IMR Press / RCM / Volume 24 / Issue 6 / DOI: 10.31083/j.rcm2406157
Open Access Review
The Transition of Cardiovascular Disease Risks from NAFLD to MAFLD
Zifeng Yang1,2,†Juan Yang3,4,†Jingjing Cai5Xiao-Jing Zhang2,6Peng Zhang2,6Zhi-Gang She1,2,*Hongliang Li1,2,4,6,7,*
Show Less
1 Department of Cardiology, Renmin Hospital of Wuhan University, 430000 Wuhan, Hubei, China
2 Institute of Model Animal, Wuhan University, 430000 Wuhan, Hubei, China
3 Department of Cardiology, Huanggang Central hospital of Yangtze University, 438000 Huanggang, Hubei, China
4 Huanggang Institute of Translational Medicine, 438000 Huanggang, Hubei, China
5 Department of Cardiology, The Third Xiangya Hospital, Central South University, 410000 Changsha, Hunan, China
6 School of Basic Medical Sciences, Wuhan University, 430000 Wuhan, Hubei, China
7 Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
*Correspondence: zgshe@whu.edu.cn (Zhi-Gang She); lihl@whu.edu.cn (Hongliang Li)
These authors contributed equally.
Rev. Cardiovasc. Med. 2023, 24(6), 157; https://doi.org/10.31083/j.rcm2406157
Submitted: 17 November 2022 | Revised: 19 January 2023 | Accepted: 31 January 2023 | Published: 31 May 2023
(This article belongs to the Special Issue Risk Factors for Cardiovascular Diseases)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

The increased burden of nonalcoholic fatty liver disease (NAFLD) parallels the increased incidence of overweight and metabolic syndrome worldwide. Because of the close relationship between metabolic disorders and fatty liver disease, a new term, metabolic-related fatty liver disease (MAFLD), was proposed by a group of experts to more precisely describe fatty liver disease resulting from metabolic disorders. According to the definitions, MAFLD and NAFLD populations have considerable discrepancies, but overlap does exist. This new definition has a nonnegligible impact on clinical practices, including diagnoses, interventions, and the risk of comorbidities. Emerging evidence has suggested that patients with MAFLD have more metabolic comorbidities and an increased risk of all-cause mortality, particularly cardiovascular mortality than patients with NAFLD. In this review, we systemically summarized and compared the risk and underlying mechanisms of cardiovascular disease (CVD) in patients with NAFLD or MAFLD.

Keywords
nonalcoholic fatty liver disease
metabolic-associated fatty liver disease
cardiovascular disease risk
1. Introduction

Nonalcoholic fatty liver disease (NAFLD) is a disease that is characterized by the accumulation of fat in the liver without excessive alcohol intake and other liver diseases [1, 2, 3]. NAFLD comprises a wide spectrum of liver diseases, ranging from simple steatosis to nonalcoholic steatohepatitis (NASH), advanced fibrosis, and cirrhosis [4]. During the past several decades, NAFLD has become one of the most prevalent chronic liver diseases and affects approximately 25% of the global population [5, 6].

Although NAFLD primarily manifests in the liver, it is a multisystemic disease affecting some extrahepatic organs [7]. As a result, NAFLD increases the risk of other diseases such as chronic kidney disease (CKD), type 2 diabetes mellitus (T2DM), and cardiovascular disease (CVD) [8, 9]. There is a strong relationship between NAFLD and CVD [10, 11, 12]. The potential mechanisms linking NAFLD and CVD, include insulin resistance, oxidative stress, chronic inflammation, hyperlipidemia, and endothelial dysfunction [13, 14, 15]. Moreover, an increasing number of studies have identified NAFLD as a risk factor for CVD [16, 17, 18].

Recently, based on the strong relationship between metabolic disorders and fatty liver disease, a new term, metabolic-associated fatty liver disease (MAFLD), has been introduced by a group of experts to more precisely describe fatty liver disease resulting from metabolic disorders [19, 20, 21]. MAFLD is defined as hepatic steatosis with any of the following conditions: overweight or obesity, presence of T2DM or metabolic disorders [22]. According to the criteria of NAFLD and MAFLD, nearly eighty percent of patients with liver steatosis can fulfill the criteria of NAFLD and MAFLD simultaneously [23, 24]. However, a number of patients who only meet one of the criteria still require consideration. For example, lean NAFLD individuals without systemic metabolic disorders cannot be diagnosed with MAFLD, and MAFLD individuals with alcoholic liver disease or other chronic liver diseases cannot be diagnosed with NAFLD [25, 26]. Therefore, individuals with hepatic steatosis can be divided into three groups, individuals with both MAFLD and NAFLD (NAFLD-MAFLD), individuals with only NAFLD but not MAFLD (NAFLD-only), and individuals with only MAFLD but not NAFLD (MAFLD-only). The transition from NAFLD to MAFLD inevitably has a significant impact on clinical practices, including the diagnosis, intervention approach, and risk of comorbidities.

Here, we review the history of NAFLD, MAFLD, and the transition from NAFLD to MAFLD. We further compare the cardiovascular risk between the NAFLD population and the MAFLD population and detail the differences in CVD risk among the NAFLD-only, MAFLD-only, and NAFLD-MAFLD overlapped groups.

2. From NAFLD to MAFLD

With the accumulation of in-depth mechanistic studies regarding the development of NAFLD, various metabolic disorders have been considered as main drivers of the occurrence and progression of NAFLD [2, 27, 28]. However, the diagnosis of NAFLD is based on the presence of excessive fat accumulation in the liver and without excessive alcohol intake and other etiologies of chronic liver disease, but it does not consider underlying metabolic disorders [29]. Therefore, a novel nomenclature that focuses mainly on systematic metabolic disorders, MAFLD, has been proposed, and MAFLD is an inclusive diagnosis [20].

2.1 History from NAFLD to MAFLD

In 1845, Addison first describe the term fatty liver. In 1964, the pathological mechanism of intrahepatic fat accumulation was first proposed [30]. In 1980, Ludwig [31] found steatohepatitis in liver biopsies from 20 individuals without alcohol abuse and other liver-damaging factors and thus named it NASH. In 1986, Schaffner and Thaler [32] proposed the concept of NAFLD and suggested that NASH should be regarded as a serious subtype of NAFLD. It was not until 1995 that NAFLD was proposed as a risk factor for CVD, which promoted an upsurge in NAFLD studies in recent decades [33]. The first NAFLD guideline was published by American scholars in 2002 [34]. Other countries and regions have also greatly increased their research interest in NAFLD and then issued corresponding guidelines [35, 36, 37]. In recent decades, with the prevalence of overweight, T2DM and metabolic dysregulation, NAFLD has been a leading cause of advanced liver diseases worldwide [38, 39, 40].

Metabolic disorders play a vital role in NAFLD, and the exclusion diagnosis strategies of NAFLD face many challenges such as the heterogeneous clinical outcomes of NAFLD and the lack of a uniform standard for the accurate calculation of alcohol intake [4, 41, 42]. In 2019, Eslam, Sanyal & George et al. [43] proposed the need for a new definition for fatty liver diseases, which foreshadowed the emergence of a novel nomenclature MAFLD the following year. The international expert group unanimously recommended redefining fatty liver disease related to metabolic disorders [20]. The proposal included using a new disease nomenclature, MAFLD, to renovate its former name NAFLD. MAFLD is diagnosed based on hepatic steatosis, similar to the diagnosis of NAFLD, but the diagnosis of MAFLD is a positive diagnosis and MAFLD can be combined with alcoholic fatty liver disease (AFLD) or other chronic liver diseases, which are common in life. Furthermore, MAFLD emphasizes the relationship between metabolic disorders and fatty liver. Currently, this name change has been endorsed by the Latin American Association for the Study of the Liver, the Asia Pacific Association for the Study of the Liver, the Chinese Society of Hepatology, and the Arabic Association for the Study of Diabetes and Metabolism [44, 45, 46, 47]. Over 1000 individuals who represent various professional institutions and doctors also support the change of terminology [48]. However, thus far, the American Association for the Study of Liver Diseases has not approved this name change [49]. In addition, a group of hepatologists, considering the current awareness of diseases among nonhepatologists, drug development, and the discovery of biomarkers, openly opposed the change of definition to MAFLD [50]. In summary, the diagnosis of MAFLD is a positive diagnosis, which emphasizes the impact of metabolic dysfunction on patients. This name change is supported by many regions and stakeholders. However, some hepatologists have expressed concern that this is a premature change in terminology. Changing the name from NAFLD to MAFLD may cause nonhepatologists to be more confused about this disease. The change may also have a negative impact on research development such as drug development and biomarker discovery. Thus, it is not clear whether the change of definition to MAFLD promotes the development of this field or leads to some unnecessary confusion and regression. Therefore, it is necessary to carefully evaluate the impact of this name change on different aspects such as disease awareness, drug development, and biomarker discovery, to judge the appropriateness of the renaming.

2.2 Comparing the Criteria of NAFLD with MAFLD

The criteria of NAFLD and MAFLD are both based on liver steatosis, but the renaming from NAFLD to MAFLD has also brought some internal changes.

NAFLD is defined by (1) fat accumulation in the liver as determined by imaging or histology, and (2) without other causes of fatty liver disease, including excessive alcohol abuse, viral infection, and hereditary disorders [51, 52]. MAFLD is diagnosed based on imaging, histological, or blood biomarker evidence of fatty liver, and the presence of at least one of the following three conditions: overweight/obesity, the presence of diabetes mellitus, or lean/normal weight with evidence of metabolic disorders [53, 54]. Metabolic disorders were defined by the presence of at least two of the following metabolic risk abnormalities: (1) waist circumference 102 cm in Caucasian men and waist circumference 88 cm in Caucasian women (or 90/80 cm in Asian men and women); (2) systolic blood pressure 130 mmHg and diastolic blood pressure 85 mmHg or hypertension drug treatment; (3) plasma triglycerides 150 mg/dL or its drug treatment; (4) plasma high-density lipoprotein cholesterol (HDL-C) <40 mg/dL for men and <50 mg/dL for women or the usage of specific drug treatment; (5) diagnosis of prediabetes or homeostasis model assessment of insulin resistance (HOMA-IR) score 2.5; and (6) plasma high-sensitivity C-reactive protein (hsCRP) level >2 mg/L [53].

The diagnostic criteria of NAFLD and MAFLD both include pathological liver steatosis and imaging features of fatty liver. In addition, the diagnosis of MAFLD can also be based on blood biomarker evidence of fatty liver. Most individuals with hepatic steatosis fulfill the diagnostic criteria of NAFLD and MAFLD [55]. There are also differences in the diagnostic criteria of NAFLD and MAFLD. NAFLD is a negative, exclusion criterion that needs to exclude liver diseases caused by alcohol and other reasons. In contrast, MAFLD is a positive, inclusion criterion that emphasizes the role of obesity, diabetes, and metabolic disorders in fatty liver, which can combine with other chronic liver diseases. The classification of individuals with hepatic steatosis can be redefined through these two different diagnostic criteria. About 80% of patients meet the diagnostic criteria of NAFLD and MAFLD, which can be classified as both NAFLD and MAFLD (NAFLD-MAFLD) [25]. About 15% of patients fulfill the criteria for MAFLD but not NAFLD, which can be classified as the MAFLD-only group [56]. This group includes individuals with hepatic steatosis who have metabolic dysregulation and other etiologies, including alcohol and viral infection [57]. In addition, about 5% of patients fulfill the criteria for NAFLD but not MAFLD, which can be classified as the NAFLD-only group [56]. This group includes lean NAFLD individuals without metabolic disorders [58]. These groups are described in Fig. 1.

Fig. 1.

Cardiovascular risk in the populations with NAFLD and MAFLD. The brown area on the left represents the population that only meets NAFLD diagnostic criteria, named the NAFLD-only group; the yellow area in the middle represents the population that meets the diagnostic criteria of NAFLD and MAFLD, termed the NAFLD-MAFLD group; the blue area on the right represents the population only meets the MAFLD diagnostic criteria, called the MAFLD-only group. The cardiovascular risk is the highest in the MAFLD-only group, followed by the NAFLD-MAFLD and the NAFLD-only groups. NAFLD, nonalcoholic fatty liver disease; MAFLD, metabolic-associated fatty liver disease.

2.3 Advantages and Disadvantages of the Conversion from NAFLD to MAFLD

Renaming from NAFLD to the new term MAFLD brings some advantages and disadvantages. These advantages and disadvantages can be described in terms of diagnosis, treatment, and prevention.

First, from the perspective of diagnosis, MAFLD criteria can better help identify patients with long-term hepatic and extrahepatic adverse consequences than the diagnostic criteria of NAFLD [59, 60, 61]. This means that patients with high risks of developing serious liver outcomes and complications can be widely screened. In addition, the new term MAFLD attaches importance to the role of overweight, metabolic disorders, and T2DM in fatty liver disease, which can enhance the awareness of fatty liver disease and the ability to diagnose fatty liver disease in the clinic [62, 63]. Second, the diagnosis of NAFLD needs to exclude other liver diseases while MAFLD can combine with other secondary liver diseases. Thus, the definition of MAFLD allows us to consider other liver diseases that may accompany NAFLD and patients can be treated more widely. Third, the term MAFLD includes “metabolic”, which may increase public awareness of the tight relationship between fatty liver and metabolism. Thus, more public attention would be given to metabolic health to prevent fatty liver.

However, the change in terminology also brings some potential disadvantages. First, the definitions of NAFLD and MAFLD are slightly different, so the individuals did not completely overlap. For example, individuals with lean NAFLD may be overlooked by MAFLD [58]. In addition, a majority of noninvasive biomarkers and scores are derived using patients with NAFLD/NASH, rather than in patients with MAFLD [64, 65]. For example, a NIS4 biomarker panel was developed using NASH patients, which leaves uncertainties in the accuracy of identifying hepatitis in patients with MAFLD [66]. Second, although there are no drugs approved by the FDA for NASH at present, some drugs, such as elafibranor, and obeticholic acid, showed encouraging results in the treatment of NASH in phase 2 or 3 clinical trials [67, 68]. MAFLD can coexist with other liver diseases. Thus, the heterogeneity of patients is higher and the efficacy of the testing reagents in ongoing clinical trials is impacted.

3. NAFLD/MAFLD is a Risk Factor for CVD

Although a causal relationship between NAFLD and CVD has not been determined, potential mechanisms linking NAFLD to CVD have been explored for over a decade [69, 70, 71]. MAFLD has been recognized as a fatty liver disease resulting from metabolic disorders. Patients who are diagnosed with MAFLD have at least two metabolic disorders or other liver diseases. Thus, individuals with MAFLD may have a higher cardiovascular risk than individuals with NAFLD (Fig. 2).

Fig. 2.

Potential mechanisms linking NAFLD/MAFLD to CVD. NAFLD/MAFLD promotes CVD through potential pathophysiological mechanisms, including insulin resistance, oxidative stress, systemic inflammation, and endothelial dysfunction. In addition to these mechanisms, patients with MAFLD may company with impairments from ethanol, viral infection, and immune dysregulation, which further increase the risk of CVD. The yellow boxes represent common mechanisms linking NAFLD/MAFLD to CVD; the red boxes represent additional mechanisms linking MAFLD to CVD. NAFLD, nonalcoholic fatty liver disease; MAFLD, metabolic-associated fatty liver disease; CVD, Cardiovascular disease; FFA, free fatty acid; VLDL, very low-density lipoprotein; ROS, reactive oxygen species; NO, nitric oxide; AFLD, alcoholic fatty liver disease; SNS, sympathetic nervous system; RAAS, renin-angiotensin-aldosterone system.

3.1 Potential Mechanisms Linking NAFLD to CVD

It has been indicated that NAFLD can promote the development of CVD independent of traditional CVD risks. Some potential pathophysiological mechanisms linking NAFLD to CVD comprise insulin resistance, oxidative stress, systemic inflammation, and endothelial dysfunction [72].

Insulin resistance is an important feature of NAFLD and plays a crucial role in CVD pathogenesis [73, 74]. Insulin resistance would cause hyperglycemia by reducing glucose uptake and can lead to the export of peripheral free fatty acids (FFAs) to the liver [75, 76]. More importantly, insulin resistance would contribute to an elevated level of insulin. Increased insulin further induces lipid accumulation in the liver through accelerating glycogenesis and de novo lipogenesis [77, 78]. At the same time, increased lipid accumulation in the liver can further deteriorate insulin resistance in individuals with NAFLD [79]. Eventually, these constitute a vicious circle, leading to the increasing accumulation of fat in the liver. Persistent hyperglycemia and insulin resistance activate inflammation and lead to abnormal lipoprotein metabolism, which induces the occurrence of atherosclerotic cardiovascular disease (ASCVD) [80]. Insulin clearance further worsens this situation in patients with NAFLD. In addition, hyperinsulinemia alters the activities of lipogenic enzymes and leads to the mobilization of subcutaneous fat to deposit in viscera [81]. Mobilized fat also increases very low-density lipoprotein (VLDL) levels and circulating FFAs, which contribute to atherosclerosis [82, 83]. Moreover, atherosclerotic dyslipidemia is also attributed to increased very low-density lipoprotein synthesis and decreased FFA oxidation and triglyceride (TG) output in individuals with NAFLD [84, 85].

Oxidative stress is also a critical mechanism linking NAFLD to CVD [86]. Excessive fat accumulates in hepatocytes inducing reactive oxygen species (ROS) overproduction in the mitochondria and endoplasmic reticulum [75]. Excessive ROS overflow into the circulation and increase circulating levels of oxidative stress markers, such as serum soluble NOX2-derived peptide (sNOX2-dp) and urinary 8-iso-prostaglandin F2α (8-iso-PGF2α), in NAFLD [87, 88]. Some studies indicate that the levels of urinary 8-iso-PGF2α and serum sNOX2-dp increase with the severity of hepatic steatosis in NAFLD patients [88]. It has been reported that urinary 8-iso-PGF2α is also an independent predictor of NAFLD [88]. Therefore, strong relationships between oxidative stress markers and NAFLD indicate that oxidative stress plays a vital role in the pathophysiology of NAFLD. Excess ROS in the circulation also damage cellular components of vascular cells, such as mitochondrial DNA and cell membrane, leading to endothelial dysfunction and atherosclerosis [16, 89, 90, 91]. Moreover, it has been reported that increased circulating levels of NOX2 and 8-iso-PGF2α are also closely associated with some CVDs such as coronary heart disease, atherosclerosis, and hypertension [92, 93].

At the same time, systemic inflammation also plays a critical role in linking NAFLD and CVD [86]. NAFLD leads to elevated levels of inflammatory mediators, such as intercellular adhesion molecule-1, P-selectin, interleukin-6, and hsCRP [94]. Increased levels of inflammatory factors would contribute to systemic inflammation, which poses a threat to the cardiovascular system [95]. Furthermore, the epicardial fat volume in patients with NAFLD is increased, which may increase the secretion of proinflammatory factors such as tumor necrosis factor-α, leptin, and interleukin 1-β [96]. These would affect the myocardium in a state of systemic inflammation [97].

Endothelial dysfunction is initiated from the early stage of atherosclerosis and is characterized by the decreased availability of nitric oxide (NO) [98]. Elevated levels of asymmetric dimethyl arginine (ADMA), an endogenous antagonist representing nitric oxide synthase, are prevalent in patients with NAFLD [99, 100]. The increase in ADMA levels results in a decrease in NO availability and endothelial dysfunction. Furthermore, the level of homocysteine is also elevated in patients with NAFLD [101]. Hyperhomocysteinemia causes oxidative stress by reducing the storage of glutathione, which is also related to a low level of NO, increased platelet activity, and vascular resistance [73].

3.2 Potential Mechanisms Linking MAFLD to CVD

Individuals with MAFLD have a higher burden from metabolic disturbances than individuals with NAFLD due to the diagnostic criteria. Thus, metabolic stress-triggered insulin resistance, oxidative stress, systemic inflammation, dyslipidemia, and endothelial dysfunction could be more conspicuous in MAFLD individuals. Additionally, the diagnosis of MAFLD does not exclude other liver diseases including AFLD, viral hepatitis, and autoimmune hepatitis. Therefore, potential mechanisms linking MAFLD to CVD are also affected by other factors, such as ethanol, viral infection, and immune dysregulation.

The cardiovascular system can be indirectly affected by chronic ethanol consumption. Chronic ethanol abuse increases the activity of the sympathetic nervous system (SNS) and the activity of the renin-angiotensin-aldosterone system (RAAS) [102]. The SNS and RAAS activation causes hypertension which increases the load on the heart and exacerbates alcoholic cardiomyopathy [102, 103]. In addition, its metabolite acetaldehyde can act as a direct toxin to cardiomyocytes. These effects can lead to cell apoptosis and mitochondrial dysfunction in myocytes, which will aggravate contractile dysfunction [104].

Viral hepatitis, such as that due to hepatitis C virus (HCV), directly or indirectly interferes with glucose and lipid metabolism, resulting in insulin resistance, steatosis, and T2DM [105, 106, 107, 108, 109, 110]. Furthermore, HCV in blood vessels directly causes a local inflammatory response, leading to the occurrence of CVD [111, 112].

Immune dysregulation in autoimmune hepatitis may also increase the risk of CVD. The enhancement of immune and inflammatory cascade reactions is related to endothelial dysfunction and ROS production [113, 114, 115]. In addition, immunity and inflammation themselves can mediate the occurrence and development of CVD [116, 117].

4. Individuals with MAFLD may be at a Higher Risk for CVD than Individuals with NAFLD

The change of definition to MAFLD is not only a change in nomenclature, but it also brings other effects, such as the different cardiovascular risks between patients with NAFLD and patients with MAFLD. Two aspects can reflect that patients with NAFLD and patients with MAFLD have different cardiovascular risks. On one hand, the cardiovascular risk is different between the NAFLD population and the MAFLD population. On the other hand, the cardiovascular risk is different among the MAFLD-only, NAFLD-MAFLD overlapped, and NAFLD-only groups. These are summarized in Table 1 (Ref. [60, 61, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128]).

Table 1.Summary of clinical studies and meta-analysis on comparing CVD risk between NAFLD and MAFLD.
Region Study design Fatty liver diagnosis Study population NAFLD and MAFLD Main results References
Comparison of cardiovascular risk differences between the NAFLD population and the MAFLD population
Japan Cross-sectional Ultrasonography 765 541 NAFLD The MAFLDs have higher BMI, LDL-c, TG, lower HDL-c, and higher risks for diabetes and hypertension than the NAFLDs. [61]
609 MAFLD
Japan Cross-sectional Ultrasonography 2306 subjects with fatty liver 1462 NAFLD MAFLD better helps identify patients with ASCVD risk than NAFLD. [118]
1859 MAFLD
Japan Cross-sectional Ultrasonography 890 subjects who underwent health checkups 268 NAFLD The MAFLDs have a higher risk of subclinical atherosclerosis than the NAFLDs. [119]
384 MAFLD
Korea Cross-sectional Fatty liver index 9,584,399 2,680,217 NAFLD The MAFLDs have a higher risk for CVD mortality than the non-MAFLDs (HR 1.46, 95% CI: 1.41–1.52); The NAFLDs have a higher risk for CVD mortality than the non-NAFLDs (HR 1.12, 95% CI: 0.96–1.30). [60]
3,573,644 MAFLD
The United States Cross-sectional Ultrasound-fatty liver index 19,617 adults 6658 NAFLD The MAFLDs and the NAFLDs have similar risks for CVD and CKD. [120]
7131 MAFLD
The United States Retrospective cohort Ultrasonography 13,083 4347 NAFLD The MAFLDs have higher BMI, HOMA-IR, lipids, and higher risks for diabetes and hypertension than the NAFLDs. [121]
3885 MAFLD
The United States Retrospective cohort Ultrasonography 12,480 3909 NAFLD The MAFLDs (HR 2.01, 95% CI: 1.66–2.64) have a higher risk for CVD-related mortality than the NAFLDs (HR 1.53, 95% CI: 1.26–1.86). [122]
3779 MAFLD
Korea Retrospective cohort Ultrasonography 2144 subjects without a history of ASCVD 995 NAFLD MAFLD criteria are better than NAFLD criteria in predicting ASCVD risk in asymptomatic subjects. [123]
891 MAFLD
Sri Lankan Prospective cohort Ultrasonography 2985 940 NAFLD The MAFLDs and the NAFLDs have similar new-onset metabolic traits and risks for CVD events. [124]
990 MAFLD
China Prospective cohort Ultrasonography 6873 2771 NAFLD The MAFLDs and the NAFLDs have similar risks for diabetes, CKD, and CVD. [125]
3212 MAFLD
Europe, Asia, and North America Meta-analysis Imaging or biopsy 22 studies, 379,801 participants Of 67,742 patients, 23,865 NAFLD. Whereas of 379,801 patients, 116,806 MAFLD The MAFLDs have higher BMI, triglycerides, lower HDL-c, and higher risks for hypertension and diabetes than the NAFLDs. [126]
Comparison of cardiovascular risk among the NAFLD-only, MAFLD-only, and NAFLD-MAFLD groups
Korea Cross-sectional Fatty liver index 9,584,399 52,747 NAFLD-only Compared to individuals without fatty liver disease, the risk for CVD events increased 2.33 (2.30–2.36) fold in the MAFLD-only group, 2.15 (2.13–2.17) fold in the NAFLD-MAFLD group, and 1.68 (1.59–1.78) fold in the NAFLD-only group. The MAFLD-only has the highest association with CVD-related death. [60]
870,818 MAFLD-only
2,625,321 NAFLD-MAFLD
Japan Cross-sectional Ultrasonography 2306 subjects with fatty liver 301 NAFLD-only The NAFLD-only have a lower incidence of CVD event than the NAFLD-MAFLD, with HR 0.70 (95%CI 0.50-0.98). The MAFLD-only has a similar risk of CVD events with the NAFLD-MAFLD, with HR 1.19 (0.89–1.58). [118]
698 MAFLD-only
1161 NAFLD-MAFLD
The United States Retrospective cohort Ultrasonography 12,480 528 NAFLD-only The risks for CVD-related mortality are different in the NAFLD-only, NAFLD-MAFLD, and MAFLD-only groups, with HRs 0.46 (0.20–1.02), 1.86 (1.51–2.28), and 2.35 (1.60–3.45), respectively. [122]
658 MAFLD-only
3251 NAFLD-MAFLD
The United States Retrospective cohort Ultrasonography 13,640 adults aged 20 years 254 NAFLD-only The MAFLD-only and NAFLD-MAFLD have more CVD risk factors than NAFLD-only. They also have a higher risk for CVD mortality than the NAFLD-only group, with HRs 9.4 (2.6–34.6) and 7.0 (2.1–23.1), respectively. [127]
503 MAFLD-only
2240 NAFLD-MAFLD
The United States Retrospective cohort Ultrasonography 7761 participants 394 NAFLD-only The MAFLD-only and the NAFLD-MAFLD groups have increased CVD risk factors compared to the NAFLD group. Compared to individuals without hepatic steatosis, the risks for CVD mortality were 2.59 (1.10–6.09), 1.95 (1.55–2.45), and 0.29 (0.10–0.86) in the MAFLD-only, NAFLD-MAFLD, and NAFLD-only groups, respectively. [128]
212 MAFLD-only
2044 NAFLD-MAFLD

CVD, Cardiovascular disease; NAFLD, nonalcoholic fatty liver disease; MAFLD, metabolic-associated fatty liver disease; BMI, body mass index; HDL-c, high-density lipoprotein cholesterol; LDL-c, low-density lipoprotein cholesterol; TG, triglyceride; HOMA-IR, homeostasis model assessment-insulin resistance; ASCVD, atherosclerotic cardiovascular disease; HR, hazard ratio; CI, confidence interval; CKD, chronic kidney disease.

4.1 Comparison of Cardiovascular Risk Differences between the NAFLD Population and the MAFLD Population

Emerging evidence from population studies has indicated that individuals with MAFLD have a higher risk for development of other traditional CVD risk factors, CVD events, and CVD death than individuals with NAFLD [61, 121, 126]. In an observational data meta-analysis involving 379,801 participants, the association between MAFLD and NAFLD in cardiovascular disease risk factors was reported [126]. MAFLD was more relevant to hypertension, diabetes, high body mass index (BMI), and high lipid levels than NAFLD. Furthermore, other studies have also reported that patients with MAFLD have higher BMI, HOMA-IR, lipid levels, and a higher possibility of having diabetes and hypertension than patients with NAFLD [61, 121].

In addition, patients with MAFLD or NAFLD also have different risks for cardiovascular events. In a single-center and cross-sectional study, 2306 subjects with fatty liver were enrolled, and ASCVD risk was estimated by noninvasive tests such as the Suita score [118]. This report indicated that MAFLD is related to worsening of the Suita score and that MAFLD criteria better help identify patients with ASCVD risk than NAFLD criteria. A cross-sectional study also showed that individuals with MAFLD have a higher probability of coronary artery calcification than individuals with NAFLD, which is one of the markers of atherosclerosis [119]. Moreover, in a cross-sectional study, 2144 individuals who had no history of ASCVD were offered a health examination at a health center [123]. ASCVD risks can be identified by MAFLD and NAFLD criteria, but MAFLD criteria can better predict the risk of ASCVD than NAFLD criteria in asymptomatic subjects. This means that individuals diagnosed with MAFLD need to further enhance their awareness of ASCVD prevention, regardless of whether they have a history of ASCVD. Whether in NAFLD or MAFLD, the risk of CVD in patients with liver fibrosis is higher than that in patients with simple hepatic steatosis [129, 130]. Moreover, MAFLD criteria can better identify patients with advanced liver fibrosis than NAFLD criteria [61]. Similar to NAFLD, liver fibrosis can also increase the risk of cardiovascular events in MAFLD [131, 132]. In a retrospective study, the risk of cardiovascular events was compared between NAFLD and MAFLD populations and among MAFLD individuals with various degrees of liver fibrosis [132]. Compared with NAFLD individuals, MAFLD individuals have significantly higher 10-year CVD risks. More importantly, the risk of cardiovascular events is increased with the severity of liver fibrosis in MAFLD patients [132]. However, this study also has some shortcomings such as a small sample size. Therefore, larger clinical trials are needed to further study the impact of liver fibrosis on CVD risks in MAFLD individuals.

The differences in cardiovascular-related mortality are also striking between the NAFLD population and the MAFLD population. It was concluded by using data from the third National Health and Nutrition Examination Survey (NHANES III) that CVD-related mortality was slightly higher in the MAFLD population (hazard ratio (HR) 2.01, 95% CI: 1.66–2.64) than in the NAFLD population (HR 1.53, 95% CI: 1.26–1.86) [122]. In addition, the conclusion was also drawn from a nationwide health information database of the National Health Insurance Service in South Korea that the MAFLD population is significantly associated with CVD-related death (HR 1.46, 95% CI: 1.41–1.52) [60]. However, there is no relationship between NAFLD and CVD-related death (HR 1.12, 95% CI: 0.96–1.30). Therefore, the risks of cardiovascular events and cardiovascular mortality are higher in individuals with MAFLD than in individuals with NAFLD. This may be because metabolic disorders are closely related to CVD and have a synergistic effect with fatty liver on CVD. In addition, other liver diseases including AFLD and viral hepatitis can also increase the risk of CVD. Other studies have yielded different results that the NAFLD population and the MAFLD population had a comparable prevalence of nonfatal and fatal CVD events or similar clinical characteristics [120, 124, 125]. However, the prevalence of NAFLD is lower than that of MAFLD in their studies, which means that more individuals with MAFLD are at risk for CVD [120, 124, 125]. In summary, the MAFLD population is at a greater risk for CVD than the NAFLD population.

4.2 Comparison of Cardiovascular Risk among the NAFLD-Only, MAFLD-Only, and NAFLD-MAFLD Groups

According to the definition of NAFLD and MAFLD, individuals with hepatic steatosis can be roughly divided into three groups: the NAFLD-only, MAFLD-only, and NAFLD-MAFLD overlapped groups. In recent years, the risk of CVD among three groups has been reported. Emerging studies have indicated that the risk for developing other traditional CVD risk factors, CVD events, and CVD death is different among the NAFLD-only, MAFLD-only, and NAFLD-MAFLD overlapped groups.

In several studies, some traditional CVD risk factors, such as high levels of lipids and increased HOMA-IR, are more common in MAFLD-only and NAFLD-MAFLD overlapped groups than in the NAFLD-only group [122, 127, 128]. Other traditional CVD risk factors, such as overweight and diabetes, only appear in MAFLD-only or NAFLD-MAFLD overlapped groups [122, 127, 128]. However, there was no obvious difference in these traditional CVD risk factors between MAFLD-only and NAFLD-MAFLD overlapped groups. To sum up, the NAFLD-only group has the lowest risk for developing other traditional CVD risk factors while the MAFLD-only and NAFLD-MAFLD overlapped groups had a similar risk for developing other traditional CVD risk factors.

Furthermore, the risk of CVD events is also different among the three groups. In a nationwide cohort study, Lee et al. [60] collected data from 9,584,399 adults aged 40–64 years who were offered health examinations from 2009 to 2010. Of these participants, 354,886 individuals were classified into three groups. The MAFLD-only group and NAFLD-MAFLD overlapped groups have the highest cumulative incidence of CVD events, followed by the NAFLD-only group. After adjusting for complex factors, compared with individuals without fatty liver disease, the HR for CVD events was 2.33 (2.30–2.36) in the MAFLD-only group, 2.15 (2.13–2.17) in the NAFLD-MAFLD group, and 1.68 (1.59–1.78) in the NAFLD-only group. Tsutsumi et al. [118] recruited 2306 subjects with fatty liver, and the worsening of the ASCVD risk score was higher in the NAFLD-MAFLD overlapped and MAFLD-only groups than in the NAFLD-only group. However, there was no statistical significance in the risk of CVD events between MAFLD-only group and NAFLD-MAFLD overlapped group. In summary, the risk of CVD events was highest in the MAFLD-only and NAFLD-MAFLD overlapped groups while the NAFLD-only group has the lowest risk of CVD events.

In addition to the different risks for developing other traditional cardiovascular risk factors and cardiovascular events, cardiovascular mortality was also different among these three groups. In a population-based study, Kim et al. [128] collected data from 7761 participants from NHANES II, of which participants in the NAFLD-MAFLD overlapped group accounted for 23.5% of total participants, those in the MAFLD-only group accounted for 2.4%, and those in the NAFLD-only group accounted for 6.1%. In univariable Model 1, compared with individuals without hepatic steatosis, the HRs (95% CI) for cardiovascular mortality in the MAFLD-only, NAFLD-MAFLD overlapped, and NAFLD-only groups were 2.59 (1.10–6.09), 1.95 (1.55–2.45), and 0.29 (0.10–0.86), respectively. This indicates that the strongest relationship with CVD-related mortality was found for the MAFLD-only group, followed by the NAFLD-MAFLD overlapped group, and then by the NAFLD-only group. This is also reflected in some other studies. Huang et al. [122] also collected 12,480 participants aged 20–74 years in NHANES III; the HR (95% CI) for CVD-related mortality was 0.46 (0.20–1.02), 1.86 (1.51–2.28), and 2.35 (1.60–3.45) in the NAFLD-only, NAFLD-MAFLD and MAFLD-only groups, respectively. Similarly, Nguyen et al. [127] and Lee et al. [60] also reported that the highest cumulative incidence of CVD-related mortality was in the MAFLD-only group while the lowest cardiovascular mortality was in the NAFLD-only group.

As discussed above, the cardiovascular risk of individuals diagnosed by the criteria of MAFLD but excluded by the criteria of NAFLD may be the highest. The cardiovascular risk of individuals with NAFLD and MAFLD is intermediate, and the cardiovascular risk of individuals diagnosed by the criterion of NAFLD and excluded by the criterion of MAFLD is the lowest. This may be because the MAFLD-only population includes people who were previously excluded by NAFLD criteria such as individuals with viral hepatitis or alcoholic liver disease. Viral infections and alcohol intake are related to the development of traditional CVD risk factors and an increased risk of CVD [133, 134, 135]. These results also indicate that MAFLD criteria can better predict the high-risk population for CVD than NAFLD criteria. Individuals captured by MAFLD criteria need to increase their awareness of CVD prevention. Additionally, physicians should be vigilant and treat such patients as early as possible to reduce the risk of CVD.

5. Conclusions and Prospective

Since NAFLD was proposed as a CVD risk factor in 1995, NAFLD has received increasing attention. At present, several large population studies have suggested that NAFLD is an independent risk factor for CVD [16, 17, 18]. NAFLD and CVD share many traditional risk factors, in addition, NAFLD promotes the development of various CVDs independent of traditional risk factors. Considering the close relationship between metabolic disorders and NAFLD, a new terminology MAFLD has been proposed. Although the majority of patients diagnosed by NAFLD could be identified using MAFLD criteria, a small number of individuals are either diagnosed by NAFLD or MAFLD alone. On average, the MAFLD population may have a higher metabolic burden and risk for CVD than the population with NAFLD. The change in terminology has a strong influence on clinical practices regarding diagnosis, intervention, prevention, and the risk of comorbidities. However, whether this change results in an improvement in patient care remains to be studied in future trials.

Author Contributions

ZFY, JY, JJC, X-JZ, PZ, Z-GS, and HLL designed the research study. ZFY, JY, and JJC provided help and advice on the table and figures. ZFY and JY analyzed the literature data. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We would like to thank all those who helped us during the writing of this manuscript.

Funding

This work was supported by grants from the National Science Foundation of China (81770053 Z-GS, 81970364 Z-GS, 82170595 X-JZ, 81970070 X-JZ, 81970011 PZ), the Innovation Platform Construction Project of Hubei Province (20204201117303072238 HL).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Suzuki A, Diehl AM. Nonalcoholic Steatohepatitis. Annual Review of Medicine. 2017; 68: 85–98.
[2]
Samuel VT, Shulman GI. Nonalcoholic Fatty Liver Disease as a Nexus of Metabolic and Hepatic Diseases. Cell Metabolism. 2018; 27: 22–41.
[3]
Deng K, Huang X, Lei F, Zhang X, Zhang P, She Z, et al. Role of hepatic lipid species in the progression of nonalcoholic fatty liver disease. American Journal of Physiology. Cell Physiology. 2022; 323: C630–C639.
[4]
Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nature Medicine. 2018; 24: 908–922.
[5]
Younossi ZM, Golabi P, de Avila L, Paik JM, Srishord M, Fukui N, et al. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis. Journal of Hepatology. 2019; 71: 793–801.
[6]
Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L, Eslam M, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nature Reviews. Gastroenterology & Hepatology. 2018; 15: 11–20.
[7]
Byrne CD, Targher G. NAFLD: a multisystem disease. Journal of Hepatology. 2015; 62: S47–S64.
[8]
Fotbolcu H, Zorlu E. Nonalcoholic fatty liver disease as a multi-systemic disease. World Journal of Gastroenterology. 2016; 22: 4079–4090.
[9]
Kasper P, Martin A, Lang S, Kütting F, Goeser T, Demir M, et al. NAFLD and cardiovascular diseases: a clinical review. Clinical Research in Cardiology: Official Journal of the German Cardiac Society. 2021; 110: 921–937.
[10]
Deprince A, Haas JT, Staels B. Dysregulated lipid metabolism links NAFLD to cardiovascular disease. Molecular Metabolism. 2020; 42: 101092.
[11]
Muzurović E, Mikhailidis DP, Mantzoros C. Non-alcoholic fatty liver disease, insulin resistance, metabolic syndrome and their association with vascular risk. Metabolism: Clinical and Experimental. 2021; 119: 154770.
[12]
Zhang L, She Z, Li H, Zhang X. Non-alcoholic fatty liver disease: a metabolic burden promoting atherosclerosis. Clinical Science. 2020; 134: 1775–1799.
[13]
Targher G, Byrne CD, Tilg H. NAFLD and increased risk of cardiovascular disease: clinical associations, pathophysiological mechanisms and pharmacological implications. Gut. 2020; 69: 1691–1705.
[14]
Galiero R, Caturano A, Vetrano E, Cesaro A, Rinaldi L, Salvatore T, et al. Pathophysiological mechanisms and clinical evidence of relationship between Nonalcoholic fatty liver disease (NAFLD) and cardiovascular disease. Reviews in Cardiovascular Medicine. 2021; 22: 755–768.
[15]
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.
[16]
Cai J, Zhang X, Ji Y, Zhang P, She Z, Li H. Nonalcoholic Fatty Liver Disease Pandemic Fuels the Upsurge in Cardiovascular Diseases. Circulation Research. 2020; 126: 679–704.
[17]
Chen Z, Liu J, Zhou F, Li H, Zhang X, She Z, et al. Nonalcoholic Fatty Liver Disease: An Emerging Driver of Cardiac Arrhythmia. Circulation Research. 2021; 128: 1747–1765.
[18]
Zhao Y, Zhao G, Chen Z, She Z, Cai J, Li H. Nonalcoholic Fatty Liver Disease: An Emerging Driver of Hypertension. Hypertension. 2020; 75: 275–284.
[19]
Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. Journal of Hepatology. 2020; 73: 202–209.
[20]
Eslam M, Sanyal AJ, George J, International Consensus Panel. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology. 2020; 158: 1999–2014.e1.
[21]
Yang C, He Q, Chen Z, Qin J, Lei F, Liu Y, et al. A Bidirectional Relationship Between Hyperuricemia and Metabolic Dysfunction-Associated Fatty Liver Disease. Frontiers in Endocrinology. 2022; 13: 821689.
[22]
Shiha G, Korenjak M, Eskridge W, Casanovas T, Velez-Moller P, Högström S, et al. Redefining fatty liver disease: an international patient perspective. The Lancet. Gastroenterology & Hepatology. 2021; 6: 73–79.
[23]
Targher G. Concordance between MAFLD and NAFLD diagnostic criteria in ‘real-world’ data. Liver International. 2020; 40: 2879–2880.
[24]
Kang SH, Cho Y, Jeong SW, Kim SU, Lee J, Korean NAFLD Study Group. From nonalcoholic fatty liver disease to metabolic-associated fatty liver disease: Big wave or ripple? Clinical and Molecular Hepatology. 2021; 27: 257–269.
[25]
Ayada I, van Kleef LA, Alferink LJM, Li P, de Knegt RJ, Pan Q. Systematically comparing epidemiological and clinical features of MAFLD and NAFLD by meta-analysis: Focusing on the non-overlap groups. Liver International. 2022; 42: 277–287.
[26]
Younossi ZM, Paik JM, Al Shabeeb R, Golabi P, Younossi I, Henry L. Are there outcome differences between NAFLD and metabolic-associated fatty liver disease? Hepatology. 2022; 76: 1423–1437.
[27]
Jarvis H, Craig D, Barker R, Spiers G, Stow D, Anstee QM, et al. Metabolic risk factors and incident advanced liver disease in non-alcoholic fatty liver disease (NAFLD): A systematic review and meta-analysis of population-based observational studies. PLoS Medicine. 2020; 17: e1003100.
[28]
Cariou B, Byrne CD, Loomba R, Sanyal AJ. Nonalcoholic fatty liver disease as a metabolic disease in humans: A literature review. Diabetes, Obesity & Metabolism. 2021; 23: 1069–1083.
[29]
Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology. 2018; 67: 328–357.
[30]
Dianzani Mu. On the Pathogenesis of the Accumulation of Fat In Hepatic Steatosis. Rassegna Medica Sarda. 1964; 66: 67–90.
[31]
Ludwig J, Viggiano TR, McGill DB, Oh BJ. Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clinic Proceedings. 1980; 55: 434–438.
[32]
Schaffner F, Thaler H. Nonalcoholic fatty liver disease. Progress in Liver Diseases. 1986; 8: 283–298.
[33]
Lonardo A, Bellini M, Tondelli E, Frazzoni M, Grisendi A, Pulvirenti M, et al. Nonalcoholic steatohepatitis and the “bright liver syndrome”: should a recently expanded clinical entity be further expanded? The American Journal of Gastroenterology. 1995; 90: 2072–2074.
[34]
American Gastroenterological Association. American Gastroenterological Association medical position statement: nonalcoholic fatty liver disease. Gastroenterology. 2002; 123: 1702–1704.
[35]
Farrell GC, Chitturi S, Lau GKK, Sollano JD, Asia-Pacific Working Party on NAFLD. Guidelines for the assessment and management of non-alcoholic fatty liver disease in the Asia-Pacific region: executive summary. Journal of Gastroenterology and Hepatology. 2007; 22: 775–777.
[36]
Loria P, Adinolfi LE, Bellentani S, Bugianesi E, Grieco A, Fargion S, et al. Practice guidelines for the diagnosis and management of nonalcoholic fatty liver disease. A decalogue from the Italian Association for the Study of the Liver (AISF) Expert Committee. Digestive and Liver Disease. 2010; 42: 272–282.
[37]
Ratziu V, Bellentani S, Cortez-Pinto H, Day C, Marchesini G. A position statement on NAFLD/NASH based on the EASL 2009 special conference. Journal of Hepatology. 2010; 53: 372–384.
[38]
Stefan N, Cusi K. A global view of the interplay between non-alcoholic fatty liver disease and diabetes. The Lancet. Diabetes & Endocrinology. 2022; 10: 284–296.
[39]
Polyzos SA, Kountouras J, Mantzoros CS. Obesity and nonalcoholic fatty liver disease: From pathophysiology to therapeutics. Metabolism: Clinical and Experimental. 2019; 92: 82–97.
[40]
Zhou J, Zhou F, Wang W, Zhang X, Ji Y, Zhang P, et al. Epidemiological Features of NAFLD From 1999 to 2018 in China. Hepatology. 2020; 71: 1851–1864.
[41]
Stefan N, Häring H, Cusi K. Non-alcoholic fatty liver disease: causes, diagnosis, cardiometabolic consequences, and treatment strategies. The Lancet. Diabetes & Endocrinology. 2019; 7: 313–324.
[42]
Eslam M, Valenti L, Romeo S. Genetics and epigenetics of NAFLD and NASH: Clinical impact. Journal of Hepatology. 2018; 68: 268–279.
[43]
Eslam M, Sanyal AJ, George J. Toward More Accurate Nomenclature for Fatty Liver Diseases. Gastroenterology. 2019; 157: 590–593.
[44]
Mendez-Sanchez N, Arrese M, Gadano A, Oliveira CP, Fassio E, Arab JP, et al. The Latin American Association for the Study of the Liver (ALEH) position statement on the redefinition of fatty liver disease. The Lancet. Gastroenterology & Hepatology. 2021; 6: 65–72.
[45]
Nan Y, An J, Bao J, Chen H, Chen Y, Ding H, et al. The Chinese Society of Hepatology position statement on the redefinition of fatty liver disease. Journal of Hepatology. 2021; 75: 454–461.
[46]
Eslam M, Sarin SK, Wong VW, Fan J, Kawaguchi T, Ahn SH, et al. The Asian Pacific Association for the Study of the Liver clinical practice guidelines for the diagnosis and management of metabolic associated fatty liver disease. Hepatology International. 2020; 14: 889–919.
[47]
Shaltout I, Alkandari H, Fouad Y, Hamed AE. Arabic Association for the Study of Diabetes and Metabolism (AASD) endorsing the MAFLD definition of fatty liver disease. Journal of Hepatology. 2022; 76: 739–740.
[48]
Méndez-Sánchez N, Bugianesi E, Gish RG, Lammert F, Tilg H, Nguyen MH, et al. Global multi-stakeholder endorsement of the MAFLD definition. The Lancet. Gastroenterology & Hepatology. 2022; 7: 388–390.
[49]
Singh SP, Anirvan P, Reddy KR, Conjeevaram HS, Marchesini G, Rinella ME, et al. Non-alcoholic fatty liver disease: Not time for an obituary just yet! Journal of Hepatology. 2021; 74: 972–974.
[50]
Younossi ZM, Rinella ME, Sanyal AJ, Harrison SA, Brunt EM, Goodman Z, et al. From NAFLD to MAFLD: Implications of a Premature Change in Terminology. Hepatology. 2021; 73: 1194–1198.
[51]
European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD), European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. Journal of Hepatology. 2016; 64: 1388–1402.
[52]
Wong VW, Chan W, Chitturi S, Chawla Y, Dan YY, Duseja A, et al. Asia-Pacific Working Party on Non-alcoholic Fatty Liver Disease guidelines 2017-Part 1: Definition, risk factors and assessment. Journal of Gastroenterology and Hepatology. 2018; 33: 70–85.
[53]
Tilg H, Effenberger M. From NAFLD to MAFLD: when pathophysiology succeeds. Nature Reviews. Gastroenterology & Hepatology. 2020; 17: 387–388.
[54]
Qu W, Ma T, Cai J, Zhang X, Zhang P, She Z, et al. Liver Fibrosis and MAFLD: From Molecular Aspects to Novel Pharmacological Strategies. Frontiers in Medicine. 2021; 8: 761538.
[55]
Xian Y, Weng J, Xu F. MAFLD vs. NAFLD: shared features and potential changes in epidemiology, pathophysiology, diagnosis, and pharmacotherapy. Chinese Medical Journal. 2020; 134: 8–19.
[56]
Yu C, Wang M, Zheng S, Xia M, Yang H, Zhang D, et al. Comparing the Diagnostic Criteria of MAFLD and NAFLD in the Chinese Population: A Population-based Prospective Cohort Study. Journal of Clinical and Translational Hepatology. 2022; 10: 6–16.
[57]
Yick F, Hajifathalian K. NAFLD or MAFLD: the data behind the debate. Hepatobiliary Surgery and Nutrition. 2022; 11: 439–442.
[58]
Huang J, Xue W, Wang M, Wu Y, Singh M, Zhu Y, et al. MAFLD Criteria May Overlook a Subtype of Patient with Steatohepatitis and Significant Fibrosis. Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy. 2021; 14: 3417–3425.
[59]
Huang S, Su H, Kao J, Tseng T, Yang H, Su T, et al. Clinical and Histologic Features of Patients with Biopsy-Proven Metabolic Dysfunction-Associated Fatty Liver Disease. Gut and Liver. 2021; 15: 451–458.
[60]
Lee H, Lee Y, Kim SU, Kim HC. Metabolic Dysfunction-Associated Fatty Liver Disease and Incident Cardiovascular Disease Risk: A Nationwide Cohort Study. Clinical Gastroenterology and Hepatology. 2021; 19: 2138–2147.e10.
[61]
Yamamura S, Eslam M, Kawaguchi T, Tsutsumi T, Nakano D, Yoshinaga S, et al. MAFLD identifies patients with significant hepatic fibrosis better than NAFLD. Liver International. 2020; 40: 3018–3030.
[62]
Fouad Y, Gomaa A, Semida N, Ghany WA, Attia D. Change from NAFLD to MAFLD increases the awareness of fatty liver disease in primary care physicians and specialists. Journal of Hepatology. 2021; 74: 1254–1256.
[63]
Kawaguchi T, Tsutsumi T, Nakano D, Torimura T. MAFLD: Renovation of clinical practice and disease awareness of fatty liver. Hepatology Research. 2022; 52: 422–432.
[64]
Loomba R, Adams LA. Advances in non-invasive assessment of hepatic fibrosis. Gut. 2020; 69: 1343–1352.
[65]
Masoodi M, Gastaldelli A, Hyötyläinen T, Arretxe E, Alonso C, Gaggini M, et al. Metabolomics and lipidomics in NAFLD: biomarkers and non-invasive diagnostic tests. Nature Reviews. Gastroenterology & Hepatology. 2021; 18: 835–856.
[66]
Harrison SA, Ratziu V, Boursier J, Francque S, Bedossa P, Majd Z, et al. A blood-based biomarker panel (NIS4) for non-invasive diagnosis of non-alcoholic steatohepatitis and liver fibrosis: a prospective derivation and global validation study. The Lancet. Gastroenterology & Hepatology. 2020; 5: 970–985.
[67]
Ratziu V, Harrison SA, Francque S, Bedossa P, Lehert P, Serfaty L, et al. Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-α and -δ, Induces Resolution of Nonalcoholic Steatohepatitis Without Fibrosis Worsening. Gastroenterology. 2016; 150: 1147–1159.e5.
[68]
Younossi ZM, Ratziu V, Loomba R, Rinella M, Anstee QM, Goodman Z, et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2019; 394: 2184–2196.
[69]
Ghouri N, Preiss D, Sattar N. Liver enzymes, nonalcoholic fatty liver disease, and incident cardiovascular disease: a narrative review and clinical perspective of prospective data. Hepatology. 2010; 52: 1156–1161.
[70]
Cai J, Xu M, Zhang X, Li H. Innate Immune Signaling in Nonalcoholic Fatty Liver Disease and Cardiovascular Diseases. Annual Review of Pathology. 2019; 14: 153–184.
[71]
Wang Z, Ye M, Zhang X, Zhang P, Cai J, Li H, et al. Impact of NAFLD and its pharmacotherapy on lipid profile and CVD. Atherosclerosis. 2022; 355: 30–44.
[72]
Stahl EP, Dhindsa DS, Lee SK, Sandesara PB, Chalasani NP, Sperling LS. Nonalcoholic Fatty Liver Disease and the Heart: JACC State-of-the-Art Review. Journal of the American College of Cardiology. 2019; 73: 948–963.
[73]
Francque SM, van der Graaff D, Kwanten WJ. Non-alcoholic fatty liver disease and cardiovascular risk: Pathophysiological mechanisms and implications. Journal of Hepatology. 2016; 65: 425–443.
[74]
Adeva-Andany MM, Fernández-Fernández C, Carneiro-Freire N, Castro-Quintela E, Pedre-Piñeiro A, Seco-Filgueira M. Insulin resistance underlies the elevated cardiovascular risk associated with kidney disease and glomerular hyperfiltration. Reviews in Cardiovascular Medicine. 2020; 21: 41–56.
[75]
Yu Y, Cai J, She Z, Li H. Insights into the Epidemiology, Pathogenesis, and Therapeutics of Nonalcoholic Fatty Liver Diseases. Advanced Science. 2018; 6: 1801585.
[76]
Hodson L, Gunn PJ. The regulation of hepatic fatty acid synthesis and partitioning: the effect of nutritional state. Nature Reviews. Endocrinology. 2019; 15: 689–700.
[77]
Geisler CE, Renquist BJ. Hepatic lipid accumulation: cause and consequence of dysregulated glucoregulatory hormones. The Journal of Endocrinology. 2017; 234: R1–R21.
[78]
Mayer AE, Löffler MC, Loza Valdés AE, Schmitz W, El-Merahbi R, Viera JT, et al. The kinase PKD3 provides negative feedback on cholesterol and triglyceride synthesis by suppressing insulin signaling. Science Signaling. 2019; 12: eaav9150.
[79]
Lim S, Oh TJ, Koh KK. Mechanistic link between nonalcoholic fatty liver disease and cardiometabolic disorders. International Journal of Cardiology. 2015; 201: 408–414.
[80]
Low Wang CC, Hess CN, Hiatt WR, Goldfine AB. Clinical Update: Cardiovascular Disease in Diabetes Mellitus: Atherosclerotic Cardiovascular Disease and Heart Failure in Type 2 Diabetes Mellitus - Mechanisms, Management, and Clinical Considerations. Circulation. 2016; 133: 2459–2502.
[81]
Worm N. Beyond Body Weight-Loss: Dietary Strategies Targeting Intrahepatic Fat in NAFLD. Nutrients. 2020; 12: 1316.
[82]
Mehta A, Shapiro MD. Apolipoproteins in vascular biology and atherosclerotic disease. Nature Reviews. Cardiology. 2022; 19: 168–179.
[83]
Zhang H, Dellsperger KC, Zhang C. The link between metabolic abnormalities and endothelial dysfunction in type 2 diabetes: an update. Basic Research in Cardiology. 2012; 107: 237.
[84]
Katsiki N, Mikhailidis DP, Mantzoros CS. Non-alcoholic fatty liver disease and dyslipidemia: An update. Metabolism: Clinical and Experimental. 2016; 65: 1109–1123.
[85]
Bai L, Li H. Innate immune regulatory networks in hepatic lipid metabolism. Journal of Molecular Medicine. 2019; 97: 593–604.
[86]
Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radical Biology & Medicine. 2020; 152: 116–141.
[87]
van der Pol A, van Gilst WH, Voors AA, van der Meer P. Treating oxidative stress in heart failure: past, present and future. European Journal of Heart Failure. 2019; 21: 425–435.
[88]
Del Ben M, Polimeni L, Carnevale R, Bartimoccia S, Nocella C, Baratta F, et al. NOX2-generated oxidative stress is associated with severity of ultrasound liver steatosis in patients with non-alcoholic fatty liver disease. BMC Gastroenterology. 2014; 14: 81.
[89]
Yu E, Calvert PA, Mercer JR, Harrison J, Baker L, Figg NL, et al. Mitochondrial DNA damage can promote atherosclerosis independently of reactive oxygen species through effects on smooth muscle cells and monocytes and correlates with higher-risk plaques in humans. Circulation. 2013; 128: 702–712.
[90]
Khatana C, Saini NK, Chakrabarti S, Saini V, Sharma A, Saini RV, et al. Mechanistic Insights into the Oxidized Low-Density Lipoprotein-Induced Atherosclerosis. Oxidative Medicine and Cellular Longevity. 2020; 2020: 5245308.
[91]
Chen Z, Yu Y, Cai J, Li H. Emerging Molecular Targets for Treatment of Nonalcoholic Fatty Liver Disease. Trends in Endocrinology and Metabolism. 2019; 30: 903–914.
[92]
Streeter J, Thiel W, Brieger K, Miller FJ. Opportunity nox: the future of NADPH oxidases as therapeutic targets in cardiovascular disease. Cardiovascular Therapeutics. 2013; 31: 125–137.
[93]
Schwedhelm E, Bartling A, Lenzen H, Tsikas D, Maas R, Brümmer J, et al. Urinary 8-iso-prostaglandin F2alpha as a risk marker in patients with coronary heart disease: a matched case-control study. Circulation. 2004; 109: 843–848.
[94]
Fricker ZP, Pedley A, Massaro JM, Vasan RS, Hoffmann U, Benjamin EJ, et al. Liver Fat Is Associated With Markers of Inflammation and Oxidative Stress in Analysis of Data From the Framingham Heart Study. Clinical Gastroenterology and Hepatology. 2019; 17: 1157–1164.e4.
[95]
Gehrke N, Schattenberg JM. Metabolic Inflammation-A Role for Hepatic Inflammatory Pathways as Drivers of Comorbidities in Nonalcoholic Fatty Liver Disease? Gastroenterology. 2020; 158: 1929–1947.e6.
[96]
Liu B, Li Y, Li Y, Liu Y, Yan Y, Luo A, et al. Association of epicardial adipose tissue with non-alcoholic fatty liver disease: a meta-analysis. Hepatology International. 2019; 13: 757–765.
[97]
Packer M. Epicardial Adipose Tissue May Mediate Deleterious Effects of Obesity and Inflammation on the Myocardium. Journal of the American College of Cardiology. 2018; 71: 2360–2372.
[98]
Tona F, Montisci R, Iop L, Civieri G. Role of coronary microvascular dysfunction in heart failure with preserved ejection fraction. Reviews in Cardiovascular Medicine. 2021; 22: 97–104.
[99]
Deanfield JE, Halcox JP, Rabelink TJ. Endothelial function and dysfunction: testing and clinical relevance. Circulation. 2007; 115: 1285–1295.
[100]
Kasumov T, Edmison JM, Dasarathy S, Bennett C, Lopez R, Kalhan SC. Plasma levels of asymmetric dimethylarginine in patients with biopsy-proven nonalcoholic fatty liver disease. Metabolism: Clinical and Experimental. 2011; 60: 776–781.
[101]
Tang Y, Chen X, Chen Q, Xiao J, Mi J, Liu Q, et al. Association of serum methionine metabolites with non-alcoholic fatty liver disease: a cross-sectional study. Nutrition & Metabolism. 2022; 19: 21.
[102]
Gardner JD, Mouton AJ. Alcohol effects on cardiac function. Comprehensive Physiology. 2015; 5: 791–802.
[103]
Ransome Y, Slopen N, Karlsson O, Williams DR. The association between alcohol abuse and neuroendocrine system dysregulation: Race differences in a National sample. Brain, Behavior, and Immunity. 2017; 66: 313–321.
[104]
Cahill PA, Redmond EM. Alcohol and cardiovascular disease–modulation of vascular cell function. Nutrients. 2012; 4: 297–318.
[105]
Wang C, Wang S, Yao W, Chang T, Chou P. Hepatitis C virus infection and the development of type 2 diabetes in a community-based longitudinal study. American Journal of Epidemiology. 2007; 166: 196–203.
[106]
Moucari R, Asselah T, Cazals-Hatem D, Voitot H, Boyer N, Ripault M, et al. Insulin resistance in chronic hepatitis C: association with genotypes 1 and 4, serum HCV RNA level, and liver fibrosis. Gastroenterology. 2008; 134: 416–423.
[107]
Bugianesi E, Salamone F, Negro F. The interaction of metabolic factors with HCV infection: does it matter? Journal of Hepatology. 2012; 56: S56–S65.
[108]
Petta S, Amato M, Cabibi D, Cammà C, Di Marco V, Giordano C, et al. Visceral adiposity index is associated with histological findings and high viral load in patients with chronic hepatitis C due to genotype 1. Hepatology. 2010; 52: 1543–1552.
[109]
Abdul-Ghani MA, Jayyousi A, DeFronzo RA, Asaad N, Al-Suwaidi J. Insulin Resistance the Link between T2DM and CVD: Basic Mechanisms and Clinical Implications. Current Vascular Pharmacology. 2019; 17: 153–163.
[110]
Adams LA, Anstee QM, Tilg H, Targher G. Non-alcoholic fatty liver disease and its relationship with cardiovascular disease and other extrahepatic diseases. Gut. 2017; 66: 1138–1153.
[111]
Adinolfi LE, Restivo L, Zampino R, Guerrera B, Lonardo A, Ruggiero L, et al. Chronic HCV infection is a risk of atherosclerosis. Role of HCV and HCV-related steatosis. Atherosclerosis. 2012; 221: 496–502.
[112]
Boddi M, Abbate R, Chellini B, Giusti B, Giannini C, Pratesi G, et al. Hepatitis C virus RNA localization in human carotid plaques. Journal of Clinical Virology. 2010; 47: 72–75.
[113]
Bernardi N, Sciatti E, Pancaldi E, Alghisi F, Drera A, Falco R, et al. Coeliac and cardiovascular disease: a possible relationship between two apparently separate conditions. Monaldi Archives for Chest Disease. 2022. (online ahead of print)
[114]
Sitia S, Tomasoni L, Atzeni F, Ambrosio G, Cordiano C, Catapano A, et al. From endothelial dysfunction to atherosclerosis. Autoimmunity Reviews. 2010; 9: 830–834.
[115]
Myers AL, Harris CM, Choe K, Brennan CA. Inflammatory production of reactive oxygen species by Drosophila hemocytes activates cellular immune defenses. Biochemical and Biophysical Research Communications. 2018; 505: 726–732.
[116]
Libby P, Mallat Z, Weyand C. Immune and inflammatory mechanisms mediate cardiovascular diseases from head to toe. Cardiovascular Research. 2021; 117: 2503–2505.
[117]
Jiang W, Wang M. New insights into the immunomodulatory role of exosomes in cardiovascular disease. Reviews in Cardiovascular Medicine. 2019; 20: 153–160.
[118]
Tsutsumi T, Eslam M, Kawaguchi T, Yamamura S, Kawaguchi A, Nakano D, et al. MAFLD better predicts the progression of atherosclerotic cardiovascular risk than NAFLD: Generalized estimating equation approach. Hepatology Research. 2021; 51: 1115–1128.
[119]
Bessho R, Kashiwagi K, Ikura A, Yamataka K, Inaishi J, Takaishi H, et al. A significant risk of metabolic dysfunction-associated fatty liver disease plus diabetes on subclinical atherosclerosis. PLoS ONE. 2022; 17: e0269265.
[120]
Zhang H, Wang Y, Chen C, Lu Y, Wang N. Cardiovascular and renal burdens of metabolic associated fatty liver disease from serial US national surveys, 1999-2016. Chinese Medical Journal. 2021; 134: 1593–1601.
[121]
Lin S, Huang J, Wang M, Kumar R, Liu Y, Liu S, et al. Comparison of MAFLD and NAFLD diagnostic criteria in real world. Liver International. 2020; 40: 2082–2089.
[122]
Huang Q, Zou X, Wen X, Zhou X, Ji L. NAFLD or MAFLD: Which Has Closer Association With All-Cause and Cause-Specific Mortality?-Results From NHANES III. Frontiers in Medicine. 2021; 8: 693507.
[123]
Kim H, Lee CJ, Ahn SH, Lee KS, Lee BK, Baik SJ, et al. MAFLD Predicts the Risk of Cardiovascular Disease Better than NAFLD in Asymptomatic Subjects with Health Check-Ups. Digestive Diseases and Sciences. 2022; 67: 4919–4928.
[124]
Niriella MA, Ediriweera DS, Kasturiratne A, De Silva ST, Dassanayaka AS, De Silva AP, et al. Outcomes of NAFLD and MAFLD: Results from a community-based, prospective cohort study. PLoS ONE. 2021; 16: e0245762.
[125]
Liang Y, Chen H, Liu Y, Hou X, Wei L, Bao Y, et al. Association of MAFLD With Diabetes, Chronic Kidney Disease, and Cardiovascular Disease: A 4.6-Year Cohort Study in China. The Journal of Clinical Endocrinology and Metabolism. 2022; 107: 88–97.
[126]
Lim GEH, Tang A, Ng CH, Chin YH, Lim WH, Tan DJH, et al. An Observational Data Meta-analysis on the Differences in Prevalence and Risk Factors Between MAFLD vs NAFLD. Clinical Gastroenterology and Hepatology. 2021. (online ahead of print)
[127]
Nguyen VH, Le MH, Cheung RC, Nguyen MH. Differential Clinical Characteristics and Mortality Outcomes in Persons With NAFLD and/or MAFLD. Clinical Gastroenterology and Hepatology. 2021; 19: 2172–2181.e6.
[128]
Kim D, Konyn P, Sandhu KK, Dennis BB, Cheung AC, Ahmed A. Metabolic dysfunction-associated fatty liver disease is associated with increased all-cause mortality in the United States. Journal of Hepatology. 2021; 75: 1284–1291.
[129]
Tamaki N, Kurosaki M, Takahashi Y, Itakura Y, Inada K, Kirino S, et al. Liver fibrosis and fatty liver as independent risk factors for cardiovascular disease. Journal of Gastroenterology and Hepatology. 2021; 36: 2960–2966.
[130]
Park J, Kim G, Kim H, Lee J, Lee Y, Jin S, et al. The association of hepatic steatosis and fibrosis with heart failure and mortality. Cardiovascular Diabetology. 2021; 20: 197.
[131]
Schonmann Y, Yeshua H, Bentov I, Zelber-Sagi S. Liver fibrosis marker is an independent predictor of cardiovascular morbidity and mortality in the general population. Digestive and Liver Disease. 2021; 53: 79–85.
[132]
Zhang P, Dong X, Zhang W, Wang S, Chen C, Tang J, et al. Metabolic-associated fatty liver disease and the risk of cardiovascular disease. Clinics and Research in Hepatology and Gastroenterology. 2023; 47: 102063.
[133]
Petta S, Maida M, Macaluso FS, Barbara M, Licata A, Craxì A, et al. Hepatitis C Virus Infection Is Associated With Increased Cardiovascular Mortality: A Meta-Analysis of Observational Studies. Gastroenterology. 2016; 150: 145–155.e4; quiz e15–e16.
[134]
Chun HS, Lee JS, Lee HW, Kim BK, Park JY, Kim DY, et al. Prevalence and Risk Factors of Cardiovascular Disease in Patients with Chronic Hepatitis B. Digestive Diseases and Sciences. 2022; 67: 3412–3425.
[135]
Xi B, Veeranki SP, Zhao M, Ma C, Yan Y, Mi J. Relationship of Alcohol Consumption to All-Cause, Cardiovascular, and Cancer-Related Mortality in U.S. Adults. Journal of the American College of Cardiology. 2017; 70: 913–922.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top