Review Article

Navigating the Link Between Non-alcoholic Fatty Liver Disease/Non-alcoholic Steatohepatitis and Cardiometabolic Syndrome

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

The global prevalence of non-alcoholic fatty liver disease (NAFLD) is nearly 25% and is increasing rapidly. The spectrum of liver damage in NAFLD ranges from simple steatosis to non-alcoholic steatohepatitis, characterised by the presence of lobular inflammation and hepatocyte ballooning degeneration, with or without fibrosis, which can further develop into cirrhosis and hepatocellular carcinoma. Not only is NAFLD a progressive liver disease, but numerous pieces of evidence also point to extrahepatic consequences. Accumulating evidence suggests that patients with NAFLD are also at increased risk of cardiovascular disease (CVD); in fact, CVDs are the most common cause of mortality in patients with NAFLD. Obesity, type 2 diabetes and higher levels of LDL are common risk factors in both NAFLD and CVD; however, how NAFLD affects the development and progression of CVD remains elusive. In this review, we comprehensively summarise current data on the key extrahepatic manifestations of NAFLD, emphasising the possible link between NAFLD and CVD, including the role of proprotein convertase substilisin/kenin type 9, extracellular vesicles, microbiota, and genetic factors.

Disclosure:MRG has received grants from Andalucía se mueve con Europa, Instituto de Salud Carlos III, Gilead, Intercept and Siemens; consulting fees from Abbvie, Alpha-sigma, Allergan, AstraZeneca, Axcella, BMS, Boehringer-Ingelheim, Gilead, Inventia, Kaleido, MSD, Novo-Nordisk, Pfizer and Proscientol; honoraria from Inventia, Novo-Nordisk, Rubió, Shionogi and Sobi; travel support from Abbvie and Gilead; and participates on an advisory board for Galmed. All other authors have no conflicts of
interest to declare.

Received:

Accepted:

Published online:

Funding:

This work was supported by Instituto de Salud Carlos III: PFIS FI20/00201 to SGZ, IFI22/00035 to VGF, Sara Borrell CD23-00024 to AGG; Junta de Andalucía: Talento Doctores DOC_00866 to RMH and Instituto de Salud Carlos III: PMP21/00078 and PI22/01342 to MRG.

Correspondence Details:Rocío Muñoz-Hernández, Calle Antonio Maura Montaner, s/n. Instituto de Biomedicina de Sevilla, lab 213, 41013 Sevilla, España. E: rociomunoz@us.es

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

Non-alcoholic fatty liver disease (NAFLD) is characterised by the presence of steatosis in more than 5% of hepatocytes, often associated with metabolic risk factors (particularly obesity and type 2 diabetes), and in the absence of excessive alcohol consumption or other chronic liver diseases.1 NAFLD serves as an umbrella term encompassing a wide range of clinicopathological findings. From a histological perspective, NAFLD represents a disease spectrum that includes steatosis, either with or without mild inflammation (known as non-alcoholic fatty liver), and a necroinflammatory subtype (known as non-alcoholic steatohepatitis [NASH]). NASH is further characterised by the presence of hepatocellular injury, marked by hepatocyte ballooning.2

NAFLD is the most common chronic liver disease in developed countries, affecting approximately 25% of the world’s adult population.3 The natural course of the disease moves towards NASH and cirrhosis, implying that NAFLD will be the leading cause of liver transplantation in the coming years.4 Sedentary behaviours and poor eating habits, in parallel with the growth of metabolic diseases, such as obesity and diabetes, are the main causes of recent increases in the prevalence of NAFLD.5 Therefore, the clinical and economic burdens of NAFLD not only depend on liver-related mortality, but also extrahepatic diseases (e.g. type 2 diabetes) and an increased risk of cardiovascular disease (CVD).6

In this sense, Stepanova et al. reported that patients with metabolic syndrome (MetS) had a 40% increased risk of NASH, and a recent meta-analysis found that 71% of patients with NASH had MetS, 82% had obesity, 44% had diabetes and 72% had hyperlipidemia.7,8 Based on these findings, the accumulation of visceral adipose tissue (VAT) that usually accompanies NAFLD may be responsible, in part, for the comorbidities, with the increased release of free fatty acids into the circulation leading to the development of hepatic insulin resistance and hepatosteatosis. Indeed, the alterations in hepatic lipid metabolism that lead to NAFLD also drive the development of atherogenic dyslipidaemia, especially elevated plasma triglyceride (TG) concentrations, remnant lipoprotein cholesterol levels and small dense LDL (sdLDL) particles that infiltrate the arterial wall and promote the development of atherosclerotic plaques.9 Furthermore, altered glucose metabolism and insulin resistance, which are also hallmarks of NAFLD, can further exacerbate CVD risk in patients with NAFLD.9 Moreover, the increased release of inflammatory cytokines from VAT and the liver observed in patients with overweight contributes to the development of insulin resistance and NAFLD.10

Although there is considerable evidence linking NAFLD to CVD, there are still unresolved questions regarding this relationship. The most important question is whether the association between the two conditions is due to shared risk factors (obesity or diabetes) or whether hepatic steatosis itself increases the risk of atherosclerosis.11,12 In addition, it remains difficult to predict which patients with NAFLD will develop severe liver complications or CVD. Therefore, there is increasing interest in identifying patient characteristics that can help predict the progression of these diseases; closer monitoring and the development of new biomarkers of subclinical atherosclerosis are needed to detect individuals at high cardiometabolic risk who are candidates for therapeutic interventions aimed at preventing the progression of NAFLD and atherosclerotic CVD.13 This review briefly summarises some of the mechanisms responsible for the association between NAFLD and CVD, including novel concepts such as extracellular vesicles (EVs), proprotein convertase subtilisin/kexin type 9 (PCSK9), the microbiome and genetics factors.

Non-alcoholic Fatty Liver Disease and the Prevalence of Clinical Cardiovascular Disease

Several recent studies have reported that the risk of cardiovascular events (fatal and non-fatal) is increased in patients with NAFLD, with either simple steatosis or NASH, independent of other cardiovascular risk factors.14–17 This suggests that NAFLD could enhance the risk already present because of the underlying cardiovascular risk factors (e.g. hypertension, diabetes and dyslipidaemia).18 Moreover, the risk of fatal cardiovascular events is increased in patients with severe NAFLD, and these events are the most common causes of morbidity and mortality in patients with NAFLD compared with other liver-related causes (incidence of 4.79 versus 0.77 per 1,000 person/years).1,9 In addition, the annual incidence of atherosclerotic CVD in patients with severe NAFLD has been estimated at around 1.1%, which, taking into account the increasing prevalence of NAFLD, is expected to grow notably in the coming years.19

According to recent literature, NAFLD is an independent risk factor for the development of MI. Indeed, Sinn et al. showed that the increased risk of MI was evident in individuals with either a high or low NAFLD fibrosis score.20 A study of 2,103 Italian outpatients with type 2 diabetes reported a significant association between NAFLD and incident CVD, defined as MI, ischaemic stroke, coronary revascularisation or cardiovascular death.21 This association appeared to be independent of a broad spectrum of risk factors, thus suggesting that NAFLD may confer an excess CVD risk over and above what would be expected because of the increased prevalence of the underlying metabolic risk factors.21 In addition, in a prospective observational study of 1,637 Japanese individuals from a health check-up program, Hamaguchi et al. observed that NAFLD was a predictor of CVD (defined as coronary heart disease, ischaemic stroke and cerebral haemorrhage) independent of conventional risk factors.22

Subclinical atherosclerosis, as an early stage of CVD, has considerable clinical significance. We previously reported that NAFLD was associated with 2.04-fold higher prevalence of pathological carotid intima–media thickness (IMT) and 4.41-fold higher risk of carotid plaques.23 Similarly, a prospective study in 3,343 patients with NAFLD diagnosed by hepatic ultrasound concluded that increased brachial–ankle pulse wave velocity was independently associated with a higher risk of NAFLD and fibrosis, whereas pathological carotid IMT was associated with incident NAFLD but not fibrosis.24

Socioeconomic and epidemiological factors, such as ethnicity and geographic differences, can influence various facets of health linked to fatty liver disease and cardiometabolic factors.25 In this context, Juonala et al. found a link between neighbourhood-related issues in Australia and less severe adiposity in children with obesity.26 That study also found that access to shopping facilities (shops or other retail services) was connected to an increased risk of dyslipidaemia and fatty liver.26 Recently, a significant correlation was established between skill level (skill level 1 covers the most basic positions; skill level 2 requires the effective comprehension of information like safety protocols and basic arithmetic; skill level 3 demands advanced abilities and specialised knowledge; skill level 4 entails solving intricate problems and making decisions grounded in extensive theoretical and experimental expertise within a specific area) and fatty liver index within a large middle-aged population, although no significant association was observed between sex and the presence of fatty liver.27

Recently, an international group of experts suggested that NAFLD be redefined as metabolic dysfunction-associated steatotic liver disease (MASLD).28 The potential effect of this change in nomenclature on the prediction of CVD risk remains uncertain. In a novel meta-analysis, a greater number of CVD events were identified in MASLD compared with NAFLD, although the risk of CVD incidents associated with either definition did not differ significantly. These findings underscore the importance of clinicians maintaining a strong suspicion that individuals with MASLD may also present with concurrent CVD.15

Mechanisms Linking Non-alcoholic Fatty Liver Disease, Endothelial Dysfunction and Atherosclerosis

The most frequent phenotype of NAFLD is associated with metabolic dysfunction, manifesting as obesity and/or diabetes and some of the features of MetS.22 To date, several dysregulated pathways have been proposed as links between NAFLD and cardiometabolic syndrome, such as changes in lipid metabolism, insulin resistance, de novo lipogenesis, endothelial dysfunction and oxidative stress, together with host genetics and changes in the gut microbiota (Figure 1).29,30

Figure 1: Mechanisms Potentially Responsible for Endothelial Dysfunction and Atherosclerosis in Non-alcoholic Fatty Liver Disease

Article image

Lipid Dysregulation

NAFLD is frequently associated with dyslipidaemia, a proatherogenic condition characterised by elevated levels of TG-rich lipoproteins (TRLs), lower HDL levels and an increase in circulating sdLDL, which are more atherogenic than normal-sized LDL.31 The increase in circulating sdLDL is a CVD risk factor and has been observed frequently in patients with obesity and MetS.32 Comparing NAFLD and NASH patients, higher sdLDL levels are found in NASH patients, who have a higher risk of CVD.32 In addition, NAFLD is associated with changes in the HDL profile, particularly smaller HDL particles that have poor functionality and are cleared more quickly, contributing to lower HDL and apolipoprotein A1 levels.33 Finally, NAFLD is associated with prolonged postprandial lipaemia, which contributes to hepatic TG content and is an independent cardiometabolic risk factor.34 Elevated levels of TRLs, particularly those containing apolipoprotein B (ApoB), are a hallmark of NAFLD and represent a biochemical marker for lipoprotein alterations and CVD risk. The impaired clearance of ApoB–TRL particles by the liver is a key factor in the development of hyperlipidaemia and hepatic fat accumulation.35 This lipid profile may also be a consequence of decreased lipoprotein lipase (LPL) activity due to increased circulating levels of the inhibitors angiopoietin-like 3 (ANGPTL3) and angiopoietin-like 4 (ANGPTL4), which are increased in patients with NAFLD.35 In summary, dyslipidaemias play a key role in the development of NAFLD; therefore, the use of appropriate lipid-lowering therapy is crucial in the therapeutic algorithm of NAFLD to arrest the progression of liver disease and the associated CVD.36

Proprotein convertase subtilisin/kexin type 9 (PCSK9), an enzyme mainly expressed in the liver, has emerged as a critical regulator of lipid homeostasis by acting as an inhibitor of LDL receptors, with a consequent increase in LDL levels and cardiovascular risk.37 In humans, PCSK9 loss-of-function mutations are associated with lower levels of cholesterol and protection against CVD.38,39 In contrast, a gain-of-function PCSK9 mutation was found to be associated with hypercholesterolaemia.38,39 Recently, PCSK9 has been linked to the severity of liver steatosis and the development of NASH, but the role of PCSK9 in NAFLD remains contentious.40 A recent study performed in NAFLD and obese patients after bariatric surgery found that the hepatic expression and circulating concentrations of PCSK9 were associated with the severity of steatosis and were inversely correlated with alanine transaminase (ALT) and aspartate transaminase (AST) levels; however, the study did not find a significant association with inflammation or ballooning.41 This supports, in agreement with other studies, the role of PCSK9 in liver fat accumulation and increased cardiovascular risk via increases in circulating cholesterol concentrations, but not in the progression of liver damage.42 Conversely, results from three cross-sectional studies from two hospitals in France with 132 patients did not find any association between circulating PCSK9 concentrations and histological liver features (Figure 2).43 Studies performed after the introduction of PCSK9 inhibitors could provide deeper insights. For example, Shafiq et al. reported a significant decrease in ALT and AST levels after 24 months of therapy with PCSK9 inhibitors in patients with NAFLD.44 In another study of 26 patients with familial hypercholesterolaemia, PCSK9 inhibitor therapy was found to significantly ameliorate steatosis in patients with low TG- HDL ratio (TG/HDL).45

Figure 2: Possible Effects of Proprotein Convertase Subtilisin/Kexin Type 9 on Non-alcoholic Fatty Liver Disease and Related Cardiovascular Disease

Article image

Insulin Resistance and De Novo Lipogenesis

Dysregulation of glucose metabolism and insulin resistance are crucial common factors in NAFLD and in the pathogenesis of CVD.46 The presence of obesity or ectopic fat deposition and impaired insulin action in adipocytes promote a failure in lipolysis suppression, adipocyte stress and the recruitment and infiltration of macrophages, with the consequent release of proinflammatory cytokines, hormones and adipocytokines.47 The hyperinsulinaemia that accompanies insulin resistance promotes de novo lipogenesis and the maintenance of high glucose concentrations, which are responsible for the atherogenic dyslipidaemia that increases the risk of CVD.48 Insulin resistance also drives hepatic de novo lipogenesis, which contributes significantly to liver TG content, together with disturbed fatty acid oxidation (FAO) or altered secretion of very low-density lipoprotein (VLDL).49

Insulin activates transcription factors such as sterol regulatory element-binding protein 1c (SREBP-1c) and carbohydrate-responsive element-binding protein (ChREBP), regulating several enzymes involved in lipogenesis and pyruvate kinase, a master regulator of glycolysis.50 Indeed, diabetes is considered the most important atherosclerotic factor in NAFLD. Moreover, some glucose-lowering drugs, such as pioglitazone, have demonstrated promising benefits in ameliorating NAFLD.51 In addition, a recent meta-analysis showed that glucagon-like peptide 1 (GLP-1) and sodium–glucose cotransporter 2 inhibitors reduced the atherosclerotic risk.52

Thromboembolic Events

Targher et al. reported that the inflammatory profile associated with NASH may also play a pathogenic role in CVD.21 So, in the presence of systemic inflammation, the liver could act as a target of and a contributor to inflammatory changes. Hepatic steatosis is associated with the increased production of interleukin-6 and tumour necrosis factor-α by hepatocytes, Kupffer cells and stellate cells, and this contributes to the local activation of nuclear factor-κB.53 The biological link between NAFLD and CVD also includes prothrombotic factors. Venous thrombosis, a complex condition with multiple contributing factors, stands out as one of the most prevalent vascular diseases. The involvement of endothelial dysfunction plays a crucial role in the development of venous thrombosis.54,55 Acute and chronic liver diseases are associated with coagulation disorders.56 In this context, patients with NAFLD can show a prothrombotic state, and previous studies reported that NASH is an independent factor for the development of deep vein thrombosis and/or pulmonary embolism among patients with cirrhosis.57,58

Oxidative Stress and Endothelial Dysfunction

Oxidative stress is another factor involved in the pathogenesis of CVD, also in close relation to other chronic diseases, such as diabetes, MetS, obesity and NAFLD. In NAFLD, the excessive load of fatty acids in hepatocytes induces upregulation of the mitochondrial respiratory chain, resulting in overproduction of reactive oxygen species (ROS), exceeding the antioxidant capacity and leading to oxidative stress.52 The accumulation of ROS can activate Kupffer cells and stellate cells in the liver, leading to fibrosis and the progression of NAFLD. Indeed, the levels of circulating biomarkers of oxidative stress, such as urinary 8-iso-prostaglandin F2α and serum Nox2-derived peptide, are correlated with the severity of NASH.59

The accumulation of plasma oxidative stress contributes to the development of CVD by promoting endothelial dysfunction. The endothelium is a vital component of the vascular wall, and endothelial dysfunction, which is a hallmark of vascular diseases, is considered a marker of early atherosclerosis.60 Risk factors for atherosclerosis, such as hyperlipidaemia, hypertension, diabetes, smoking and infections, are common in patients with NAFLD. These factors can directly or indirectly stress the arterial endothelium, resulting in dysfunction or damage.61 Several pieces of evidence show that markers of endothelial dysfunction, such as flow-mediated dilatation percentage, are significantly impaired in patients with NAFLD, with a significant gradient according to the severity of liver disease.62 In addition, mean IMT, used as a non-invasive marker of endothelial dysfunction and arteriopathy, was significantly increased in patients with than without NAFLD.62 Patients with NAFLD had more carotid atherosclerosis than those without NAFLD, with enlarged mean IMT and higher plaque prevalence.63 This suggests that patients with NAFLD have an increased risk of CVD due to endothelial dysfunction.

Dysregulated Hepatokines

Emerging research has highlighted the role of dysregulated hepatokines in the pathogenesis of CVD in individuals with NAFLD.64 Fetuin-A and fibroblast growth factor 21 (FGF21) are implicated in insulin resistance, which is a major risk factor for both NAFLD and CVD.65,66 Conversely, FGF21 may reduce the risk of atherosclerosis by lowering inflammation, regulating lipid metabolism and adiponectin expression.67 ANGPTL3 can inhibit LPL activity.68 LPL activity encourages the healthy storage of TGs in the gluteofemoral compartment rather than in the pathological visceral compartment, thereby enhancing lipid metabolism and insulin sensitivity.69 Thus, elevated ANGPTL3 may contribute to cardiometabolic risk by inhibiting LPL. Finally, it has been shown that serum hepatocyte growth factor concentrations are elevated in patients with essential hypertension and extreme obesity, suggesting the role of hepatocyte growth factor in the pathophysiology of MetS and insulin resistance, and therefore in the increased cardiovascular risk.70

Genetics

Several pieces of evidence suggest that the NAFLD phenotype and differences in its progression are the result of complex interactions between the environment and an individual’s genetic pool. Recent studies have investigated the role of genetic polymorphisms associated with insulin signalling, type 2 diabetes, hypertension and lipid metabolism, all of which are shared risk factors for both NAFLD and CVD.71

The I148M variant of patatin-like phospholipase domain-containing protein 3 (PNPLA3) is the most known inherited determinant of NAFLD because it is associated with the progression of NAFLD, NASH and NAFLD-related hepatocellular carcinoma.72,73 This polymorphism induces a loss of function in enzyme activity, resulting in the accumulation of triacylglycerol in the liver and the entrapment of TG in lipid droplets in hepatocytes and hepatic stellate cells.69,74 This leads to accumulation of TG in liver cells, but a reduced secretion of VLDL into the circulation, which may reduce the deposition of lipids in the wall of blood vessels, reducing cardiovascular mortality.67 Supporting this scenario, extensive genomic research has linked the PNPLA3 I148M variant to a reduced risk of CVD.75

The transmembrane 6 superfamily member 2 (TM6SF2) E167K variant has been associated with higher TG levels in the liver and a greater risk of advanced fibrosis. It has also been reported to reduce ApoB lipidation, promoting a reduction in VLDL secretion from the liver; thus, controversially, patients harbouring this polymorphism showed a reduced cardiovascular risk.69,76 However, TM6SF2 upregulation may protect against cardiovascular events by reducing liver fat and increasing levels of circulating lipoproteins.77

Membrane-bound O-acyltransferase domain-containing (MBOAT7) is a gene implicated in the remodelling of phosphatidylinositol (and other phospholipids) via the incorporation of arachidonic acid and other unsaturated fatty acids into lysophospholipids. The common genetic variant leads to downregulation of MBOAT7 activity, and consequently to the accumulation of lysophosphatidylinositol in hepatocytes; this leads to a higher synthesis of TGs in the liver and NAFLD.69,78 Results regarding MBOAT7 and CVD are contentious; although Ismaiel and Dumitrascu reported no evidence of an association between MBOAT7 and cardiovascular risk, Xu et al. recently reported that MBOAT7 rs641738 (C>T) was associated with reduced metabolic traits and type 2 diabetes in elderly Chinese with NAFLD.79,80

In summary, several lines of evidence support that genetic variants associated with NAFLD are linked to a decreased risk of CVD or, at the very least, do not increase susceptibility to CVD. The observed connection between fatty liver and cardiovascular complications in epidemiological studies is likely primarily mediated by dyslipidaemia and insulin resistance, which are well-established risk factors for atherosclerosis. To definitively determine whether NAFLD genetics can impact CVD, we should take into account other genetic variations that predispose to NAFLD without affecting lipid secretion and plasma cholesterol and TG concentrations.81

Gut Microbiota

The gut microbiota is a complex ecosystem formed by different microorganisms, including bacteria, viruses, fungi and protozoa, that coexist in our gut through intricate relationships that also involve interactions with different organ systems of the host. The way in which these microorganisms behave, the metabolites they produce and the relative abundance of certain taxa have lately been related to health and disease.82 Disturbances to this symbiosis, known as dysbiosis, can alter the metabolic capacity of the system and impact the immune function of the host.83 Moreover, the gut microbiota can cause liver lipotoxicity and translocation of dysbiotic microorganisms or their products by increasing the permeability of the intestine barrier, inducing changes in the proinflammatory and anti-inflammatory balance in the liver, which has been demonstrated to be essential for the development of NASH.84 Indeed, disruption of the gut barrier seems to be a link between NAFLD and CVD because bacterial DNA was found in human atherosclerotic plaques.85 In addition, lipopolysaccharide, a component of the wall of Gram-negative bacteria whose levels are increased in NASH patients, can bind the endothelial surface, inducing an inflammatory response and endothelial dysfunction.86,87

Under normal conditions, up to 90% of the gut microbiota is formed by members of Bacteroidetes and Firmicutes, but an increase in the Firmicutes/Bacteroidetes ratio has been linked to CVD and NAFLD.88,89 The abundance of different taxa in the gut microbiota has been linked to CVD. On the one hand, Roseburia intestinalis and Faecalibacterium prausnitzii are both related to the amelioration of atherosclerosis, with R. intestinalis increasing fatty acid metabolism and reducing the inflammatory response in mice.90 On the other hand, Enterobacteriaceae, Ruminococcus gnavus and Eggerthella lenta are increased in individuals with atherosclerosis.91 Several taxa have also been shown to be related to NAFLD and its progression.92 However, technical and methodological challenges make comparisons between studies unreliable in most cases for analysing specific taxa. Bigger and better-designed studies in the future will allow a common nexus in the development of NAFLD and CVD to be found and, more importantly, how the functionality of the microbiota impact disease risk to be determined.

In this sense, metabolites produced by the gut microbiota can pass into the circulation, featuring systemic bioactive effects with inflammatory and metabolic properties. According to several studies, high circulating concentrations of trimethylamine N-oxide (TMAO) increase the risk of CVD, diabetes, NAFLD and other metabolic diseases.87,93,95 Trimethylamine (TMA), which is produced via metabolism of choline or l-carnitine by the gut microbiota, is oxidised to TMAO in the liver.95 TMAO has a proinflammatory effect, and studies have demonstrated that it is associated with cardiovascular events, NASH diagnosis and all-cause mortality.96 TMAO induces proatherogenic and prothrombotic mechanisms by promoting macrophage foam cell formation, decreasing liver cholesterol metabolism, modifying reverse cholesterol transport and causing endothelial dysfunction and platelet activation and aggregation.86 TMAO is related to endothelial cell dysfunction by elevating interleukin 6, C-reactive protein, tumour necrosis factor-α and ROS, while reducing nitric oxide production.97 Controversially, choline, from which TMA is produced, is essential for VLDL production and choline deficiency can lead to NAFLD by inducing hepatosteatosis and oxidative stress.98

Other microbially derived metabolites released by the microbiota after metabolising complex carbohydrates, such as short-chain fatty acids, can have an impact on the risk of CVD. These metabolites have been shown to improve lipid metabolism (decreasing LDL and TGs while increasing HDL), reduce inflammation and improve insulin sensitivity, all of which are risk factors for CVD and associated with NAFLD.99 In addition, by binding G protein-coupled receptor 41 and G protein-coupled receptor 43, these metabolites can activate the release of GLP-1 and peptide YY, regulating glucose homeostasis and appetite.100

Endothelium-derived Microvesicles as Biomarkers of Endothelial Dysfunction and Atherosclerosis in Patients With NAFLD

EVs, which are important mediators of intercellular communication, are lipid bilayer-delimited particles naturally released from almost all cell types that contribute to the pathogenesis, initiation and progression of several liver diseases and could be one of the mechanisms linking steatosis to the progression of atherosclerosis and endothelial dysfunction in patients with NAFLD.101 Growing evidence suggests that endothelium-derived microvesicles (EMVs; accounting for ~5–15% of all microvesicles) may be useful as novel biomarkers for CVD and metabolic diseases in adults, dyslipidaemia, endothelial dysfunction and MetS.102–105 In this context, Amabile et al. found that plasma EMV concentrations are associated with several cardiovascular risk factors, including higher TG concentrations, hypertension and MetS.103 We previously reported that plasma EMV concentrations decreased and endothelial dysfunction (measured by laser Doppler flowmetry) improved after liver disease improved in patients with chronic hepatitis C.106 In this context, EVs have been proposed as molecular mechanisms linking steatosis to the progression of atherosclerosis in patients with NAFLD. Jiang et al. isolated, quantified and characterised EVs from steatotic hepatocytes and demonstrated an inflammatory effect on endothelial cells via miR-1.107 These findings provide insights into the important role of EVs in the interaction between the liver and the vasculature. In agreement with the study of Jiang et al., Chen et al. isolated and characterised EVs from steatotic hepatocytes and concluded that these EVs promote foam cell formation and atherogenesis via the miR-30a-3p/ATP-binding cassette subfamily A member 1 transporter axis.108

Non-pharmacological Interventions and Their Potential Impact on Cardiometabolic Disease in Patients With NAFLD

A low-quality diet with elevated consumption of saturated fats and processed meat is a shared risk factor for the development of NAFLD, metabolic disorder and cardiovascular and all-cause mortality, confirming the complex interconnection between diet, metabolism, metabolic liver diseases and the cardiovascular system.109–111

In the absence of an approved effective drug for the treatment of NAFLD, the current first-line treatment for NAFLD consists of lifestyle modifications, including dietary, physical activity and weight-loss interventions.112 Following a calorie-restricted diet for a long period is linked to a reduction in cardiovascular risk and mobilisation of liver fat.113 However, only a low percentage of patients manage to stay on such a diet for a long period of time.114 Nevertheless, patients eating a Mediterranean diet showed improvements in steatosis independent of weight loss.115 In addition, another study found a reduced incidence of CVD in patients on a Mediterranean diet supplemented with either extra virgin olive oil or nuts compared with patients on a low-fat diet.116 Despite not achieving the goal of weight loss, a benefit on serum liver enzymes and the extension of steatosis have been reported with exercise alone.117 Most patients with NAFLD have a higher risk of MetS due to their lack of adherence to physical activity. Physical activity seems to be inversely associated with the prevalence of NAFLD, whereas BMI category is directly linked to NAFLD.118 Accordingly, a study in a Korean population showed a lower prevalence of or an improvement in NAFLD when patients performed moderate exercise five or more times per week.119 Furthermore, smoking should be avoided given that smoking is a risk factor for CVD and has been related to the progression of NAFLD.120 Thus far, merging diet and physical activity changes is the best advice for weight reduction in patients with NAFLD, also decreasing the risk of MetS.115 This approach boosts the reduction in CVD risk due to amelioration of the atherogenic risk and myocardial structure and function.121

Pharmacological Interventions and Their Potential Impact on Cardiometabolic Disease in Patients With Non-alcoholic Fatty Liver Disease

Several pharmaceutical drugs have been evaluated for the treatment of NAFLD. However, no single therapy has been approved thus far, although clinical trials are ongoing. Many of these clinical trials are based on peroxisome proliferator-activated receptor (PPAR) agonists.51,122–124 There are three types of PPAR nuclear receptors, namely PPARα, PPARδ and PPARγ, with PPARα being highly expressed in the liver.125 Activation of PPARα receptors stimulates FAO, thus protecting hepatocytes against oxidative stress-induced damage.125 In contrast, PPARγ is mainly located in the pancreas and in adipose tissue, with activation of PPARγ receptors promoting adipocyte differentiation, FAO levels in the liver and insulin sensitivity.125 PPARδ is ubiquitously expressed, and its function is to inhibit the oxidation of non-esterified fatty acids and lipogenesis. Moreover, PPARδ enhances anti-inflammatory outcomes.125 In a Phase 4 clinical trial, 58% of patients with NASH and diabetes achieved the primary outcome (reduction of at least 2 points in 2 histologic categories of the NAS without worsening of fibrosis after 18 months of therapy) after being treated pioglitazone, a first-generation PPARγ agonist (thiazolidinedione).51 Pioglitazone treatment improved hepatic steatosis, inflammation and swelling while decreasing TG concentrations and increasing HDL concentrations.51 The most important mechanism responsible for this improvement was the redistribution of lipids towards the subcutaneous adipose tissue.126 Pirfenidone, a PPARα agonist, inhibits adipogenesis and fibrosis.122 In the PROMETEO clinical trial (Phase 2), more than one-third of patients exhibited an improvement in fibrosis.123 Activation of PPARα has been related to improvements in atherosclerotic dyslipidaemia, inhibition of vascular inflammation and macrophage foam cell formation.121 This means that PPARα activation can prevent atherosclerotic progression and is why treatments for NAFLD also have effects on atherosclerotic cardiovascular disease (ASCVD).121

There are some treatments that include molecules acting on more than one receptor, and they are more potent. Lanifibranor, a pan-PPAR agonist, is able to activate PPARα, PPARδ and PPARγ. In a randomised control trial, lanifibranor treatment reduced steatohepatitis activity and improved fibrosis.124 Liver enzymes decreased and lipid biomarkers, inflammation and fibrosis were ameliorated.124 PPAR agonists also have benefits in the regulation of lipid metabolism, which means that not only can they be used for patients with NAFLD, but they can also be used to reduce ASCVD risk.121

Conclusion

NAFLD is a public health problem affecting up to one-third of the world’s adult population and is considered a multisystemic disease associated with extrahepatic alterations. NAFLD, especially in its more severe form (NASH), is associated with higher risk of CVD given the close association with cardiometabolic risk factors, with CVD being the leading cause of death among patients with NASH. There have been many studies investigating whether NAFLD contributes to cardiovascular risk, and it is plausible that treatment of liver disease may ameliorate the cardiovascular risk. Because there is no approved pharmacotherapy for NAFLD, these patients should be closely monitored and regularly screened for cardiometabolic risk factors. Treatment should focus on lifestyle interventions and lipid-lowering therapies, which will not only improve the liver, but also ameliorate the cardiovascular risk factors.

As described in this review, there are multiple factors in the aetiopathogenesis of cardiometabolic alterations associated with liver disease that need to be considered to determine the risk of cardiovascular disease in patients with NAFLD: genetic variants that serve as predictors of NAFLD are linked to a decreased risk of cardiovascular disease; TMAO, which is produced by the gut microbiota and oxidised in the liver, reduces nitric oxide production, enhancing endothelial dysfunction; and recent studies highlight the role of EVs in the progression of NAFLD-driven atherosclerosis. The development of precision medicine based on the development of biomarkers and therapeutic approaches considering proteomics, metabolomics, the microbiome and genetics (all of which are involved in the development of NAFLD and cardiovascular disease) would improve the management of patients with NAFLD.

References

  1. European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD), European Association for the Study of Obesity (EASO). EASL–EASD–EASO clinical practice guidelines for the management of non-alcoholic fatty liver disease. J Hepatol 2016;64:1388–402. 
    Crossreff | PubMed
  2. Powell EE, Wong VW, Rinella M. Non-alcoholic fatty liver disease. Lancet 2021;397:2212–24. 
    Crossreff | PubMed
  3. Younossi Z, Anstee QM, Marietti M, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol 2018;15:11–20. 
    Crossreff | PubMed
  4. Charlton MR, Burns JM, Pedersen RA, et al. Frequency and outcomes of liver transplantation for nonalcoholic steatohepatitis in the United States. Gastroenterology 2011;141:1249–53. 
    Crossreff | PubMed
  5. Ballestri S, Zona S, Targher G, et al. Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome. Evidence from a systematic review and meta-analysis. J Gastroenterol Hepatol 2016;31:936–44. 
    Crossreff | PubMed
  6. Musso G, Cassader M, Rosina F, Gambino R. Impact of current treatments on liver disease, glucose metabolism and cardiovascular risk in non-alcoholic fatty liver disease (NAFLD): a systematic review and meta-analysis of randomised trials. Diabetologia 2012;55:885–904. 
    Crossreff | PubMed
  7. Stepanova M, Rafiq N, Younossi ZM. Components of metabolic syndrome are independent predictors of mortality in patients with chronic liver disease: a population-based study. Gut 2010;59:1410–5. 
    Crossreff | PubMed
  8. Younossi ZM, Koenig AB, Abdelatif D, et al. Global epidemiology of nonalcoholic fatty liver disease – meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016;64:73–84. 
    Crossreff | PubMed
  9. Deprince A, Haas JT, Staels B. Dysregulated lipid metabolism links NAFLD to cardiovascular disease. Mol Metab 2020;42:101092. 
    Crossreff | PubMed
  10. Hanlon CL, Yuan L. Nonalcoholic fatty liver disease: the role of visceral adipose tissue. Clin Liver Dis (Hoboken) 2022;19:106–10. 
    Crossreff | PubMed
  11. Stefan N, Cusi K. A global view of the interplay between non-alcoholic fatty liver disease and diabetes. Lancet Diabetes Endocrinol 2022;10:284–96. 
    Crossreff | PubMed
  12. Liu H, Lu HY. Nonalcoholic fatty liver disease and cardiovascular disease. World J Gastroenterol 2014;20:8407–15. 
    Crossreff | PubMed
  13. Janssen A, Grobbee DE, Dendale P. Non-alcoholic fatty liver disease, a new and growing risk indicator for cardiovascular disease. Eur J Prev Cardiol 2020;27:1059–63. 
    Crossreff | PubMed
  14. Mantovani A, Csermely A, Petracca G, et al. Non-alcoholic fatty liver disease and risk of fatal and non-fatal cardiovascular events: an updated systematic review and meta-analysis. Lancet Gastroenterol Hepatol 2021;6:903–13. 
    Crossreff | PubMed
  15. Mantovani A, Csermely A, Tilg H, et al. Comparative effects of non-alcoholic fatty liver disease and metabolic dysfunction-associated fatty liver disease on risk of incident cardiovascular events: a meta-analysis of about 13 million individuals. Gut 2023;72:1433–6. 
    Crossreff | PubMed
  16. Liu Y, Zhong GC, Tan HY, et al. Nonalcoholic fatty liver disease and mortality from all causes, cardiovascular disease, and cancer: a meta-analysis. Sci Rep 2019;9:11124. 
    Crossreff | PubMed
  17. Anstee QM, Targher G, Day CP. Progression of NAFLD to diabetes mellitus, cardiovascular disease or cirrhosis. Nat Rev Gastroenterol Hepatol 2013;10:330–44. 
    Crossreff | PubMed
  18. Targher G, Marra F, Marchesini G. Increased risk of cardiovascular disease in non-alcoholic fatty liver disease: causal effect or epiphenomenon? Diabetologia 2008;51:1947–53. 
    Crossreff | PubMed
  19. Targher G, Byrne CD, Lonardo A, et al. Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: a meta-analysis. J Hepatol 2016;65:589–600. 
    Crossreff | PubMed
  20. Sinn DH, Kang D, Chang Y, et al. Non-alcoholic fatty liver disease and the incidence of myocardial infarction: a cohort study. J Gastroenterol Hepatol 2020;35:833–9. 
    Crossreff | PubMed
  21. Targher G, Bertolini L, Rodella S, et al. Nonalcoholic fatty liver disease is independently associated with an increased incidence of cardiovascular events in type 2 diabetic patients. Diabetes Care 2007;30:2119–21. 
    Crossreff | PubMed
  22. Hamaguchi M, Kojima T, Takeda N, et al. Nonalcoholic fatty liver disease is a novel predictor of cardiovascular disease. World J Gastroenterol 2007;13:1579–84. 
    Crossreff | PubMed
  23. Ampuero J, Gallego-Durán R, Romero-Gómez M. Association of NAFLD with subclinical atherosclerosis and coronary-artery disease: meta-analysis. Rev Esp Enferm Dig 2015;107:10–6 
    PubMed
  24. Xin Z, Zhu Y, Wang S, et al. Associations of subclinical atherosclerosis with nonalcoholic fatty liver disease and fibrosis assessed by non-invasive score. Liver Int 2020;40:806–14. 
    Crossreff | PubMed
  25. Kivimäki M, Vahtera J, Tabák AG, et al. Neighbourhood socioeconomic disadvantage, risk factors, and diabetes from childhood to middle age in the Young Finns Study: a cohort study. Lancet Public Health 2018;3:e365–73. 
    Crossreff | PubMed
  26. Juonala M, Harcourt BE, Saner C, et al. Neighbourhood socioeconomic circumstances, adiposity and cardiometabolic risk measures in children with severe obesity. Obes Res Clin Pract 2019;13:345–51. 
    Crossreff | PubMed
  27. Sadeghianpour Z, Cheraghian B, Farshchi HR, Asadi-Lari M. Non-alcoholic fatty liver disease and socioeconomic determinants in an Iranian cohort study. BMC Gastroenterol 2023;23:350. 
    Crossreff | PubMed
  28. Rinella ME, Lazarus JV, Ratziu V, et al. A multisociety Delphi consensus statement on new fatty liver disease nomenclature. Hepatology 2023;78:1966–86. 
    Crossreff | PubMed
  29. Villanova N, Moscatiello S, Ramilli S, et al. Endothelial dysfunction and cardiovascular risk profile in nonalcoholic fatty liver disease. Hepatology 2005;42:473–80. 
    Crossreff | PubMed
  30. Schindhelm RK, Diamant M, Bakker SJ, et al. Liver alanine aminotransferase, insulin resistance and endothelial dysfunction in normotriglyceridaemic subjects with type 2 diabetes mellitus. Eur J Clin Investig 2005;35:369–74. 
    Crossreff | PubMed
  31. Peng K, Mo Z, Tian G. Serum lipid abnormalities and nonalcoholic fatty liver disease in adult males. Am J Med Sci 2017;353:236–41. 
    Crossreff | PubMed
  32. Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty liver disease and dyslipidemia: an update. Metabolism 2016;65:1109–23. 
    Crossreff | PubMed
  33. Yang MH, Sung J, Gwak GY. The associations between apolipoprotein B, A1, and the B/A1 ratio and nonalcoholic fatty liver disease in both normal-weight and overweight Korean population. J Clin Lipidol 2016;10:289–98. 
    Crossreff | PubMed
  34. Velenosi TJ, Ben-Yakov G, Podszun MC, et al. Postprandial plasma lipidomics reveal specific alteration of hepatic-derived diacylglycerols in nonalcoholic fatty liver disease. Gastroenterology 2022;162:1990–2003. 
    Crossreff | PubMed
  35. Harada M, Yamakawa T, Kashiwagi R, et al. Association between ANGPTL3, 4, and 8 and lipid and glucose metabolism markers in patients with diabetes. PLoS One 2021;16:e0255147. 
    Crossreff | PubMed
  36. Anwar SD, Foster C, Ashraf A. Lipid disorders and metabolic-associated fatty liver disease. Endocrinol Metab Clin North Am 2023;52:445–57. 
    Crossreff | PubMed
  37. Seidah NG, Abifadel M, Prost S, et al. The proprotein convertases in hypercholesterolemia and cardiovascular diseases: emphasis on proprotein convertase subtilisin/kexin 9. Pharmacol Rev 2017;69:33–52. 
    Crossreff | PubMed
  38. Lambert G, Sjouke B, Choque B, et al. The PCSK9 decade. J Lipid Res 2012;53:2515–24. 
    Crossreff | PubMed
  39. Cohen JC, Boerwinkle E, Mosley TH Jr, Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med 2006;354:1264–72. 
    Crossreff | PubMed
  40. Momtazi-Borojeni AA, Banach M, Ruscica M, Sahebkar A. The role of PCSK9 in NAFLD/NASH and therapeutic implications of PCSK9 inhibition. Expert Rev Clin Pharmacol 2022;15:1199–208. 
    Crossreff | PubMed
  41. Emma MR, Giannitrapani L, Cabibi D, et al. Hepatic and circulating levels of PCSK9 in morbidly obese patients: relation with severity of liver steatosis. Biochim Biophys Acta Mol Cell Biol Lipids 2020;1865:158792. 
    Crossreff | PubMed
  42. Ruscica M, Ferri N, Macchi C, et al. Liver fat accumulation is associated with circulating PCSK9. Ann Med 2016;48:384–91. 
    Crossreff | PubMed
  43. Wargny M, Ducluzeau PH, Petit JM, et al. Circulating PCSK9 levels are not associated with the severity of hepatic steatosis and NASH in a high-risk population. Atherosclerosis 2018;278:82–90. 
    Crossreff | PubMed
  44. Shafiq M, Walmann T, Nutalapati V, et al. Effects of proprotein convertase subtilisin/kexin type-9 inhibitors on fatty liver. World J Hepatol 2020;12:1258–66. 
    Crossreff | PubMed
  45. Scicali R, Di Pino A, Urbano F, et al. Analysis of steatosis biomarkers and inflammatory profile after adding on PCSK9 inhibitor treatment in familial hypercholesterolemia subjects with nonalcoholic fatty liver disease: A single lipid center real-world experience. Nutr Metab Cardiovasc Dis 2021;31:869–79. 
    Crossreff | PubMed
  46. Stahl EP, Dhindsa DS, Lee SK, et al. Nonalcoholic fatty liver disease and the heart: JACC state-of-the-art review. J Am Coll Cardiol 2019;73:948–63. 
    Crossreff | PubMed
  47. Jepsen P, Vilstrup H, Mellemkjaer L, et al. Prognosis of patients with a diagnosis of fatty liver – a registry-based cohort study. Hepatogastroenterology 2003;50:2101–4
    PubMed
  48. Dongiovanni P, Rametta R, Meroni M, Valenti L. The role of insulin resistance in nonalcoholic steatohepatitis and liver disease development – a potential therapeutic target? Expert Rev Gastroenterol Hepatol 2016;10:229–42. 
    Crossreff | PubMed
  49. Nguyen P, Leray V, Diez M, et al. Liver lipid metabolism. J Anim Physiol Anim Nutr (Berl) 2008;92:272–83. 
    Crossreff | PubMed
  50. Dentin R, Girard J, Postic C. Carbohydrate responsive element binding protein (ChREBP) and sterol regulatory element binding protein-1c (SREBP-1c): two key regulators of glucose metabolism and lipid synthesis in liver. Biochimie 2005;87:81–6. 
    Crossreff | PubMed
  51. Cusi K, Orsak B, Bril F, et al. Long-term pioglitazone treatment for patients with nonalcoholic steatohepatitis and prediabetes or type 2 diabetes mellitus: a randomized trial. Ann Intern Med 2016;165:305–15. 
    Crossreff | PubMed
  52. Palmer SC, Tendal B, Mustafa RA, et al. Sodium–glucose cotransporter protein-2 (SGLT-2) inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists for type 2 diabetes: systematic review and network meta-analysis of randomised controlled trials. BMJ 2021;372:m4573. 
    Crossreff | PubMed
  53. Sabio G, Das M, Mora A, et al. A stress signaling pathway in adipose tissue regulates hepatic insulin resistance. Science 2008;322:1539–43. 
    Crossreff | PubMed
  54. Koliaki C, Szendroedi J, Kaul K, et al. Adaptation of hepatic mitochondrial function in humans with non-alcoholic fatty liver is lost in steatohepatitis. Cell Metab 2015;21:739–46. 
    Crossreff | PubMed
  55. Thomas DP, Merton RE, Wood RD, Hockley DJ. The relationship between vessel wall injury and venous thrombosis: an experimental study. Br J Haematol 1985;59:449–57. 
    Crossreff | PubMed
  56. Amitrano L, Guardascione MA, Brancaccio V, Balzano A. Coagulation disorders in liver disease. Semin Liver Dis 2002;22:83–96. 
    Crossreff | PubMed
  57. Tripodi A, Fracanzani AL, Primignani M, et al. Procoagulant imbalance in patients with non-alcoholic fatty liver disease. J Hepatol 2014;61:148–54. 
    Crossreff | PubMed
  58. Stine JG, Niccum BA, Zimmet AN, et al. Increased risk of venous thromboembolism in hospitalized patients with cirrhosis due to non-alcoholic steatohepatitis. Clin Transl Gastroenterol 2018;9:140. 
    Crossreff | PubMed
  59. Del Ben M, Polimeni L, Carnevale R, et al. NOX2-generated oxidative stress is associated with severity of ultrasound liver steatosis in patients with non-alcoholic fatty liver disease. BMC Gastroenterol 2014;14:81. 
    Crossreff | PubMed
  60. Hooglugt A, Klatt O, Huveneers S. Vascular stiffening and endothelial dysfunction in atherosclerosis. Curr Opin Lipidol 2022;33:353–63. 
    Crossreff | PubMed
  61. Al Rifai M, Silverman MG, Nasir K, et al. The association of nonalcoholic fatty liver disease, obesity, and metabolic syndrome, with systemic inflammation and subclinical atherosclerosis: the Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis 2015;239:629–33. 
    Crossreff | PubMed
  62. Mohammadi A, Sedani HH, Ghasemi-Rad M. Evaluation of carotid intima–media thickness and flow-mediated dilatation in middle-aged patients with nonalcoholic fatty liver disease. Vasc Health Risk Manag 2011;7:661–5. 
    Crossreff | PubMed
  63. Kumari S, Porwal YC, Gupta R. Correlation between carotid intima media thickness and non-alcoholic fatty liver disease. J Assoc Physicians India 2022;70:11–2PubMed
  64. Stefan N, Schick F, Birkenfeld AL, et al. The role of hepatokines in NAFLD. Cell Metab 2023;35:236–52. 
    Crossreff | PubMed
  65. Sardana O, Goyal R, Bedi O. Molecular and pathobiological involvement of fetuin-A in the pathogenesis of NAFLD. Inflammopharmacology 2021;29:1061–74. 
    Crossreff | PubMed
  66. Falamarzi K, Malekpour M, Tafti MF, et al. The role of FGF21 and its analogs on liver associated diseases. Front Med (Lausanne) 2022;9:967375. 
    Crossreff | PubMed
  67. Tabari FS, Karimian A, Parsian H, et al. The roles of FGF21 in atherosclerosis pathogenesis. Rev Endocr Metab Disord 2019;20:103–14. 
    Crossreff | PubMed
  68. Jensen-Cody SO, Potthoff MJ. Hepatokines and metabolism: deciphering communication from the liver. Mol Metab 2021;44:101138. 
    Crossreff | PubMed
  69. Tall AR. Increasing lipolysis and reducing atherosclerosis. N Engl J Med 2017;377:280–3. 
    Crossreff | PubMed
  70. Hiratsuka A, Adachi H, Fujiura Y, et al. Strong association between serum hepatocyte growth factor and metabolic syndrome. J Clin Endocrinol Metab 2005;90:2927–31. 
    Crossreff | PubMed
  71. Jonas W, Schürmann A. Genetic and epigenetic factors determining NAFLD risk. Mol Metab 2021;50:101111. 
    Crossreff | PubMed
  72. Dongiovanni P, Romeo S, Valenti L. Genetic factors in the pathogenesis of nonalcoholic fatty liver and steatohepatitis. BioMed Res Int 2015;2015:460190. 
    Crossreff | PubMed
  73. Eslam M, Valenti L, Romeo S. Genetics and epigenetics of NAFLD and NASH: clinical impact. J Hepatol 2018;68:268–79. 
    Crossreff | PubMed
  74. Huang Y, Cohen JC, Hobbs HH. Expression and characterization of a PNPLA3 protein isoform (I148M) associated with nonalcoholic fatty liver disease. J Biol Chem 2011;286:37085–93. 
    Crossreff | PubMed
  75. Meffert PJ, Repp KD, Völzke H, et al. 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;68:858–60. 
    Crossreff | PubMed
  76. Choudhary NS, Duseja A. Genetic and epigenetic disease modifiers: non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD). Transl Gastroenterol Hepatol 2021;6:2. 
    Crossreff | PubMed
  77. Romero S, Sanyal A, Valenti L. Leveraging human genetics to identify potential new treatments for fatty liver disease. Cell Metab 2020;31:35–45. 
    Crossreff | PubMed
  78. Trépo E, Valenti L. Update on NAFLD genetics: from new variants to the clinic. J Hepatol 2020;72:1196–209. 
    Crossreff | PubMed
  79. Ismaiel A, Dumitrascu DL. Genetic predisposition in metabolic-dysfunction-associated fatty liver disease and cardiovascular outcomes – systematic review. Eur J Clin Investig 2020;50:e13331. 
    Crossreff | PubMed
  80. Xu X, Xu H, Liu X, et al. MBOAT7 rs641738 (C>T) is associated with NAFLD progression in men and decreased ASCVD risk in elder Chinese population. Front Endocrinol (Lausanne) 2023;14:1199429. 
    Crossreff | PubMed
  81. Dongiovanni P, Paolini E, Corsini A, et al. Nonalcoholic fatty liver disease or metabolic dysfunction-associated fatty liver disease diagnoses and cardiovascular diseases: from epidemiology to drug approaches. Eur J Clin Investig 2021;51:e13519. 
    Crossreff | PubMed
  82. Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J 2017;474:1823–36. 
    Crossreff | PubMed
  83. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell 2014;157:121–41. 
    Crossreff | PubMed
  84. Mouries J, Brescia P, Silvestri A, et al. Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development. J Hepatol 2019;71:1216–28. 
    Crossreff | PubMed
  85. Koren O, Spor A, Felin J, et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc Natl Acad Sci USA 2011;108(Suppl 1):4592–8. 
    Crossreff | PubMed
  86. Carpino G, Del Ben M, Pastori D, et al. Increased liver localization of lipopolysaccharides in human and experimental NAFLD. Hepatology 2020;72:470–85. 
    Crossreff | PubMed
  87. Violi F, Cammisotto V, Bartimoccia S, et al. Gut-derived low-grade endotoxaemia, atherothrombosis and cardiovascular disease. Nat Rev Cardiol 2023;20:24–37. 
    Crossreff | PubMed
  88. Zhou W, Cheng Y, Zhu P, et al. Implication of gut microbiota in cardiovascular diseases. Oxid Med Cell Longev 2020;2020:5394096. 
    Crossreff | PubMed
  89. Gill SR, Pop M, Deboy RT, et al. Metagenomic analysis of the human distal gut microbiome. Science 2006;312:1355–9. 
    Crossreff | PubMed
  90. Kasahara K, Krautkramer KA, Org E, et al. Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat Microbiol 2018;3:1461–71. 
    Crossreff | PubMed
  91. Jie Z, Xia H, Zhong SL, et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun 2017;8:845. 
    Crossreff | PubMed
  92. Boursier J, Mueller O, Barret M, et al. The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 2016;63:764–75. 
    Crossreff | PubMed
  93. Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011;472:57–63. 
    Crossreff | PubMed
  94. Tang WH, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 2013;368:1575–84. 
    Crossreff | PubMed
  95. Targher G, Byrne CD, Tilg H. H. NAFLD and increased risk of cardiovascular disease: clinical associations, pathophysiological mechanisms and pharmacological implications. Gut 2020;69:1691–705. 
    Crossreff | PubMed
  96. León-Mimila P, Villamil-Ramírez H, Li XS, et al. Trimethylamine N-oxide levels are associated with NASH in obese subjects with type 2 diabetes. Diabetes Metab 2021;47:101183. 
    Crossreff | PubMed
  97. Canyelles M, Borràs C, Rotllan N, et al. Gut microbiota-derived TMAO: A causal factor promoting atherosclerotic cardiovascular disease? Int J Mol Sci 2023;24:1940. 
    Crossreff | PubMed
  98. Corbin KD, Zeisel SH. Choline metabolism provides novel insights into nonalcoholic fatty liver disease and its progression. Curr Opin Gastroenterol 2012;28:159–65. 
    Crossreff | PubMed
  99. den Besten G, van Eunen K, Groen AK, et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res 2013;54:2325–40. 
    Crossreff | PubMed
  100. Zhao L, Zhang F, Ding X, et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 2018;359:1151–6. 
    Crossreff | PubMed
  101. Kornek M, Lynch M, Mehta SH, et al. Circulating microparticles as disease-specific biomarkers of severity of inflammation in patients with hepatitis C or nonalcoholic steatohepatitis. Gastroenterology 2012;143:448–58. 
    Crossreff | PubMed
  102. Helbing T, Olivier C, Bode C, et al. Role of microparticles in endothelial dysfunction and arterial hypertension. World J Cardiol 2014;6:1135–9. 
    Crossreff | PubMed
  103. Amabile N, Cheng S, Renard JM, et al. Association of circulating endothelial microparticles with cardiometabolic risk factors in the Framingham Heart Study. Eur Heart J 2014;35:2972–9. 
    Crossreff | PubMed
  104. Esposito K, Ciotola M, Schisano B, et al. Endothelial microparticles correlate with endothelial dysfunction in obese women. J Clin Endocrinol Metab 2006;91:3676–9. 
    Crossreff | PubMed
  105. Agouni A, Lagrue-Lak-Hal AH, Ducluzeau PH, et al. Endothelial dysfunction caused by circulating microparticles from patients with metabolic syndrome. Am J Pathol 2008;173:1210–9. 
    Crossreff | PubMed
  106. Muñoz-Hernández R, Ampuero J, Millán R, et al. Hepatitis C virus clearance by direct-acting antivirals agents improves endothelial dysfunction and subclinical atherosclerosis: HEPCAR study. Clin Transl Gastroenterol 2020;11:e00203. 
    Crossreff | PubMed
  107. Jiang F, Chen Q, Wang W, et al. Hepatocyte-derived extracellular vesicles promote endothelial inflammation and atherogenesis via microRNA-1. J Hepatol 2020;72:156–66. 
    Crossreff | PubMed
  108. Chen X, Chen S, Pang J, et al. Hepatic steatosis aggravates atherosclerosis via small extracellular vesicle-mediated inhibition of cellular cholesterol efflux. J Hepatol 2023;79:1491–501. 
    Crossreff | PubMed
  109. Nseir W, Hellou E, Assy N. Role of diet and lifestyle changes in nonalcoholic fatty liver disease. World J Gastroenterol 2014;20:9338–44. 
    Crossreff | PubMed
  110. Asgari-Taee F, Zerafati-Shoae N, Dehghani M, et al. Association of sugar sweetened beverages consumption with non-alcoholic fatty liver disease: a systematic review and meta-analysis. Eur J Nutr 2019;58:1759–69. 
    Crossreff | PubMed
  111. Seidelmann SB, Claggett B, Cheng S, et al. Dietary carbohydrate intake and mortality: a prospective cohort study and meta-analysis. Lancet Public Health 2018;3:e419–28. 
    Crossreff | PubMed
  112. Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology 2015;149:367–78.e5. 
    Crossreff | PubMed
  113. Fontana L, Meyer TE, Klein S, Holloszy JO. Long-term calorie restriction is highly effective in reducing the risk for atherosclerosis in humans. Proc Natl Acad Sci USA 2004;101:6659–63. 
    Crossreff | PubMed
  114. Tana C, Ballestri S, Ricci F, et al. Cardiovascular risk in non-alcoholic fatty liver disease: mechanisms and therapeutic implications. Int J Environ Res Public Health 2019;16:3104. 
    Crossreff | PubMed
  115. Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018;67:328–57. 
    Crossreff | PubMed
  116. Estruch R, Ros E, Salas-Salvadó J, et al. Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. N Engl J Med 2018;378:e34. 
    Crossreff | PubMed
  117. Katsagoni CN, Georgoulis M, Papatheodoridis GV, et al. Effects of lifestyle interventions on clinical characteristics of patients with non-alcoholic fatty liver disease: A meta-analysis. Metabolism 2017;68:119–32. 
    Crossreff | PubMed
  118. Church TS, Kuk JL, Ross R, et al. Association of cardiorespiratory fitness, body mass index, and waist circumference to nonalcoholic fatty liver disease. Gastroenterology 2006;130:2023–30. 
    Crossreff | PubMed
  119. Sung KC, Ryu S, Lee JY, et al. Effect of exercise on the development of new fatty liver and the resolution of existing fatty liver. J Hepatol 2016;65:791–7. 
    Crossreff | PubMed
  120. Jung HS, Chang Y, Kwon MJ, et al. Smoking and the risk of non-alcoholic fatty liver disease: a cohort study. Am J Gastroenterol 2019;114:453–63. 
    Crossreff | PubMed
  121. Ballestri S, Lonardo A, Bonapace S, et al. Risk of cardiovascular, cardiac and arrhythmic complications in patients with non-alcoholic fatty liver disease. World J Gastroenterol 2014;20:1724–45. 
    Crossreff | PubMed
  122. Sandoval-Rodriguez A, Monroy-Ramirez HC, Meza-Rios A, et al. Pirfenidone is an agonistic ligand for PPARα and improves NASH by activation of SIRT1/LKB1/pAMPK. Hepatol Commun 2020;4:434–49. 
    Crossreff | PubMed PMCID: PMC7049672.
  123. Poo JL, Torre A, Aguilar-Ramírez JR, et al. Benefits of prolonged-release pirfenidone plus standard of care treatment in patients with advanced liver fibrosis: PROMETEO study. Hepatol Int 2020;14:817–27. 
    Crossreff | PubMed
  124. Francque SM, Bedossa P, Ratziu V, et al. A randomized, controlled trial of the pan-PPAR agonist lanifibranor in NASH. N Engl J Med 2021;385:1547–58. 
    Crossreff | PubMed
  125. Wang Z, Ye M, Zhang XJ, et al. Impact of NAFLD and its pharmacotherapy on lipid profile and CVD. Atherosclerosis 2022;355:30–44. 
    Crossreff | PubMed
  126. Stefan N. Causes, consequences, and treatment of metabolically unhealthy fat distribution. Lancet Diabetes Endocrinol 2020;8:616–27. 
    Crossreff | PubMed