of liver diseases, from simple steatosis or liver fat accumulation—which usually occurs early in the disease course—to more advanced stages of hepatic disease, including non-alcoholic steatohepatitis or NASH, cirrhosis, and liver cancer. Simple steatosis can progress to inflammation and necrosis (i.e., to NASH), and subsequently evolve into cirrhosis and hepatocellular carcinoma [13]. The diagnosis of NAFLD is that of exclusion and requires identification of hepatic steatosis by histology (defined as liver fat infiltration comprising >5% of hepatocytes) or imaging (bright liver echo-pattern), and exclusion of secondary causes of CLD, such as alcohol (<30 g/day for men and <20 g/day for women), congenital or autoimmune diseases, viruses, and hepatotoxic drugs [14].
The diagnosis of NAFLD can be achieved by several techniques. Although liver biopsy and demonstration of steatosis and other histology hallmarks remain the gold standard for NAFLD diagnosis, other less-invasive techniques such as ultrasonography (US), computed tomography (CT), and magnetic resonance imaging are widely utilized clinically because of a better safety profile and availability, lesser cost, and good sensitivity and specificity compared to the gold standard [13,15,16]. Moreover, several European associations, including the European Association for the Study of the Liver, recommend US as the first-line diagnostic procedure for NAFLD in most patients [13]. However, accounting for the limitations of each modality is crucial for proper NAFLD diagnosis. For instance, the sensitivity of US for NAFLD decreases when <30% of hepatocytes are involved, but this modality may also lose sensitivity in morbidly obese individuals [15,16].
Several newer noninvasive approaches have been investigated for the diagnosis of NAFLD, and some of them have been introduced in clinical practice (Table 1).
The fatty liver index (FLI) [22] is calculated based on levels of triglycerides, body mass index (BMI), waist circumference, and γ-glutamyl-transpeptidase. A FLI < 30 rules out NAFLD whereas a FLI ≥ 60 rules in hepatic steatosis. Scores between 30 and 59 are considered undetermined. The ultrasound fatty liver index (US-FLI) [29] is a different score based on US parameters: a score > 2 suggests the presence of steatosis. Another accurate method to quantify hepatic steatosis is represented by assessment of the controlled attenuation parameter (CAP) on hepatic Fibroscan [30]. In NAFLD, Fibroscan has also been used to assess liver stiffness, a measure of liver fibrosis, by transient elastography (TE). The NAFLD Fat Score is another score that can be used to predict NAFLD and liver fat content (AUROC = 0.88) [23]. It is based on presence of MetS, T2DM, fasting serum (fs) insulin, fs-aspartate aminotransferase (AST), and AST/alanine aminotransferase ratio.
Staging of fibrosis in NAFLD can also be accurately estimated using the NAFLD fibrosis score (NFS) [27] calculated on the combination of several parameters: age, BMI, altered glucose metabolism, AST/ALT ratio, platelet count, and albumin levels. Significant liver fibrosis (equivalent to F3-F4 fibrosis on liver biopsy) is highly suspected when the NFS is >0.675. This measurement needs a diagnosis of NAFLD done with other tests/assays. Two other methods have been used to assess probability of cirrhosis in NAFLD, including the AST to Platelet ratio index (APRI test) [31,32] and the FIB-4 test [25,33].

3. NAFLD as a Systemic Disorder

As numerous compelling evidence shows NAFLD is a systemic disease, rather than confined to the liver, the increasing clinical and economic burden of NAFLD stems from both hepatic and extrahepatic complications. The hepatic complications of NAFLD include NASH, cirrhosis, and hepatocellular carcinoma, and hence it currently represents the second main indication for liver transplantation, projected to become the main one over the next ten years [34,35]. NAFLD affects many other systems and has been shown to increase the risk of developing a number of diseases, including many cardiovascular diseases (CVD), as well as T2DM, chronic kidney disease, and colorectal and other cancers [36,37,38,39,40,41].
CVDs represent the main cause of mortality in patients with NAFLD, accounting for 40–45% of total deaths, followed by extrahepatic cancers (20%) and liver-related complications (10%) [36,42,43,44,45].

4. NAFLD and Cardiovascular Disease: Current Understanding

The relationship between NAFLD and CVD is complex (Figure 1). For instance, NAFLD is associated with many CVDs and the total CVD risk is almost double in patients with NAFLD compared to those without [46]. Moreover, as both conditions share many risk factors, most notably T2DM, dyslipidemia, and obesity, it could be conferred that the same factors account for both diseases and the association being merely a result of these common risk factors [47,48]. However, there is ample evidence showing that a direct mechanistic relationship exists between NAFLD and CVD, and that the former is an independent risk factor for the latter [36,41,42,46,48,49,50]. Due to the association with CVD, recent European clinical practice guidelines mandate screening of the CV system in all patients with NAFLD, at least by detailed risk factor assessment and a comprehensive clinical exam [13].
A number of studies suggest NAFLD association with subclinical and clinical CVD, both closely related to atherosclerosis and inflammation. Subclinical CVD is commonly inferred from carotid intima-media thickening (CIMT), increased coronary calcium score (CAC), and abdominal aortic calcification (AAC), whereas clinical CVD includes most commonly ischemic heart disease (IHD) and atrial fibrillation (AF) [41]. In one metanalysis with 3497 subjects, NAFLD was found to be significantly associated with CIMT and NAFLD patients demonstrated a 13% increase in CIMT compared to controls [51]. Similarly, in a large cohort study with 8020 adult men without carotid atherosclerosis (CA) at baseline followed up for eight years, NAFLD regression was associated with a decreased risk of subclinical CA development (HR 0.82) compared to those with persistent NAFLD [52]. The authors explained this association by metabolic factors, which could mediate the effect of NAFLD [52].
Several studies demonstrated an association between NAFLD and CAC, thus implying an increased risk of coronary events in patients with NAFLD [53,54,55,56,57,58,59,60]. In one meta-analysis of 16 cross-sectional studies with 16,433 NAFLD patients and 41,717 controls, it was shown that NAFLD was significantly associated with CAC > 0 and CAC > 100 independent of traditional risk factors [61]. In a second study on 4731 participants with no history of CVD or liver disease and approximately 4 years of follow-up, NAFLD was shown to be associated with CAC development independent of metabolic or CV risk factors [62]. These associations were further established in a recent large meta-analysis on 85,395 participants (including 29,493 patients with NAFLD), where NAFLD was associated with an increased risk of CIMT (OR 1.74), arterial stiffness (OR 1.56), CAC (OR 1.4), and endothelial dysfunction (OR 3.73) [42].
Clinical CVD, most importantly IHD, is the leading cause of death in patients with NAFLD [45,63]. NAFLD patients seem to be at a higher risk of developing IHD [56]. In one study with 445 patients, high risk coronary plaques—assessed by coronary CT angiography—were more likely to occur in patients with NAFLD compared to controls (59.3% vs. 19.0%, respectively) [56]. Similar results were observed in other studies, which concluded higher prevalence of coronary plaques among NAFLD patients independent of other metabolic risk factors [64,65,66]. A striking finding was the observation that higher NAFLD severity directly correlated with higher CVD risk [67]. In one study involving 360 patients with ST-segment elevation myocardial infarction (STEMI), higher NAFLD grade (as assessed by liver US) correlated with higher in-hospital and three-year mortality rates, and the authors concluded recommending NAFLD screening in patients with STEMI [68]. In another study examining 186 non-diabetic patients with STEMI undergone percutaneous coronary intervention (PCI), NAFLD was associated with higher odds of myocardial reperfusion failure, no ST-segment resolution, and higher in hospital major adverse cardiac events (MACE) [69].
AF is the most common sustainable cardiac arrhythmia [36]; the health and economic burden of atrial fibrillation is mainly due to its association with stroke and increased mortality [50]. AF was shown in many studies to be associated with NAFLD [70,71,72,73]. For instance, Targher et al. demonstrated, over a 10-year follow-up, an increased incidence of AF in patients with T2DM and NAFLD compared to patients with T2DM without NAFLD (OR 4.49) [72]. These results were confirmed by a cohort study by Käräjämäki et al. involving 958 hypertensive patients, where they found higher odds of developing AF in patients with NAFLD, independent of T2DM (adjusted OR 1.88) [74].
Several studies suggested NAFLD association with other CVD, including structural and functional cardiac dysfunction and heart valve sclerosis [75]. Some studies reported that NAFLD patients had thicker left ventricular walls during both systole and diastole [76], lower early diastolic relaxation velocity and higher left ventricle (LV) filling pressures [77] and greater LV myocardial mass [78] than patients without NAFLD. In one study assessing cardiac function of 606 T2DM patients according to NAFLD presence, LV diastolic dysfunction was significantly more prevalent in those with NAFLD (59.7% vs. 49%), with stronger association in patients with liver fibrosis independent of insulin resistance and other metabolic risk factors [79]. Moreover, several studies indicated an association between NAFLD and valvular heart disease [80], most commonly aortic valve sclerosis (AVS). In one study with 2212 participants, it was found that patients with NAFLD had 32% higher odds of having AVS than patients without NAFLD, after adjusting for major confounders [81]. This finding was also replicated in T2DM patients with no history of liver or cardiac disease [82,83].

5. NAFLD and Cardiovascular Disease: Pathogenic Mechanisms

Data on possible mechanisms underlying the interplay between liver and heart in NAFLD are limited and still not completely understood (Figure 2) [84]. Endothelial damage was demonstrated to be an early step towards atherosclerosis, and a number of studies showed that NAFLD is associated with endothelial dysfunction [85,86]. Possible mechanisms for endothelial dysfunction include increased levels of asymmetric dimethyl arginine (ADMA), which is an endogenous antagonist of nitric oxide synthase, which in turn is protective against CVD [87,88]. Moreover, levels of endothelial progenitor cells (EPCs)—which participate in endothelial repair—were shown to be reduced, and attenuated in function, in NAFLD [89].
In addition, NAFLD is associated with increased oxidative stress and altered lipid profile, which in turn may contribute to CVD development [84,90]. It was shown that patients with NAFLD have elevated levels of homocysteine [91,92,93]. Homocysteine, in turn, triggers oxidative stress and endothelial dysfunction, impairs redox status, and enhances platelet activation, all contributing to CV effects [94]. NAFLD severity associates with the extent of serum lipid profile changes, with greater alterations in NASH [84]. Patients with NAFLD have abnormally high triglyceride (TG), very low-density lipoprotein (VLDL), and low-density lipoprotein (LDL) levels, as well as decreased high-density lipoprotein (HDL) levels [95,96,97], a combination resulting in more atherogenic lipid ratios [98]. Similar but more pronounced reduction in HDL and increase in LDL levels were found in NASH, as compared to NAFLD, which hints at similar CV risks [99]. In addition to their levels, lipid composition, subclass, surface apolipoproteins, and phospholipids are important mediators of CV risk [100] and NAFLD severity [97]. Individuals with NASH were shown to have lower levels of larger LDL (LDL1) but increased small dense LDL (LDL3 and LDL4), as compared to those with NAFLD [97].
Insulin resistance, which associates with both CVD and NAFLD, comprises the ability of adipocytes to store fat, resulting in release of free fatty acids into the circulation, and thus exposes the liver to higher levels of free fatty acids [101]. These fatty acids are taken up by the hepatocytes by FATP5 and CD36 receptors, which also get upregulated in obesity [102,103], leading to higher triglyceride synthesis and impaired insulin signalling [104]. Hepatocytes contribute themselves to steatosis accumulation by de novo lipogenesis (DNL), the enzymes of which are upregulated by insulin and glucose [105]. DNL can contribute as much as 25% of the hepatic lipid stores and is believed to be an important contributor of NAFLD development [106,107]. Specifically, 40% of the lipid that builds up in steatosis derive from dietary sugar and fat with the remaining 60% deriving from dysfunctional adipose tissue [106].
The liver is an essential player in systemic inflammation and immunity. For instance, it harbors the largest number of resident macrophages along with a high number of several other immune cells [108]. It also generates and interacts with various inflammatory hormones/cytokines secreted from places like visceral adipose tissue, macrophages, and endothelial cells, which are associated with CVD initiation and progression [109,110]. Higher pro-inflammatory states associate with worse metabolic, histological, and hemodynamic features in NAFLD [111]. C-reactive protein (CRP), an inflammatory marker mostly produced in the liver, has been implicated as an independent risk factor for CVD in many studies [112]. Compared to patients with no steatosis, patients with NASH were found to have significantly higher levels of CRP, fibrinogen, and plasminogen activator inhibitor-1 (PAI-1) activity. NASH severity on histology correlated independently of other variables with levels of these markers [113].
Diseased liver was shown to secrete increased levels of cytokines systemically, which are associated with CVD and drive systemic inflammation [84,114]. Indeed, patients with NAFLD were shown to have increased markers of systemic inflammation including interleukin 6, high sensitivity CRP, interleukin 1b, tumor necrosis factor [TNF]-α, chemokine [C-C motif] ligand 3, soluble intracellular adhesion molecule 1, and macrophage phenotype 1/2 ratio [M1/M2] [111,115,116,117,118]. Systemic inflammation increases CVD risk by causing endothelial dysfunction and oxidative stress, and altering vascular tone [114,119]. In one study, NASH patients were found to have increased expression of several genes linked to inflammation and plaque formation, as compared to simple steatosis [120].
Overall, these studies show that the observed systemic inflammation and immune dysfunction are directly contributed to by the liver, and this chronic inflammation drives CVD initiation and/or progression.
Similarly, the liver is a very important, if not exclusive, source of both pro- and anticoagulant factors [121]. Several studies demonstrated increased pro-coagulant and decreased anticoagulant factor production in NAFLD [122,123], which is one mechanism postulated to link CVD and NAFLD. Kotronen et al. demonstrated that the activity of factors VIII, IX, XI, and XII were increased in patients with NAFLD, independent of other variables like BMI and age, as compared to those without NAFLD [122].
Furthermore, several studies investigated plasminogen inhibitor activator 1 (PAI-1) level, which is secreted by the liver and functions to inhibit fibrinolysis and thus activate the coagulation system, leading to an increased atherosclerotic state [123]. Liver steatosis was shown to be an independent determinant of PAI-1 levels, with levels of this factor progressively rising with increasing degrees of steatosis [124]. Another study by Verrijken et al. showed that fibrinogen, factor VII, von Willebrand factor and PAI-1 were increased, while antithrombin III was decreased. Interestingly, PAI-1 levels were significantly correlated with histological severity of NAFLD [125]. In another study by Song et al., the authors suggested a direct association between PAI-1 levels and coronary heart disease risk [126]. Increased pro-coagulant and decreased anti-coagulant factors boost risk for atherosclerosis and CVD [127].
Recent evidences point to a significant role of the intestinal microbiota in NAFLD pathogenesis and progression. In their study, Loomba et al. analyzed the gut microbiota of 86 patients with biopsy-proven NAFLD and demonstrated a positive association of specific bacterial species, such as Proteobacteria phylum, with progression of NAFLD from mild/moderate to advanced fibrosis. They also used data to construct a robust model that was able to accurately (AUROC 0.936) diagnose advanced fibrosis [128]. Other studies similarly demonstrated increased Proteobacteria phylum with increased Enterobacteriaceae and decreased Rikenellaceae and Ruminococcaceae families in patients with NAFLD or NASH compared with healthy controls [129]. Moreover, the gut microbiome can systemically secrete several molecules such as secondary bile-acids, trimethylamine (TMA), and short chain fatty acids [84]. These molecules were demonstrated to affect energy balance and insulin sensitivity and thus could affect both NAFLD and CVD [130]. A clearer link is found between CVD, NAFLD, and trimethylamine-N-oxide, which is thought to be a pro-atherogenic compound [131,132]. Furthermore, incretins such as glucagon like peptide 1 (GLP-1) are hormones secreted by the gastrointestinal tract and function in regulating postprandial glucose metabolism [84]. The cardioprotective effects of GLP-1 are well described [133] and GLP-1 agonists such as exenatide were shown to improve NASH, vessel inflammation, and plaque size [134]. Despite these results, large discrepancies are found with divergent results for phylum, family, genus, and species across many studies investigating microbiome and NAFLD [129].
Genetic polymorphisms were also studied as predisposing factors for NAFLD, and several alleles were found to be associated with NAFLD and the progression of hepatic fibrosis. In one meta-analysis by Singal et al., a single nucleotide polymorphism—PNPLA3—was associated with increased risk of fibrosis (OR 1.23) and HCC (OR 1.67) in patients with NAFLD [135]. In another study, Liu et al. demonstrated an association between NAFLD and the stage of hepatic fibrosis with TM6SF2 allele, independent of PNPLA3 [136]. Other genetic variants including MBOAT7 and glucokinase regulatory protein were suggested to be associated with increased risk of steatosis, NASH and increased fibrosis stage [137,138]. Much less is known regarding the association of these alleles with CVD. In one twin study investigating heritability of NAFLD and associated CVD, a correlation was established between hepatic steatosis and monozygotic, rather than dizygotic twins, even after adjusting for other possible confounders [139]. However, another twin study failed to demonstrate heritability of NAFLD or its associated CVD [140]. Moreover, in several studies, mutation in TM6SF2 was associated with LDL, VLDL, and TG reduction [141,142,143,144,145]. Indeed, in a recent large exome-wide association study including >300,000 individuals, both polymorphisms in TM6SF2 and PNLPA3 were associated with lower lipid levels and a lower risk of CAD, but an increased risk of fatty liver and T2DM [146]. A finding also confirmed with a 4081 adult cohort followed up for 11.3 years, where PNPLA3 allele was found to be associated with a fourfold increase in the hazard of liver disease-related mortality but reduced risk of death from CVD and overall mortality [147].

6. Nonclinical Experimental Studies on CVD and NAFLD Association

The associations between heart disease and metabolic steatosis were also demonstrated in animal models. In rodents, a high-fat diet resulted in hepatic steatosis and up-regulation of NF-kB, which increased hepatic production of inflammatory cytokines such as IL-6, IL-1β, and TNF-α, along with activation of Kupffer cells and macrophages [148]. The role of TNF-α in NAFLD initiation and progression was also investigated in mice. For that purpose, Kakino et al. analyzed NAFLD progression in TNF-α knockout NAFLD mice models, where they found that these mice significantly regressed steatosis and fibrosis and improved glucose tolerance at 20 weeks, as compared to wild NAFLD mice [149]. Moreover, the effect of IL-1β on steatosis was also studied, where in one study, IL-1 depletion from liver Kupffer cells in obese mice significantly reduced hepatic steatosis, triglyceride levels, and lipogenic enzyme expressions with similar results achieved with utilizing IL-1 receptor antagonists [150].
Moreover, and similar to data from clinical studies, plasma PAI-1 levels in mice with NAFLD were shown to be significantly elevated and associated with the degree of steatosis and hepatic expression of PAI-1 [151]. It was also shown that PAI-1 modulates the development of atherosclerosis in mice [152,153] and that obese mice had around 2-fold higher PAI-1 liver expression than lean mice [154]. Other mechanisms of cardiac disease in NAFLD spectrum includes changes in collagen isoforms expression in the heart. One study demonstrated that cirrhotic rats had increased type 1 collagen content, leading to increased ventricular stiffness and diastolic dysfunction, as compared to sham animals [155].
The effect of some medications was also investigated in animals with steatosis. In one such study, angiotensin-II receptor blocker (ARB) Losartan’s effect was studied in mice models of NASH. Results showed that after eight weeks of treatment, ARB inhibited liver fibrosis development along with suppression of activated hepatic stellate cells and TGF-β and also suppression of TLR4 and NF-κB expressions [156]. In another study, the effect of nonsteroidal anti-inflammatory drugs (NSAIDs) was investigated in mice NASH models. After six months of treatment, mice who received Aspirin had significantly reduced steatosis, and all four tested NSAIDs (meclofenamate, mefenamate, flufenamate, and aspirin) were able to stop steatosis progress to steatohepatitis [157].

7. Effect of Cardiovascular Diseases and Their Treatments on NAFLD

Emerging data on the interplay between NAFLD and CVD show a complex two-way relationship between the two conditions [158,159,160,161]. On one hand, and as previously stated, NAFLD was suggested to be a major risk factor for many CVD [44,45,52,66]. On the other hand, other studies investigated the reverse relationship: the effect of cardiac pathology, mainly acute and chronic heart failure, on hepatic disease [158,159,160,161]. Liver disease caused by heart failure includes congestive hepatopathy, or chronic hepatic passive congestion, and cardiogenic ischemic hepatitis, a rapid and acute cardiogenic liver injury [159].
Due to the strong association between NAFLD and CVD, and the increased risk of mortality from CV events of patients with NAFLD, a number of primary and secondary prevention strategies were recommended by the American College of Cardiology, the American Heart Association, the European Association for the Study of the Liver, and the Italian Association for the Study of the Liver. Primary prevention strategies for NAFLD also target the traditional risk factors for CVD: healthy dietary pattern, moderate or vigorous exercise, and optimal body weight achievement [13,162,163].
Lifestyle modifications remain a cornerstone in both the treatment of NAFLD and the prevention of CV complications, considering that there is a lack of definitive treatments [164]. For instance, in one prospective study on 293 patients with NASH, lifestyle modifications that are beneficial for CVD (decrease in calorie intake and increase in exercise) achieved regression of fibrosis and resolution of steatohepatitis in 19% and 25% of patients, respectively [165]. To date, a number of drugs have been studied as possible treatments of NAFLD and secondary prevention of CV complications [164]. For example—whose CV benefits are clearly established—were shown to be inversely correlated with NAFLD and liver fibrosis (OR 0.57 and 0.47, respectively) [166]. In another study with 1600 patients over three years, atorvastatin was shown to improve liver function tests and decrease cardiovascular events (from 10 to 3.2 events per 100 patient-years) in NAFLD patients who took it compared to those who did not [167].
Other drugs including metformin, thiazolidinediones (TZD), and aspirin have been investigated as potential NAFLD treatments. In one meta-analysis of randomized controlled trials (RCTs) involving a total of 417 participants, metformin was shown to improve liver function in patients with NAFLD, but failed to significantly improve histological response [168]. Moreover, metformin was shown to be significantly associated with weight loss, improved insulin sensitivity [169] and lipid profiles (decreased LDL and increased HDL levels) in NAFLD patients [170,171], which might in turn decrease the CV risk related to NAFLD.
In another meta-analysis including 8 RCTs and a total of 516 patients evaluating TZD effect on histology of biopsy-proven NASH, TZD treatment (5 RCTs evaluating pioglitazone; 3 evaluating rosiglitazone) was associated with improved fibrosis (OR 3.15) and NASH resolution (OR 3.22)—a trend seen in patients with or without T2DM [172]. However, TZD therapy was associated with weight gain and limb edema [169,172], and because of the short duration of the trials, there was no report of congestive heart failure or increased CV mortality [169]. RCTs on TZD therapy, in general, had small sample sizes, short duration, and evaluated the impact of TZD on histological rather clinical response [172,173].
In a recent prospective cohort study with 361 adults with biopsy-confirmed NAFLD, with nine-year follow-up, daily aspirin use was associated with significantly lower odds for NASH (OR 0.68) and fibrosis (OR 0.54), with greatest benefit with at least four years of aspirin use—an association not seen with other non-aspirin NSAIDs [174]. Furthermore, a cross-sectional study including 11,416 patients showed an inverse correlation between regular aspirin use (defined as ≥15 times in the prior month) and prevalent NAFLD (OR 0.62), although this was limited to older men (>60 years) [175]. Aspirin, and not ibuprofen use, showed association with lower indices of liver fibrosis among adults with chronic viral hepatitis, suspected alcoholic liver disease, and NAFLD [176]. The suggested mechanisms of action of platelets in NAFLD stem from the growing evidence of the active role of platelets in liver disease and inflammation [177,178,179]. Indeed, in a model of viral hepatitis, activated platelets were found to contribute to cytotoxic T-lymphocyte-mediated liver damage [180,181]. Moreover, blocking platelet activity with drugs such as aspirin or clopidogrel hampered T cell influx and subsequent liver damage and tumorigenesis in viral hepatitis [182]. Another recently suggested role for platelet inhibition by aspirin in NAFLD is activation of the PPARδ-AMPK-PGC-1α pathway, which in turn inhibits lipid synthesis and elevates catabolic metabolism, and modulation of mannose receptor and CCR2 in macrophages, all of which are suggested to ameliorate NAFLD and atherosclerosis [183]. Despite the favorable results these studies show, further investigations with larger sample sizes are required to assess the impact of these drugs, among others, on NAFLD inception and progression.
Another interesting topic to be addressed is the effect of newer anti-diabetes and anti-hypertensive drugs on NAFLD inception or progression, with or without concomitant CVD. GLP-1 receptor agonists (GLP-1Ras) and Renin-Angiotensin-Aldosterone System Inhibitors (angiotensin converting enzyme inhibitors (ACEi) and angiotensin receptor blockers (ARBs)) were investigated as potential treatments for NAFLD. Early studies demonstrated decreasing and normalizing AST levels in T2DM patients receiving exenatide with elevated AST levels at baseline, compared to those who did not receive it [184]. Recent meta-analyses showed association of GLP-1Ras, mainly exenatide and liraglutide, with reduced body mass index (BMI) and waist circumference (WC) [185,186], and liver fat fraction [185]. Moreover, in subgroup analyses, patients receiving exenatide had improvements in liver enzyme levels (AST and ALT) [185]. In a recent clinical trial, the LEAN (Liraglutide Efficacy and Action in NASH) by Armstrong et al., 52 overweight patients with NASH were randomized to receive liraglutide or placebo (26 in each group). The results of this small pilot study showed that patients in the experimental treatment arm had 4.3 times higher chance of histologically-proven NASH resolution [187].
SGLT2 inhibitors are another class of diabetes and CVD drugs with recent implications in NAFLD treatment. In one phase III randomized controlled trial by Frías et al., 695 patients with T2DM were randomized to receive exenatide plus placebo, dapagliflozin plus placebo or a combination of the two drugs and followed up for 28 weeks. The results of this study showed that all groups had a decrease in the traditional CV risk factors, such as blood pressure, HbA1c and glucose levels, that were more pronounced and superior in the group receiving both drugs [188]. A recent post hoc analysis of the same study evaluated the effects of the tested therapies on non-invasive markers of hepatic steatosis (FLI, NAFLD Fat Score, FIB-4 index, NAFLD Fibrosis Score and liver enzymes) and found that both drugs had stronger effects than each drug alone in ameliorating markers of hepatic steatosis and fibrosis in patients with T2DM [189]. Another study showed that NAFLD patients with T2DM who received dapagliflozin alone had a significant decrease in CAP, liver stiffness, and AST and GGT levels compared with controls [190]. Other studies also demonstrated the potential of dapagliflozin monotherapy to reduce liver fat assessed by MRI, liver injury biomarkers such as enzyme levels [191], and achieve histological improvement with fibrosis regression [192] in NAFLD patients with T2DM.
RAAS activation was shown to be upregulated in NAFLD [193] and to play a role in development of inflammation and insulin resistance [194], both of which are possible risk factors for NAFLD. Pellusi et al. studied the effect of ACEi and ARBs in an observational cohort of 118 diabetic patients with a median follow-up of 36 months. The authors found that treatment with ACEi or ARBs was associated with decreased histological fibrosis progression [195]. Other studies employing ARBs found improvement in both AST levels and histological stage of patients with NAFLD [196,197]. Moreover, authors investigated the role of selective mineralocorticoid receptor (MR) antagonists, such as spironolactone and eplerenone. Indeed, some clinical trials found that combined low-dose spironolactone plus vitamin E decreased NAFLD liver fat score, an index of steatosis, along with insulin levels and homeostasis model assessment of insulin resistance (HOMA-IR) [198,199]. In addition, eplerenone was suggested to prevent NASH development and improve metabolic abnormalities in mice by inhibiting inflammatory responses in both Kupffer cells and macrophages [199,200].
Beta blockers (BB)—a heterogeneous class of drugs—are commonly used for multiple CVD and other medical indications. BB are known to worsen metabolic parameters: increase weight [201], fasting glucose levels, TG, LDL, and decrease HDL and energy expenditure [202,203,204,205,206,207,208,209] with most of these adverse events observed with the conventional non-selective BB with no intrinsic sympathomimetic activity [210,211]. However, these adverse metabolic side-effects were not shown in newer generation BB with vasodilator properties such as carvedilol and nebivolol [212,213]. The role of BB in NAFLD remains unclear and inconclusive due to scarcity of data. In one study on mice as NASH models, propranolol seemed to enhance liver injury as evident by higher necrosis scores in mice that took it and activate the cell-death pathway by increasing the release of lactate dehydrogenase, FAS-L, and TNF-α [214]. In another study on rats as NAFLD models, carvedilol was shown to improve liver enzymes, lipids, and histology [215].
Bariatric surgery—an effective treatment strategy for severe obesity—has been shown to cause a significant decrease in liver transaminases and histology improvement in NAFLD patients [216]. Moreover, in one prospective study on 109 morbidly obese patients with NASH who underwent bariatric surgery, NASH disappeared in 85% of them and levels of liver transaminases significantly decreased [217]. Moreover, bariatric surgery was shown to improve the traditional CV risk factors. In a systematic review of 73 studies including a total of 19,534 patients with about 58 months of follow-up, 73% of subjects had improvement in T2DM, 65% in hyperlipidemia, and 63% in hypertension [218].
Vitamin E was also studied as a potential treatment for NAFLD due to its anti-oxidative properties. In one meta-analysis of three trials analyzing vitamin E supplementation in 242 patients with NASH, vitamin E use was associated with improved ALT levels, steatosis, lobular inflammation, and ballooning but not fibrosis of the liver [219]. However, vitamin E supplementation was shown in other studies to have no apparent effects on CV outcomes [220].
Other drugs, including obeticholic acid, saroglitazar, and elafibranor are currently being investigated for NAFLD in large clinical trials. Obeticholic acid is a bile acid derivative that can bind to and activate farnesoid X receptors, which in turn can increase insulin sensitivity, decrease hepatic gluconeogenesis, and protect against cholestasis liver injury [221,222]. In one trial on 283 patients with NASH, those taking obeticholic acid were more likely to have improved liver histology than those taking placebo at 72 weeks (RR 1.9) [223].
Elafibranor is a dual PPAR-α/δ agonist—both receptors being implicated in the activation of inflammatory changes within the liver [224,225]. In a preliminary study, Ratziu et al. observed that elafibranor resolved NASH without fibrosis worsening [226]. Saroglitazar is a dual PPARα/γ agonist indicated mainly for the treatment of diabetic dyslipidemia and hypertriglyceridemia not controlled by statins [227]. This drug is currently being investigated as a potential treatment for NAFLD in an ongoing phase 2 trial [224]. Further studies are needed before more stringent recommendations can be done on the use of obeticholic acid, saroglitazar, and elafibranor.
The relationship between CVD treatments and NAFLD can also encompass a potentially detrimental effect of some molecules on liver disease inception or progression. Indeed, several CV drugs have been linked to hepatic steatosis, both microvesicular and macrovesicular. Drugs potentially causing microvesicular steatosis include aspirin, nonsteroidal anti-inflammatory drugs (NSAIDS), and valproic acid, among others, which can lead to lipid accumulation in hepatocytes and subsequent steatosis [228]. On the other hand, drugs implicated in macrovesicular steatosis include amiodarone, a frequently used antiarrhythmic agent with a wide range of hepatic and thyroid adverse events. Up to 30% of patients taking amiodarone can develop elevated liver enzymes, with a proportion of 1–2% developing overt steatohepatitis [228]. Furthermore, a recent meta-analysis concluded that patients taking amiodarone have a higher risk of hepatic adverse events (RR 2.27), even at low doses, as compared to placebo [229].

8. Effect of NAFLD on Progression of CVD

In addition to the association of NAFLD with CVD onset, a role has been hypothesized for NAFLD on the progression of CVD, in terms of development of cardiomyopathy (both ischemic and tachycardia-related cardiomyopathy), heart failure (HF) and worsening HF stage.
Despite the growing evidence showing association of NAFLD with CVD, the relationship is still uncertain for many reasons [48,53]. First, a number of studies, two of which carried out by Kim et al. and Lazo et al., featuring a large number of participants (n = 11,154 and 11,371, respectively), concluded that there was no association between increased CV mortality and NAFLD [230,231]. Second, conclusions of meta-analyses have been questioned because of the variability of the included articles in these studies, mostly related to the different diagnostic modalities utilized for NAFLD diagnosis [232]. Moreover, the observational nature of the individual studies demonstrating association between CVD and NAFLD leaves room for confounding bias; and thus interpreting these results require caution and does not allow to draw definitive causal inferences [43,232].
Lastly, and to our knowledge, no study questioned the risk of developing NAFLD in patients presenting with a first episode of IHD or AF. Meanwhile, articles focusing on liver disease caused by cardiac pathology included only congestive hepatopathy and ischemic hepatitis, both well described in the literature. Moreover, we could not find a study quantifying the de novo incidence of NAFLD in patients with IHD as most articles investigate the reverse relationship: incidence of IHD/CVD in patients with known NAFLD.

9. Conclusions

The association of NAFLD with the CV system is quite complex and incompletely understood. NAFLD is suggested to be implicated in many CVDs and seems to worsen the prognosis of patients with CVD. On the other hand, the effect of CVD on liver pathology is not well studied. No studies to our knowledge investigated a possible relationship between NAFLD inception and progression due to CVD or the incidence of NAFLD in patients with CVD. Moreover, as there is no definitive treatment for NAFLD yet, many CV drugs show promising results in NAFLD treatment both biochemically and histologically with fibrosis regression; others were shown to be associated with increased steatosis. Despite the suggested results, further studies are needed to better understand the two-way liver-heart interplay and the roles of drugs in the pathophysiology and treatment of NAFLD.

Author Contributions

M.S.R. reviewed the literature and drafted the manuscript. E.D.-M. and R.Z. provided the idea of writing this review and gave substantial contributions to the conception, methodology, and writing of this review. V.R. and G.N. helped in literature review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Younossi, Z.M.; Stepanova, M.; Afendy, M.; Fang, Y.; Younossi, Y.; Mir, H.; Srishord, M. Changes in the prevalence of the most common causes of chronic liver diseases in the United States from 1988 to 2008. Clin. Gastroenterol. Hepatol. 2011, 9, 524–530.e1. [Google Scholar] [CrossRef] [PubMed]
  2. Kotronen, A.; Yki-Järvinen, H. Fatty liver: A novel component of the metabolic syndrome. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 27–38. [Google Scholar] [CrossRef] [PubMed]
  3. Sperling, L.S.; Mechanick, J.I.; Neeland, I.J.; Herrick, C.J.; Després, J.P.; Ndumele, C.E.; Vijayaraghavan, K.; Handelsman, Y.; Puckrein, G.A.; Araneta, M.R.; et al. The CardioMetabolic Health Alliance: Working Toward a New Care Model for the Metabolic Syndrome. J. Am. Coll. Cardiol. 2015, 66, 1050–1067. [Google Scholar] [CrossRef] [Green Version]
  4. Marchesini, G.; Brizi, M.; Bianchi, G.; Tomassetti, S.; Bugianesi, E.; Lenzi, M.; McCullough, A.J.; Natale, S.; Forlani, G.; Melchionda, N. Nonalcoholic fatty liver disease: A feature of the metabolic syndrome. Diabetes 2001, 50, 1844–1850. [Google Scholar] [CrossRef] [Green Version]
  5. Bedogni, G.; Miglioli, L.; Masutti, F.; Tiribelli, C.; Marchesini, G.; Bellentani, S. Prevalence of and risk factors for nonalcoholic fatty liver disease: The Dionysos nutrition and liver study. Hepatology 2005, 42, 44–52. [Google Scholar] [CrossRef] [PubMed]
  6. Zelber-Sagi, S.; Nitzan-Kaluski, D.; Halpern, Z.; Oren, R. Prevalence of primary non-alcoholic fatty liver disease in a population-based study and its association with biochemical and anthropometric measures. Liver Int. 2006, 26, 856–863. [Google Scholar] [CrossRef]
  7. Williams, C.D.; Stengel, J.; Asike, M.I.; Torres, D.M.; Shaw, J.; Contreras, M.; Landt, C.L.; Harrison, S.A. Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle-aged population utilizing ultrasound and liver biopsy: A prospective study. Gastroenterology 2011, 140, 124–131. [Google Scholar] [CrossRef]
  8. World Health Organization, Obesity: Preventing and Managing the Global Epidemic; Report of a WHO Consultation, WHO Technical Report Series; 2000; Volume 894, pp. 1–253.
  9. Portillo-Sanchez, P.; Bril, F.; Maximos, M.; Lomonaco, R.; Biernacki, D.; Orsak, B.; Subbarayan, S.; Webb, A.; Hecht, J.; Cusi, K. High Prevalence of Nonalcoholic Fatty Liver Disease in Patients With Type 2 Diabetes Mellitus and Normal Plasma Aminotransferase Levels. J. Clin. Endocrinol. Metab. 2015, 100, 2231–2238. [Google Scholar] [CrossRef]
  10. Leite, N.C.; Salles, G.F.; Araujo, A.L.; Villela-Nogueira, C.A.; Cardoso, C.R. Prevalence and associated factors of non-alcoholic fatty liver disease in patients with type-2 diabetes mellitus. Liver Int. 2009, 29, 113–119. [Google Scholar] [CrossRef]
  11. Eslam, M.; Sanyal, A.J.; George, J. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014. [Google Scholar] [CrossRef] [PubMed]
  12. Vacca, M.; Allison, M.; Griffin, J.L.; Vidal-Puig, A. Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism: Implications in NAFLD. Semin. Liver Dis. 2015, 35, 250–261. [Google Scholar] [CrossRef]
  13. EASL–EASD–EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J. Hepatol. 2016, 64, 1388–1402. [CrossRef] [PubMed]
  14. Ratziu, V.; Bellentani, S.; Cortez-Pinto, H.; Day, C.; Marchesini, G. A position statement on NAFLD/NASH based on the EASL 2009 special conference. J. Hepatol. 2010, 53, 372–384. [Google Scholar] [CrossRef] [Green Version]
  15. Saadeh, S.; Younossi, Z.M.; Remer, E.M.; Gramlich, T.; Ong, J.P.; Hurley, M.; Mullen, K.D.; Cooper, J.N.; Sheridan, M.J. The utility of radiological imaging in nonalcoholic fatty liver disease. Gastroenterology 2002, 123, 745–750. [Google Scholar] [CrossRef]
  16. Wieckowska, A.; McCullough, A.J.; Feldstein, A.E. Noninvasive diagnosis and monitoring of nonalcoholic steatohepatitis: Present and future. Hepatology 2007, 46, 582–589. [Google Scholar] [CrossRef] [PubMed]
  17. Brunt, E.M.; Wong, V.W.S.; Nobili, V.; Day, C.P.; Sookoian, S.; Maher, J.J.; Bugianesi, E.; Sirlin, C.B.; Neuschwander-Tetri, B.A.; Rinella, M.E. Nonalcoholic fatty liver disease. Nat. Rev. Dis. Prim. 2015, 1, 15080. [Google Scholar] [CrossRef] [PubMed]
  18. Park, C.C.; Nguyen, P.; Hernandez, C.; Bettencourt, R.; Ramirez, K.; Fortney, L.; Hooker, J.; Sy, E.; Savides, M.T.; Alquiraish, M.H.; et al. Magnetic Resonance Elastography vs Transient Elastography in Detection of Fibrosis and Noninvasive Measurement of Steatosis in Patients With Biopsy-Proven Nonalcoholic Fatty Liver Disease. Gastroenterology 2017, 152, 598–607. [Google Scholar] [CrossRef] [Green Version]
  19. Hernaez, R.; Lazo, M.; Bonekamp, S.; Kamel, I.; Brancati, F.L.; Guallar, E.; Clark, J.M. Diagnostic accuracy and reliability of ultrasonography for the detection of fatty liver: A meta-analysis. Hepatology 2011, 54, 1082–1090. [Google Scholar] [CrossRef] [Green Version]
  20. Lee, S.S.; Park, S.H.; Kim, H.J.; Kim, S.Y.; Kim, M.-Y.; Kim, D.Y.; Suh, D.J.; Kim, K.M.; Bae, M.H.; Lee, J.Y.; et al. Non-invasive assessment of hepatic steatosis: Prospective comparison of the accuracy of imaging examinations. J. Hepatol. 2010, 52, 579–585. [Google Scholar] [CrossRef]
  21. Ballestri, S.; Nascimbeni, F.; Baldelli, E.; Marrazzo, A.; Romagnoli, D.; Targher, G.; Lonardo, A. Ultrasonographic fatty liver indicator detects mild steatosis and correlates with metabolic/histological parameters in various liver diseases. Metabolism 2017, 72, 57–65. [Google Scholar] [CrossRef] [PubMed]
  22. Bedogni, G.; Bellentani, S.; Miglioli, L.; Masutti, F.; Passalacqua, M.; Castiglione, A.; Tiribelli, C. The Fatty Liver Index: A simple and accurate predictor of hepatic steatosis in the general population. BMC Gastroenterol. 2006, 6, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kotronen, A.; Peltonen, M.; Hakkarainen, A.; Sevastianova, K.; Bergholm, R.; Johansson, L.M.; Lundbom, N.; Rissanen, A.; Ridderstråle, M.; Groop, L.; et al. Prediction of Non-Alcoholic Fatty Liver Disease and Liver Fat Using Metabolic and Genetic Factors. Gastroenterology 2009, 137, 865–872. [Google Scholar] [CrossRef]
  24. Karlas, T.; Petroff, D.; Sasso, M.; Fan, J.-G.; Mi, Y.-Q.; de Lédinghen, V.; Kumar, M.; Lupsor-Platon, M.; Han, K.-H.; Cardoso, A.C.; et al. Individual patient data meta-analysis of controlled attenuation parameter (CAP) technology for assessing steatosis. J. Hepatol. 2017, 66, 1022–1030. [Google Scholar] [CrossRef] [PubMed]
  25. Sterling, R.K.; Lissen, E.; Clumeck, N.; Sola, R.; Correa, M.C.; Montaner, J.S.; Sulkowski, M.; Torriani, F.J.; Dieterich, D.T.; Thomas, D.L.; et al. Development of a simple noninvasive index to predict significant fibrosis in patients with HIV/HCV coinfection. Hepatology 2006, 43, 1317–1325. [Google Scholar] [CrossRef]
  26. Afdhal, N.H.; Bacon, B.R.; Patel, K.; Lawitz, E.J.; Gordon, S.C.; Nelson, D.R.; Challies, T.L.; Nasser, I.; Garg, J.; Wei, L.-J.; et al. Accuracy of Fibroscan, Compared with Histology, in Analysis of Liver Fibrosis in Patients With Hepatitis B or C: A United States Multicenter Study. Clin. Gastroenterol. Hepatol. 2015, 13, 772–779. [Google Scholar] [CrossRef]
  27. Angulo, P.; Hui, J.M.; Marchesini, G.; Bugianesi, E.; George, J.; Farrell, G.C.; Enders, F.; Saksena, S.; Burt, A.D.; Bida, J.P.; et al. The NAFLD fibrosis score: A noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology 2007, 45, 846–854. [Google Scholar] [CrossRef]
  28. Khan, D.A.; Fatima Tuz, Z.; Khan, F.A.; Mubarak, A. Evaluation of diagnostic accuracy of APRI for prediction of fibrosis in hepatitis C patients. J. Ayub Med. Coll. Abbottabad 2008, 20, 122–126. [Google Scholar]
  29. Ballestri, S.; Lonardo, A.; Romagnoli, D.; Carulli, L.; Losi, L.; Day, C.P.; Loria, P. Ultrasonographic fatty liver indicator, a novel score which rules out NASH and is correlated with metabolic parameters in NAFLD. Liver Int. 2012, 32, 1242–1252. [Google Scholar] [CrossRef]
  30. Petta, S.; Wong, V.W.; Cammà, C.; Hiriart, J.B.; Wong, G.L.; Marra, F.; Vergniol, J.; Chan, A.W.; Di Marco, V.; Merrouche, W.; 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. [Google Scholar] [CrossRef] [PubMed]
  31. Lin, Z.H.; Xin, Y.N.; Dong, Q.J.; Wang, Q.; Jiang, X.J.; Zhan, S.H.; Sun, Y.; Xuan, S.Y. 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. [Google Scholar] [CrossRef] [PubMed]
  32. Wai, C.T.; Greenson, J.K.; Fontana, R.J.; Kalbfleisch, J.D.; Marrero, J.A.; Conjeevaram, H.S.; Lok, A.S. A simple noninvasive index can predict both significant fibrosis and cirrhosis in patients with chronic hepatitis C. Hepatology 2003, 38, 518–526. [Google Scholar] [CrossRef] [Green Version]
  33. McPherson, S.; Hardy, T.; Dufour, J.F.; Petta, S.; Romero-Gomez, M.; Allison, M.; Oliveira, C.P.; Francque, S.; Van Gaal, L.; Schattenberg, J.M.; et al. Age as a Confounding Factor for the Accurate Non-Invasive Diagnosis of Advanced NAFLD Fibrosis. Am. J. Gastroenterol. 2017, 112, 740–751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Charlton, M.R.; Burns, J.M.; Pedersen, R.A.; Watt, K.D.; Heimbach, J.K.; Dierkhising, R.A. Frequency and outcomes of liver transplantation for nonalcoholic steatohepatitis in the United States. Gastroenterology 2011, 141, 1249–1253. [Google Scholar] [CrossRef]
  35. Wong, R.J.; Aguilar, M.; Cheung, R.; Perumpail, R.B.; Harrison, S.A.; Younossi, Z.M.; Ahmed, A. Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United States. Gastroenterology 2015, 148, 547–555. [Google Scholar] [CrossRef] [PubMed]
  36. Anstee, Q.M.; Mantovani, A.; Tilg, H.; Targher, G. Risk of cardiomyopathy and cardiac arrhythmias in patients with nonalcoholic fatty liver disease. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 425–439. [Google Scholar] [CrossRef]
  37. Chitturi, S.; Abeygunasekera, S.; Farrell, G.C.; Holmes-Walker, J.; Hui, J.M.; Fung, C.; Karim, R.; Lin, R.; Samarasinghe, D.; Liddle, C.; et al. NASH and insulin resistance: Insulin hypersecretion and specific association with the insulin resistance syndrome. Hepatology 2002, 35, 373–379. [Google Scholar] [CrossRef]
  38. Byrne, C.D.; Targher, G. NAFLD: A multisystem disease. J. Hepatol. 2015, 62, S47–S64. [Google Scholar] [CrossRef] [Green Version]
  39. Targher, G.; Chonchol, M.; Zoppini, G.; Abaterusso, C.; Bonora, E. Risk of chronic kidney disease in patients with non-alcoholic fatty liver disease: Is there a link? J. Hepatol. 2011, 54, 1020–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Targher, G.; Chonchol, M.B.; Byrne, C.D. CKD and nonalcoholic fatty liver disease. Am. J. Kidney Dis. 2014, 64, 638–652. [Google Scholar] [CrossRef] [PubMed]
  41. Adams, L.A.; Anstee, Q.M.; Tilg, H.; Targher, G. Non-alcoholic fatty liver disease and its relationship with cardiovascular disease and other extrahepatic diseases. Gut 2017, 66, 1138–1153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zhou, Y.Y.; Zhou, X.D.; Wu, S.J.; Fan, D.H.; Van Poucke, S.; Chen, Y.P.; Fu, S.W.; Zheng, M.H. Nonalcoholic fatty liver disease contributes to subclinical atherosclerosis: A systematic review and meta-analysis. Hepatol. Commun. 2018, 2, 376–392. [Google Scholar] [CrossRef]
  43. Targher, G.; Byrne, C.D.; Lonardo, A.; Zoppini, G.; Barbui, C. Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: A meta-analysis. J. Hepatol. 2016, 65, 589–600. [Google Scholar] [CrossRef] [Green Version]
  44. Adams, L.A.; Lymp, J.F.; St Sauver, J.; Sanderson, S.O.; Lindor, K.D.; Feldstein, A.; Angulo, P. The natural history of nonalcoholic fatty liver disease: A population-based cohort study. Gastroenterology 2005, 129, 113–121. [Google Scholar] [CrossRef]
  45. Rafiq, N.; Bai, C.; Fang, Y.; Srishord, M.; McCullough, A.; Gramlich, T.; Younossi, Z.M. Long-term follow-up of patients with nonalcoholic fatty liver. Clin. Gastroenterol. Hepatol. 2009, 7, 234–238. [Google Scholar] [CrossRef] [PubMed]
  46. Mahfood Haddad, T.; Hamdeh, S.; Kanmanthareddy, A.; Alla, V.M. Nonalcoholic fatty liver disease and the risk of clinical cardiovascular events: A systematic review and meta-analysis. Diabetes Metab. Syndr. 2017, 11 (Suppl. 1), S209–S216. [Google Scholar] [CrossRef]
  47. Arslan, U.; Yenerçağ, M. Relationship between non-alcoholic fatty liver disease and coronary heart disease. World J. Clin. Cases 2020, 8, 4688–4699. [Google Scholar] [CrossRef] [PubMed]
  48. Ismaiel, A.; Dumitraşcu, D.L. Cardiovascular Risk in Fatty Liver Disease: The Liver-Heart Axis-Literature Review. Front. Med. 2019, 6, 202. [Google Scholar] [CrossRef] [Green Version]
  49. 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 Res. Int. 2015, 2015, 213737. [Google Scholar] [CrossRef] [Green Version]
  50. Ismaiel, A.; Colosi, H.A.; Rusu, F.; Dumitrașcu, D.L. Cardiac Arrhythmias and Electrocardiogram Modifications in Non-Alcoholic Fatty Liver Disease. A Systematic Review. J. Gastrointest. Liver Dis. 2019, 28, 483–493. [Google Scholar] [CrossRef] [PubMed]
  51. Sookoian, S.; Pirola, C.J. Non-alcoholic fatty liver disease is strongly associated with carotid atherosclerosis: A systematic review. J. Hepatol. 2008, 49, 600–607. [Google Scholar] [CrossRef] [PubMed]
  52. Sinn, D.H.; Cho, S.J.; Gu, S.; Seong, D.; Kang, D.; Kim, H.; Yi, B.K.; Paik, S.W.; Guallar, E.; Cho, J.; et al. Persistent Nonalcoholic Fatty Liver Disease Increases Risk for Carotid Atherosclerosis. Gastroenterology 2016, 151, 481–488. [Google Scholar] [CrossRef] [PubMed]
  53. Wu, R.; Hou, F.; Wang, X.; Zhou, Y.; Sun, K.; Wang, Y.; Liu, H.; Wu, J.; Zhao, R.; Hu, J. Nonalcoholic Fatty Liver Disease and Coronary Artery Calcification in a Northern Chinese Population: A Cross Sectional Study. Sci. Rep. 2017, 7, 9933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Wong, V.W.; Wong, G.L.; Yeung, J.C.; Fung, C.Y.; Chan, J.K.; Chang, Z.H.; Kwan, C.T.; Lam, H.W.; Limquiaco, J.; Chim, A.M.; et al. Long-term clinical outcomes after fatty liver screening in patients undergoing coronary angiogram: A prospective cohort study. Hepatology 2016, 63, 754–763. [Google Scholar] [CrossRef] [Green Version]
  55. Sung, K.C.; Wild, S.H.; Kwag, H.J.; Byrne, C.D. Fatty liver, insulin resistance, and features of metabolic syndrome: Relationships with coronary artery calcium in 10,153 people. Diabetes Care 2012, 35, 2359–2364. [Google Scholar] [CrossRef] [Green Version]
  56. Puchner, S.B.; Lu, M.T.; Mayrhofer, T.; Liu, T.; Pursnani, A.; Ghoshhajra, B.B.; Truong, Q.A.; Wiviott, S.D.; Fleg, J.L.; Hoffmann, U.; et al. High-risk coronary plaque at coronary CT angiography is associated with nonalcoholic fatty liver disease, independent of coronary plaque and stenosis burden: Results from the ROMICAT II trial. Radiology 2015, 274, 693–701. [Google Scholar] [CrossRef] [Green Version]
  57. Park, H.E.; Kwak, M.S.; Kim, D.; Kim, M.K.; Cha, M.J.; Choi, S.Y. Nonalcoholic Fatty Liver Disease Is Associated With Coronary Artery Calcification Development: A Longitudinal Study. J. Clin. Endocrinol. Metab. 2016, 101, 3134–3143. [Google Scholar] [CrossRef] [Green Version]
  58. Kim, J.; Lee, D.Y.; Park, S.E.; Park, C.-Y.; Lee, W.-Y.; Oh, K.-W.; Park, S.-W.; Rhee, E.-J. Increased risk for development of coronary artery calcification in subjects with non-alcoholic fatty liver disease and systemic inflammation. PLoS ONE 2017, 12, e0180118. [Google Scholar] [CrossRef] [Green Version]
  59. Kim, D.; Choi, S.Y.; Park, E.H.; Lee, W.; Kang, J.H.; Kim, W.; Kim, Y.J.; Yoon, J.H.; Jeong, S.H.; Lee, D.H.; et al. Nonalcoholic fatty liver disease is associated with coronary artery calcification. Hepatology 2012, 56, 605–613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Ishiba, H.; Sumida, Y.; Kataoka, S.; Kuroda, M.; Akabame, S.; Tomiyasu, K.; Tanaka, M.; Arai, M.; Taketani, H.; Seko, Y.; et al. Association of coronary artery calcification with liver fibrosis in Japanese patients with non-alcoholic fatty liver disease. Hepatol. Res. 2016, 46, 1107–1117. [Google Scholar] [CrossRef] [PubMed]
  61. Jaruvongvanich, V.; Wirunsawanya, K.; Sanguankeo, A.; Upala, S. Nonalcoholic fatty liver disease is associated with coronary artery calcification: A systematic review and meta-analysis. Dig. Liver Dis. 2016, 48, 1410–1417. [Google Scholar] [CrossRef] [PubMed]
  62. Sinn, D.H.; Kang, D.; Chang, Y.; Ryu, S.; Gu, S.; Kim, H.; Seong, D.; Cho, S.J.; Yi, B.K.; Park, H.D.; et al. Non-alcoholic fatty liver disease and progression of coronary artery calcium score: A retrospective cohort study. Gut 2017, 66, 323–329. [Google Scholar] [CrossRef] [PubMed]
  63. Targher, G.; Day, C.P.; Bonora, E. Risk of cardiovascular disease in patients with nonalcoholic fatty liver disease. N. Engl. J. Med. 2010, 363, 1341–1350. [Google Scholar] [CrossRef] [Green Version]
  64. Assy, N.; Djibre, A.; Farah, R.; Grosovski, M.; Marmor, A. Presence of coronary plaques in patients with nonalcoholic fatty liver disease. Radiology 2010, 254, 393–400. [Google Scholar] [CrossRef] [Green Version]
  65. Patil, R.; Sood, G.K. Non-alcoholic fatty liver disease and cardiovascular risk. World J. Gastrointest. Pathophysiol. 2017, 8, 51–58. [Google Scholar] [CrossRef]
  66. Wong, V.W.; Wong, G.L.; Yip, G.W.; Lo, A.O.; Limquiaco, J.; Chu, W.C.; Chim, A.M.; Yu, C.M.; Yu, J.; Chan, F.K.; et al. Coronary artery disease and cardiovascular outcomes in patients with non-alcoholic fatty liver disease. Gut 2011, 60, 1721–1727. [Google Scholar] [CrossRef] [PubMed]
  67. Lee, S.H.; Yun, S.J.; Kim, D.H.; Jo, H.H.; Park, Y.S. Severity of nonalcoholic fatty liver disease on sonography and risk of coronary heart disease. J. Clin. Ultrasound 2017, 45, 391–399. [Google Scholar] [CrossRef]
  68. Keskin, M.; Hayıroğlu, M.; Uzun, A.O.; Güvenç, T.S.; Şahin, S.; Kozan, Ö. Effect of Nonalcoholic Fatty Liver Disease on In-Hospital and Long-Term Outcomes in Patients With ST-Segment Elevation Myocardial Infarction. Am. J. Cardiol. 2017, 120, 1720–1726. [Google Scholar] [CrossRef]
  69. Emre, A.; Terzi, S.; Celiker, E.; Sahin, S.; Yazıcı, S.; Erdem, A.; Ceylan, U.S.; Asik, M.; Yesilcimen, K. Impact of Nonalcoholic Fatty Liver Disease on Myocardial Perfusion in Nondiabetic Patients Undergoing Primary Percutaneous Coronary Intervention for ST-Segment Elevation Myocardial Infarction. Am. J. Cardiol. 2015, 116, 1810–1814. [Google Scholar] [CrossRef]
  70. Mantovani, A. Nonalcoholic Fatty Liver Disease (NAFLD) and Risk of Cardiac Arrhythmias: A New Aspect of the Liver-heart Axis. J. Clin. Transl. Hepatol. 2017, 5, 134–141. [Google Scholar] [CrossRef] [Green Version]
  71. Targher, G.; Mantovani, A.; Pichiri, I.; Rigolon, R.; Dauriz, M.; Zoppini, G.; Morani, G.; Vassanelli, C.; Bonora, E. Non-alcoholic fatty liver disease is associated with an increased prevalence of atrial fibrillation in hospitalized patients with type 2 diabetes. Clin. Sci. 2013, 125, 301–309. [Google Scholar] [CrossRef] [Green Version]
  72. Targher, G.; Valbusa, F.; Bonapace, S.; Bertolini, L.; Zenari, L.; Rodella, S.; Zoppini, G.; Mantovani, W.; Barbieri, E.; Byrne, C.D. Non-alcoholic fatty liver disease is associated with an increased incidence of atrial fibrillation in patients with type 2 diabetes. PLoS ONE 2013, 8, e57183. [Google Scholar] [CrossRef]
  73. Käräjämäki, A.J.; Hukkanen, J.; Ukkola, O. The association of non-alcoholic fatty liver disease and atrial fibrillation: A review. Ann. Med. 2018, 50, 371–380. [Google Scholar] [CrossRef]
  74. Käräjämäki, A.J.; Pätsi, O.-P.; Savolainen, M.; Kesäniemi, Y.A.; Huikuri, H.; Ukkola, O. Non-Alcoholic Fatty Liver Disease as a Predictor of Atrial Fibrillation in Middle-Aged Population (OPERA Study). PLoS ONE 2015, 10, e0142937. [Google Scholar] [CrossRef] [Green Version]
  75. Targher, G.; Byrne, C.D.; Tilg, H. NAFLD and increased risk of cardiovascular disease: Clinical associations, pathophysiological mechanisms and pharmacological implications. Gut 2020, 69, 1691–1705. [Google Scholar] [CrossRef]
  76. Hallsworth, K.; Hollingsworth, K.G.; Thoma, C.; Jakovljevic, D.; MacGowan, G.A.; Anstee, Q.M.; Taylor, R.; Day, C.P.; Trenell, M.I. Cardiac structure and function are altered in adults with non-alcoholic fatty liver disease. J. Hepatol. 2013, 58, 757–762. [Google Scholar] [CrossRef]
  77. VanWagner, L.B.; Wilcox, J.E.; Colangelo, L.A.; Lloyd-Jones, D.M.; Carr, J.J.; Lima, J.A.; Lewis, C.E.; Rinella, M.E.; Shah, S.J. Association of nonalcoholic fatty liver disease with subclinical myocardial remodeling and dysfunction: A population-based study. Hepatology 2015, 62, 773–783. [Google Scholar] [CrossRef] [PubMed]
  78. Trovato, F.M.; Martines, G.F.; Catalano, D.; Musumeci, G.; Pirri, C.; Trovato, G.M. Echocardiography and NAFLD (non-alcoholic fatty liver disease). Int. J. Cardiol. 2016, 221, 275–279. [Google Scholar] [CrossRef] [PubMed]
  79. Lee, H.; Kim, G.; Choi, Y.J.; Huh, B.W.; Lee, B.W.; Kang, E.S.; Cha, B.S.; Lee, E.J.; Lee, Y.H.; Huh, K.B. Association between Non-Alcoholic Steatohepatitis and Left Ventricular Diastolic Dysfunction in Type 2 Diabetes Mellitus. Diabetes Metab. J. 2020, 44, 267–276. [Google Scholar] [CrossRef] [PubMed]
  80. Mari, A.; Khoury, T.; Said Ahmad, H.; Abu Baker, F.; Kadah, A.; Sbeit, W.; Pellicano, R.; Mahamid, M. The association between non-alcoholic fatty liver disease and valvular heart disease. Minerva Cardioangiol. 2020, 68, 42–46. [Google Scholar] [CrossRef] [PubMed]
  81. Markus, M.R.P.; Baumeister, S.E.; Stritzke, J.; Dörr, M.; Wallaschofski, H.; Völzke, H.; Lieb, W. Hepatic Steatosis Is Associated With Aortic Valve Sclerosis in the General Population. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 1690–1695. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Bonapace, S.; Valbusa, F.; Bertolini, L.; Pichiri, I.; Mantovani, A.; Rossi, A.; Zenari, L.; Barbieri, E.; Targher, G. Nonalcoholic fatty liver disease is associated with aortic valve sclerosis in patients with type 2 diabetes mellitus. PLoS ONE 2014, 9, e88371. [Google Scholar] [CrossRef] [PubMed]
  83. Mantovani, A.; Pernigo, M.; Bergamini, C.; Bonapace, S.; Lipari, P.; Valbusa, F.; Bertolini, L.; Zenari, L.; Pichiri, I.; Dauriz, M.; et al. Heart valve calcification in patients with type 2 diabetes and nonalcoholic fatty liver disease. Metabolism 2015, 64, 879–887. [Google Scholar] [CrossRef] [Green Version]
  84. Francque, S.M.; van der Graaff, D.; Kwanten, W.J. Non-alcoholic fatty liver disease and cardiovascular risk: Pathophysiological mechanisms and implications. J. Hepatol. 2016, 65, 425–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Vanhoutte, P.M. Endothelial dysfunction: The first step toward coronary arteriosclerosis. Circ. J. 2009, 73, 595–601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Villanova, N.; Moscatiello, S.; Ramilli, S.; Bugianesi, E.; Magalotti, D.; Vanni, E.; Zoli, M.; Marchesini, G. Endothelial dysfunction and cardiovascular risk profile in nonalcoholic fatty liver disease. Hepatology 2005, 42, 473–480. [Google Scholar] [CrossRef] [PubMed]
  87. Deanfield, J.E.; Halcox, J.P.; Rabelink, T.J. Endothelial Function and Dysfunction. Circulation 2007, 115, 1285–1295. [Google Scholar] [CrossRef] [PubMed]
  88. Kasumov, T.; Edmison, J.M.; Dasarathy, S.; Bennett, C.; Lopez, R.; Kalhan, S.C. Plasma levels of asymmetric dimethylarginine in patients with biopsy-proven nonalcoholic fatty liver disease. Metab. Clin. Exp. 2011, 60, 776–781. [Google Scholar] [CrossRef] [Green Version]
  89. Chiang, C.H.; Huang, P.H.; Chung, F.P.; Chen, Z.Y.; Leu, H.B.; Huang, C.C.; Wu, T.C.; Chen, J.W.; Lin, S.J. Decreased circulating endothelial progenitor cell levels and function in patients with nonalcoholic fatty liver disease. PLoS ONE 2012, 7, e31799. [Google Scholar] [CrossRef] [Green Version]
  90. Madamanchi, N.R.; Vendrov, A.; Runge, M.S. Oxidative stress and vascular disease. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 29–38. [Google Scholar] [CrossRef] [Green Version]
  91. Gulsen, M.; Yesilova, Z.; Bagci, S.; Uygun, A.; Ozcan, A.; Ercin, C.N.; Erdil, A.; Sanisoglu, S.Y.; Cakir, E.; Ates, Y.; et al. Elevated plasma homocysteine concentrations as a predictor of steatohepatitis in patients with non-alcoholic fatty liver disease. J. Gastroenterol. Hepatol. 2005, 20, 1448–1455. [Google Scholar] [CrossRef]
  92. Bravo, E.; Palleschi, S.; Aspichueta, P.; Buqué, X.; Rossi, B.; Cano, A.; Napolitano, M.; Ochoa, B.; Botham, K.M. High fat diet-induced non alcoholic fatty liver disease in rats is associated with hyperhomocysteinemia caused by down regulation of the transsulphuration pathway. Lipids Health Dis. 2011, 10, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. De Carvalho, S.C.R.; Muniz, M.T.C.; Siqueira, M.D.V.; Siqueira, E.R.F.; Gomes, A.V.; Silva, K.A.; Bezerra, L.C.L.; D’Almeida, V.; de Oliveira, C.P.M.S.; Pereira, L.M.M.B. Plasmatic higher levels of homocysteine in Non-alcoholic fatty liver disease (NAFLD). Nutr. J. 2013, 12, 37. [Google Scholar] [CrossRef]
  94. Santilli, F.; Davì, G.; Patrono, C. Homocysteine, methylenetetrahydrofolate reductase, folate status and atherothrombosis: A mechanistic and clinical perspective. Vasc. Pharm. 2016, 78, 1–9. [Google Scholar] [CrossRef]
  95. DeFilippis, A.P.; Blaha, M.J.; Martin, S.S.; Reed, R.M.; Jones, S.R.; Nasir, K.; Blumenthal, R.S.; Budoff, M.J. Nonalcoholic fatty liver disease and serum lipoproteins: The Multi-Ethnic Study of Atherosclerosis. Atherosclerosis 2013, 227, 429–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Siddiqui, M.S.; Fuchs, M.; Idowu, M.O.; Luketic, V.A.; Boyett, S.; Sargeant, C.; Stravitz, R.T.; Puri, P.; Matherly, S.; Sterling, R.K.; et al. Severity of nonalcoholic fatty liver disease and progression to cirrhosis are associated with atherogenic lipoprotein profile. Clin. Gastroenterol. Hepatol. Off. Clin. Prac. J. Am. Gastroenterol. Assoc. 2015, 13, 1000–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Sonmez, A.; Nikolic, D.; Dogru, T.; Ercin, C.N.; Genc, H.; Cesur, M.; Tapan, S.; Karslioğlu, Y.; Montalto, G.; Banach, M.; et al. Low- and high-density lipoprotein subclasses in subjects with nonalcoholic fatty liver disease. J. Clin. Lipidol. 2015, 9, 576–582. [Google Scholar] [CrossRef] [PubMed]
  98. Alkhouri, N.; Tamimi, T.A.; Yerian, L.; Lopez, R.; Zein, N.N.; Feldstein, A.E. The inflamed liver and atherosclerosis: A link between histologic severity of nonalcoholic fatty liver disease and increased cardiovascular risk. Dig. Dis. Sci. 2010, 55, 2644–2650. [Google Scholar] [CrossRef] [PubMed]
  99. Gottlieb, A.; Leven, A.S.; Sowa, J.P.; Borucki, K.; Link, A.; Yilmaz, E.; Aygen, S.; Canbay, A.; Porsch-Özcürümez, M. Lipoprotein and Metabolic Profiles Indicate Similar Cardiovascular Risk of Liver Steatosis and NASH. Digestion 2020. [Google Scholar] [CrossRef] [PubMed]
  100. Ouweneel, A.B.; Van Eck, M. Lipoproteins as modulators of atherothrombosis: From endothelial function to primary and secondary coagulation. Vasc. Pharmacol. 2016, 82, 1–10. [Google Scholar] [CrossRef] [PubMed]
  101. Maher, J.J.; Leon, P.; Ryan, J.C. Beyond insulin resistance: Innate immunity in nonalcoholic steatohepatitis. Hepatology 2008, 48, 670–678. [Google Scholar] [CrossRef] [Green Version]
  102. Mitsuyoshi, H.; Yasui, K.; Harano, Y.; Endo, M.; Tsuji, K.; Minami, M.; Itoh, Y.; Okanoue, T.; Yoshikawa, T. Analysis of hepatic genes involved in the metabolism of fatty acids and iron in nonalcoholic fatty liver disease. Hepatol. Res. 2009, 39, 366–373. [Google Scholar] [CrossRef] [PubMed]
  103. He, J.; Lee, J.H.; Febbraio, M.; Xie, W. The emerging roles of fatty acid translocase/CD36 and the aryl hydrocarbon receptor in fatty liver disease. Exp. Biol. Med. 2011, 236, 1116–1121. [Google Scholar] [CrossRef] [PubMed]
  104. Samuel, V.T.; Liu, Z.X.; Wang, A.; Beddow, S.A.; Geisler, J.G.; Kahn, M.; Zhang, X.M.; Monia, B.P.; Bhanot, S.; Shulman, G.I. Inhibition of protein kinase Cepsilon prevents hepatic insulin resistance in nonalcoholic fatty liver disease. J. Clin. Investig. 2007, 117, 739–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Postic, C.; Girard, J. Contribution of de novo fatty acid synthesis to hepatic steatosis and insulin resistance: Lessons from genetically engineered mice. J. Clin. Investig. 2008, 118, 829–838. [Google Scholar] [CrossRef] [Green Version]
  106. Donnelly, K.L.; Smith, C.I.; Schwarzenberg, S.J.; Jessurun, J.; Boldt, M.D.; Parks, E.J. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin. Investig. 2005, 115, 1343–1351. [Google Scholar] [CrossRef] [Green Version]
  107. Lambert, J.E.; Ramos-Roman, M.A.; Browning, J.D.; Parks, E.J. Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology 2014, 146, 726–735. [Google Scholar] [CrossRef]
  108. Racanelli, V.; Rehermann, B. The liver as an immunological organ. Hepatology 2006, 43, S54–S62. [Google Scholar] [CrossRef]
  109. Lafontan, M.; Girard, J. Impact of visceral adipose tissue on liver metabolism. Part I: Heterogeneity of adipose tissue and functional properties of visceral adipose tissue. Diabetes Metab. 2008, 34, 317–327. [Google Scholar] [CrossRef]
  110. De Ferranti, S.; Mozaffarian, D. The Perfect Storm: Obesity, Adipocyte Dysfunction, and Metabolic Consequences. Clin. Chem. 2008, 54, 945–955. [Google Scholar] [CrossRef] [Green Version]
  111. Vonghia, L.; Magrone, T.; Verrijken, A.; Michielsen, P.; Van Gaal, L.; Jirillo, E.; Francque, S. Peripheral and Hepatic Vein Cytokine Levels in Correlation with Non-Alcoholic Fatty Liver Disease (NAFLD)-Related Metabolic, Histological, and Haemodynamic Features. PLoS ONE 2015, 10, e0143380. [Google Scholar] [CrossRef]
  112. Lavie, C.J.; Milani, R.V.; Verma, A.; O’Keefe, J.H. C-Reactive Protein and Cardiovascular Diseases—Is it Ready for Primetime? Am. J. Med. Sci. 2009, 338, 486–492. [Google Scholar] [CrossRef]
  113. Targher, G.; Bertolini, L.; Rodella, S.; Lippi, G.; Franchini, M.; Zoppini, G.; Muggeo, M.; Day, C.P. NASH predicts plasma inflammatory biomarkers independently of visceral fat in men. Obesity 2008, 16, 1394–1399. [Google Scholar] [CrossRef]
  114. Stoner, L.; Lucero, A.A.; Palmer, B.R.; Jones, L.M.; Young, J.M.; Faulkner, J. Inflammatory biomarkers for predicting cardiovascular disease. Clin. Biochem. 2013, 46, 1353–1371. [Google Scholar] [CrossRef] [PubMed]
  115. Wolfs, M.G.M.; Gruben, N.; Rensen, S.S.; Verdam, F.J.; Greve, J.W.; Driessen, A.; Wijmenga, C.; Buurman, W.A.; Franke, L.; Scheja, L.; et al. Determining the association between adipokine expression in multiple tissues and phenotypic features of non-alcoholic fatty liver disease in obesity. Nutr. Diabetes 2015, 5, e146. [Google Scholar] [CrossRef]
  116. Yoneda, M.; Mawatari, H.; Fujita, K.; Iida, H.; Yonemitsu, K.; Kato, S.; Takahashi, H.; Kirikoshi, H.; Inamori, M.; Nozaki, Y.; et al. High-sensitivity C-reactive protein is an independent clinical feature of nonalcoholic steatohepatitis (NASH) and also of the severity of fibrosis in NASH. J. Gastroenterol. 2007, 42, 573–582. [Google Scholar] [CrossRef] [PubMed]
  117. Hamirani, Y.S.; Katz, R.; Nasir, K.; Zeb, I.; Blaha, M.J.; Blumenthal, R.S.; Kronmal, R.N.; Budoff, M.J. Association between inflammatory markers and liver fat: The Multi-Ethnic Study of Atherosclerosis. J. Clin. Exp. Cardiolog. 2014, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Wieckowska, A.; Papouchado, B.G.; Li, Z.; Lopez, R.; Zein, N.N.; Feldstein, A.E. Increased hepatic and circulating interleukin-6 levels in human nonalcoholic steatohepatitis. Am. J. Gastroenterol. 2008, 103, 1372–1379. [Google Scholar] [CrossRef] [PubMed]
  119. Kofler, S.; Nickel, T.; Weis, M. Role of cytokines in cardiovascular diseases: A focus on endothelial responses to inflammation. Clin. Sci. 2005, 108, 205–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Sookoian, S.; Gianotti, T.F.; Rosselli, M.S.; Burgueño, A.L.; Castaño, G.O.; Pirola, C.J. Liver transcriptional profile of atherosclerosis-related genes in human nonalcoholic fatty liver disease. Atherosclerosis 2011, 218, 378–385. [Google Scholar] [CrossRef] [PubMed]
  121. Tripodi, A.; Mannucci, P.M. The Coagulopathy of Chronic Liver Disease. N. Engl. J. Med. 2011, 365, 147–156. [Google Scholar] [CrossRef] [Green Version]
  122. Kotronen, A.; Joutsi-Korhonen, L.; Sevastianova, K.; Bergholm, R.; Hakkarainen, A.; Pietiläinen, K.H.; Lundbom, N.; Rissanen, A.; Lassila, R.; Yki-Järvinen, H. Increased coagulation factor VIII, IX, XI and XII activities in non-alcoholic fatty liver disease. Liver Int. 2011, 31, 176–183. [Google Scholar] [CrossRef]
  123. Bhatia, L.S.; Curzen, N.P.; Calder, P.C.; Byrne, C.D. Non-alcoholic fatty liver disease: A new and important cardiovascular risk factor? Eur. Heart J. 2012, 33, 1190–1200. [Google Scholar] [CrossRef] [Green Version]
  124. Barbato, A.; Iacone, R.; Tarantino, G.; Russo, O.; Sorrentino, P.; Avallone, S.; Galletti, F.; Farinaro, E.; Della Valle, E.; Strazzullo, P. Relationships of PAI-1 levels to central obesity and liver steatosis in a sample of adult male population in southern Italy. Intern. Emerg. Med. 2009, 4, 315–323. [Google Scholar] [CrossRef]
  125. Verrijken, A.; Francque, S.; Mertens, I.; Prawitt, J.; Caron, S.; Hubens, G.; Van Marck, E.; Staels, B.; Michielsen, P.; Van Gaal, L. Prothrombotic factors in histologically proven nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Hepatology 2014, 59, 121–129. [Google Scholar] [CrossRef]
  126. Song, C.; Burgess, S.; Eicher, J.D.; O’Donnell, C.J.; Johnson, A.D. Causal Effect of Plasminogen Activator Inhibitor Type 1 on Coronary Heart Disease. J. Am. Heart Assoc. 2017, 6. [Google Scholar] [CrossRef] [PubMed]
  127. Loeffen, R.; Spronk, H.M.H.; Ten Cate, H. The impact of blood coagulability on atherosclerosis and cardiovascular disease. J. Thromb. Haemost. 2012, 10, 1207–1216. [Google Scholar] [CrossRef] [PubMed]
  128. Loomba, R.; Seguritan, V.; Li, W.; Long, T.; Klitgord, N.; Bhatt, A.; Dulai, P.S.; Caussy, C.; Bettencourt, R.; Highlander, S.K.; et al. Gut Microbiome-Based Metagenomic Signature for Non-invasive Detection of Advanced Fibrosis in Human Nonalcoholic Fatty Liver Disease. Cell Metab. 2017, 25, 1054–1062. [Google Scholar] [CrossRef] [PubMed]
  129. Aron-Wisnewsky, J.; Vigliotti, C.; Witjes, J.; Le, P.; Holleboom, A.G.; Verheij, J.; Nieuwdorp, M.; Clément, K. Gut microbiota and human NAFLD: Disentangling microbial signatures from metabolic disorders. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 279–297. [Google Scholar] [CrossRef]
  130. Brown, J.M.; Hazen, S.L. The gut microbial endocrine organ: Bacterially derived signals driving cardiometabolic diseases. Annu. Rev. Med. 2015, 66, 343–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; DuGar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.-M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef] [Green Version]
  132. Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Kim, J.; Samson, S.L. Cardiovascular effects of incretin therapy in diabetes care. Met. Syndr. Relat. Dis. 2014, 12, 303–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Wang, Y.; Parlevliet, E.T.; Geerling, J.J.; van der Tuin, S.J.; Zhang, H.; Bieghs, V.; Jawad, A.H.; Shiri-Sverdlov, R.; Bot, I.; de Jager, S.C.; et al. Exendin-4 decreases liver inflammation and atherosclerosis development simultaneously by reducing macrophage infiltration. Br. J. Pharmacol. 2014, 171, 723–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Singal, A.G.; Manjunath, H.; Yopp, A.C.; Beg, M.S.; Marrero, J.A.; Gopal, P.; Waljee, A.K. The effect of PNPLA3 on fibrosis progression and development of hepatocellular carcinoma: A meta-analysis. Am. J. Gastroenterol. 2014, 109, 325–334. [Google Scholar] [CrossRef] [Green Version]
  136. Liu, Y.L.; Reeves, H.L.; Burt, A.D.; Tiniakos, D.; McPherson, S.; Leathart, J.B.; Allison, M.E.; Alexander, G.J.; Piguet, A.C.; Anty, R.; et al. TM6SF2 rs58542926 influences hepatic fibrosis progression in patients with non-alcoholic fatty liver disease. Nat. Commun. 2014, 5, 4309. [Google Scholar] [CrossRef] [Green Version]
  137. Mancina, R.M.; Dongiovanni, P.; Petta, S.; Pingitore, P.; Meroni, M.; Rametta, R.; Borén, J.; Montalcini, T.; Pujia, A.; Wiklund, O.; et al. The MBOAT7-TMC4 Variant rs641738 Increases Risk of Nonalcoholic Fatty Liver Disease in Individuals of European Descent. Gastroenterology 2016, 150, 1219–1230. [Google Scholar] [CrossRef] [Green Version]
  138. Petta, S.; Miele, L.; Bugianesi, E.; Cammà, C.; Rosso, C.; Boccia, S.; Cabibi, D.; Di Marco, V.; Grimaudo, S.; Grieco, A.; et al. Glucokinase regulatory protein gene polymorphism affects liver fibrosis in non-alcoholic fatty liver disease. PLoS ONE 2014, 9, e87523. [Google Scholar] [CrossRef] [Green Version]
  139. Loomba, R.; Schork, N.; Chen, C.H.; Bettencourt, R.; Bhatt, A.; Ang, B.; Nguyen, P.; Hernandez, C.; Richards, L.; Salotti, J.; et al. Heritability of Hepatic Fibrosis and Steatosis Based on a Prospective Twin Study. Gastroenterology 2015, 149, 1784–1793. [Google Scholar] [CrossRef] [Green Version]
  140. Tarnoki, A.D.; Tarnoki, D.L.; Bata, P.; Littvay, L.; Osztovits, J.; Jermendy, G.; Karlinger, K.; Lannert, A.; Preda, I.; Kiss, R.G.; et al. Heritability of non-alcoholic fatty liver disease and association with abnormal vascular parameters: A twin study. Liver Int. 2012, 32, 1287–1293. [Google Scholar] [CrossRef]
  141. Holmen, O.L.; Zhang, H.; Fan, Y.; Hovelson, D.H.; Schmidt, E.M.; Zhou, W.; Guo, Y.; Zhang, J.; Langhammer, A.; Løchen, M.L.; et al. Systematic evaluation of coding variation identifies a candidate causal variant in TM6SF2 influencing total cholesterol and myocardial infarction risk. Nat. Genet. 2014, 46, 345–351. [Google Scholar] [CrossRef]
  142. Zhou, Y.; Llauradó, G.; Orešič, M.; Hyötyläinen, T.; Orho-Melander, M.; Yki-Järvinen, H. Circulating triacylglycerol signatures and insulin sensitivity in NAFLD associated with the E167K variant in TM6SF2. J. Hepatol. 2015, 62, 657–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Speliotes, E.K.; Yerges-Armstrong, L.M.; Wu, J.; Hernaez, R.; Kim, L.J.; Palmer, C.D.; Gudnason, V.; Eiriksdottir, G.; Garcia, M.E.; Launer, L.J.; et al. Genome-wide association analysis identifies variants associated with nonalcoholic fatty liver disease that have distinct effects on metabolic traits. PLoS Genet. 2011, 7, e1001324. [Google Scholar] [CrossRef] [PubMed]
  144. Willer, C.J.; Schmidt, E.M.; Sengupta, S.; Peloso, G.M.; Gustafsson, S.; Kanoni, S.; Ganna, A.; Chen, J.; Buchkovich, M.L.; Mora, S.; et al. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 2013, 45, 1274–1283. [Google Scholar] [CrossRef] [Green Version]
  145. Kahali, B.; Liu, Y.-L.; Daly, A.K.; Day, C.P.; Anstee, Q.M.; Speliotes, E.K. TM6SF2: Catch-22 in the Fight Against Nonalcoholic Fatty Liver Disease and Cardiovascular Disease? Gastroenterology 2015, 148, 679–684. [Google Scholar] [CrossRef] [PubMed]
  146. Liu, D.J.; Peloso, G.M.; Yu, H.; Butterworth, A.S.; Wang, X.; Mahajan, A.; Saleheen, D.; Emdin, C.; Alam, D.; Alves, A.C.; et al. Exome-wide association study of plasma lipids in >300,000 individuals. Nat. Genet. 2017, 49, 1758–1766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Meffert, P.J.; Repp, K.D.; Völzke, H.; Weiss, F.U.; Homuth, G.; Kühn, J.P.; Lerch, M.M.; Aghdassi, A.A. The PNPLA3 SNP rs738409:G allele is associated with increased liver disease-associated mortality but reduced overall mortality in a population-based cohort. J. Hepatol. 2018, 68, 858–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Cai, D.; Yuan, M.; Frantz, D.F.; Melendez, P.A.; Hansen, L.; Lee, J.; Shoelson, S.E. Local and systemic insulin resistance resulting from hepatic activation of IKK-beta and NF-kappaB. Nat. Med. 2005, 11, 183–190. [Google Scholar] [CrossRef]
  149. Kakino, S.; Ohki, T.; Nakayama, H.; Yuan, X.; Otabe, S.; Hashinaga, T.; Wada, N.; Kurita, Y.; Tanaka, K.; Hara, K.; et al. Pivotal Role of TNF-α in the Development and Progression of Nonalcoholic Fatty Liver Disease in a Murine Model. Horm. Metab. Res. 2018, 50, 80–87. [Google Scholar] [CrossRef]
  150. Negrin, K.A.; Roth Flach, R.J.; DiStefano, M.T.; Matevossian, A.; Friedline, R.H.; Jung, D.; Kim, J.K.; Czech, M.P. IL-1 signaling in obesity-induced hepatic lipogenesis and steatosis. PLoS ONE 2014, 9, e107265. [Google Scholar] [CrossRef] [Green Version]
  151. Alessi, M.-C.; Bastelica, D.; Mavri, A.; Morange, P.; Berthet, B.; Grino, M.; Juhan-Vague, I. Plasma PAI-1 Levels Are More Strongly Related to Liver Steatosis Than to Adipose Tissue Accumulation. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1262–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Eitzman, D.T.; Westrick, R.J.; Xu, Z.; Tyson, J.; Ginsburg, D. Plasminogen activator inhibitor-1 deficiency protects against atherosclerosis progression in the mouse carotid artery. Blood 2000, 96, 4212–4215. [Google Scholar] [CrossRef] [PubMed]
  153. Luttun, A.; Lupu, F.; Storkebaum, E.; Hoylaerts, M.F.; Moons, L.; Crawley, J.; Bono, F.; Poole, A.R.; Tipping, P.; Herbert, J.M.; et al. Lack of plasminogen activator inhibitor-1 promotes growth and abnormal matrix remodeling of advanced atherosclerotic plaques in apolipoprotein E-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2002, 22, 499–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Samad, F.; Loskutoff, D.J. Tissue distribution and regulation of plasminogen activator inhibitor-1 in obese mice. Mol. Med. 1996, 2, 568–582. [Google Scholar] [CrossRef] [Green Version]
  155. Glenn, T.K.; Honar, H.; Liu, H.; ter Keurs, H.E.D.J.; Lee, S.S. Role of cardiac myofilament proteins titin and collagen in the pathogenesis of diastolic dysfunction in cirrhotic rats. J. Hepatol. 2011, 55, 1249–1255. [Google Scholar] [CrossRef]
  156. Shirai, Y.; Yoshiji, H.; Noguchi, R.; Kaji, K.; Aihara, Y.; Douhara, A.; Moriya, K.; Namisaki, T.; Kawaratani, H.; Fukui, H. Cross talk between toll-like receptor-4 signaling and angiotensin-II in liver fibrosis development in the rat model of non-alcoholic steatohepatitis. J. Gastroenterol. Hepatol. 2013, 28, 723–730. [Google Scholar] [CrossRef]
  157. Madrigal-Perez, V.M.; García-Rivera, A.; Rodriguez-Hernandez, A.; Ceja-Espiritu, G.; Briseño-Gomez, X.G.; Galvan-Salazar, H.R.; Soriano-Hernandez, A.D.; Guzman-Esquivel, J.; Martinez-Fierro, M.L.; Newton-Sanchez, O.A.; et al. Preclinical analysis of nonsteroidal anti-inflammatory drug usefulness for the simultaneous prevention of steatohepatitis, atherosclerosis and hyperlipidemia. Int. J. Clin. Exp. Med. 2015, 8, 22477–22483. [Google Scholar] [PubMed]
  158. Møller, S.; Dümcke, C.W.; Krag, A. The heart and the liver. Expert Rev. Gastroenterol. Hepatol. 2009, 3, 51–64. [Google Scholar] [CrossRef]
  159. Correale, M.; Tarantino, N.; Petrucci, R.; Tricarico, L.; Laonigro, I.; Di Biase, M.; Brunetti, N.D. Liver disease and heart failure: Back and forth. Eur. J. Intern. Med. 2018, 48, 25–34. [Google Scholar] [CrossRef]
  160. Ford, R.M.; Book, W.; Spivey, J.R. Liver disease related to the heart. Transplant. Rev. 2015, 29, 33–37. [Google Scholar] [CrossRef]
  161. El Hadi, H.; Di Vincenzo, A.; Vettor, R.; Rossato, M. Relationship between Heart Disease and Liver Disease: A Two-Way Street. Cells 2020, 9, 567. [Google Scholar] [CrossRef] [Green Version]
  162. Eckel, R.H.; Jakicic, J.M.; Ard, J.D.; Jesus, J.M.d.; Miller, N.H.; Hubbard, V.S.; Lee, I.-M.; Lichtenstein, A.H.; Loria, C.M.; Millen, B.E.; et al. 2013 AHA/ACC Guideline on Lifestyle Management to Reduce Cardiovascular Risk. Circulation 2014, 129, S76–S99. [Google Scholar] [CrossRef] [Green Version]
  163. AISF position paper on nonalcoholic fatty liver disease (NAFLD): Updates and future directions. Dig. Liver Dis. 2017, 49, 471–483. [CrossRef] [PubMed]
  164. 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]
  165. Vilar-Gomez, E.; Martinez-Perez, Y.; Calzadilla-Bertot, L.; Torres-Gonzalez, A.; Gra-Oramas, B.; Gonzalez-Fabian, L.; Friedman, S.L.; Diago, M.; Romero-Gomez, M. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology 2015, 149, 367–378. [Google Scholar] [CrossRef] [PubMed]
  166. Nascimbeni, F.; Aron-Wisnewsky, J.; Pais, R.; Tordjman, J.; Poitou, C.; Charlotte, F.; Bedossa, P.; Poynard, T.; Clément, K.; Ratziu, V. Statins, antidiabetic medications and liver histology in patients with diabetes with non-alcoholic fatty liver disease. BMJ Open Gastroenterol. 2016, 3, e000075. [Google Scholar] [CrossRef] [PubMed]
  167. Athyros, V.G.; Tziomalos, K.; Gossios, T.D.; Griva, T.; Anagnostis, P.; Kargiotis, K.; Pagourelias, E.D.; Theocharidou, E.; Karagiannis, A.; Mikhailidis, D.P. 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. [Google Scholar] [CrossRef]
  168. Li, Y.; Liu, L.; Wang, B.; Wang, J.; Chen, D. Metformin in non-alcoholic fatty liver disease: A systematic review and meta-analysis. Biomed. Rep. 2013, 1, 57–64. [Google Scholar] [CrossRef] [Green Version]
  169. Musso, G.; Gambino, R.; Cassader, M.; Pagano, G. A meta-analysis of randomized trials for the treatment of nonalcoholic fatty liver disease. Hepatology 2010, 52, 79–104. [Google Scholar] [CrossRef]
  170. Haukeland, J.W.; Konopski, Z.; Eggesbø, H.B.; von Volkmann, H.L.; Raschpichler, G.; Bjøro, K.; Haaland, T.; Løberg, E.M.; Birkeland, K. Metformin in patients with non-alcoholic fatty liver disease: A randomized, controlled trial. Scand. J. Gastroenterol. 2009, 44, 853–860. [Google Scholar] [CrossRef]
  171. Loomba, R.; Lutchman, G.; Kleiner, D.E.; Ricks, M.; Feld, J.J.; Borg, B.B.; Modi, A.; Nagabhyru, P.; Sumner, A.E.; Liang, T.J.; et al. Clinical trial: Pilot study of metformin for the treatment of non-alcoholic steatohepatitis. Aliment. Pharmacol. Ther. 2009, 29, 172–182. [Google Scholar] [CrossRef]
  172. Musso, G.; Cassader, M.; Paschetta, E.; Gambino, R. Thiazolidinediones and Advanced Liver Fibrosis in Nonalcoholic Steatohepatitis: A Meta-analysis. JAMA Intern. Med. 2017, 177, 633–640. [Google Scholar] [CrossRef]
  173. Blazina, I.; Selph, S. Diabetes drugs for nonalcoholic fatty liver disease: A systematic review. Syst. Rev. 2019, 8, 295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Simon, T.G.; Henson, J.; Osganian, S.; Masia, R.; Chan, A.T.; Chung, R.T.; Corey, K.E. Daily Aspirin Use Associated with Reduced Risk For Fibrosis Progression In Patients With Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2019, 17, 2776–2784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Shen, H.; Shahzad, G.; Jawairia, M.; Bostick, R.M.; Mustacchia, P. Association between aspirin use and the prevalence of nonalcoholic fatty liver disease: A cross-sectional study from the Third National Health and Nutrition Examination Survey. Aliment. Pharmacol. Ther. 2014, 40, 1066–1073. [Google Scholar] [CrossRef] [PubMed]
  176. Jiang, Z.G.; Feldbrügge, L.; Tapper, E.B.; Popov, Y.; Ghaziani, T.; Afdhal, N.; Robson, S.C.; Mukamal, K.J. Aspirin use is associated with lower indices of liver fibrosis among adults in the United States. Aliment. Pharmacol. Ther. 2016, 43, 734–743. [Google Scholar] [CrossRef]
  177. Ripoche, J. Blood platelets and inflammation: Their relationship with liver and digestive diseases. Clin. Res. Hepatol. Gastroenterol. 2011, 35, 353–357. [Google Scholar] [CrossRef]
  178. Fujita, K.; Nozaki, Y.; Wada, K.; Yoneda, M.; Endo, H.; Takahashi, H.; Iwasaki, T.; Inamori, M.; Abe, Y.; Kobayashi, N.; et al. Effectiveness of antiplatelet drugs against experimental non-alcoholic fatty liver disease. Gut 2008, 57, 1583–1591. [Google Scholar] [CrossRef] [PubMed]
  179. Chauhan, A.; Adams, D.H.; Watson, S.P.; Lalor, P.F. Platelets: No longer bystanders in liver disease. Hepatology 2016, 64, 1774–1784. [Google Scholar] [CrossRef]
  180. Iannacone, M.; Sitia, G.; Isogawa, M.; Marchese, P.; Castro, M.G.; Lowenstein, P.R.; Chisari, F.V.; Ruggeri, Z.M.; Guidotti, L.G. Platelets mediate cytotoxic T lymphocyte-induced liver damage. Nat. Med. 2005, 11, 1167–1169. [Google Scholar] [CrossRef] [Green Version]
  181. Iannacone, M.; Sitia, G.; Narvaiza, I.; Ruggeri, Z.M.; Guidotti, L.G. Antiplatelet Drug Therapy Moderates Immune-Mediated Liver Disease and Inhibits Viral Clearance in Mice Infected with a Replication-Deficient Adenovirus. Clin. Vaccin. Immunol. 2007, 14, 1532–1535. [Google Scholar] [CrossRef] [Green Version]
  182. Sitia, G.; Aiolfi, R.; Di Lucia, P.; Mainetti, M.; Fiocchi, A.; Mingozzi, F.; Esposito, A.; Ruggeri, Z.M.; Chisari, F.V.; Iannacone, M.; et al. Antiplatelet therapy prevents hepatocellular carcinoma and improves survival in a mouse model of chronic hepatitis B. Proc. Natl. Acad. Sci. USA 2012, 109, E2165–E2172. [Google Scholar] [CrossRef] [Green Version]
  183. Han, Y.M.; Lee, Y.J.; Jang, Y.N.; Kim, H.M.; Seo, H.S.; Jung, T.W.; Jeong, J.H. Aspirin Improves Nonalcoholic Fatty Liver Disease and Atherosclerosis through Regulation of the PPARδ-AMPK-PGC-1α Pathway in Dyslipidemic Conditions. BioMed Res. Int. 2020, 2020, 7806860. [Google Scholar] [CrossRef] [Green Version]
  184. Buse, J.B.; Klonoff, D.C.; Nielsen, L.L.; Guan, X.; Bowlus, C.L.; Holcombe, J.H.; Maggs, D.G.; Wintle, M.E. Metabolic effects of two years of exenatide treatment on diabetes, obesity, and hepatic biomarkers in patients with type 2 diabetes: An interim analysis of data from the open-label, uncontrolled extension of three double-blind, placebo-controlled trials. Clin. Ther. 2007, 29, 139–153. [Google Scholar] [CrossRef]
  185. Fan, S.; Shi, X.; Yao, J.; Zhong, M.; Feng, P. The efficacy of glucagon-like peptide 1 receptor agonists in patients with non-alcoholic fatty liver disease: A systematic review and meta-analysis of randomized controlled trials. Rev. Esp. Enferm. Dig. 2020, 112, 627–635. [Google Scholar] [CrossRef]
  186. Nowrouzi-Sohrabi, P.; Rezaei, S.; Jalali, M.; Ashourpour, M.; Ahmadipour, A.; Keshavarz, P.; Akbari, H. The effects of glucagon-like peptide-1 receptor agonists on glycemic control and anthropometric profiles among diabetic patients with non-alcoholic fatty liver disease: A systematic review and meta-analysis of randomized controlled trials. Eur. J. Pharmacol. 2020, 173823. [Google Scholar] [CrossRef]
  187. Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; Abouda, G.; Aldersley, M.A.; 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. [Google Scholar] [CrossRef] [Green Version]
  188. Frías, J.P.; Guja, C.; Hardy, E.; Ahmed, A.; Dong, F.; Öhman, P.; Jabbour, S.A. Exenatide once weekly plus dapagliflozin once daily versus exenatide or dapagliflozin alone in patients with type 2 diabetes inadequately controlled with metformin monotherapy (DURATION-8): A 28 week, multicentre, double-blind, phase 3, randomised controlled trial. Lancet Diav. Endocrinol. 2016, 4, 1004–1016. [Google Scholar] [CrossRef]
  189. Gastaldelli, A.; Repetto, E.; Guja, C.; Hardy, E.; Han, J.; Jabbour, S.A.; Ferrannini, E. Exenatide and dapagliflozin combination improves markers of liver steatosis and fibrosis in patients with type 2 diabetes. Diabetes Obes. Metab. 2020, 22, 393–403. [Google Scholar] [CrossRef] [PubMed]
  190. Shimizu, M.; Suzuki, K.; Kato, K.; Jojima, T.; Iijima, T.; Murohisa, T.; Iijima, M.; Takekawa, H.; Usui, I.; Hiraishi, H.; 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 Obes. Metab. 2019, 21, 285–292. [Google Scholar] [CrossRef] [PubMed]
  191. Eriksson, J.W.; Lundkvist, P.; Jansson, P.A.; Johansson, L.; Kvarnström, M.; Moris, L.; Miliotis, T.; Forsberg, G.B.; Risérus, U.; Lind, L.; et al. Effects of dapagliflozin and n-3 carboxylic acids on non-alcoholic fatty liver disease in people with type 2 diabetes: A double-blind randomised placebo-controlled study. Diabetologia 2018, 61, 1923–1934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Akuta, N.; Kawamura, Y.; Watanabe, C.; Nishimura, A.; Okubo, M.; Mori, Y.; Fujiyama, S.; Sezaki, H.; Hosaka, T.; Kobayashi, M.; et al. Impact of sodium glucose cotransporter 2 inhibitor on histological features and glucose metabolism of non-alcoholic fatty liver disease complicated by diabetes mellitus. Hepatol. Res. 2019, 49, 531–539. [Google Scholar] [CrossRef] [PubMed]
  193. Tang, M.; Jia, H.; Chen, S.; Yang, B.; Patpur, B.K.; Song, W.; Chang, Y.; Li, J.; Yang, C. Significance of MR/OPN/HMGB1 axis in NAFLD-associated hepatic fibrogenesis. Life Sci. 2021, 264, 118619. [Google Scholar] [CrossRef]
  194. Giacchetti, G.; Sechi, L.A.; Rilli, S.; Carey, R.M. The renin-angiotensin-aldosterone system, glucose metabolism and diabetes. Trends Endocrinol. Metab. 2005, 16, 120–126. [Google Scholar] [CrossRef]
  195. Pelusi, S.; Petta, S.; Rosso, C.; Borroni, V.; Fracanzani, A.L.; Dongiovanni, P.; Craxi, A.; Bugianesi, E.; Fargion, S.; Valenti, L. Renin-Angiotensin System Inhibitors, Type 2 Diabetes and Fibrosis Progression: An Observational Study in Patients with Nonalcoholic Fatty Liver Disease. PLoS ONE 2016, 11, e0163069. [Google Scholar] [CrossRef] [PubMed]
  196. Georgescu, E.F.; Ionescu, R.; Niculescu, M.; Mogoanta, L.; Vancica, L. Angiotensin-receptor blockers as therapy for mild-to-moderate hypertension-associated non-alcoholic steatohepatitis. World J. Gastroenterol. 2009, 15, 942–954. [Google Scholar] [CrossRef]
  197. Yokohama, S.; Yoneda, M.; Haneda, M.; Okamoto, S.; Okada, M.; Aso, K.; Hasegawa, T.; Tokusashi, Y.; Miyokawa, N.; Nakamura, K. Therapeutic efficacy of an angiotensin II receptor antagonist in patients with nonalcoholic steatohepatitis. Hepatology 2004, 40, 1222–1225. [Google Scholar] [CrossRef]
  198. Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S.; Polymerou, V.; Katsinelos, P. Effects of combined low-dose spironolactone plus vitamin E vs vitamin E monotherapy on insulin resistance, non-invasive indices of steatosis and fibrosis, and adipokine levels in non-alcoholic fatty liver disease: A randomized controlled trial. Diabetes Obes. Metab. 2017, 19, 1805–1809. [Google Scholar] [CrossRef]
  199. Polyzos, S.A.; Kountouras, J.; Zafeiriadou, E.; Patsiaoura, K.; Katsiki, E.; Deretzi, G.; Zavos, C.; Tsarouchas, G.; Rakitzi, P.; Slavakis, A. Effect of spironolactone and vitamin E on serum metabolic parameters and insulin resistance in patients with nonalcoholic fatty liver disease. J. Renin Angiotensin Aldosterone Syst. 2011, 12, 498–503. [Google Scholar] [CrossRef]
  200. Pizarro, M.; Solís, N.; Quintero, P.; Barrera, F.; Cabrera, D.; Rojas-de Santiago, P.; Arab, J.P.; Padilla, O.; Roa, J.C.; Moshage, H.; et al. Beneficial effects of mineralocorticoid receptor blockade in experimental non-alcoholic steatohepatitis. Liver Int. 2015, 35, 2129–2138. [Google Scholar] [CrossRef] [Green Version]
  201. Sharma, A.M.; Pischon, T.; Hardt, S.; Kunz, I.; Luft, F.C. Hypothesis: Beta-adrenergic receptor blockers and weight gain: A systematic analysis. Hypertension 2001, 37, 250–254. [Google Scholar] [CrossRef] [Green Version]
  202. Lithell, H.O. Effect of antihypertensive drugs on insulin, glucose, and lipid metabolism. Diabetes Care 1991, 14, 203–209. [Google Scholar] [CrossRef]
  203. Lithell, H.O. Hyperinsulinemia, insulin resistance, and the treatment of hypertension. Am. J. Hypertens. 1996, 9, 150s–154s. [Google Scholar] [CrossRef] [Green Version]
  204. Lind, L.; Pollare, T.; Berne, C.; Lithell, H. Long-term metabolic effects of antihypertensive drugs. Am. Heart J. 1994, 128, 1177–1183. [Google Scholar] [CrossRef]
  205. Rabkin, S.W. Mechanisms of action of adrenergic receptor blockers on lipids during antihypertensive drug treatment. J. Clin. Pharmacol. 1993, 33, 286–291. [Google Scholar] [CrossRef] [PubMed]
  206. Dornhorst, A.; Powell, S.H.; Pensky, J. Aggravation by propranolol of hyperglycaemic effect of hydrochlorothiazide in type II diabetics without alteration of insulin secretion. Lancet 1985, 1, 123–126. [Google Scholar] [CrossRef]
  207. Wolinsky, H. The effects of beta-adrenergic blocking agents on blood lipid levels. Clin. Cardiol. 1987, 10, 561–566. [Google Scholar] [CrossRef] [PubMed]
  208. Day, J.L.; Metcalfe, J.; Simpson, N.; Lowenthal, L. Adrenergic mechanisms in the control of plasma lipids in man. Am. J. Med. 1984, 76, 94–96. [Google Scholar] [CrossRef]
  209. Kunz, I.; Schorr, U.; Klaus, S.; Sharma, A.M. Resting Metabolic Rate and Substrate Use in Obesity Hypertension. Hypertension 2000, 36, 26–32. [Google Scholar] [CrossRef] [Green Version]
  210. Siegel, D.; Swislocki, A.L. Effects of antihypertensives on glucose metabolism. Metab. Syndr. Relat. Disord. 2007, 5, 211–219. [Google Scholar] [CrossRef] [Green Version]
  211. Kovacić, D.; Marinsek, M.; Gobec, L.; Lainscak, M.; Podbregar, M. Effect of selective and non-selective beta-blockers on body weight, insulin resistance and leptin concentration in chronic heart failure. Clin. Res. Cardiol. 2008, 97, 24–31. [Google Scholar] [CrossRef] [PubMed]
  212. Bakris, G.L.; Fonseca, V.; Katholi, R.E.; McGill, J.B.; Messerli, F.H.; Phillips, R.A.; Raskin, P.; Wright, J.T.; Oakes, R.; Lukas, M.A.; et al. Metabolic Effects of Carvedilol vs Metoprolol in Patients With Type 2 Diabetes Mellitus and HypertensionA Randomized Controlled Trial. JAMA 2004, 292, 2227–2236. [Google Scholar] [CrossRef] [Green Version]
  213. Schmidt, A.C.; Graf, C.; Brixius, K.; Scholze, J. Blood pressure-lowering effect of nebivolol in hypertensive patients with type 2 diabetes mellitus: The YESTONO study. Clin. Drug. Investig. 2007, 27, 841–849. [Google Scholar] [CrossRef] [PubMed]
  214. McKee, C.; Soeda, J.; Asilmaz, E.; Sigalla, B.; Morgan, M.; Sinelli, N.; Roskams, T.; Oben, J.A. Propranolol, a β-adrenoceptor antagonist, worsens liver injury in a model of non-alcoholic steatohepatitis. Biochem. Biophys. Res. Commun. 2013, 437, 597–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Soliman, G.F.; Rashed, L.A.; Morsi, H.; Ibrahim, W.; Abdallah, H.; Bastawy, N.; Abdel Maksoud, O.M. Interrelation of liver vascularity to non-alcoholic fatty liver through a comparative study of the vasodilator effect of carvedilol or nicorandil in rats. Life Sci. 2019, 222, 175–182. [Google Scholar] [CrossRef]
  216. Bower, G.; Toma, T.; Harling, L.; Jiao, L.R.; Efthimiou, E.; Darzi, A.; Athanasiou, T.; Ashrafian, H. Bariatric Surgery and Non-Alcoholic Fatty Liver Disease: A Systematic Review of Liver Biochemistry and Histology. Obes. Surg. 2015, 25, 2280–2289. [Google Scholar] [CrossRef] [PubMed]
  217. Lassailly, G.; Caiazzo, R.; Buob, D.; Pigeyre, M.; Verkindt, H.; Labreuche, J.; Raverdy, V.; Leteurtre, E.; Dharancy, S.; Louvet, A.; et al. Bariatric Surgery Reduces Features of Nonalcoholic Steatohepatitis in Morbidly Obese Patients. Gastroenterology 2015, 149, 379–388. [Google Scholar] [CrossRef] [Green Version]
  218. Vest, A.R.; Heneghan, H.M.; Agarwal, S.; Schauer, P.R.; Young, J.B. Bariatric surgery and cardiovascular outcomes: A systematic review. Heart 2012, 98, 1763–1777. [Google Scholar] [CrossRef]
  219. Said, A.; Akhter, A. Meta-Analysis of Randomized Controlled Trials of Pharmacologic Agents in Non-alcoholic Steatohepatitis. Ann. Hepatol. 2017, 16, 538–547. [Google Scholar] [CrossRef]
  220. Yusuf, S.; Dagenais, G.; Pogue, J.; Bosch, J.; Sleight, P. Vitamin E supplementation and cardiovascular events in high-risk patients. N. Engl. J. Med. 2000, 342, 154–160. [Google Scholar] [CrossRef]
  221. Porez, G.; Prawitt, J.; Gross, B.; Staels, B. Bile acid receptors as targets for the treatment of dyslipidemia and cardiovascular disease. J. Lipid Res. 2012, 53, 1723–1737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Mudaliar, S.; Henry, R.R.; Sanyal, A.J.; Morrow, L.; Marschall, H.U.; Kipnes, M.; Adorini, L.; Sciacca, C.I.; Clopton, P.; Castelloe, E.; et al. Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology 2013, 145, 574–582. [Google Scholar] [CrossRef] [PubMed]
  223. Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; Van Natta, M.L.; Abdelmalek, M.F.; Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): A multicentre, randomised, placebo-controlled trial. Lancet 2015, 385, 956–965. [Google Scholar] [CrossRef] [Green Version]
  224. Sumida, Y.; Yoneda, M. Current and future pharmacological therapies for NAFLD/NASH. J. Gastroenterol. 2018, 53, 362–376. [Google Scholar] [CrossRef] [Green Version]
  225. Hong, F.; Xu, P.; Zhai, Y. The Opportunities and Challenges of Peroxisome Proliferator-Activated Receptors Ligands in Clinical Drug Discovery and Development. Int. J. Mol. Sci. 2018, 19, 2189. [Google Scholar] [CrossRef] [Green Version]
  226. Ratziu, V.; Harrison, S.A.; Francque, S.; Bedossa, P.; Lehert, P.; Serfaty, L.; Romero-Gomez, M.; Boursier, J.; Abdelmalek, M.; Caldwell, S.; et al. Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-α and -δ, Induces Resolution of Nonalcoholic Steatohepatitis Without Fibrosis Worsening. Gastroenterology 2016, 150, 1147–1159. [Google Scholar] [CrossRef] [Green Version]
  227. Joshi, S.R. Saroglitazar for the treatment of dyslipidemia in diabetic patients. Expert Opin. Pharmacother. 2015, 16, 597–606. [Google Scholar] [CrossRef] [PubMed]
  228. Patel, V.; Sanyal, A.J. Drug-induced steatohepatitis. Clin. Liver Dis. 2013, 17, 533. [Google Scholar] [CrossRef] [Green Version]
  229. Ruzieh, M.; Moroi, M.K.; Aboujamous, N.M.; Ghahramani, M.; Naccarelli, G.V.; Mandrola, J.; Foy, A.J. Meta-Analysis Comparing the Relative Risk of Adverse Events for Amiodarone Versus Placebo. Am. J. Cardiol. 2019, 124, 1889–1893. [Google Scholar] [CrossRef] [PubMed]
  230. Lazo, M.; Hernaez, R.; Bonekamp, S.; Kamel, I.R.; Brancati, F.L.; Guallar, E.; Clark, J.M. Non-alcoholic fatty liver disease and mortality among US adults: Prospective cohort study. BMJ 2011, 343, d6891. [Google Scholar] [CrossRef] [Green Version]
  231. Kim, D.; Kim, W.R.; Kim, H.J.; Therneau, T.M. Association between noninvasive fibrosis markers and mortality among adults with nonalcoholic fatty liver disease in the United States. Hepatology 2013, 57, 1357–1365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Stahl Eric, P.; Dhindsa Devinder, S.; Lee Suegene, K.; Sandesara Pratik, B.; Chalasani Naga, P.; Sperling Laurence, S. Nonalcoholic Fatty Liver Disease and the Heart. J. Am. Coll. Cardiol. 2019, 73, 948–963. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Summary of Mutual Exacerbation of Hepatic and Cardiac Disease Severity. Nonalcoholic fatty liver is associated with several cardiac diseases, including heart failure, cardiomyopathy, ischemic heart disease, and arrythmias. In contrast, there is a paucity of studies evaluating cardiac diseases leading to hepatic dysfunction.
Figure 1. Summary of Mutual Exacerbation of Hepatic and Cardiac Disease Severity. Nonalcoholic fatty liver is associated with several cardiac diseases, including heart failure, cardiomyopathy, ischemic heart disease, and arrythmias. In contrast, there is a paucity of studies evaluating cardiac diseases leading to hepatic dysfunction.
Jcm 10 01569 g001
Figure 2. Summary of suggested pathophysiological mechanisms underlying the cardio hepatic interactions in NAFLD, ischemic hepatitis and congestive hepatopathy with cardiovascular diseases. ADMA, asymmetric dimethyl arginine; CCL3, chemokine ligand 3; CO, cardiac output; CRP, C-reactive protein; EPCs, endothelial progenitor cells; HDL, high-density lipoproteins; HF, heart failure; HTN, hypertension; IL-1, interleukin 1; IL-6, interleukin 6; LDL, low-density lipoprotein; M1/M2, macrophage phenotype 1/2 ratio; NAFLD: non-alcoholic fatty liver disease; OxLDL oxidized low-density lipoprotein; sICAM, soluble intracellular adhesion molecule; TG, Triglycerides; TNF-α, tumor necrosis factor α; VLDL, very low-density lipoproteins.
Figure 2. Summary of suggested pathophysiological mechanisms underlying the cardio hepatic interactions in NAFLD, ischemic hepatitis and congestive hepatopathy with cardiovascular diseases. ADMA, asymmetric dimethyl arginine; CCL3, chemokine ligand 3; CO, cardiac output; CRP, C-reactive protein; EPCs, endothelial progenitor cells; HDL, high-density lipoproteins; HF, heart failure; HTN, hypertension; IL-1, interleukin 1; IL-6, interleukin 6; LDL, low-density lipoprotein; M1/M2, macrophage phenotype 1/2 ratio; NAFLD: non-alcoholic fatty liver disease; OxLDL oxidized low-density lipoprotein; sICAM, soluble intracellular adhesion molecule; TG, Triglycerides; TNF-α, tumor necrosis factor α; VLDL, very low-density lipoproteins.
Jcm 10 01569 g002
Table 1. Current strategies to diagnose and stage NAFLD.
Table 1. Current strategies to diagnose and stage NAFLD.
TestTypeInvasivenessAccuracy ^ in Steatosis Detection Liver Fat Detection Liver Fibrosis StagingReference
Liver biopsyHistology+++>99%[17]
MRI-PDFFImaging-98%[18]
USImaging-91% *, 93% **[19]
DGE-MRIImaging+83% *, 94% ***[20]
CTImaging+67% *, 90% ***[20]
US-FLIImaging-90%[21]
Fatty Liver IndexScore based on biochemical parameters-84%[22]
NAFLD Fat ScoreScore based on biochemical parameter-76%[23]
CAPImaging-76%[24]
Fib-4Score based on biochemical parameters-90%[25]
VCTEImaging-89%[26]
NAFLD Fibrosis Score Score based on biochemical parameters-86%[27]
APRIScore based on biochemical parameters-48%, 66%[28]
APRI, AST to Platelet ratio index; CT, Computed tomography; DGE-MRI, dual gradient echo magnetic resonance imaging; US, Ultrasonography; MRI-PDFF, MRI-based proton-density fat fraction; US-FLI, ultrasonographic fatty liver indicator; VCTE, Vibration-controlled transient elastography; CAP, Fibroscan Controlled Attenuation Parameter. ^ Accuracy was calculated as Accuracy = ((Sensitivity) * (Prevalence)) + ((Specificity) * (1 − Prevalence)) based on data derived from the referenced publications. * detection of ≥5% of steatosis ** detection of ≥10% of steatosis *** detection of ≥30% of steatosis.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ramadan, M.S.; Russo, V.; Nigro, G.; Durante-Mangoni, E.; Zampino, R. Interplay between Heart Disease and Metabolic Steatosis: A Contemporary Perspective. J. Clin. Med. 2021, 10, 1569. https://doi.org/10.3390/jcm10081569

AMA Style

Ramadan MS, Russo V, Nigro G, Durante-Mangoni E, Zampino R. Interplay between Heart Disease and Metabolic Steatosis: A Contemporary Perspective. Journal of Clinical Medicine. 2021; 10(8):1569. https://doi.org/10.3390/jcm10081569

Chicago/Turabian Style

Ramadan, Mohammad Said, Vincenzo Russo, Gerardo Nigro, Emanuele Durante-Mangoni, and Rosa Zampino. 2021. "Interplay between Heart Disease and Metabolic Steatosis: A Contemporary Perspective" Journal of Clinical Medicine 10, no. 8: 1569. https://doi.org/10.3390/jcm10081569

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