Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 5574 2022-12-22 19:58:39 |
2 format Meta information modification 5574 2022-12-23 03:09:46 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Josloff, K.;  Beiriger, J.;  Khan, A.;  Gawel, R.J.;  Kirby, R.S.;  Kendrick, A.D.;  Rao, A.K.;  Wang, R.X.;  Schafer, M.M.;  Pearce, M.E.; et al. Cardiovascular Disease Risk in Nonalcoholic Fatty Liver Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/39117 (accessed on 31 July 2024).
Josloff K,  Beiriger J,  Khan A,  Gawel RJ,  Kirby RS,  Kendrick AD, et al. Cardiovascular Disease Risk in Nonalcoholic Fatty Liver Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/39117. Accessed July 31, 2024.
Josloff, Kevan, Jacob Beiriger, Adnan Khan, Richard J. Gawel, Richard S. Kirby, Aaron D. Kendrick, Abhinav K. Rao, Roy X. Wang, Michelle M. Schafer, Margaret E. Pearce, et al. "Cardiovascular Disease Risk in Nonalcoholic Fatty Liver Disease" Encyclopedia, https://encyclopedia.pub/entry/39117 (accessed July 31, 2024).
Josloff, K.,  Beiriger, J.,  Khan, A.,  Gawel, R.J.,  Kirby, R.S.,  Kendrick, A.D.,  Rao, A.K.,  Wang, R.X.,  Schafer, M.M.,  Pearce, M.E.,  Chauhan, K.,  Shah, Y.B.,  Marhefka, G.D., & Halegoua-Demarzio, D. (2022, December 22). Cardiovascular Disease Risk in Nonalcoholic Fatty Liver Disease. In Encyclopedia. https://encyclopedia.pub/entry/39117
Josloff, Kevan, et al. "Cardiovascular Disease Risk in Nonalcoholic Fatty Liver Disease." Encyclopedia. Web. 22 December, 2022.
Cardiovascular Disease Risk in Nonalcoholic Fatty Liver Disease
Edit

Nonalcoholic Fatty Liver Disease (NAFLD) is a growing global phenomenon, and its damaging effects in terms of cardiovascular disease (CVD) risk are becoming more apparent. NAFLD is estimated to affect around one quarter of the world population and is often comorbid with other metabolic disorders including diabetes mellitus, hypertension, coronary artery disease, and metabolic syndrome.

NAFLD CVD MAFLD

1. Hypertension

Hypertension is estimated to affect more than 31% of adults across the world and is the most common risk factor for cardiovascular disease (CVD) [1][2]. Recent evidence suggests a significant association between Nonalcoholic fatty liver disease (NAFLD) and hypertension. An observational study estimated the prevalence of NAFLD to be nearly 50% among patients with hypertension [3]. Both in this patient cohort and in a larger population study, hypertension was significantly more prevalent among patients with NAFLD than in patients without NAFLD [4]. Additional cross-sectional studies have demonstrated a similar association between NAFLD and increased blood pressure among normotensive individuals with elevated blood pressure and pre-hypertensive individuals [5][6]. A 2022 meta-analysis of more than 390,000 patients indicated that NAFLD is associated with an approximately 1.6-fold increased risk for developing hypertension [7]. While the association between NAFLD and arterial hypertension is well supported by clinical evidence, the precise relationship in terms of causality between the two remains unclear.
The 2017 American College of Cardiology/American Heart Association High Blood Pressure Guidelines do not list NAFLD as a cause of hypertension [8]. However, emerging evidence suggests that preexisting NAFLD may contribute to the development of hypertension [9]. In a large prospective analysis of more than 22,000 non-hypertensive males with varying degrees of baseline NAFLD diagnosed by ultrasound, patients with NAFLD were more likely to develop hypertension after 5 years of follow-up than patients without NAFLD [10]. This association was more significant in patients with moderate to severe NAFLD compared to mild NAFLD (hazard ratio, 1.14 vs. 1.07), suggesting that the development of hypertension might be more strongly associated with more severe NAFLD. A large retrospective cohort analysis found similar results after 5 years of follow-up, demonstrating that NAFLD was associated with the development of incident hypertension (odds ratio, 1.36) [11]. Resolution of fatty liver at follow-up was not associated with increased incidence of hypertension, further supporting that NAFLD is likely an independent risk factor for developing hypertension. Additionally, NAFLD has been found to independently increase the risk of developing elevated systolic blood pressure among normotensive individuals (odds ratio, 2.13) [3]. The pathophysiology of NAFLD as a causative contributor to incident hypertension is still a topic of investigation, but preliminary studies suggest etiological factors may include NAFLD-induced systemic inflammation, insulin resistance, oxidative stress, increased vasoconstriction, decreased vasodilation, and arterial stiffness [9][12][13][14][15].
Conversely, multiple studies have demonstrated that hypertension may contribute to the development of NAFLD. Two large prospective cohort studies have found hypertension to be an independent predictor of incident NAFLD (relative risk and hazard ratio, 1.31 and 1.75, respectively) [16][17]. Another small prospective cohort study showed hypertension to be an independent risk factor for the progression of liver fibrosis (odds ratio, 4.8) [18]. In a retrospective study of patients with NAFLD diagnosed using transient elastography, the prevalence NAFLD was progressively increased among higher tiers of blood pressure, suggesting a correlation between blood pressure severity and risk of developing NAFLD [19]. Further evidence suggests that NAFLD and hypertension might be independent risk factors for each other. A prospective study involving a subgroup for participants from the Framingham Heart Study demonstrated a bidirectional relationship between NAFLD and hypertension after a follow up period of six years [20]. Specifically, NAFLD was identified as a risk factor for developing hypertension (odds ratio, 1.42), while hypertension was shown in a parallel analysis to be a risk factor for incident NAFLD (odds ratio, 3.34). Two additional studies demonstrated similar bidirectional relationships between NAFLD and hypertension [21][22]. This is due to a cyclical progression of disease, such that increasing blood pressure status facilitates liver damage and vice versa. Additional prospective investigation is needed to further elucidate the precise relationship between these two diseases.
Evidence-based approaches for managing coexistent NAFLD and hypertension remain limited, and there are no current recommendation guidelines for pharmacologic or nonpharmacological treatment for these individuals. Lifestyle adjustments, including nutritional changes, improved physical activity, and weight loss continue to be the cornerstone of NAFLD therapy [23][24]. Numerous trials of specialized dietary programs including the Dietary Approaches to Stop Hypertension (DASH) Diet and the Mediterranean Dietary Pattern (MedDiet) have shown success in reducing hypertension risk, thereby reducing the risk of developing hepatic steatosis NAFLD and improving other cardiovascular comorbidities among individuals with NAFLD [25][26][27][28]. However, prospective investigation into the efficacy of the DASH diet for managing NAFLD is limited. One randomized controlled trial demonstrated that overweight/obese subjects with NAFLD in a DASH diet group had higher degrees of improvements in liver enzymes, insulin sensitivity, and body weight compared to matched subjects in a low-energy diet group [29].
Regarding pharmacologic therapy for coexistent NAFLD and hypertension, renin-angiotensin-aldosterone system (RAAS) inhibitors have shown some favorability. This is predicated on the current understanding that upregulation of the RAA system seems to be involved in the pathogenesis of NAFLD [30]. One small study of seven patients with coexistent NASH and hypertension treated for 48 weeks with losartan demonstrated improved blood markers of hepatic fibrosis and improved hepatic necroinflammation and fibrosis as determined by biopsy [31]. Larger studies have shown similar favorable results in improving hepatic fibrosis, liver stiffness, and other markers of NAFLD after treatment with either angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) [32][33][34].
Several newer biologic therapies have been evaluated in phase II and phase III clinical trials [35]. Modulators of the farnesoid X receptor (FXR) are thought to be involved in the pathogenesis of hepatic fibrosis and are the topic of several clinical trials. Obeticholic acid is an FXR agonist currently under investigation in two Phase 3 clinical trials to evaluate the efficacy in improving hepatic fibrosis [36][37]. A previous trial has demonstrated that obeticholic acid improved systolic blood pressure and reduced the degree of necroinflammation in patients with NASH [38]. Analogs of fibroblast growth factor 19/21 (FGF 19/21) are also currently undergoing phase II clinical trials to evaluate utility in improving fibrosis in NASH patients [39][40]. FGF21 is thought to reduce progression of NAFLD, and positive results have already been demonstrated in a small phase II trial of the PEGylated FGF21 analog Pegbelfermin [41]. Additional agents of interest in managing hepatic fibrosis were PPARα/δ agonist Elafibranor and the CCR2/5 antagonist cenicriviroc. Unfortunately, neither drug’s clinical trial results warranted continuing the study, and both were terminated early. Cenicriviroc was shown in a randomized placebo-controlled trial to improve fibrosis without progressing to steatohepatitis, and a preclinical study has demonstrated that a different experimental CCR2 inhibitor improved elevated blood pressure in mice [42][43]. Further preclinical and clinical investigations are ongoing and required to evaluate the efficacy of these and other biologic agents for their utility in managing coexistent NAFLD and hypertension.

2. Diabetes Mellitus

The global prevalence of type 2 diabetes mellitus (T2DM) has continued to rise and is projected to increase to 7079 individuals per 100,000 worldwide by the year 2030 [44]. The association between NAFLD and T2DM is well established. A 2015 meta-analysis of 729 studies including more than 8.5 million individuals demonstrated that the global prevalence of T2DM was 22.5% among patients with radiographically diagnosed NAFLD and greater than 43.6% among NAFLD diagnosed via biopsy [45]. A 2019 meta-analysis revealed that the prevalence of biopsy-proven NAFLD in patients with T2DM is greater than 55%, more than 2-fold higher than the general population [46]. Among the major components of metabolic syndrome, including obesity, hypertension, and hyperlipidemia, T2DM appears to be the most important risk factor for the predicting NAFLD-associated sequela, such as hepatic fibrosis [47][48]. Furthermore, T2DM has also been demonstrated to increase the risk of hepatocellular carcinoma, liver-related mortality, and overall mortality among NAFLD patients [46][49][50][51].
Multiple studies have demonstrated that the presence of NAFLD in individuals with T2DM increases the risk of CVD, independent of other components of metabolic syndrome. Among these, a prospective case–control study demonstrated a significantly higher risk for developing CVD (including MI, coronary revascularization procedures, ischemic stroke, or cardiovascular-related death) among T2DM individuals with ultrasound diagnosed NAFLD compared to those without evidence of NAFLD (odds ratio, 1.84) [52]. A larger cross-sectional study of nearly 3000 T2DM individuals conducted by the same researchers demonstrated an increased prevalence of coronary (26.6 vs. 18.3%), cerebral (20.0 vs. 13.3%), and peripheral (15.4 vs. 10.0%) vascular disease among NAFLD patients compared to those without NAFLD [53]. More recently, among a Turkish cohort of individuals with NAFLD, patients with T2DM compared to non-diabetic patients were shown to have higher mean carotid intima-media thickness, the study’s endpoint for assessment of cardiovascular risk [54]. Additionally, NAFLD was shown to be associated with an increase in the development of atrial fibrillation in patients with T2DM [55]. The results of these studies were significant after adjusting for other CVD risk factors, demonstrating that coexistent T2DM and NAFLD are independently associated with multiple subtypes of CVD.
Numerous studies have demonstrated that NAFLD increases the risk of developing T2DM, with some estimating the risk is increased approximately 2-fold to 3-fold compared to the general population [56][57][58]. One large 2010 prospective study with a mean follow-up of 11.5 years found that unadjusted NAFLD significantly increased the risk of incident T2DM (19% vs. 6%), though this became statistically nonsignificant after adjusting for confounders [59]. Multiple meta-analyses performed within the past decade have demonstrated an increased NAFLD-associated risk for developing T2DM after adjusting for confounding variables. Among these, two meta-analyses demonstrated ultrasonographic-diagnosed NAFLD increased the risk for incident T2DM (odds ratio, 3.51 and 1.86, respectively) [60][61]. A larger 2018 meta-analysis of nearly 300,000 individuals from 19 studies indicated that individuals with NAFLD have a greater than 2-fold increased risk for developing incident T2DM [62]. Within this cohort of studies, individuals with more severe ultrasonographic steatosis and greater degree of fibrosis tended to have a higher risk of developing T2DM than individuals with less severe liver disease [63][64].
At present, there is far less conclusive evidence indicating whether T2DM is a significant contributor to the development of NAFLD. In a cross-sectional analysis of nondiabetic Korean individuals, ultrasound diagnosed NAFLD was associated with increased HbA1c level and insulin resistance, independent of obesity and other metabolic confounders [65]. These authors concluded that abnormal glucose tolerance and the phenomenon of pre-diabetes might contribute to the development or progression of steatosis in patients who do not meet criteria for a formal diagnosis of diabetes. Evidence has also suggested that the percentage of liver fat is higher in patients with T2DM compared to patients without T2DM, supporting the authors’ theories [66]. One recent study showed that among overweight and obese patients with NAFLD, T2DM significantly increased the risk of steatosis (48.3% vs. 17.4% and 79.9% vs. 57.6%, respectively) and moderate-to-high risk fibrosis (31.8% vs. 20.1%) [67].
Despite the high coexistence of NAFLD and T2DM and increased risk of cardiovascular sequela among these individuals, evidence does not currently support screening for NASH or NAFLD in patients with T2DM. Due to the lack of quality evidence-based therapeutic options and inconsistent efficacy of diagnostic modalities, a recent analysis demonstrated that screening for NAFLD in these patients is not currently cost effective [24][68]. In its 2017 practice guideline, the American Association for the Study of Liver Diseases did not recommend routine NAFLD screening for high-risk patients in primary care, diabetes, or obesity clinics [24]. No pharmacological agents are currently approved for treating coexistent NAFLD and T2DM, though utility of pharmacologic and non-pharmacologic therapies is under investigation. Given the current understanding of the pathophysiological interplay between insulin resistance in both T2DM and NAFLD, most therapeutic strategies tend to target insulin resistance. At present, lifestyle modification appears to be the most effective strategy in reducing adverse cardiovascular outcomes among individuals with coexistent NAFLD and T2DM [69]. Among T2DM patients with NAFLD, one study demonstrated that a 15% decrease in BMI through aerobic exercise and diet effectively improved liver function, marked by a reduction in liver enzymes [70]. Other researchers showed that aerobic exercise alone without change in body weight also improved liver enzymes in patients with concomitant T2DM and NAFLD and that intense exercise is more effective than moderate exercise [24][71]. However, a major limitation of lifestyle modifications is the poor adherence to exercise and weight loss, particularly among overweight and obese individuals, in whom the risk of NAFLD is greatest [72].
While evidence supporting the use of antihyperglycemic agents remains limited, multiple classes of agents have shown promise [73]. Several studies showed metformin improved liver enzymes, reduced histologic liver damage, and improved weight loss in T2DM patients with NAFLD [74][75], though a small randomized controlled trial between metformin and a placebo showed no significant improvement in liver enzymes or histopathology [76]. The experimental mitochondrial pyruvate carrier agent MSD-0602K has shown similar efficacy in reducing NAFLD histopathology after 12 months, though additional trials evaluating its longer-term efficacy in managing NAFLD and NASH are ongoing [77]. Incretin modulators, including agents from both the glucagon-like peptide-1 (GLP-1) agonist and dipeptidyl peptidase-4 (DPP-4) inhibitor classes, have recently shown promise in potentially reducing the progression of NAFLD. To date, the GLP-1 agonist liraglutide has been most widely investigated for this purpose. In a small prospective study of obese individuals with known T2DM on stable doses of metformin and evidence of hepatic steatosis, those treated for 6 months with either liraglutide or the GLP-1 agonist exenatide demonstrated reduced hepatic steatosis and decreased liver enzyme levels [78]. Studies have reported similar effectiveness of exenatide, liraglutide, and the DPP-4 inhibitor sitagliptin

3. Metabolic Syndrome

The metabolic syndrome (MetS) is characterized by a cluster of metabolic derangements including elevated blood pressure, atherogenic dyslipidemia, impaired glucose tolerance and insulin resistance, and abdominal obesity [79]. Multiple sets of varying criteria are commonly used to diagnose MetS, including those described by the World Health Organization (WHO), the American Association of Clinical Endocrinologists, the National Cholesterol Education Program’s Adult Treatment Panel III report (ATP III), and the International Diabetes Foundation [80][81][82]. The association of MetS with an increased risk of cardiovascular-associated morbidity and mortality is well established [83][84][85]. The prevalence of MetS varies based on which organizational criteria is used for diagnosis, but prevalence rates of the non-obese international population range from 26% to more than 35% [86][87]. Among obese individuals, prevalence has been estimated to be as high as 65% in women and 78% in men, with hypertension being the most frequently occurring factor contributing to MetS prevalence [88].
NAFLD is widely considered the hepatic manifestation of MetS given the large degree of metabolic overlap [89]. In one study, 88% of individuals with NAFLD had at least one component of MetS, and approximately one third had three or more MetS components [90]. Furthermore, MetS is considerably associated with the prevalence and severity of NAFLD. In a large cohort study of nearly 12,000 individuals, the prevalence of NAFLD diagnosed with ultrasound and the NAFLD Fibrosis score (NFS) was 43% greater in those with MetS (odds ratio, 11.5) [91]. These investigators also showed that the prevalence of NAFLD increased with the number of MetS components, such that the prevalence of NAFLD among individuals with all five MetS components was 67% greater than that of the general population. Additionally, the presence of MetS has been shown to increase morbidity and mortality risk in NAFLD patients. Among a cohort of NAFLD patients from the National Health and Nutrition Examination Survey (NHANES III), the presence of coexistent MetS was found to be an independent predictor of all-cause, liver-specific, and cardiovascular mortality, whereas NAFLD without MetS conferred no increased risk [92]. In another cross-sectional study, the presence of at least one MetS component increased the risk of mortality of NAFLD patients compared to individuals without MetS after both 8 and 16 years of follow-up (4.7% vs. 2.6% and 11.9% vs. 6%, respectively) [93].
Due to the multifactorial and multicomponent nature of MetS, precisely evaluating the causal relationship between MetS and NAFLD is challenging. Nevertheless, numerous studies have demonstrated that MetS and its individual components independently contribute to the development and progression of NAFLD [18][94]. The number of metabolic abnormalities also seems to be positively associated with NAFLD risk. Adjusted for lifestyle risk factors, one study associated NAFLD with either one, two, or three components of MetS (hazard ratio, 1.92, 2.64, 3.51, respectively) compared to individuals without MetS [18]. Among these metabolic abnormalities, obesity and hyperlipidemia were more highly associated with NAFLD compared to hypertension or hyperglycemia. In a 2020 study, Kim et al. demonstrated that MetS, regardless of obesity, was associated with increased progression of fibrosis among patients with NAFLD [94].
Unfortunately, the diagnosis of NAFLD remains challenging. While the gold standard for diagnosing NAFLD is liver biopsy, most studies evaluating and risk stratifying NAFLD employ noninvasive techniques, including ultrasound, magnetic resonance elastography, or laboratory-based diagnostics. The FIB-4, NFS, and other scoring systems such as the AST/platelet ratio index (APRI) have also been used to risk stratify for liver-related morbidity in NAFLD patients [23][95]. This can complicate evaluating the causal relationship between NAFLD and MetS, particularly when NAFLD progression is assessed using one of these scoring systems, given that both NAFLD and MetS can independently elevate liver enzymes.
Evidence suggests that the relationship between MetS and NAFLD is likely bidirectional, such that preexisting NAFLD can contribute to the development and worsening of metabolic derangements and MetS [96]. Among a cohort of nearly 18,000 Chinese individuals without baseline MetS, researchers found that after 6 years of follow-up, NAFLD was an independent risk factor for the development of MetS (hazard ratio, 1.55) [97]. Additional studies have also found that NAFLD is an independent risk factor for MetS [98][99][100].
Lifestyle modification remains the primary management strategy for reducing cardiovascular risk among individuals with MetS and NAFLD. Weight loss, either due to diet alone or diet combined with physical activity, has been shown to decrease progression of hepatic steatosis, inflammation, and fibrosis [101]. Additional studies have shown that physical exercise alone without weight loss can decrease liver enzymes and steatosis [102]. Among patients with NAFLD, these lifestyle modifications have considerably decreased the risk of cardiovascular and cardiac complications [103]. Managing individual components of MetS continues to be the mainstay of reducing cardiovascular risk in NAFLD individuals.
A 2016 non-systematic review suggested that bariatric surgery might be helpful in managing NAFLD in patients with MetS [104]. Multiple studies showed that bariatric surgery improved multiple metabolic comorbidities and resulted in decreased lobular inflammation, hepatic ballooning, and steatosis. Similar results have been demonstrated in small prospective studies [105]. However, these studies secondarily evaluated NAFLD and MetS outcomes in patients who otherwise had indications for bariatric surgery. A 2019 meta-analysis of 32 studies reported that bariatric surgery decreased ballooning degeneration, fibrosis, and inflammation and showed biopsy-confirmed resolution of steatosis in 66% of patients [106]. While bariatric surgery is not currently recommended as a means of managing coexistent MetS and NAFLD, further investigation with larger prospective clinical trials may show utility in select populations.

4. Coronary Artery Disease

Despite NAFLD being a liver pathology, the most common cause of death among NAFLD patients stems from CVD, mainly ischemic heart disease [107][108][109]. Previous literature has shown the prevalence of coronary heart disease (CHD), including coronary atherosclerosis and established coronary artery disease (CAD), to be around 47% in NAFLD patients [110]. A 2021 meta-analysis from Toh et al. demonstrated a pooled prevalence of CAD of 44.6% (95% CI: 36.0–53.6%) among 67,070 patients with NAFLD [111]. Moderate to severe steatosis had a higher prevalence of CHD compared to mild steatosis (37.5%, 95% CI: 15.0–67.2% vs. 29.6%, 95% CI: 13.1–54.0%) [112].
Myriad factors contribute to the association between CAD and NAFLD including dyslipidemias, systemic inflammation, insulin resistance, endothelial dysfunction, oxidative stress, and disturbances in gut microbiota [107][113]. NAFLD is also associated with increased arterial thickness, epicardial fat thickness, and presence of calcified and non-calcified coronary plaques, all contributing to CAD [114][115]. As mentioned, there seems to be a correlation between the degree of NAFLD and prevalence of CAD. One study showed that as the severity of NAFLD increases, the percentage of non-CAD patients decreases (p < 0.001) [116]. One way of defining the severity of CAD is using the Coronary Artery Diseases Reporting and Data System (CAD-RADS) [117]. Multiple studies have shown that more severe NAFLD was associated with multiple vessel coronary disease and more severe CAD scores [109][118][119][120][121].
While robust evidence connects NAFLD and CHD, the relationship between NAFLD and mortality is less clear. Several cohort studies and a 2016 meta-analysis from Targher et al. of 34,000 patients demonstrated that NAFLD was associated with an increase in all-cause mortality and fatal and/or non-fatal cardiovascular events [122][123][124][125]. However, several other studies have shown differing results. A cohort study in the United States using the NHANES III: 1988–1994 demonstrated that NAFLD was not associated with increased risk of all-cause mortality [126]. A 2016 meta-analysis by Wu et al. also did not show increased cardiovascular or all-cause mortality in NAFLD patients [127]. However, Wu et al. (2016) did show an increased incidence and prevalence of CVD in NAFLD patients, including CAD and atherosclerosis [127]. A large 2016 meta-analysis showed a non-significant pooled incidence rate ratio (IRR) for overall mortality between NAFLD and non-NAFLD patients of 1.05 (95% CI: 0.70–1.56) but a significantly increased adjusted hazard ratio for overall mortality in NAFLD patients of 1.04 (95% CI: 1.03–1.04) [45]. If only studies that identified NAFLD by imaging (versus imaging and serum enzymes) were included, the pooled IRR for cardiovascular mortality became significant with an IRR of 1.37 (95% CI: 1.23–1.54). Similarly, the relationship between NAFLD and acute MI is poorly characterized. Several cohort studies showed that NAFLD was associated with an increased risk of MI compared to non-NAFLD patients [128][129][130]. However, a cohort study from 2019 consisting of 18 million patients from Italy, the Netherlands, Spain, and the United Kingdom demonstrated that NAFLD was not associated with increased MI (HR of 1.01, 95% CI: 0.91–1.12) and stroke (HR: 1.04, 95% CI: 0.99–1.09) after controlling for covariates such as blood pressure, type 2 diabetes status, cholesterol level, statin use, and hypertension [131].
A 2020 meta-analysis including approximately 21 million patients identified NAFLD as an independent risk factor for acute coronary syndrome (ACS) in Asian populations, but this association conflicts with American and European studies [109]. Further, Ismaiel et al. characterized NAFLD as an independent predictor of all-cause and CV mortality as well as in-hospital major adverse cardiac events in ACS patients [109]. The impact of sex on NAFLD and CVD is poorly studied [113][132]. Female sex was associated with protection against ischemic cardiac events; however, female patients with NAFLD lose this benefit and experience increased CVD and mortality compared to men [133]. A 2020 meta-analysis by Khalid et al. demonstrated that in a population of 108,711 patients with NAFLD, with 44% being women with weighted mean age of 50 years, female sex was associated with increased all-cause mortality with odds ratio of 1.65 (95% CI: 1.12–2.43, p < 0.012) and increased CV events and mortality with OR of 2.12 (95% CI 1.65–2.73, p < 0.001) [134]. Meta-regression showed that women experienced higher mortality with advancing age beginning at age 42 [134]. Further studies are needed to elucidate the interplay of NAFLD, sex, CVD, and mortality.
Modifying risk factors remains critical for preventing and managing NAFLD [24][135][136]. Many of the same lifestyle modifications, such as diet, weight loss, and exercise, are also beneficial for managing CAD [137][138][139][140]. Weight loss of about 5% has been shown to decrease hepatic steatosis, while weight loss of around 10% is required to improve non-alcoholic steatohepatitis and fibrosis [23][24]. Bariatric surgery is known to have cardiovascular benefits and may also have a role in managing NAFLD [141][142][143]. Adherence to a calorie restricted diet can improve both NAFLD and atherogenesis [137][138][139]. Refs. [143][144][145][146][147] Particularly, a caloric reduction of at least 30% (750–1000 kcal/day) can improve both insulin resistance and hepatic steatosis. [148]. The MedDiet and DASH diet have both had positive effects in NAFLD patients, with the MedDiet improving hepatic steatosis [24][144][145][146][147]. Sedentary behavior increases mortality and may predispose patients to NAFLD [149][150]. While exercise has been shown to improve NAFLD, the specific parameters relating to the type of exercise and duration are unclear.
Ref. [113] Several studies, including the Pioglitazone versus Vitamin E versus Placebo for the Treatment of Nondiabetic Patients with Nonalcoholic Steatohepatitis (PIVENS) trial and a 2017 meta-analysis by Musso et al., showed that pioglitazone improved fibrosis even in non-diabetic patients with NASH [151][152][153]. Pioglitazone also has cardiovascular benefits, including reducing coronary disease, which may be beneficial in limiting mortality and cardiovascular events in NAFLD patients [154][155][156]. However, these medications have fallen to the background with the advent of GLP-1 agonists and SGLT-2 inhibitors. Liraglutide, a glucagon like peptide-1 analog, has been shown to reduce serum liver enzymes and fibrosis, improve steatosis, and reduce adverse cardiovascular events [157][158][159][160][161]. Vitamin E has also been suggested as a treatment for NAFLD given its anti-oxidative properties. Early studies of vitamin E were confounded by differing doses and formulations that could affect bioavailability [24]. However, vitamin E can improve serum liver enzymes, inflammation, and steatosis [24][162][163][164]. Previous concerns about increased all-cause mortality related to vitamin E have been unable to be reproduced in large meta-analyses [165][166][167]. Concerns remain about vitamin E and an increased risk of prostate cancer in limited patient populations [168][169][170].
Statins, which limit cholesterol biosynthesis through inhibition of 3-hydroxy-3-methylglutaryl coenzyme A reductase, have been shown to improve steatosis and NAFLD activity scores across multiple small studies, although robust, high-quality evidence is lacking, which limits their indication in managing liver disease in NAFLD [171][172][173][174][175][176]. Researchers have hypothesized that the beneficial effects of statins in NAFLD may in part be mediated through alterations in gut microbiota [177]. In contrast, statins have a definitive role in treating dyslipidemias and improving cardiovascular outcomes [178][179]. Despite some concern about statin use in patients with pre-existing liver disease, multiple studies have established the safety of statins in patients with NAFLD regardless of elevated baseline liver enzymes, and statins should be started in patients with NAFLD who meet criterion [180][181][182][183].

5. Heart Disease

Extensive evidence associates NAFLD with myocardial changes, including support for a graded relationship between functional and structural myocardial abnormalities and severity of NAFLD histology [184][185][186][187]. A cohort study of 181 patients showed that the prevalence of NAFLD in heart failure (HF) with preserved rejection fraction (HFpEF) may reach 50% [188]. Another cohort study of 102 patients found that the prevalence of NAFLD in heart failure with reduced ejection fraction (HFrEF) was 36.3% [189]. A Korean cohort study of 3300 patients by Chung et al. revealed that the prevalence of left ventricular (LV) diastolic dysfunction increased with NAFLD fibrosis grade (30.4% in non-NAFLD patients, 35.2% in NAFLD patients without advanced fibrosis, and 57.4% in NAFLD patients with advanced fibrosis, p < 0.001) [190].
Researchers have hypothesized that the relationship between NAFLD and structural cardiac changes stems primarily from systemic inflammation and other processes including insulin resistance, oxidative stresses, activation of the renin-angiotensin aldosterone system, and gut microbiota [191][192]. Inflammatory cytokines such as tumor necrosis factor-alpha (TNF-a), nuclear factor kappa-B (NF-kB), and interleukin-6 may result in inflammatory responses in myocardium and ventricular remodeling [192][193]. Multiple studies have confirmed these radiographic findings of cardiac dysfunction including increased myocardial, pericardial, and epicardial fat by magnetic resonance imaging and reduced early diastolic and systolic velocities in NAFLD patients [186][194]. In an analysis of patients from the multicenter, community-based Coronary Artery Risk Development in Young Adults (CARDIA) study, VanWagner et al. found that patients with NAFLD had lower early diastolic relaxation (e’) velocity (10.8 ± 2.6 vs. 11.9 ± 2.8 cm/s), higher LV filling pressure (E/e’ ratio: 7.7 ± 2.6 vs. 7.0 ± 2.3), and worse absolute GLS (14.2 ± 2.4% vs. 15.2 ± 2.4%) (p < 0.001 for all) suggestive of early subclinical LV systolic dysfunction [184]. Prior studies support the association between severity of NAFLD and severity of LV diastolic dysfunction [187][190][195]. Chung et al. demonstrated a significant incremental increase in risk of LV diastolic dysfunction in non-obese patients with NAFLD compared to patients without NAFLD (OR: 1.40, 95% CI: 1.06–1.84 for NAFLD without advanced fibrosis, OR: 1.44, 95% CI: 0.95–2.17 for NAFLD with advanced fibrosis, p = 0.022) [190].
Most investigations examining the relationship between NAFLD and HF have primarily combined HFpEF with HFrEF or looked at HFpEF alone, and strong evidence supports this association [188][189][196][197][198]. In a study of 870,535 Medicare beneficiaries, Fudim et al. found that the cumulative incidence and hazard ratio in Cox models were only significant for HFpEF, but not for HFrEF, when comparing patients with NAFLD vs. without NAFLD [199]. Recent studies have attempted to classify NAFLD and HFpEF into different subgroups through phenomapping, a clustering analysis that utilizes dense phenotypic data to identify phenotypically distinct classifications of HFpEF [200][201][202]. Salah et al. proposed three distinct phenotypes: obstructive HFpEF, metabolic HFpEF, and advanced liver disease/cirrhosis HFpEF (PMID: 34869957). Due to physiological differences between these phenotypes, additional investigation is required to further characterize mortality and optimal management of these phenotypic groups. A growing body of evidence supports the negative association between NAFLD and outcomes of patients with HF, including in-patient and post-discharge mortality as well as hospital readmissions [196][203][204][205]. In a cohort of elderly patients, Valbusa reported a 5-fold increase in 1-year all-cause re-hospitalization rate in NAFLD patients admitted for acute HF compared to non-NAFLD patients admitted for acute HF (HR: 5.05, 95% CI: 2.78–9.10, p < 0.001) [196]. In a large cohort study of about 3.5 million patients, Minhas et al. reported NAFLD was associated with higher mortality for both HFrEF and HFpEF (HFrEF, aOR: 1.84, 95% CI 1.66–2.04, p < 0.001, HFpEF, aOR: 1.65, 95% CI 1.43–1.9, p < 0.001) as well as increased length of stay and cost compared to non-NAFLD patients with HFrEF and HFpEF [205]. In an analysis of the SwedeHF registry, Ergatoudes showed that liver disease may be associated with worse outcomes in HFrEF compared to HFpEF (HR: 2.13, 95% CI 1.83–2.47 vs. HR 1.42, 95% CI 1.09–1.85, p = 0.02) [206]. This highlights the need for further work comparing the impact of NAFLD on HFrEF and HFpEF.
Lifestyle management remains an integral part of preventing and treating NAFLD and HF [23][24][207][208]. As discussed, weight loss has been shown to improve NAFLD, and prior studies demonstrated that weight loss ameliorated myocardial structural changes in obesity [23][24][209]. Therefore, weight loss of at least 5% is recommended in patients with NAFLD for hepatic and cardiac benefits [23][24]. Weight reduction via diet or bariatric surgery was shown to have these benefits [210][211][212][213]. Another benefit of bariatric surgery is that it may be used as a bridge for weight loss in morbidly obese patients ineligible for cardiac transplantation [214]. The visceral fat deposits commonly seen in obese patients may be resistant to mild changes in diet-induced weight reductions [198][213]. In a small randomized controlled trial studying dietary interventions in obese subjects, de las Fuentes observed partial weight regain at 24 months (average percent weight loss at 3: 7.3 ± 4.0%, 6: 9.2 ± 5.6%, 12: 7.8 ± 6.6%, 24: 3.8 ± 7.9% months) [213]. Although the maximal beneficial effects of weight loss were reduced, structural cardiovascular parameters still showed significant improvement compared to baseline [213]. From a dietary perspective, it is recommended that NAFLD patients adhere to a hypocaloric diet with a daily consumption of 1200–1800 kcal or daily deficit of 500–1000 kcal from baseline [23][215][216]. The DASH diet and MedDiet have been shown to improve NAFLD and may improve cardiovascular risks as well [29][147]. The MedDiet is thought to modulate hepatic and cardiovascular risks through alterations in insulin resistance, serum triglyceride levels, intrahepatic triglyceride levels, and serum cholesterol levels [217][218]. Given these benefits, multiple societies recommend the MedDiet for patients with NAFLD [23][215][216]. As discussed, exercise offers numerous benefits including decreased intrahepatic fat, improved serum metabolic markers, and cardiac function parameters such as early diastolic filling [219][220]. Several studies have shown no difference in aerobic vs. resistance training in reducing intrahepatic fat content in NAFLD patients [221][222][223]. However, a 2021 meta-analysis by Xiong et al. showed that aerobic exercise reduced more metabolic indicators compared to resistance exercise and high-intensity interval training in NAFLD patients specifically [224]. Additionally, robust evidence supports the association between the aggressiveness of weight loss intervention and improvement in NAFLD [146][225][226][227]. A paradoxical relationship exists between obesity and cardiac dysfunction [208]. When patients have clinically symptomatic HF, higher BMI has a protective mortality benefit [228][229][230][231]. Thus, patients with advanced cardiac dysfunction must be cautious before beginning weight loss regimens.
Multiple medications have been investigated for treating NAFLD and the ability to concomitantly manage HF symptoms. Pioglitazone was previously mentioned as having beneficial effects related to ischemic CVD; however, pioglitazone can lead to weight gain and increased peripheral edema in a dose-dependent fashion in approximately 5–10% of patients [232][233]. The concern for fluid retention and possible triggering of HF limits the use of pioglitazone in these patients. GLP1-receptor agonists are rapidly becoming of interest, given weight loss benefits and improved insulin resistance. In several studies, including the Liraglutide Effect and Action in Diabetes (LEAD) program, LEAD-2 study, and Liraglutide Efficacy and Action in NASH (LEAN) trial, liraglutide improved hepatic steatosis and resolution of NASH in biochemistry-based or in biochemistry-defined, imaging-defined, and biopsy-defined NAFLD, although liraglutide failed to improve hepatic fibrosis [158][159][234][235]. Semaglutide, another GLP-1 receptor agonist, has also shown promising results in several trials [234]. A 2019 meta-analysis of eight studies by Kristensen et al. demonstrated that GLP-1 receptor agonists reduced hospitalizations for heart failure by 9% [160]. This evidence supports the use of these medications in managing diabetes in patients with comorbid heart failure. SGLT2-inhibitors have also demonstrated extraordinary promise in NAFLD patients with heart failure. Multiple meta-analyses have demonstrated that SGLT2-inhibitors improved liver function tests and hepatic fat as assessed by imaging techniques [159][236][237][238]. Many meta-analyses and trials, including the Empagliflozin Outcome Trial in Patients With Chronic Heart Failure With Reduced Ejection Fraction (EMPEROR-Reduced), Empagliflozin Outcome Trial in Patients With Chronic Heart Failure With Preserved Ejection Fraction (EMPEROR-Preserved), and Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure (DAPA-HF), showed that SGLT2 inhibitors can decrease hospitalization for HF in both HFrEF and HFpEF patients regardless of diabetes status [239][240][241][242]. A 2021 meta-analysis by Salah et al. quantified this effect and demonstrated a 31% reduction in hospitalization for HF in patients taking SGLT2 inhibitors [243].
Novel agents that can treat NAFLD and reverse fibrosis continue to be developed. Four medications are currently farthest along in development, undergoing phase III trials [38][208][244]. Current evidence does not reveal any impact of obetacholic acid on HF [38][113]. Elafibranor, a peroxisome proliferator−activated receptor-alpha and peroxisome proliferator−activated receptor-delta agonist, improves steatohepatitis without worsening fibrosis [244]. Elafibranor did not cause weight gain or increase cardiac events [244]. Cenicrivoric, a dual chemokine receptor (CCR) 2 and 5 antagonist, has been shown to improve liver fibrosis without worsening steatohepatitis [42]. While the trial did not report cardiovascular benefits, prior studies using mouse models have shown CCR2 blockade may decrease atherosclerotic lesion development, possibly portending cardiovascular benefit in humans [42][245]. Selonsertib is an apoptosis signal-regulating kinase 1 (ASK1) inhibitor that has also been shown to improve liver fibrosis [246]. This trial also did not report cardiovascular benefits, but ASK1 is known to affect the cellular stress response and apoptosis pathway, contributing to diseases such as heart failure and ischemia/reperfusion injury [246][247][248][249][250]. ASK1 deficient mice displayed reduced levels of cardiomyocyte apoptosis, hypertrophy, and interstitial fibrosis [251].

References

  1. Mills, K.T.; Bundy, J.D.; Kelly, T.N.; Reed, J.; Kearney, P.M.; Reynolds, K.; Chen, J.; He, J. Global Disparities of Hypertension Prevalence and Control: A Systematic Analysis of Population-Based Studies From 90 Countries. Circulation 2016, 134, 441–450.
  2. Chobufo, M.D.; Gayam, V.; Soluny, J.; Rahman, E.U.; Enoru, S.; Foryoung, J.B.; Agbor, V.N.; Dufresne, A.; Nfor, T. Prevalence and control rates of hypertension in the USA: 2017–2018. Int. J. Cardiol. Hypertens. 2020, 6, 100044.
  3. Lopez-Suarez, A.; Guerrero, J.M.R.; Elvira-González, J.; Beltrán-Robles, M.; Cañas-Hormigo, F.; Bascuñana-Quirell, A. Nonalcoholic fatty liver disease is associated with blood pressure in hypertensive and nonhypertensive individuals from the general population with normal levels of alanine aminotransferase. Eur. J. Gastroenterol. Hepatol. 2011, 23, 1011–1017.
  4. Feng, R.N.; Du, S.-S.; Wang, C.; Li, Y.-C.; Liu, L.-Y.; Guo, F.-C.; Sun, C.-H. Lean-non-alcoholic fatty liver disease increases risk for metabolic disorders in a normal weight Chinese population. World J. Gastroenterol. 2014, 20, 17932–17940.
  5. Qian, L.Y.; Zou, H. Association of blood pressure level with nonalcoholic fatty liver disease in nonhypertensive population: Normal is not the new normal. Medicine 2016, 95, e4293.
  6. Wu, S.J.; Zou, H.; Zhu, G.-Q.; Wang, L.-R.; Zhang, Q.; Shi, K.-Q.; Han, J.-B.; Huang, W.-J.; Braddock, M.; Chen, Y.-P.; et al. Increased levels of systolic blood pressure within the normal range are associated with significantly elevated risks of nonalcoholic fatty liver disease. Medicine 2015, 94, e842.
  7. Ciardullo, S.; Grassi, G.; Mancia, G.; Perseghin, G. Nonalcoholic fatty liver disease and risk of incident hypertension: A systematic review and meta-analysis. Eur. J. Gastroenterol. Hepatol. 2022, 34, 365–371.
  8. Whelton, P.K.; Carey, R.M.; Aronow, W.S.; Casey, D.E., Jr.; Collins, K.J.; Himmelfarb, C.D.; DePalma, S.M.; Gidding, S.; Jamerson, K.A.; Jones, D.W.; et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 2018, 71, e127–e248.
  9. Zhao, Y.-C.; Zhao, G.-J.; Chen, Z.; She, Z.G.; Cai, J.; Li, H. Nonalcoholic Fatty Liver Disease: An Emerging Driver of Hypertension. Hypertension 2020, 75, 275–284.
  10. Ryoo, J.H.; Suh, Y.J.; Shin, H.C.; Cho, Y.K.; Choi, J.-M.; Park, S.K. Clinical association between non-alcoholic fatty liver disease and the development of hypertension. J. Gastroenterol. Hepatol. 2014, 29, 1926–1931.
  11. Sung, K.C.; Wild, S.H.; Byrne, C.D. Development of new fatty liver, or resolution of existing fatty liver, over five years of follow-up, and risk of incident hypertension. J. Hepatol. 2014, 60, 1040–1045.
  12. Oikonomou, D.; Georgiopoulos, G.E.; Katsi, V.; Kourek, C.; Tsioufis, C.; Alexopoulou, A.; Koutli, E.; Tousoulis, D. Non-alcoholic fatty liver disease and hypertension: Coprevalent or correlated? Eur. J. Gastroenterol. Hepatol. 2018, 30, 979–985.
  13. Houghton, D.; Zalewski, P.; Hallsworth, K.; Cassidy, S.; Thoma, C.; Avery, L.; Slomko, J.; Hardy, T.; Burt, A.D.; Tiniakos, D.; et al. The degree of hepatic steatosis associates with impaired cardiac and autonomic function. J. Hepatol. 2019, 70, 1203–1213.
  14. Yu, Y.; Cai, J.; She, Z.; Li, H. Insights into the Epidemiology, Pathogenesis, and Therapeutics of Nonalcoholic Fatty Liver Diseases. Adv. Sci. 2019, 6, 1801585.
  15. Li, N.; Zhang, G.-W.; Zhang, J.-R.; Jin, D.; Li, Y.; Liu, T.; Wang, R.-T. Non-alcoholic fatty liver disease is associated with progression of arterial stiffness. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 218–223.
  16. Tsuneto, A.; Hida, A.; Sera, N.; Imaizumi, M.; Ichimaru, S.; Nakashima, E.; Seto, S.; Maemura, K.; Akahoshi, M. Fatty liver incidence and predictive variables. Hypertens. Res. 2010, 33, 638–643.
  17. Zhang, T.; Zhang, C.; Zhang, Y.; Tang, F.; Li, H.; Zhang, Q.; Lin, H.; Wu, S.; Liu, Y.; Xue, F. Metabolic syndrome and its components as predictors of nonalcoholic fatty liver disease in a northern urban Han Chinese population: A prospective cohort study. Atherosclerosis 2015, 240, 144–148.
  18. Sorrentino, P.; Terracciano, L.; D’Angelo, S.; Ferbo, U.; Bracigliano, A.; Vecchione, R. Predicting fibrosis worsening in obese patients with NASH through parenchymal fibronectin, HOMA-IR, and hypertension. Am. J. Gastroenterol. 2010, 105, 336–344.
  19. Wang, Y.; Zeng, Y.; Lin, C.; Chen, Z. Hypertension and non-alcoholic fatty liver disease proven by transient elastography. Hepatol. Res. 2016, 46, 1304–1310.
  20. Ma, J.; Hwang, S.-J.; Pedley, A.; Massaro, J.M.; Hoffmann, U.; Chung, R.T.; Benjamin, E.J.; Levy, D.; Fox, C.S.; Long, M.T. Bi-directional analysis between fatty liver and cardiovascular disease risk factors. J. Hepatol. 2017, 66, 390–397.
  21. Liu, P.; Tang, Y.; Guo, X.; Zhu, X.; He, M.; Yuan, J.; Wang, Y.; Wei, S.; Chen, W.; Zhang, X.; et al. Bidirectional association between nonalcoholic fatty liver disease and hypertension from the Dongfeng-Tongji cohort study. J. Am. Soc. Hypertens. 2018, 12, 660–670.
  22. Zhang, Y.; Zhang, T.; Zhang, C.; Tang, F.; Zhong, N.; Li, H.; Song, X.; Lin, H.; Liu, Y.; Xue, F. Identification of reciprocal causality between non-alcoholic fatty liver disease and metabolic syndrome by a simplified Bayesian network in a Chinese population. BMJ Open 2015, 5, e008204.
  23. European Association for the Study of The Liver; European Association for the Study of Diabetes. EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J. Hepatol. 2016, 64, 1388–1402.
  24. 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.
  25. Hekmatdoost, A.; Shamsipour, A.; Meibodi, M.; Gheibizadeh, N.; Eslamparast, T.; Poustchi, H. Adherence to the Dietary Approaches to Stop Hypertension (DASH) and risk of Nonalcoholic Fatty Liver Disease. Int. J. Food Sci. Nutr. 2016, 67, 1024–1029.
  26. Maskarinec, G.; Lim, U.; Jacobs, S.; Monroe, K.R.; Ernst, T.; Buchthal, S.D.; Shepherd, J.A.; Wilkens, L.R.; Le Marchand, L.; Boushey, C.J. Diet Quality in Midadulthood Predicts Visceral Adiposity and Liver Fatness in Older Ages: The Multiethnic Cohort Study. Obesity 2017, 25, 1442–1450.
  27. Park, S.Y.; Noureddin, M.; Boushey, C.; Wilkens, L.R.; Setiawan, V.W. Diet Quality Association with Nonalcoholic Fatty Liver Disease by Cirrhosis Status: The Multiethnic Cohort. Curr. Dev. Nutr. 2020, 4, nzaa024.
  28. Akhlaghi, M.; Ghasemi-Nasab, M.; Riasatian, M. Mediterranean diet for patients with non-alcoholic fatty liver disease, a systematic review and meta-analysis of observational and clinical investigations. J. Diabetes Metab. Disord. 2020, 19, 575–584.
  29. Razavi Zade, M.; Telkabadi, M.H.; Bahmani, F.; Salehi, B.; Farshbaf, S.; Asemi, Z. The effects of DASH diet on weight loss and metabolic status in adults with non-alcoholic fatty liver disease: A randomized clinical trial. Liver Int. 2016, 36, 563–571.
  30. Yang, M.; Ma, X.; Xuan, X.; Deng, H.; Chen, Q.; Yuan, L. Liraglutide Attenuates Non-Alcoholic Fatty Liver Disease in Mice by Regulating the Local Renin-Angiotensin System. Front. Pharmacol. 2020, 11, 432.
  31. 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.
  32. 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.
  33. Hirata, T.; Tomita, K.; Kawai, T.; Yokoyama, H.; Shimada, A.; Kikuchi, M.; Hirose, H.; Ebinuma, H.; Irie, J.; Ojiro, K.; et al. Effect of Telmisartan or Losartan for Treatment of Nonalcoholic Fatty Liver Disease: Fatty Liver Protection Trial by Telmisartan or Losartan Study (FANTASY). Int. J. Endocrinol. 2013, 2013, 587140.
  34. Goh, G.B.; Pagadala, M.R.; Dasarathy, J.; Unalp-Arida, A.; Sargent, R.; Hawkins, C.; Sourianarayanane, A.; Khiyami, A.; Yerian, L.; Pai, R.; et al. Renin-angiotensin system and fibrosis in non-alcoholic fatty liver disease. Liver Int. 2015, 35, 979–985.
  35. Chen, M.M.; Cai, J.-J.; Yu, Y.; She, Z.-G.; Li, H. Current and Emerging Approaches for Nonalcoholic Steatohepatitis Treatment. Gene Expr. 2019, 19, 175–185.
  36. Verzijl, C.R.C.; van de Peppel, I.; Struik, D.; Jonker, J.W. Pegbelfermin (BMS-986036): An investigational PEGylated fibroblast growth factor 21 analogue for the treatment of nonalcoholic steatohepatitis. Expert Opin. Investig. Drugs 2020, 29, 125–133.
  37. Ratziu, V.; Sanyal, A.J.; Loomba, R.; Rinella, M.; Harrison, S.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; MacConell, L.; Shringarpure, R.; et al. REGENERATE: Design of a pivotal, randomised, phase 3 study evaluating the safety and efficacy of obeticholic acid in patients with fibrosis due to nonalcoholic steatohepatitis. Contemp. Clin. Trials 2019, 84, 105803.
  38. 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.
  39. Abdelmalek, M.F.; Charles, E.D.; Sanyal, A.J.; Harrison, S.A.; Neuschwander-Tetri, B.A.; Goodman, Z.; Ehman, R.A.; Karsdal, M.; Nakajima, A.; Du, S.; et al. The FALCON program: Two phase 2b randomized, double-blind, placebo-controlled studies to assess the efficacy and safety of pegbelfermin in the treatment of patients with nonalcoholic steatohepatitis and bridging fibrosis or compensated cirrhosis. Contemp. Clin. Trials 2021, 104, 106335.
  40. Sanyal, A.J.; Ling, L.; Beuers, U.; DePaoli, A.M.; Lieu, H.D.; Harrison, S.A.; Hirschfield, G.M. Potent suppression of hydrophobic bile acids by aldafermin, an FGF19 analogue, across metabolic and cholestatic liver diseases. JHEP Rep. 2021, 3, 100255.
  41. Sanyal, A.; Charles, E.D.; Neuschwander-Tetri, B.A.; Loomba, R.; Harrison, S.A.; Abdelmalek, M.F.; Lawitz, E.J.; Halegoua-DeMarzio, D.; Kundu, S.; Noviello, S.; et al. Pegbelfermin (BMS-986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: A randomised, double-blind, placebo-controlled, phase 2a trial. Lancet 2019, 392, 2705–2717.
  42. Friedman, S.L.; Ratziu, V.; Harrison, S.A.; Abdelmalek, M.F.; Aithal, G.P.; Caballeria, J.; Francque, S.; Farrell, G.; Kowdley, K.V.; Craxi, A.; et al. A randomized, placebo-controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology 2018, 67, 1754–1767.
  43. Chan, C.T.; Moore, J.P.; Budzyn, K.; Guida, E.; Diep, H.; Vinh, A.; Jones, E.S.; Widdop, R.; Armitage, J.; Sakkal, S.; et al. Reversal of vascular macrophage accumulation and hypertension by a CCR2 antagonist in deoxycorticosterone/salt-treated mice. Hypertension 2012, 60, 1207–1212.
  44. Khan, M.A.B.; Hashim, M.J.; King, J.K.; Govender, R.D.; Mustafa, H.; Al Kaabi, J. Epidemiology of Type 2 Diabetes-Global Burden of Disease and Forecasted Trends. J. Epidemiol. Glob. Health 2020, 10, 107–111.
  45. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84.
  46. Younossi, Z.M.; Golabi, P.; de Avila, L.; Paik, J.M.; Srishord, M.; Fukui, N.; Qiu, Y.; Burns, L.; Afendy, A.; Nader, F. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. J. Hepatol. 2019, 71, 793–801.
  47. Hossain, N.; Afendy, A.; Stepanova, M.; Nader, F.; Srishord, M.; Rafiq, N.; Goodman, Z.; Younossi, Z. Independent predictors of fibrosis in patients with nonalcoholic fatty liver disease. Clin. Gastroenterol. Hepatol. 2009, 7, 1224–1229, 1229.e1-2.
  48. Stepanova, M.; Rafiq, N.; Makhlouf, H.; Agrawal, R.; Kaur, I.; Younoszai, Z.; McCullough, A.; Goodman, Z.; Younossi, Z.M. Predictors of all-cause mortality and liver-related mortality in patients with non-alcoholic fatty liver disease (NAFLD). Dig. Dis. Sci. 2013, 58, 3017–3023.
  49. Baffy, G.; Brunt, E.M.; Caldwell, S.H. Hepatocellular carcinoma in non-alcoholic fatty liver disease: An emerging menace. J. Hepatol. 2012, 56, 1384–1391.
  50. 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.
  51. Stepanova, M.; Rafiq, N.; Younossi, Z.M. Components of metabolic syndrome are independent predictors of mortality in patients with chronic liver disease: A population-based study. Gut 2010, 59, 1410–1415.
  52. Targher, G.; Bertolini, L.; Poli, F.; Rodella, S.; Scala, L.; Tessari, R.; Zenari, L.; Falezza, G. Nonalcoholic fatty liver disease and risk of future cardiovascular events among type 2 diabetic patients. Diabetes 2005, 54, 3541–3546.
  53. Targher, G.; Bertolini, L.; Padovani, R.; Rodella, S.; Tessari, R.; Zenari, L.; Day, C.; Arcaro, G. Prevalence of nonalcoholic fatty liver disease and its association with cardiovascular disease among type 2 diabetic patients. Diabetes Care 2007, 30, 1212–1218.
  54. Cakir, E.; Ozbek, E.; Colak, N.; Cakal, E.; Delıbaşi, T. Is NAFLD an independent risk factor for increased IMT in T2DM? Minerva Endocrinol. 2012, 37, 187–193.
  55. 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.
  56. Hazlehurst, J.M.; Woods, C.; Marjot, T.; Cobbold, J.F.; Tomlinson, J.W. Non-alcoholic fatty liver disease and diabetes. Metabolism 2016, 65, 1096–1108.
  57. Anstee, Q.M.; Targher, G.; Day, C.P. Progression of NAFLD to diabetes mellitus, cardiovascular disease or cirrhosis. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 330–344.
  58. Morrison, A.E.; Zaccardi, F.; Khunti, K.; Davies, M.J. Causality between non-alcoholic fatty liver disease and risk of cardiovascular disease and type 2 diabetes: A meta-analysis with bias analysis. Liver Int. 2019, 39, 557–567.
  59. Adams, L.A.; Harmsen, S.; Sauver, J.S.; Charatcharoenwitthaya, P.; Enders, F.; Therneau, T.; Angulo, P. Nonalcoholic fatty liver disease increases risk of death among patients with diabetes: A community-based cohort study. Am. J. Gastroenterol. 2010, 105, 1567–1573.
  60. Musso, G.; Gambino, R.; Cassader, M.; Pagano, G. Meta-analysis: Natural history of non-alcoholic fatty liver disease (NAFLD) and diagnostic accuracy of non-invasive tests for liver disease severity. Ann. Med. 2011, 43, 617–649.
  61. Ballestri, S.; Zona, S.; Targher, G.; Romagnoli, D.; Baldelli, E.; Nascimbeni, F.; Roverato, A.; Guaraldi, G.; Lonardo, A. Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome. Evidence from a systematic review and meta-analysis. J. Gastroenterol. Hepatol. 2016, 31, 936–944.
  62. Mantovani, A.; Byrne, C.D.; Bonora, E.; Targher, G. Nonalcoholic Fatty Liver Disease and Risk of Incident Type 2 Diabetes: A Meta-analysis. Diabetes Care 2018, 41, 372–382.
  63. Kim, C.H.; Park, J.-Y.; Lee, K.-U.; Kim, J.-H.; Kim, H.-K. Fatty liver is an independent risk factor for the development of Type 2 diabetes in Korean adults. Diabet. Med. 2008, 25, 476–481.
  64. Park, S.K.; Seo, M.H.; Shin, H.C.; Ryoo, J.-H. Clinical availability of nonalcoholic fatty liver disease as an early predictor of type 2 diabetes mellitus in Korean men: 5-year prospective cohort study. Hepatology 2013, 57, 1378–1383.
  65. Bae, J.C.; Cho, Y.K.; Lee, W.Y.; Seo, H.I.; Rhee, E.J.; Park, S.E.; Park, C.Y.; Oh, K.W.; Sung, K.C.; Kim, B.I. Impact of nonalcoholic fatty liver disease on insulin resistance in relation to HbA1c levels in nondiabetic subjects. Am. J. Gastroenterol. 2010, 105, 2389–2395.
  66. Kotronen, A.; Juurinen, L.; Hakkarainen, A.; Westerbacka, J.; Cornér, A.; Bergholm, R.; Yki-Järvinen, H. Liver fat is increased in type 2 diabetic patients and underestimated by serum alanine aminotransferase compared with equally obese nondiabetic subjects. Diabetes Care 2008, 31, 165–169.
  67. Barb, D.; Repetto, E.M.; Stokes, M.E.; Shankar, S.S.; Cusi, K. Type 2 diabetes mellitus increases the risk of hepatic fibrosis in individuals with obesity and nonalcoholic fatty liver disease. Obesity 2021, 29, 1950–1960.
  68. Corey, K.E.; Klebanoff, M.J.; Tramontano, A.C.; Chung, R.T.; Hur, C. Screening for Nonalcoholic Steatohepatitis in Individuals with Type 2 Diabetes: A Cost-Effectiveness Analysis. Dig. Dis. Sci. 2016, 61, 2108–2117.
  69. Perry, R.J.; Samuel, V.T.; Petersen, K.F.; Shulman, G.I. The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes. Nature 2014, 510, 84–91.
  70. Al-Jiffri, O.; Alsharif, F.M.; El-Kader, S.M.A.; Ashmawy, E.M. Weight reduction improves markers of hepatic function and insulin resistance in type-2 diabetic patients with non-alcoholic fatty liver. Afr. Health Sci. 2013, 13, 667–672.
  71. Kistler, K.D.; Brunt, E.M.; Clark, J.M.; Diehl, A.M.; Sallis, J.F.; Schwimmer, J.B.; for the NASH CRN Research Group. Physical activity recommendations, exercise intensity, and histological severity of nonalcoholic fatty liver disease. Am. J. Gastroenterol. 2011, 106, 460–468, quiz 469.
  72. Stewart, K.E.; Haller, D.L.; Sargeant, C.; Levenson, J.L.; Puri, P.; Sanyal, A.J. Readiness for behaviour change in non-alcoholic fatty liver disease: Implications for multidisciplinary care models. Liver Int. 2015, 35, 936–943.
  73. Gastaldelli, A.; Cusi, K. From NASH to diabetes and from diabetes to NASH: Mechanisms and treatment options. JHEP Rep. 2019, 1, 312–328.
  74. Duseja, A.; Das, A.; Dhiman, R.K.; Chawla, Y.K.; Thumburu, K.K.; Bhadada, S.; Bhansali, A. Metformin is effective in achieving biochemical response in patients with nonalcoholic fatty liver disease (NAFLD) not responding to lifestyle interventions. Ann. Hepatol. 2007, 6, 222–226.
  75. Shyangdan, D.; Clar, C.; Ghouri, N.; Henderson, R.; Gurung, T.; Preiss, D.; Sattar, N.; Fraser, A.; Waugh, N. Insulin sensitisers in the treatment of non-alcoholic fatty liver disease: A systematic review. Health Technol. Assess. 2011, 15, 1–110.
  76. 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.
  77. Colca, J.R.; McDonald, W.G.; Adams, W.J. MSDC-0602K, a metabolic modulator directed at the core pathology of non-alcoholic steatohepatitis. Expert Opin. Investig. Drugs 2018, 27, 631–636.
  78. Cuthbertson, D.J.; Irwin, A.; Gardner, C.J.; Daousi, C.; Purewal, T.; Furlong, N.; Goenka, N.; Thomas, E.L.; Adams, V.L.; Pushpakom, S.P.; et al. Improved glycaemia correlates with liver fat reduction in obese, type 2 diabetes, patients given glucagon-like peptide-1 (GLP-1) receptor agonists. PLoS ONE 2012, 7, e50117.
  79. Eckel, R.H.; Grundy, S.M.; Zimmet, P.Z. The metabolic syndrome. Lancet 2005, 365, 1415–1428.
  80. Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wong, V.W.-S.; Dufour, J.-F.; Schattenberg, J.M.; et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J. Hepatol. 2020, 73, 202–209.
  81. Grundy, S.M.; Cleeman, J.I.; Daniels, S.R.; Donato, K.A.; Eckel, R.H.; Franklin, B.A.; Gordon, D.J.; Krauss, R.M.; Savage, P.J.; SmithJr, S.C.; et al. Diagnosis and management of the metabolic syndrome: An American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation 2005, 112, 2735–2752.
  82. Alberti, K.G.M.M.; Zimmet, P.; Shaw, J.; IDF Epidemiology Task Force Consensus Group. The metabolic syndrome—A new worldwide definition. Lancet 2005, 366, 1059–1062.
  83. Isomaa, B.; Almgren, P.; Tuomi, T.; Forsén, B.; Lahti, K.; Nissén, M.; Taskinen, M.-R.; Groop, L. Cardiovascular morbidity and mortality associated with the metabolic syndrome. Diabetes Care 2001, 24, 683–689.
  84. Lakka, H.M.; Laaksonen, D.E.; Lakka, T.; Niskanen, L.K.; Kumpusalo, E.; Tuomilehto, J.; Salonen, J.T. The metabolic syndrome and total and cardiovascular disease mortality in middle-aged men. JAMA 2002, 288, 2709–2716.
  85. Girman, C.J.; Rhodes, T.; Mercuri, M.; Pyörälä, K.; Kjekshus, J.; Pedersen, T.R.; Beere, P.A.; Gotto, A.M.; Clearfield, M. The metabolic syndrome and risk of major coronary events in the Scandinavian Simvastatin Survival Study (4S) and the Air Force/Texas Coronary Atherosclerosis Prevention Study (AFCAPS/TexCAPS). Am. J. Cardiol. 2004, 93, 136–141.
  86. Moore, J.X.; Chaudhary, N.; Akinyemiju, T. Metabolic Syndrome Prevalence by Race/Ethnicity and Sex in the United States, National Health and Nutrition Examination Survey, 1988–2012. Prev. Chronic. Dis. 2017, 14, E24.
  87. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12.
  88. van Vliet-Ostaptchouk, J.V.; Nuotio, M.-L.; Slagter, S.N.; Doiron, D.; Fischer, K.; Foco, L.; Gaye, A.; Heier, M.; Hiekkalinna, T.; Joensuu, A.; et al. The prevalence of metabolic syndrome and metabolically healthy obesity in Europe: A collaborative analysis of ten large cohort studies. BMC Endocr. Disord. 2014, 14, 9.
  89. Rector, R.S.; Thyfault, J.P.; Wei, Y.; Ibdah, J.A. Non-alcoholic fatty liver disease and the metabolic syndrome: An update. World J. Gastroenterol. 2008, 14, 185–192.
  90. Marchesini, G.; Bugianesi, E.; Forlani, G.; Cerrelli, F.; Lenzi, M.; Manini, R.; Natale, S.; Vanni, E.; Villanova, N.; Melchionda, N.; et al. Nonalcoholic fatty liver, steatohepatitis, and the metabolic syndrome. Hepatology 2003, 37, 917–923.
  91. Jinjuvadia, R.; Antaki, F.; Lohia, P.; Liangpunsakul, S. The Association Between Nonalcoholic Fatty Liver Disease and Metabolic Abnormalities in The United States Population. J. Clin. Gastroenterol. 2017, 51, 160–166.
  92. Younossi, Z.M.; Otgonsuren, M.; Venkatesan, C.; Mishra, A. In patients with non-alcoholic fatty liver disease, metabolically abnormal individuals are at a higher risk for mortality while metabolically normal individuals are not. Metabolism 2013, 62, 352–360.
  93. Golabi, P.; Otgonsuren, M.; de Avila, L.; Sayiner, M.; Rafiq, N.; Younossi, Z.M. Components of metabolic syndrome increase the risk of mortality in nonalcoholic fatty liver disease (NAFLD). Medicine 2018, 97, e0214.
  94. Kim, D.; Kim, W.; Joo, S.K.; Han, J.; Kim, J.H.; Harrison, S.A.; Younossi, Z.M.; Ahmed, A. Association between body size-metabolic phenotype and nonalcoholic steatohepatitis and significant fibrosis. J. Gastroenterol. 2020, 55, 330–341.
  95. Siddiqui, M.S.; Yamada, G.; Vuppalanchi, R.; Van Natta, M.; Loomba, R.; Guy, C.; Brandman, D.; Tonascia, J.; Chalasani, N.; Neuschwander-Tetri, B.; et al. Diagnostic Accuracy of Noninvasive Fibrosis Models to Detect Change in Fibrosis Stage. Clin. Gastroenterol. Hepatol. 2019, 17, 1877–1885.e5.
  96. Wainwright, P.; Byrne, C.D. Bidirectional Relationships and Disconnects between NAFLD and Features of the Metabolic Syndrome. Int. J. Mol. Sci. 2016, 17, 367.
  97. Zhang, T.; Zhang, Y.; Zhang, C.; Tang, F.; Li, H.; Zhang, Q.; Lin, H.; Wu, S.; Liu, Y.; Xue, F. Prediction of metabolic syndrome by non-alcoholic fatty liver disease in northern urban Han Chinese population: A prospective cohort study. PLoS ONE 2014, 9, e96651.
  98. Adams, L.A.; Waters, O.R.; Knuiman, M.W.; Elliott, R.R.; Olynyk, J.K. NAFLD as a risk factor for the development of diabetes and the metabolic syndrome: An eleven-year follow-up study. Am. J. Gastroenterol. 2009, 104, 861–867.
  99. Hamaguchi, M.; Kojima, T.; Itoh, Y.; Harano, Y.; Fujii, K.; Nakajima, T.; Kato, T.; Takeda, N.; Okuda, J.; Ida, K.; et al. The severity of ultrasonographic findings in nonalcoholic fatty liver disease reflects the metabolic syndrome and visceral fat accumulation. Am. J. Gastroenterol. 2007, 102, 2708–2715.
  100. Mohan, V.; Farooq, S.; Deepa, M.; Ravikumar, R.; Pitchumoni, C. Prevalence of non-alcoholic fatty liver disease in urban south Indians in relation to different grades of glucose intolerance and metabolic syndrome. Diabetes Res. Clin. Pract. 2009, 84, 84–91.
  101. 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.e5, quiz e14-5.
  102. Katsagoni, C.N.; Georgoulis, M.; Papatheodoridis, G.V.; Panagiotakos, D.B.; Kontogianni, M.D. Effects of lifestyle interventions on clinical characteristics of patients with non-alcoholic fatty liver disease: A meta-analysis. Metabolism 2017, 68, 119–132.
  103. Ballestri, S.; Lonardo, A.; Bonapace, S.; Byrne, C.D.; Loria, P.; Targher, G. Risk of cardiovascular, cardiac and arrhythmic complications in patients with non-alcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 1724–1745.
  104. Aguilar-Olivos, N.E.; Almeda-Valdes, P.; Aguilar-Salinas, C.A.; Uribe, M.; Méndez-Sánchez, N. The role of bariatric surgery in the management of nonalcoholic fatty liver disease and metabolic syndrome. Metabolism 2016, 65, 1196–1207.
  105. 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, quiz e15-6.
  106. Lee, Y.; Doumouras, A.G.; Yu, J.; Brar, K.; Banfield, L.; Gmora, S.; Anvari, M.; Hong, D. Complete Resolution of Nonalcoholic Fatty Liver Disease After Bariatric Surgery: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2019, 17, 1040–1060.e11.
  107. 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.
  108. Byrne, C.D.; Targher, G. NAFLD: A multisystem disease. J. Hepatol. 2015, 62 (Suppl. 1), S47–S64.
  109. Ismaiel, A.; Popa, S.L.; Dumitrascu, D.L. Acute Coronary Syndromes and Nonalcoholic Fatty Liver Disease: “Un Affaire de Coeur”. Can J. Gastroenterol. Hepatol. 2020, 2020, 8825615.
  110. Baharvand-Ahmadi, B.; Sharifi, K.; Namdari, M. Prevalence of non-alcoholic fatty liver disease in patients with coronary artery disease. ARYA Atheroscler. 2016, 12, 201–205.
  111. Akuta, N.; Kawamura, Y.; Arase, Y.; Saitoh, S.; Fujiyama, S.; Sezaki, H.; Hosaka, T.; Kobayashi, M.; Kobayashi, M.; Suzuki, Y.; et al. PNPLA3 genotype and fibrosis-4 index predict cardiovascular diseases of Japanese patients with histopathologically-confirmed NAFLD. BMC Gastroenterol. 2021, 21, 434.
  112. Toh, J.Z.K.; Pan, X.-H.; Tay, P.W.L.; Ng, C.H.; Yong, J.N.; Xiao, J.; Koh, J.H.; Tan, E.Y.; Tan, E.X.X.; Dan, Y.Y.; et al. A Meta-Analysis on the Global Prevalence, Risk factors and Screening of Coronary Heart Disease in Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2021.
  113. 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.
  114. Sunbul, M.; Agirbasli, M.; Durmus, E.; Kivrak, T.; Akin, H.; Aydin, Y.; Ergelen, R.; Yilmaz, Y. Arterial stiffness in patients with non-alcoholic fatty liver disease is related to fibrosis stage and epicardial adipose tissue thickness. Atherosclerosis 2014, 237, 490–493.
  115. 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.
  116. Gholoobi, A.; Gifani, M.; Gholoobi, A.; Akhlaghi, S.; Pezeshki Rad, M.; Baradaran Rahimi, V. Relationship between the prevalence and severity of non-alcoholic fatty liver disease and coronary artery disease: Findings from a cross-sectional study of a referral center in northeast Iran. JGH Open 2022, 6, 330–337.
  117. Cury, R.C.; Abbara, S.; Achenbach, S.; Achenbach, A.; Berman, D.S.; Budoff, M.J.; Dill, K.E.; Jacobs, J.E.; Maroules, C.D.; Rubin, G.D.; et al. Coronary Artery Disease-Reporting and Data System (CAD-RADS): An Expert Consensus Document of SCCT, ACR and NASCI: Endorsed by the ACC. JACC Cardiovasc. Imaging 2016, 9, 1099–1113.
  118. Chen, L.-Z.; Jing, X.-B.; Wu, C.-F.; Zeng, Y.-C.; Xie, Y.-C.; Wang, M.-Q.; Chen, W.-X.; Hu, X.; Zhou, Y.-N.; Cai, X.-B. Nonalcoholic Fatty Liver Disease-Associated Liver Fibrosis Is Linked with the Severity of Coronary Artery Disease Mediated by Systemic Inflammation. Dis. Markers 2021, 2021, 6591784.
  119. Choi, D.H.; Lee, S.J.; Kang, C.D.; Park, M.O.; Choi, N.W.; Kim, T.S.; Lee, W.; Cho, B.R.; Kim, Y.H.; Lee, B.-K.; et al. Nonalcoholic fatty liver disease is associated with coronary artery disease in Koreans. World J. Gastroenterol. 2013, 19, 6453–6457.
  120. Alper, A.T.; Hasdemir, H.; Sahin, S.; Ontürk, E.; Akyol, A.; Nurkalem, Z.; Cakmak, N.; Erdinler, I.; Gürkan, K. The relationship between nonalcoholic fatty liver disease and the severity of coronary artery disease in patients with metabolic syndrome. Turk Kardiyol Dern Ars 2008, 36, 376–381.
  121. Acikel, M.; Sunay, S.; Koplay, M.; Gündoğdu, F.; Karakelleoğlu, S. Evaluation of ultrasonographic fatty liver and severity of coronary atherosclerosis, and obesity in patients undergoing coronary angiography. Anadolu Kardiyol Derg 2009, 9, 273–279.
  122. 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.
  123. Mantovani, A.; Scorletti, E.; Mosca, A.; Alisi, A.; Byrne, C.D.; Targher, G. Complications, morbidity and mortality of nonalcoholic fatty liver disease. Metabolism 2020, 111S, 154170.
  124. Adams, L.A.; Lymp, J.F.; Sauver, J.S.; 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.
  125. Ong, J.P.; Pitts, A.; Younossi, Z.M. Increased overall mortality and liver-related mortality in non-alcoholic fatty liver disease. J. Hepatol. 2008, 49, 608–612.
  126. 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.
  127. Wu, S.; Wu, F.; Ding, Y.; Hou, J.; Bi, J.; Zhang, Z. Association of non-alcoholic fatty liver disease with major adverse cardiovascular events: A systematic review and meta-analysis. Sci. Rep. 2016, 6, 33386.
  128. Ozturk, H.; Gumrukcuoglu, H.A.; Yaman, M.; Akyol, A.; Öztürk; Akdag, S.; Şimşek, H.; Sahin, M.; Günaydın, Z.Y.; Simsek, H.; et al. Hepatosteatosis and carotid intima-media thickness in patients with myocardial infarction. J. Med. Ultrason. 2016, 43, 77–82.
  129. Labenz, C.; Huber, Y.; Michel, M.; Nagel, M.; Galle, P.R.; Kostev, K.; Schattenberg, J.M. Impact of NAFLD on the Incidence of Cardiovascular Diseases in a Primary Care Population in Germany. Dig. Dis. Sci. 2020, 65, 2112–2119.
  130. Sinn, D.H.; Kang, D.; Chang, Y.; Ryu, S.; Cho, S.J.; Paik, S.W.; Bin Song, Y.; Pastor-Barriuso, R.; Guallar, E.; Cho, J.; et al. Non-alcoholic fatty liver disease and the incidence of myocardial infarction: A cohort study. J. Gastroenterol. Hepatol. 2020, 35, 833–839.
  131. Alexander, M.; Loomis, A.K.; Van Der Lei, J.; Duarte-Salles, T.; Prieto-Alhambra, D.; Ansell, D.; Pasqua, A.; Lapi, F.; Rijnbeek, P.; Mosseveld, M.; et al. Non-alcoholic fatty liver disease and risk of incident acute myocardial infarction and stroke: Findings from matched cohort study of 18 million European adults. BMJ 2019, 367, l5367.
  132. Lonardo, A.; Nascimbeni, F.; Ballestri, S.; Fairweather, D.; Win, S.; Than, T.A.; Abdelmalek, M.F.; Suzuki, A. Sex Differences in Nonalcoholic Fatty Liver Disease: State of the Art and Identification of Research Gaps. Hepatology 2019, 70, 1457–1469.
  133. Allen, A.M.; Therneau, T.M.; Mara, K.C.; Larson, J.J.; Watt, K.D.; Hayes, S.N.; Kamath, P.S. Women With Nonalcoholic Fatty Liver Disease Lose Protection Against Cardiovascular Disease: A Longitudinal Cohort Study. Am. J. Gastroenterol. 2019, 114, 1764–1771.
  134. Khalid, Y.S.; Dasu, N.R.; Suga, H.; Dasu, K.N.; Reja, D.; Shah, A.; McMahon, D.; Levine, A. Increased cardiovascular events and mortality in females with NAFLD: A meta-analysis. Am. J. Cardiovasc. Dis. 2020, 10, 258–271.
  135. Kasper, P.; Martin, A.; Lang, S.; Kütting, F.; Goeser, T.; Demir, M.; Steffen, H.-M. NAFLD and cardiovascular diseases: A clinical review. Clin. Res. Cardiol. 2021, 110, 921–937.
  136. Sao, R.; Aronow, W.S. Association of non-alcoholic fatty liver disease with cardiovascular disease and subclinical atherosclerosis. Arch. Med. Sci. 2018, 14, 1233–1244.
  137. Pedersen, L.R.; Olsen, R.; Anholm, C.; Astrup, A.; Eugen-Olsen, J.; Fenger, M.; Simonsen, L.; Walzem, R.L.; Haugaard, S.B.; Prescott, E. Effects of 1 year of exercise training versus combined exercise training and weight loss on body composition, low-grade inflammation and lipids in overweight patients with coronary artery disease: A randomized trial. Cardiovasc. Diabetol. 2019, 18, 127.
  138. Makarewicz-Wujec, M.; Henzel, J.; Kruk, M.; Kępka, C.; Wardziak; Trochimiuk, P.; Parzonko, A.; Demkow, M.; Kozłowska-Wojciechowska, M. DASH diet decreases CXCL4 plasma concentration in patients diagnosed with coronary atherosclerotic lesions. Nutr. Metab. Cardiovasc. Dis. 2020, 30, 56–59.
  139. Yang, Z.Q.; Yang, Z.; Duan, M.L. Dietary approach to stop hypertension diet and risk of coronary artery disease: A meta-analysis of prospective cohort studies. Int. J. Food Sci. Nutr. 2019, 70, 668–674.
  140. Sekhar, A.; Kuttan, A.; Borges, J.C.; Rajachandran, M. Food for Thought or Feeding a Dogma? Diet and Coronary Artery Disease: A Clinician’s Perspective. Curr. Cardiol. Rep. 2021, 23, 127.
  141. Mathurin, P.; Hollebecque, A.; Arnalsteen, L.; Buob, D.; Leteurtre, E.; Caiazzo, R.; Pigeyre, M.; Verkindt, H.; Dharancy, S.; Louvet, A.; et al. Prospective study of the long-term effects of bariatric surgery on liver injury in patients without advanced disease. Gastroenterology 2009, 137, 532–540.
  142. Chavez-Tapia, N.C.; Tellez-Avila, F.I.; Barrientos-Gutierrez, T.; Mendez-Sanchez, N.; Lizardi-Cervera, J.; Uribe, M. Bariatric surgery for non-alcoholic steatohepatitis in obese patients. Cochrane Database Syst. Rev. 2010, 1, CD007340.
  143. Bower, G.; Athanasiou, T.; Isla, A.M.; Harling, L.; Li, J.; Holmes, E.; Efthimiou, E.; Darzi, A.; Ashrafian, H. Bariatric surgery and nonalcoholic fatty liver disease. Eur. J. Gastroenterol. Hepatol. 2015, 27, 755–768.
  144. Kontogianni, M.D.; Tileli, N.; Margariti, A.; Georgoulis, M.; Deutsch, M.; Tiniakos, D.; Fragopoulou, E.; Zafiropoulou, R.; Manios, Y.; Papatheodoridis, G. Adherence to the Mediterranean diet is associated with the severity of non-alcoholic fatty liver disease. Clin. Nutr. 2014, 33, 678–683.
  145. Trovato, F.M.; Catalano, D.; Martines, C.F.; Pace, P.; Trovato, G.M. Mediterranean diet and non-alcoholic fatty liver disease: The need of extended and comprehensive interventions. Clin. Nutr. 2015, 34, 86–88.
  146. Romero-Gomez, M.; Zelber-Sagi, S.; Trenell, M. Treatment of NAFLD with diet, physical activity and exercise. J. Hepatol. 2017, 67, 829–846.
  147. Doustmohammadian, A.; Clark, C.C.T.; Maadi, M.; Motamed, N.; Sobhrakhshankhah, E.; Ajdarkosh, H.; Mansourian, M.R.; Esfandyari, S.; Hanjani, N.A.; Nikkhoo, M.; et al. Favorable association between Mediterranean diet (MeD) and DASH with NAFLD among Iranian adults of the Amol Cohort Study (AmolCS). Sci. Rep. 2022, 12, 2131.
  148. Caussy, C.; Aubin, A.; Loomba, R. The Relationship Between Type 2 Diabetes, NAFLD, and Cardiovascular Risk. Curr. Diab. Rep. 2021, 21, 15.
  149. Grontved, A.; Hu, F.B. Television viewing and risk of type 2 diabetes, cardiovascular disease, and all-cause mortality: A meta-analysis. JAMA 2011, 305, 2448–2455.
  150. Church, T.S.; Kuk, J.L.; Ross, R.; Priest, E.L.; Biltoff, E.; Blair, S.N. Association of cardiorespiratory fitness, body mass index, and waist circumference to nonalcoholic fatty liver disease. Gastroenterology 2006, 130, 2023–2030.
  151. Aithal, G.P.; Thomas, J.; Kaye, P.V.; Lawson, A.; Ryder, S.D.; Spendlove, I.; Austin, A.S.; Freeman, J.G.; Morgan, L.; Webber, J. Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology 2008, 135, 1176–1184.
  152. Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685.
  153. 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.
  154. Dormandy, J.A.; Charbonnel, B.; Eckland, D.J.A.; Erdmann, E.; Massi-Benedetti, M.; Moules, I.K.; Skene, A.M.; Tan, M.H.; Lefèbvre, P.J.; Murray, G.D.; et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): A randomised controlled trial. Lancet 2005, 366, 1279–1289.
  155. Erdmann, E.; Dormandy, J.A.; Charbonnel, B.; Massi-Benedetti, M.; Moules, I.K.; Skene, A.M. The effect of pioglitazone on recurrent myocardial infarction in 2,445 patients with type 2 diabetes and previous myocardial infarction: Results from the PROactive (PROactive 05) Study. J. Am. Coll. Cardiol. 2007, 49, 1772–1780.
  156. Liao, H.W.; Saver, J.L.; Wu, Y.-L.; Chen, T.-H.; Lee, M.; Ovbiagele, B. Pioglitazone and cardiovascular outcomes in patients with insulin resistance, pre-diabetes and type 2 diabetes: A systematic review and meta-analysis. BMJ Open 2017, 7, e013927.
  157. Eguchi, Y.; Kitajima, Y.; Hyogo, H.; Takahashi, H.; Kojima, M.; Ono, M.; Araki, N.; Tanaka, K.; Yamaguchi, M.; Matsuda, Y.; et al. Pilot study of liraglutide effects in non-alcoholic steatohepatitis and non-alcoholic fatty liver disease with glucose intolerance in Japanese patients (LEAN-J). Hepatol. Res. 2015, 45, 269–278.
  158. 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.
  159. Mantovani, A.; Byrne, C.; Scorletti, E.; Mantzoros, C.; Targher, G. Efficacy and safety of anti-hyperglycaemic drugs in patients with non-alcoholic fatty liver disease with or without diabetes: An updated systematic review of randomized controlled trials. Diabetes Metab. 2020, 46, 427–441.
  160. Kristensen, S.L.; Rørth, R.; Jhund, P.S.; Docherty, K.F.; Sattar, N.; Preiss, D.; Køber, L.; Petrie, M.C.; McMurray, J.J.V. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: A systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol. 2019, 7, 776–785.
  161. Zhu, J.; Yu, X.; Zheng, Y.; Li, J.; Wang, Y.; Lin, Y.; He, Z.; Zhao, W.; Chen, C.; Qiu, K.; et al. Association of glucose-lowering medications with cardiovascular outcomes: An umbrella review and evidence map. Lancet Diabetes Endocrinol. 2020, 8, 192–205.
  162. Xu, R.; Tao, A.; Zhang, S.; Deng, Y.; Chen, G. Association between vitamin E and non-alcoholic steatohepatitis: A meta-analysis. Int. J. Clin. Exp. Med. 2015, 8, 3924–3934.
  163. Sato, K.; Gosho, M.; Yamamoto, T.; Kobayashi, Y.; Ishii, N.; Ohashi, T.; Nakade, Y.; Ito, K.; Fukuzawa, Y.; Yoneda, M. Vitamin E has a beneficial effect on nonalcoholic fatty liver disease: A meta-analysis of randomized controlled trials. Nutrition 2015, 31, 923–930.
  164. Lavine, J.E.; Schwimmer, J.B.; Van Natta, M.L.; Molleston, J.P.; Murray, K.F.; Rosenthal, P.; Abrams, S.H.; Scheimann, A.O.; Sanyal, A.J.; Chalasani, N.; et al. Effect of vitamin E or metformin for treatment of nonalcoholic fatty liver disease in children and adolescents: The TONIC randomized controlled trial. JAMA 2011, 305, 1659–1668.
  165. Miller III, E.R.; Pastor-Barriuso, R.; Dalal, D.; Riemersma, R.A.; Appel, L.J.; Guallar, E. Meta-analysis: High-dosage vitamin E supplementation may increase all-cause mortality. Ann. Intern. Med. 2005, 142, 37–46.
  166. Gerss, J.; Kopcke, W. The questionable association of vitamin E supplementation and mortality--inconsistent results of different meta-analytic approaches. Cell. Mol. Biol. 2009, 55, OL1111-20.
  167. Abner, E.L.; Schmitt, F.A.; Mendiondo, M.S.; Marcum, J.; Kryscio, R.J. Vitamin E and all-cause mortality: A meta-analysis. Curr. Aging Sci. 2011, 4, 158–170.
  168. Klein, E.A.; Thompson, I.; Tangen, C.M.; Lucia, M.S.; Goodman, P.; Minasian, L.M.; Ford, L.G.; Parnes, H.L.; Gaziano, J.M.; Karp, D.D.; et al. Vitamin E and the risk of prostate cancer: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 2011, 306, 1549–1556.
  169. Kristal, A.R.; Darke, A.K.; Morris, J.S.; Tangen, C.M.; Goodman, P.J.; Thompson, I.M.; Meyskens, F.L., Jr.; Goodman, G.E.; Minasian, L.M.; Parnes, H.L.; et al. Baseline selenium status and effects of selenium and vitamin e supplementation on prostate cancer risk. J. Natl. Cancer Inst. 2014, 106, djt456.
  170. Chan, J.M.; Darke, A.K.; Penney, K.L.; Tangen, C.M.; Goodman, P.J.; Lee, G.-S.M.; Sun, T.; Peisch, S.; Tinianow, A.M.; Rae, J.M.; et al. Selenium- or Vitamin E-Related Gene Variants, Interaction with Supplementation, and Risk of High-Grade Prostate Cancer in SELECT. Cancer Epidemiol. Biomarkers Prev. 2016, 25, 1050–1058.
  171. de Keyser, C.E.; Koehler, E.M.; Schouten, J.N.L.; Visser, L.E.; Hofman, A.; Janssen, H.L.A.; Stricker, B.H. Statin therapy is associated with a reduced risk of non-alcoholic fatty liver in overweight individuals. Dig. Liver Dis. 2014, 46, 720–725.
  172. Kiyici, M.; Gulten, M.; Gurel, S.; Nak, S.G.; Dolar, E.; Savci, G.; Adim, S.B.; Yerci, O.; Memik, F. Ursodeoxycholic acid and atorvastatin in the treatment of nonalcoholic steatohepatitis. Can J. Gastroenterol. 2003, 17, 713–718.
  173. Foster, T.; Budoff, M.J.; Saab, S.; Ahmadi, N.; Gordon, C.; Guerci, A.D. Atorvastatin and antioxidants for the treatment of nonalcoholic fatty liver disease: The St Francis Heart Study randomized clinical trial. Am. J. Gastroenterol. 2011, 106, 71–77.
  174. Kimura, Y.; Hyogo, H.; Yamagishi, S.-I.; Takeuchi, M.; Ishitobi, T.; Nabeshima, Y.; Arihiro, K.; Chayama, K. Atorvastatin decreases serum levels of advanced glycation endproducts (AGEs) in nonalcoholic steatohepatitis (NASH) patients with dyslipidemia: Clinical usefulness of AGEs as a biomarker for the attenuation of NASH. J. Gastroenterol. 2010, 45, 750–757.
  175. Ekstedt, M.; Franzén, L.E.; Mathiesen, U.L.; Holmqvist, M.; Bodemar, G.; Kechagias, S. Statins in non-alcoholic fatty liver disease and chronically elevated liver enzymes: A histopathological follow-up study. J. Hepatol. 2007, 47, 135–141.
  176. Athyros, V.G.; Alexandrides, T.K.; Bilianou, H.; Cholongitas, E.; Doumas, M.; Ganotakis, E.S.; Goudevenos, J.; Elisaf, M.S.; Germanidis, G.; Giouleme, O.; et al. The use of statins alone, or in combination with pioglitazone and other drugs, for the treatment of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis and related cardiovascular risk. An Expert Panel Statement. Metabolism 2017, 71, 17–32.
  177. Kim, J.; Lee, H.; An, J.; Song, Y.; Lee, C.-K.; Kim, K.; Kong, H. Alterations in Gut Microbiota by Statin Therapy and Possible Intermediate Effects on Hyperglycemia and Hyperlipidemia. Front. Microbiol. 2019, 10, 1947.
  178. Tikkanen, M.J.; Fayyad, R.; Faergeman, O.; Olsson, A.G.; Wun, C.-C.; Laskey, R.; Kastelein, J.J.; Holme, I.; Pedersen, T.R. Effect of intensive lipid lowering with atorvastatin on cardiovascular outcomes in coronary heart disease patients with mild-to-moderate baseline elevations in alanine aminotransferase levels. Int. J. Cardiol. 2013, 168, 3846–3852.
  179. 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.
  180. Lewis, J.H.; Mortensen, M.E.; Zweig, S.; Fusco, M.J.; Medoff, J.R.; Belder, R.; Pravastatin in Chronic Liver Disease Study Investigators. Efficacy and safety of high-dose pravastatin in hypercholesterolemic patients with well-compensated chronic liver disease: Results of a prospective, randomized, double-blind, placebo-controlled, multicenter trial. Hepatology 2007, 46, 1453–1463.
  181. Chalasani, N.; Aljadhey, H.; Kesterson, J.; Murray, M.D.; Hall, S.D. Patients with elevated liver enzymes are not at higher risk for statin hepatotoxicity. Gastroenterology 2004, 126, 1287–1292.
  182. Sigler, M.A.; Congdon, L.; Edwards, K.L. An Evidence-Based Review of Statin Use in Patients With Nonalcoholic Fatty Liver Disease. Clin. Med. Insights Gastroenterol. 2018, 11, 1179552218787502.
  183. Stone, N.J.; Robinson, J.G.; Lichtenstein, A.H.; Bairey Merz, C.N.; Blum, C.B.; Eckel, R.H.; Goldberg, A.C.; Gordon, D.; Levy, D.; Lloyd-Jones, D.M.; et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J. Am. Coll. Cardiol. 2014, 63, 2889–2934.
  184. 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.
  185. Jung, J.Y.; Park, S.K.; Ryoo, J.; Oh, C.; Kang, J.G.; Lee, J.; Choi, J. Effect of non-alcoholic fatty liver disease on left ventricular diastolic function and geometry in the Korean general population. Hepatol. Res. 2017, 47, 522–532.
  186. Graner, M.; Nyman, K.; Siren, R.; Pentikäinen, M.O.; Lundbom, J.; Hakkarainen, A.; Lauerma, K.; Lundbom, N.; Nieminen, M.S.; Taskinen, M.-R. Ectopic fat depots and left ventricular function in nondiabetic men with nonalcoholic fatty liver disease. Circ. Cardiovasc. Imaging 2015, 8, e001979.
  187. Petta, S.; Argano, C.; Colomba, D.; Cammà, C.; Di Marco, V.; Cabibi, D.; Tuttolomondo, A.; Marchesini, G.; Pinto, A.; Licata, G.; et al. Epicardial fat, cardiac geometry and cardiac function in patients with non-alcoholic fatty liver disease: Association with the severity of liver disease. J. Hepatol. 2015, 62, 928–933.
  188. Miller, A.; McNamara, J.; Hummel, S.L.; Konerman, M.C.; Tincopa, M.A. Prevalence and staging of non-alcoholic fatty liver disease among patients with heart failure with preserved ejection fraction. Sci. Rep. 2020, 10, 12440.
  189. Zhang, Z.; Wang, P.; Guo, F.; Liu, X.; Luo, T.; Guan, Y.; Chen, H.; Wang, Z.; Zhao, L.; Ma, X.; et al. Chronic heart failure in patients with nonalcoholic fatty liver disease: Prevalence, clinical features, and relevance. J. Int. Med. Res. 2018, 46, 3959–3969.
  190. Chung, G.E.; Lee, J.-H.; Lee, H.; Kim, M.K.; Yim, J.Y.; Choi, S.-Y.; Kim, Y.J.; Yoon, J.-H.; Kim, D. Nonalcoholic fatty liver disease and advanced fibrosis are associated with left ventricular diastolic dysfunction. Atherosclerosis 2018, 272, 137–144.
  191. Stahl, E.P.; Dhindsa, D.S.; Lee, S.K.; Sandesara, P.K.; Chalasani, N.P.; Sperling, L.S. Nonalcoholic Fatty Liver Disease and the Heart: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 948–963.
  192. Zhou, J.; Bai, L.; Zhang, X.; Li, H.; Cai, J. Nonalcoholic Fatty Liver Disease and Cardiac Remodeling Risk: Pathophysiological Mechanisms and Clinical Implications. Hepatology 2021, 74, 2839–2847.
  193. Cai, J.; Xu, M.; Zhang, X.; Li, H. Innate Immune Signaling in Nonalcoholic Fatty Liver Disease and Cardiovascular Diseases. Annu. Rev. Pathol. 2019, 14, 153–184.
  194. Fotbolcu, H.; Yakar, T.; Duman, D.; Karaahmet, T.; Tigen, K.; Cevik, C.; Kurtoglu, U.; Dindar, I. Impairment of the left ventricular systolic and diastolic function in patients with non-alcoholic fatty liver disease. Cardiol. J. 2010, 17, 457–463.
  195. Lee, Y.H.; Kim, K.J.; Yoo, M.E.; Kim, G.; Yoon, H.-J.; Jo, K.; Youn, J.-C.; Yun, M.; Park, J.Y.; Shim, C.Y.; et al. Association of non-alcoholic steatohepatitis with subclinical myocardial dysfunction in non-cirrhotic patients. J. Hepatol. 2018, 68, 764–772.
  196. Valbusa, F.; Bonapace, S.; Agnoletti, D.; Scala, L.; Grillo, C.; Arduini, P.; Turcato, E.; Mantovani, A.; Zoppini, G.; Arcaro, G.; et al. Nonalcoholic fatty liver disease and increased risk of 1-year all-cause and cardiac hospital readmissions in elderly patients admitted for acute heart failure. PLoS ONE 2017, 12, e0173398.
  197. Yoshihisa, A.; Sato, Y.; Yokokawa, T.; Sato, T.; Suzuki, S.; Oikawa, M.; Kobayashi, A.; Yamaki, T.; Kunii, H.; Nakazato, K.; et al. Liver fibrosis score predicts mortality in heart failure patients with preserved ejection fraction. ESC Heart Fail. 2018, 5, 262–270.
  198. Packer, M. Atrial Fibrillation and Heart Failure With Preserved Ejection Fraction in Patients With Nonalcoholic Fatty Liver Disease. Am. J. Med. 2020, 133, 170–177.
  199. Fudim, M.; Zhong, L.; Patel, K.V.; Khera, R.; Abdelmalek, M.F.; Diehl, A.M.; McGarrah, R.W.; Molinger, J.; Moylan, C.A.; Rao, V.N.; et al. Nonalcoholic Fatty Liver Disease and Risk of Heart Failure Among Medicare Beneficiaries. J. Am. Heart Assoc. 2021, 10, e021654.
  200. Cohen, J.B.; Schrauben, S.J.; Zhao, L.; Basso, M.D.; Cvijic, M.E.; Li, Z.; Yarde, M.; Wang, Z.; Bhattacharya, P.T.; Chirinos, D.A.; et al. Clinical Phenogroups in Heart Failure With Preserved Ejection Fraction: Detailed Phenotypes, Prognosis, and Response to Spironolactone. JACC Heart Fail. 2020, 8, 172–184.
  201. Shah, S.J.; Katz, D.H.; Selvaraj, S.; Burke, M.A.; Yancy, C.W.; Gheorghiade, M.; Bonow, R.O.; Huang, C.-C.; Deo, R.C. Phenomapping for novel classification of heart failure with preserved ejection fraction. Circulation 2015, 131, 269–279.
  202. Salah, H.M.; Katz, D.; Selvaraj, S.; Burke, M.A.; Yancy, C.W.; Gheorghiade, M.; Bonow, R.O.; Huang, C.-C.; Deo, R.C. Relationship of Nonalcoholic Fatty Liver Disease and Heart Failure With Preserved Ejection Fraction. JACC Basic Transl. Sci. 2021, 6, 918–932.
  203. Takahashi, T.; Watanabe, T.; Shishido, T.; Watanabe, K.; Sugai, T.; Toshima, T.; Kinoshita, D.; Yokoyama, M.; Tamura, H.; Nishiyama, S.; et al. The impact of non-alcoholic fatty liver disease fibrosis score on cardiac prognosis in patients with chronic heart failure. Heart Vessels 2018, 33, 733–739.
  204. Valbusa, F.; Agnoletti, D.; Scala, L.; Grillo, C.; Arduini, P.; Bonapace, S.; Calabria, S.; Scaturro, G.; Mantovani, A.; Zoppini, G.; et al. Non-alcoholic fatty liver disease and increased risk of all-cause mortality in elderly patients admitted for acute heart failure. Int. J. Cardiol. 2018, 265, 162–168.
  205. Minhas, A.M.K.; Bhopalwala, H.M.; Dewaswala, N.; Salah, H.M.; Khan, M.S.; Shahid, I.; Biegus, J.; Lopes, R.D.; Pandey, A.; Fudim, M. Association of Non-Alcoholic Fatty Liver Disease With in-Hospital Outcomes in Primary Heart Failure Hospitalizations With Reduced or Preserved Ejection Fraction. Curr. Probl. Cardiol. 2022, 101199.
  206. Ergatoudes, C.; Schaufelberger, M.; Andersson, B.; Pivodic, A.; Dahlström, U.; Fu, M. Non-cardiac comorbidities and mortality in patients with heart failure with reduced vs. preserved ejection fraction: A study using the Swedish Heart Failure Registry. Clin. Res. Cardiol. 2019, 108, 1025–1033.
  207. Mantovani, A.; Byrne, C.D.; Benfari, G.; Bonapace, S.; Simon, T.G.; Targher, G. Risk of Heart Failure in Patients With Nonalcoholic Fatty Liver Disease: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2022, 79, 180–191.
  208. 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.
  209. Fenk, S.; Fischer, M.; Strack, C.; Schmitz, G.; Loew, T.; Lahmann, C.; Baessler, A. Successful weight reduction improves left ventricular diastolic function and physical performance in severe obesity. Int. Heart J. 2015, 56, 196–202.
  210. McDowell, K.; Petrie, M.; Raihan, N.A.; Logue, J. Effects of intentional weight loss in patients with obesity and heart failure: A systematic review. Obes. Rev. 2018, 19, 1189–1204.
  211. Poirier, P.; Cornier, M.-A.; Mazzone, T.; Stiles, C.; Klein, S.; McCullough, P.A.; Fielding, C.R.; Franklin, B.A. Bariatric surgery and cardiovascular risk factors: A scientific statement from the American Heart Association. Circulation 2011, 123, 1683–1701.
  212. Karimian, S.; Stein, J.; Bauer, B.; Teupe, C. Improvement of impaired diastolic left ventricular function after diet-induced weight reduction in severe obesity. Diabetes Metab. Syndr. Obes. 2017, 10, 19–25.
  213. Fuentes, L.D.L.; Waggoner, A.D.; Mohammed, B.S.; Stein, R.I.; Miller, B.V.; Foster, G.D.; Wyatt, H.R.; Klein, S.; Davila-Roman, V.G. Effect of moderate diet-induced weight loss and weight regain on cardiovascular structure and function. J. Am. Coll. Cardiol. 2009, 54, 2376–2381.
  214. Kindel, T.L.; Strande, J.L. Bariatric surgery as a treatment for heart failure: Review of the literature and potential mechanisms. Surg. Obes. Relat. Dis. 2018, 14, 117–122.
  215. Younossi, Z.M.; Corey, K.E.; Lim, J.K. AGA Clinical Practice Update on Lifestyle Modification Using Diet and Exercise to Achieve Weight Loss in the Management of Nonalcoholic Fatty Liver Disease: Expert Review. Gastroenterology 2021, 160, 912–918.
  216. Arab, J.P.; Dirchwolf, M.; Álvares-Da-Silva, M.R.; Barrera, F.; Benítez, C.; Castellanos-Fernandez, M.; Castro-Narro, G.; Chavez-Tapia, N.; Chiodi, D.; Cotrim, H.; et al. Latin American Association for the study of the liver (ALEH) practice guidance for the diagnosis and treatment of non-alcoholic fatty liver disease. Ann. Hepatol. 2020, 19, 674–690.
  217. Ryan, M.C.; Itsiopoulos, C.; Thodis, T.; Ward, G.; Trost, N.; Hofferberth, S.; O’Dea, K.; Desmond, P.V.; Johnson, N.A.; Wilson, A.M. The Mediterranean diet improves hepatic steatosis and insulin sensitivity in individuals with non-alcoholic fatty liver disease. J. Hepatol. 2013, 59, 138–143.
  218. Pinto, X.; Fanlo-Maresma, M.; Corbella, E.; Corbella, X.; Mitjavila, M.T.; Moreno, J.J.; Casas, R.; Estruch, R.; Corella, D.; Bulló, M.; et al. A Mediterranean Diet Rich in Extra-Virgin Olive Oil Is Associated with a Reduced Prevalence of Nonalcoholic Fatty Liver Disease in Older Individuals at High Cardiovascular Risk. J. Nutr. 2019, 149, 1920–1929.
  219. Hallsworth, K.; Thoma, C.; Hollingsworth, K.G.; Cassidy, S.; Anstee, Q.M.; Day, C.P.; Trenell, M. Modified high-intensity interval training reduces liver fat and improves cardiac function in non-alcoholic fatty liver disease: A randomized controlled trial. Clin. Sci. 2015, 129, 1097–1105.
  220. Lin, J.S.; O’Connor, E.; Evans, C.V.; Senger, C.A.; Rowland, M.G.; Groom, H. Behavioral counseling to promote a healthy lifestyle in persons with cardiovascular risk factors: A systematic review for the U.S. Preventive Services Task Force. Ann. Intern. Med. 2014, 161, 568–578.
  221. Hashida, R.; Kawaguchi, T.; Bekki, M.; Omoto, M.; Matsuse, H.; Nago, T.; Takano, Y.; Ueno, T.; Koga, H.; George, J.; et al. Aerobic vs. resistance exercise in non-alcoholic fatty liver disease: A systematic review. J. Hepatol. 2017, 66, 142–152.
  222. Orci, L.A.; Gariani, K.; Oldani, G.; Delaune, V.; Morel, P.; Toso, C. Exercise-based Interventions for Nonalcoholic Fatty Liver Disease: A Meta-analysis and Meta-regression. Clin. Gastroenterol. Hepatol. 2016, 14, 1398–1411.
  223. Bacchi, E.; Negri, C.; Targher, G.; Faccioli, N.; Lanza, M.; Zoppini, G.; Zanolin, E.; Schena, F.; Bonora, E.; Moghetti, P. Both resistance training and aerobic training reduce hepatic fat content in type 2 diabetic subjects with nonalcoholic fatty liver disease (the RAED2 Randomized Trial). Hepatology 2013, 58, 1287–1295.
  224. Xiong, Y.; Peng, Q.; Cao, C.; Xu, Z.; Zhang, B. Effect of Different Exercise Methods on Non-Alcoholic Fatty Liver Disease: A Meta-Analysis and Meta-Regression. Int. J. Environ. Res. Public Health 2021, 18, 3242.
  225. Golabi, P.; Locklear, C.T.; Austin, P.; Afdhal, S.; Byrns, M.; Gerber, L.; Younossi, Z.M. Effectiveness of exercise in hepatic fat mobilization in non-alcoholic fatty liver disease: Systematic review. World J. Gastroenterol. 2016, 22, 6318–6327.
  226. Koutoukidis, D.A.; Astbury, N.M.; Tudor, K.E.; Morris, E.; Henry, J.A.; Noreik, M.; Jebb, S.A.; Aveyard, P. Association of Weight Loss Interventions With Changes in Biomarkers of Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-analysis. JAMA Intern. Med. 2019, 179, 1262–1271.
  227. Katsagoni, C.N.; Papatheodoridis, G.V.; Ioannidou, P.; Deutsch, M.; Alexopoulou, A.; Papadopoulos, N.; Papageorgiou, M.-V.; Fragopoulou, E.; Kontogianni, M. Improvements in clinical characteristics of patients with non-alcoholic fatty liver disease, after an intervention based on the Mediterranean lifestyle: A randomised controlled clinical trial. Br. J. Nutr. 2018, 120, 164–175.
  228. Oreopoulos, A.; Padwal, R.; Kalantar-Zadeh, K.; Fonarow, G.C.; Norris, C.M.; McAlister, F.A. Body mass index and mortality in heart failure: A meta-analysis. Am. Heart J. 2008, 156, 13–22.
  229. Bozkurt, B.; Deswal, A. Obesity as a prognostic factor in chronic symptomatic heart failure. Am. Heart J. 2005, 150, 1233–1239.
  230. Horwich, T.B.; Fonarow, G.C.; Hamilton, M.A.; MacLellan, W.R.; Woo, M.A.; Tillisch, J.H. The relationship between obesity and mortality in patients with heart failure. J. Am. Coll. Cardiol. 2001, 38, 789–795.
  231. Gupta, P.P.; Fonarow, G.C.; Horwich, T.B. Obesity and the obesity paradox in heart failure. Can J. Cardiol. 2015, 31, 195–202.
  232. Francque, S.; Szabo, G.; Abdelmalek, M.F.; Byrne, C.D.; Cusi, K.; Dufour, J.-F.; Roden, M.; Sacks, F.; Tacke, F. Nonalcoholic steatohepatitis: The role of peroxisome proliferator-activated receptors. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 24–39.
  233. Zanchi, A.; Maillard, M.; Jornayvaz, F.R.; Vinciguerra, M.; Deleaval, P.; Nussberger, J.; Burnier, M.; Pechere-Bertschi, A. Effects of the peroxisome proliferator-activated receptor (PPAR)-gamma agonist pioglitazone on renal and hormonal responses to salt in diabetic and hypertensive individuals. Diabetologia 2010, 53, 1568–1575.
  234. Newsome, P.N.; Buchholtz, K.; Cusi, K.; Linder, M.; Okanoue, T.; Ratziu, V.; Sanyal, A.J.; Sejling, A.-S.; Harrison, S.A. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2021, 384, 1113–1124.
  235. Mantovani, A.; Petracca, G.; Beatrice, G.; Csermely, A.; Lonardo, A.; Targher, G. Glucagon-Like Peptide-1 Receptor Agonists for Treatment of Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis: An Updated Meta-Analysis of Randomized Controlled Trials. Metabolites 2021, 11, 73.
  236. Mantovani, A.; Petracca, G.; Csermely, A.; Beatrice, G.; Targher, G. Sodium-Glucose Cotransporter-2 Inhibitors for Treatment of Nonalcoholic Fatty Liver Disease: A Meta-Analysis of Randomized Controlled Trials. Metabolites 2020, 11, 22.
  237. Coelho, F.D.S.; Borges-Canha, M.; Hafe, M.; Neves, J.S.; Vale, C.; Leite, A.R.; Carvalho, D.; Leite-Moreira, A. Effects of sodium-glucose co-transporter 2 inhibitors on liver parameters and steatosis: A meta-analysis of randomized clinical trials. Diabetes Metab. Res. Rev. 2021, 37, e3413.
  238. Xing, B.; Zhao, Y.; Dong, B.; Zhou, Y.; Lv, W.; Zhao, W. Effects of sodium-glucose cotransporter 2 inhibitors on non-alcoholic fatty liver disease in patients with type 2 diabetes: A meta-analysis of randomized controlled trials. J. Diabetes Investig. 2020, 11, 1238–1247.
  239. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424.
  240. McMurray, J.J.V.; Solomon, S.D.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; Anand, I.S.; Bělohlávek, J.; et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2019, 381, 1995–2008.
  241. Brown, E.; Heerspink, H.J.L.; Cuthbertson, D.J.; Wilding, J.P.H. SGLT2 inhibitors and GLP-1 receptor agonists: Established and emerging indications. Lancet 2021, 398, 262–276.
  242. Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Bocchi, E.; Böhm, M.; Brunner–La Rocca, H.-P.; Choi, D.-J.; Chopra, V.; Chuquiure-Valenzuela, E.; et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N. Engl. J. Med. 2021, 385, 1451–1461.
  243. Salah, H.M.; Al’Aref, S.J.; Khan, M.S.; Al-Hawwas, M.; Vallurupalli, S.; Mehta, J.L.; Mounsey, J.P.; Greene, S.J.; McGuire, D.K.; Lopes, R.D.; et al. Effect of sodium-glucose cotransporter 2 inhibitors on cardiovascular and kidney outcomes-Systematic review and meta-analysis of randomized placebo-controlled trials. Am. Heart J. 2021, 232, 10–22.
  244. Ratziu, V.; Harrison, S.A.; Francque, S.; Bedossa, P.; Lehert, P.; Serfaty, L.; Romero-Gómez, M.; Boursier, J.; Abdelmalek, M.; Caldwell, S.; et al. Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-alpha and -delta, Induces Resolution of Nonalcoholic Steatohepatitis Without Fibrosis Worsening. Gastroenterology 2016, 150, 1147–1159.e5.
  245. Zivkovic, L.; Asare, Y.; Bernhagen, J.; Dichgans, M.; Georgakis, M.K. Pharmacological Targeting of the CCL2/CCR2 Axis for Atheroprotection: A Meta-Analysis of Preclinical Studies. Arterioscler. Thromb. Vasc. Biol. 2022, 42, e131–e144.
  246. Loomba, R.; Lawitz, E.; Mantry, P.S.; Jayakumar, S.; Caldwell, S.H.; Arnold, H.; Diehl, A.M.; Djedjos, C.S.; Han, L.; Myers, R.P.; et al. The ASK1 inhibitor selonsertib in patients with nonalcoholic steatohepatitis: A randomized, phase 2 trial. Hepatology 2018, 67, 549–559.
  247. Lanier, M.; Pickens, J.; Bigi, S.V.; Bradshaw-Pierce, E.L.; Chambers, A.; Cheruvallath, Z.S.; Cole, D.; Dougan, D.R.; Ermolieff, J.; Gibson, T.; et al. Structure-Based Design of ASK1 Inhibitors as Potential Agents for Heart Failure. ACS Med. Chem. Lett. 2017, 8, 316–320.
  248. Izumiya, Y.; Kim, S.; Izumi, Y.; Yoshida, K.; Yoshiyama, M.; Matsuzawa, A.; Ichijo, H.; Iwao, H. Apoptosis signal-regulating kinase 1 plays a pivotal role in angiotensin II-induced cardiac hypertrophy and remodeling. Circ. Res. 2003, 93, 874–883.
  249. Shiizaki, S.; Naguro, I.; Ichijo, H. Activation mechanisms of ASK1 in response to various stresses and its significance in intracellular signaling. Adv. Biol. Regul. 2013, 53, 135–144.
  250. Tibbles, L.A.; Woodgett, J.R. The stress-activated protein kinase pathways. Cell. Mol. Life Sci. 1999, 55, 1230–1254.
  251. Hattori, K.; Naguro, I.; Runchel, C.; Ichijo, H. The roles of ASK family proteins in stress responses and diseases. Cell Commun. Signal. 2009, 7, 9.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , , , , ,
View Times: 345
Revisions: 2 times (View History)
Update Date: 23 Dec 2022
1000/1000
Video Production Service