Next Article in Journal
Should Grain-Based Staple Foods Be Included in Admonitions to “Avoid Processed and Ultra-Processed Food”?
Previous Article in Journal
Postbiotics Formulation and Therapeutic Effect in Inflammation: A Systematic Review
Previous Article in Special Issue
Refining Nutritional Assessment Methods for Older Adults: A Pilot Study on Sicilian Long-Living Individuals
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Steatotic Liver Disease in Older Adults: Clinical Implications and Unmet Needs

1
Department of Gastroenterology, Alfred Health, Melbourne 3004, Australia
2
School of Translational Medicine, Monash University, Melbourne 3004, Australia
3
Department of Gastroenterology, Eastern Health, Box Hill 3128, Australia
4
Department of Geriatrics and General Medicine, Werribee Mercy Hospital, Werribee 3030, Australia
5
Department of Aged Care and Medicine, The Royal Melbourne Hospital, Melbourne Medical School, The University of Melbourne, Parkville 3050, Australia
6
Department of Gastroenterology, Northern Health, Epping 3076, Australia
7
School of Health and Biomedical Science, RMIT University, Melbourne 3083, Australia
8
Eastern Clinical School, Monash University, Box Hill 3128, Australia
9
School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Australia
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(13), 2189; https://doi.org/10.3390/nu17132189
Submission received: 27 May 2025 / Revised: 24 June 2025 / Accepted: 28 June 2025 / Published: 30 June 2025
(This article belongs to the Special Issue Dietary Intake and Health Status in Older Adults—2nd Edition)

Abstract

Metabolic dysfunction-associated steatotic liver disease (MASLD) is the commonest cause of chronic liver disease worldwide. Its incidence has been increasing rapidly, alongside the growing epidemics of type 2 diabetes mellitus and overweight/obesity. Global population age has also been increasing in parallel, and predictions indicate there will be more than 2 billion persons aged over 65 by the year 2050. The interplay between MASLD and other health conditions of older persons has been a focus of recent research. In this narrative review, we aim to describe its prevalence; clinical and sociodemographic associations; and outcomes for older persons, all of which are of significant importance when considering public health messaging as well as screening and counselling individual older adults.

Graphical Abstract

1. Introduction

1.1. Background and Aetiology

Hepatic steatosis—defined as intrahepatic triacylglycerol rich lipid droplets of at least 5% of liver weight or 5% of hepatocytes containing lipid vacuoles—was first described by Thomas Addison in 1836 [1] in the context of excessive alcohol intake. It took another 150 years for non-alcoholic fatty liver disease (NAFLD) [2] and non-alcoholic steatohepatitis (NASH) [3] to be recognised as distinct disease entities unrelated to alcohol intake, and instead associated with insulin resistance, obesity and type 2 diabetes mellitus (T2DM) [4]. Since this time, there has been increased recognition of NAFLD, rising in parallel with the global epidemics of obesity [5] and T2DM [6], such that it is estimated that up to 38% of people globally are living with NAFLD [7].
To better recognise the link between non-alcoholic hepatic steatosis and aspects of the metabolic syndrome [8,9], a global Delphi process recently reclassified NAFLD to metabolic dysfunction-associated steatotic liver disease (MASLD) [10]. This new definition utilises similar criteria to the previous NAFLD definition—hepatic steatosis in the absence of excessive alcohol intake or steatogenic medication use—with the addition of at least one positive metabolic criterion (including dyslipidaemia, overweight/obesity, impaired fasting glucose/T2DM, or hypertension) [10]. The change in nomenclature has had minimal impact on disease epidemiology, with ~99% of NAFLD cases re-classified as MASLD [11,12]. The metabolic diseases used as criteria are not only important for linking disease aetiology with nomenclature—they also impact risk stratification and outcomes. It is well recognised that those living with both MASLD and T2DM have worse outcomes than those with either MASLD or T2DM alone [13,14]. MASLD is causally linked with a multitude of additional metabolic diseases, including hypertension [15] as well as dysregulated lipid metabolism [16].
Regardless of definition, MASLD represents a significant global burden of disease. Along with its more rapidly progressive form, metabolic dysfunction-associated steatohepatitis (MASH), MASLD is the most common chronic liver disease worldwide [17], and consequently an increasingly important cause of liver-related morbidity and mortality [18,19,20]. In Australia, it is estimated that from 2019 to 2030 there will be an 85% increase in MASLD liver deaths [21]; similar rates are predicted in the USA [22], with a tripling of MASLD-related deaths from 2020 to 2050. While these outcomes are linked to liver fibrosis resultant from the progression of liver diseases [23,24,25,26], the number of MASLD cases is increasing so rapidly that even low rates of progressive fibrosis will be associated with significantly increasing negative health outcomes worldwide. In spite of this, a recent study evaluating global preparedness for this rapidly increasing disease showed that none of the 102 countries evaluated had a national or sub-national strategy for MASLD, and almost one third of countries scored 0/100 on a preparedness index [27]. This raises significant concerns about the public health approach to this epidemic of chronic liver disease.

1.2. An Ageing Population

Rising in tandem with this epidemic of metabolic diseases is the ageing global population. It is estimated that, by 2050, one in six persons will be aged over 65 (compared to one in 11 in 2019), representing a doubling of older persons from 703 million to 1.5 billion globally [28]. This increase is driven by a combination of both reductions in premature mortality associated with non-communicable diseases (such as tobacco-related diseases and cardiovascular disease [CVD] [29]) as well as through improved sanitation [30,31] and increasing access to healthcare [32].
Despite this increasing number and proportion of older persons, societal ageism remains a significant barrier to older person care and community engagement [33]—and may contribute to the known relative lack of clinical trial research including older persons [34,35]. This is critically important when interpreting research in the context of population ageing, as without targeted studies, there is an inability to provide appropriate care and advice to this growing portion of the population. Importantly, without their participation in research, we also miss capturing what health outcomes are seen as important by the ageing population.

1.3. The Ageing Liver

The effects of ageing are not solely due to the accumulation of disease and the cumulative effect of risk factors; indeed, chronological age is a poor guide to ageing, and the ageing process is heterogenous between individuals and within the individual by organ system [36]. This accentuates the need for specific ageing-related research on common conditions such as MASLD, as generalizability from studies of younger adults is fraught.
Specific to the liver, there are anatomical and physiological alterations that result in functional changes and possible susceptibility to disease [37]. Hepatic blood flow and liver mass decreases, more so in females [38], and ageing is also associated with alterations in liver enzyme activity [39,40]. Senescent cells—cells that are non-functional but also not prone to apoptosis or necrosis—accumulate in a non-linear manner in the aged liver [41]. While these cells are non-functional, they are resistant to fibrosis [41]. In keeping with this understanding of hepatocyte senescence, prior work has shown that the ageing liver may also be characterised by fewer remnant functional hepatocytes, not just smaller liver volumes [42].
Adding further to this, it is known that ageing, while heterogeneously experienced, is not isolated to single organ systems; the bidirectional relationships between ageing organ systems leads to additional physiologic and functional changes [36].
Despite these known hepatic changes, there is still a relative paucity of liver-related research in older persons. While there have been some steps to address ageing-specific changes in other organ systems—for example, the development of a cardiovascular risk calculator specifically for people aged ≥65 years (SCORE2-OP [43])—further work is needed to improve our understanding of the whether there should be differences in the hepatological care we provide older persons. As such, this review aims to highlight the evidence—and gaps—that exist regarding MASLD in older persons, as well as identify key unmet needs and opportunities for future research.

2. Materials and Methods

In order to develop an overview of the impact of MASLD on older persons and related outcomes, databases were accessed using multiple keywords in combination, including: geriatric, ageing, aged, and elderly; MASLD and metabolic dysfunction-associated steatotic liver disease; NAFLD and non-alcoholic fatty liver disease; MAFLD and metabolic dysfunction-associated steatotic liver disease; MASH and metabolic dysfunction-associated steatohepatitis; NASH and non-alcoholic steatohepatitis; non-invasive testing; lifestyle; diet and diet score; sociodemography; non-communicable diseases; exercise; death and mortality; cardiovascular disease; malignancy; frailty; physical disability; cognition, cognitive decline, and dementia; senescence. Studies were restricted to humans unless a non-human animal or other pre-clinical model was felt necessary to expand on concepts. Additional material was identified through the reference section of key papers and personal knowledge of the literature. Databases accessed included PubMed and Google Scholar. No date restrictions were imposed, and papers up to 22 June 2025 were included.

3. MASLD and Fibrosis Identification

MASLD is, at its core, a histologic diagnosis [44]. However, due to the inherent risks of obtaining liver tissue via invasive biopsy as well as sampling error and inter-/intra-rater reporting variability [45,46], liver biopsy is not commonly performed [17]. In older persons this is especially important, as they may have more risks associated with biopsy (such as increasing rates of anticoagulation use [47]), and are less likely to tolerate complications due to reduced physiologic reserve [48,49]. As such, surrogate measures exist, which can be broadly stratified into radiological and score-based options.
Radiologically, ultrasonography (USS) [50,51], computed tomography (CT) [50,52], magnetic resonance imaging (MRI) [50,52,53], and the USS-derived controlled attenuation parameter (CAP) measurement [54] can all be used to identify hepatosteatosis. While their accuracy compared to biopsy is imperfect, they are commonly used in clinical practice [17], and there are no age-specific reasons for reduced accuracy compared to younger persons. Additionally, a variety of new and device-specific radiological algorithms to improve USS accuracy are being developed [55], although their relative accuracy and applicability to older persons specifically is unknown.
Score-based options use a composite of laboratory tests, anthropometric variables, and clinical parameters, with differences in their ability to identify hepatosteatosis in older persons. Several scores exist and have been predominantly validated in middle-aged populations, including the Fatty Liver Index (FLI) [56], Framingham Steatosis Index (FSI) [57], Dallas Steatosis Index (DSI) [58], Hepatic Steatosis Index (HSI) [59], Lipid Accumulation Product (LAP) [44], Visceral Adiposity Index (VAI) [60] and the ZJU Index (ZJU) [61]. Of all these scores, FLI is the only one validated against ultrasound in older persons with NAFLD [62]. None have been validated against biopsy in older persons. The components of these scores can be seen in Table 1.
Our previous observational study, using the FLI as the ‘gold standard’ epidemiological score for older persons, given its previous validation [62] and recommended use in local guidelines [64], established FSI as the next most accurate score for identifying MASLD [63]. Other non-FLI scores have significant limitations in older persons, largely due to (a) their derivation populations (recognising that a healthy BMI range in older persons is different to that in younger persons [65] and is ethnicity-specific [66,67]), and (b) the recognised change in alanine aminotransferase (ALT) values, along with a changing ALT/aspartate aminotransferase (AST) ratio with increasing age [40,68].
Although liver biopsy remains the gold standard for identifying liver fibrosis (a major determinant of liver- and cardiovascular-related outcomes in MASLD and other liver diseases) [23,24,25,26], the emergence of non-invasive tools to assess fibrosis has transformed clinical practice. Such tests include radiology-based tests (magnetic resonance elastography [MRE] [69,70] and vibration controlled transient elastography [VCTE] [70,71,72,73]), simple laboratory-based scores (including the Fibrosis-4 [FIB-4]; BMI, AST/ALT Ratio, Diabetes score [BARD]; AST to Platelet Ratio Index [APRI]; NAFLD Fibrosis; BMI and AST [BIMAST]; Metabolic Dysfunction-Associated Fibrosis 5 [MAF-5]; and Steatosis-Associated Fibrosis Estimator [SAFE] scores) [74,75,76,77,78], composite scores such as the MEFIB (combining MRE with FIB-4) [79], and proprietary direct-fibrosis laboratory tests (such as the Enhanced Liver Fibrosis [ELF] test [80]). The components of these scores can be seen in Table 2.
However, much like the steatosis indices, these scores have almost exclusively been validated in middle-aged populations, or with a predominantly middle-aged cohort [77], without considering potential differences that could occur with age. Indeed, the FIB-4 was initially age-limited (validated only between 35–65 years), and a new threshold was subsequently proposed for use in older adults [81]. Despite this, studies have still shown poorer discriminatory ability in those aged over 65 [82]. Similarly, the SAFE score has been shown to be markedly less useful as a screening test in those aged 60–80 years compared with middle-aged adults [83], with similar results seen for several other composite scores [84]. Few studies have focused on validating the ELF score in older persons; one reported no significant difference in biopsy-stratified ELF scores in those older than 60 compared with those younger than 60 [85], although there was a non-significant increase in ELF values with increasing age within each fibrosis-stage stratum. In keeping with this, in the general population, there is an age-dependent increase in ELF scores [86,87]. TIMP-1 may change with differing extra-hepatic clinical and nutritional state [88], and there is a small trend to changing PIIINP levels with increasing age (though insufficient to alter reference intervals) [89]. Additionally, most non-proprietary scores use the ALT (which falls with age) [40,68], and three of the four also use platelets (which may change with age in a sex-dependent manner) [90]. Given this, it could be hypothesized these indices perform less well in older persons than in their middle-aged derivation cohorts, although, apart from an altered FIB-4 upper limit of normal, there is no reference to this in current guidelines [17,91].
Recommendations for Future Research
  • Identify a preferred steatosis index that is older-person specific, rather than a re-purposed index based on middle-aged persons
  • Identify a preferred non-proprietary fibrosis index that is older-person specific, and consider re-evaluation of (a) the age variable and (b) ALT/AST cut-offs or ratios in pre-existing scores
  • Specifically evaluate the utility of the ELF and similar proprietary scores in older persons
  • Evaluate the utility of novel device-specific sonographic techniques for MASLD identification in older persons

4. Lifestyle and Sociodemographic Considerations

4.1. Diet and Lifestyle

MASLD is, at its core, a non-communicable disease (NCD) [92], intrinsically linked to at least three other major NCDs (CVD [93], cancer [94], and T2DM [95,96]). Like other NCDs, there are multiple lifestyle and environmental risk-factors associated with the development of MASLD, including unhealthy diets [97,98,99,100], infrequent physical activity and/or increased sedentary time [101,102,103], metabolic dysregulation-related conditions (including dyslipidaemia, insulin resistance and hypertension) [8,104,105], obesity [106,107], and even air pollution [108]. These considerations are even more important in the context of MASLD in older adults, due to the increasing prevalence of metabolic diseases in older persons [109], reduced physical activity compared to younger persons [110], and an increasing risk of poorer diet quality over time [111].
Many of these changes are due to a constellation of ageing-related processes, including dietary changes in the context of altered living circumstances, bereavement, isolation, and access to transportation [112], and reduced physical function due to co-morbid medical conditions, a lack of interest, a poor understanding of physical activity requirements, and musculoskeletal pain [113,114,115]. Guidelines to reduce the progression or prevent the development of MASLD in older persons need to focus on practical strategies to overcome these unique barriers to NCD reduction in older persons, with a focus on attainable dietary change, physical activity engagement, and appropriate management of co-morbid metabolic diseases.
Recommendations for Future Research
  • Evaluate the role of healthy lifestyle promotion (diet, physical activity, social engagement) in ameliorating the development or progression of MASLD along with other co-morbid cardiometabolic conditions in older persons
  • Evaluate both individual and structural barriers to the implementation of public policy-level interventions for non-communicable diseases, including MASLD, in older persons

4.2. Sociodemographic Influences

Many NCDs are considered to be related to both the aforementioned ‘lifestyle’ conditions and other social determinants of health, including socioeconomic disadvantage, psychosocial stress, educational attainment, and housing conditions [116,117], as well as ethnicity [118], indigenous status [119], and living remotely [120,121]. Many of these have been shown to be related to the presence of MASLD [122,123], including some in older persons specifically [124]. Despite this, a recent scoping review did not find any prospective studies evaluating social determinants of health in MASLD patients in the clinic [125]. Neither of the recent European [17] or North American [91] guidelines reference social determinants of health, the recognition of which has important public policy implications in the management of MASLD more generally—and in older adults specifically.
Recommendations for Future Research
  • Prospectively evaluate the presence of and outcomes related to social determinants of health in older persons with MASLD to guide future targeted interventions

5. MASLD and Clinical Outcomes in Older Persons

5.1. MASLD and Cardiovascular Disease

Traditionally, CVD has been described as the most common cause of death amongst adults with MASLD [126,127]. However, a more granular evaluation of the data raises several questions about these conclusions. Firstly, in a recent meta-analysis, it was noted that many studies adjust for covariates that include dyslipidaemia (rather than individual cholesterol parameters) and the presence/absence of hypertension (rather than blood pressure in mmHg) [126]. However, this is not how usual cardiovascular risk assessment is carried out. Most risk calculators (including the older-person specific SCORE2-OP) [43] utilise total cholesterol, high density lipoprotein (HDL) cholesterol, and systolic blood pressure. In our study looking at rates of fatal and non-fatal major adverse cardiovascular events (MACE) in older adults, we found an ‘independent’ relationship between MASLD and MACE when adjusting for categorical risk factors—a relationship that was lost when including multiple known predictors of CVD in older persons [128,129]. These findings are concordant with another meta-analysis, whereby MASLD was not associated with CVD-related mortality [130]. Similarly, some of the more commonly cited MASLD-related single nucleotide polymorphisms (for example PNPLA3) seem to confer a reduced risk of CVD [131], implying heterogeneity in CVD risk for different patients with MASLD.
Given these results, it is recommended that older individuals with known MASLD undergo CVD risk-stratification using an age-appropriate CVD risk calculator. However, it is not clear that MASLD itself is an independent or synergistic risk for MACE in older persons, nor that targeting MASLD specifically would reduce atherosclerotic cardiovascular events or CVD-related deaths beyond concomitant improvements in other cardiometabolic risk factors.
In contrast, there are data to support an independent relationship between MASLD and incident atrial fibrillation (AF), including in our own ASPREE cohort [128]. While the Rotterdam study showed that liver stiffness (rather than steatosis alone) was associated with AF [132], this is contradicted by another study conducted in older persons [128] as well as by a meta-analysis of observational studies of more general cohorts [133]. While this does not imply a need for AF-related screening in older persons with MASLD, it is a risk that could be discussed in clinic, and self-reports of arrhythmia or tachycardia should prompt further investigation.
Recommendations for Future Research
  • Assess the risks of MASLD on CVD outcomes in older persons including the impact of hepatic steatosis and fibrosis severity while adjusting for all known contributors to risk
  • Ascertain whether MASLD patients (irrespective of CVD risk profile) benefit from primary prevention for CVD with statins and other therapies
  • Determine whether there are specific markers of excess CVD risk in older persons with MASLD (such as advanced fibrosis, or MASLD with specific monogenic/polygenic findings)

5.2. MASLD and Extra-Hepatic Malignancy

Extra-hepatic malignancy is a significant concern in persons of all ages with MASLD. While there are conflicting data around its role in overall cancer-related mortality in the general population [130,134], MASLD is thought to confer an increased risk of extra-hepatic malignancy in middle-aged persons, particularly cancers arising from the gastrointestinal tract, breast, and genitourinary systems [94]. However, there is less cancer-focused research in the older adult MASLD population. Of the 10 studies cited in a recent meta-analysis [94], none had an average participant age of ≥65 years. While one observational Japanese study focused on persons aged ≥60 years, it compared MASLD-related chronic liver disease vs. hepatitis C-related chronic liver disease, rather than older adults with MASLD vs. no-MASLD [135].
This remains an important knowledge gap in the literature. Screening programs for common cancers (such as colorectal cancer) often recommended an ‘end date’ based on age or are otherwise recommended to be individualised based on perceived risks and benefits [136,137]. While MASLD may confer an increased risk of colorectal cancer—and thus potentially imply increased benefit to screening in older adults with MASLD—it is also associated with an increased prevalence of frailty and future physical disability [124,138,139]. A better understanding of risks and benefits of screening for such malignancy in older persons specifically would help guide clinical decision making, inform guidelines, and educate patients.
Recommendations for Future Research
  • Evaluate whether MASLD confers an increased risk of extra-hepatic malignancy in older persons
  • Evaluate whether screening for extra-hepatic malignancy in older persons with MASLD yields meaningful improvements in quality or length of life and disability-adjusted life-years saved

5.3. MASLD and Diverticulosis

Colonic diverticulosis is extremely common—over two thirds of adults eventually develop it—and is associated with both increasing age and a ‘Western’ lifestyle [140]. Indeed, in the USA, one study has reported a prevalence of over 70% in persons aged 80 years or older [141]. As we have noted previously, MASLD is also a lifestyle-related disease, and there are reasons to believe there may be shared underlying aetiological factors and pathophysiological drivers [142,143]. Indeed, obesity (which is inextricably linked with MASLD [106,107]) is also linked with the development of diverticular disease [142]. Additionally, central adiposity and increased visceral adipose tissue are more strongly linked with diverticulosis than generalised overweight [142,144], and these differences are also seen in those with MASLD [145].
Another such shared pathway includes the gut microbiome. While a detailed exploration of the microbiome is outside the scope of this review, there are known links between gastrointestinal ‘dysbiosis’ and the development of metabolic disorders such as MASLD [146,147,148]. Similarly, there are alterations in the microbiome of people living with diverticular disease, and alterations also exist between those with and without complications associated with their diverticulosis (i.e., incidentally identified diverticulosis compared with symptomatic uncomplications or complicated diverticular disease) [142].
However, despite these links, it remains uncertain whether targeting MASLD in older persons reduces the risk of either developing diverticular disease or conversion from asymptomatic to symptomatic disease.
Recommendations for Future Research
  • Evaluate whether treating MASLD in older persons reduces the risk of developing complicated diverticular disease

5.4. MASLD and Infectious Complications

It has been estimated that approximately one-third of deaths in older age are attributable to infections [149]. There are known changes to the immune system with ageing [150], including impaired memory B cells and antibody production [151], impaired T cell function (which may also predispose to autoimmune disease and cancer) [152], and impaired innate immunity [149]; all of these may increase an affected individual’s susceptibility to infection. Additionally, MASLD has been identified as being associated with an increased risk of serious infections [153], including in those with ‘only’ simple steatosis through to those with cirrhosis [154]. One of the larger studies evaluating this link found that, while the risk of infection did increase with age, biopsy-proven MASLD remained associated with an overall increased risk of infection [154]. While the mechanisms leading to this remain speculative, it is possible that a combination of chronic low-grade inflammation, a disrupted intestinal barrier function, and altered adaptive and innate immunity may contribute [155,156,157,158,159]. Indeed, immunosenescence has previously been linked to the progression of MASLD in those with T2DM [160], and more general cellular senescence is also linked with metabolic disorders [161]. Better understanding the potential dual impact of ageing and MASLD on immune function and predisposition to specific infections would assist with the clinical management of impacted individuals.
Recommendations for Future Research
  • Determine the overall impact of serious infections on older adults with MASLD
  • Better understand the dual impacts of ageing and MASLD on immune function

5.5. MASLD Associations with Frailty and Physical Disability

Sarcopenia and frailty are common problems in advanced chronic liver disease across populations of all ages and are associated with increased risk of complications and short-term mortality [162,163,164]. This risk is also bidirectional—those with increased baseline frailty are at increased risk of incident MASLD, cirrhosis, and other liver-related events [165]. The relationship between MASLD and frailty appears to be independent of cirrhosis status, though is more severe in those with advanced chronic liver disease than those without [166].
The relationship between frailty and MASLD has been studied predominantly in older persons, with recent work building on that evaluating the impact of MASLD on longer-term physical function [138]—although the MASLD–frailty relationship was also seen in at least one middle-aged cohort [167]. Our paper showed a significantly increased risk of developing persistent physical disability (defined as the loss of one of the basic activities of daily living [ADL] for at least six months, or placement in a residential care facility due to the loss of an ADL), despite adjustment for multiple potential contributors, including gait speed and grip strength [138]. The mechanism underlying this is uncertain, although it may provide the opportunity for intervention if it were better understood. This may then allow for improving the health span and well-being of older persons with MASLD.
Recommendations for Future Research
  • Determine whether MASLD is an epiphenomenon or causative factor in the development of frailty and persistent physical disability (e.g., through metabolomic factors)
  • Determine if a targeted intervention for frailty improves quality of life or health span in older persons with MASLD

5.6. MASLD and Cognitive Outcomes

Multiple studies have examined dementia risk and brain-ageing in middle-aged persons with MASLD, often finding an increased risk of cognitive decline [168,169,170]. This relationship is less clear, however, in older adults. While one Swedish study (in which participants had a median age of 70 years) found a relationship between MASLD and dementia [171], this study did not adjust for baseline cognitive performance. Other studies involving older persons have suggested either no relationship between MASLD and dementia [172], or even an inverse relationship, whereby baseline MASLD was associated with a reduced risk of incident dementia [138,173].
The reason for this discrepancy is unclear; it is possible that ‘survivors’ to older age with MASLD have a less cognitively harmful phenotype of their liver disease. It is also possible that—given the known accelerated weight loss that occurs prior to and during the development of dementia [174,175]—even if MASLD increases the overall risk of dementia in older persons, their steatotic liver disease may have regressed prior to the formal diagnosis of dementia. In either case, when counselling older persons, although baseline cognitive function testing may assist with overall risk stratification, MASLD in isolation does not seem to clearly increase the risk of dementia.
Understanding this discrepancy in findings is important. It is well known that dementia is one of the greatest concerns people have as they age [176,177] and this fear can act as a barrier to being evaluated for cognitive decline, despite concerns patients or caregivers have [178]. While debate persists around the potential benefits and harms of screening for dementia [179,180], one strategy may be to specifically target at-risk groups. While various models exist for dementia screening [181], it remains to be seen whether there are specific subgroups of patients with MASLD who would potentially benefit from screening—and if screening the middle-aged, whether stopping as people transition to later life is appropriate.
Recommendations for Future Research
  • Determine if there are differences between middle-aged and older MASLD that influence the development of cognitive decline/dementia
  • Evaluate (a) the cognitive decline in the middle-aged/older adult transition and (b) biomarkers of dementia progression in older persons with MASLD and no MASLD to assist in understanding causality/reverse causality in these groups

5.7. MASLD and Mortality

MASLD is frequently cited as a cause of increased mortality in middle-aged adults [134,182,183]. Given this, and other known complications of the disease, case-finding and consideration of pharmacologic intervention has been proposed in higher-risk groups [17]. However, there is less certainty that MASLD leads to increased mortality in older persons. Key studies have shown either no excess mortality risk [128,184] or a markedly attenuated risk [185] in older persons (depending on age cut-off). While the study based on the ASPREE (ASPirin in Reducing Events in the Elderly) cohort [128] may have some inherent bias due to exclusion of individuals with known pre-existing CVD and an a priori presumed low 5-year mortality risk (a key inclusion criterion for the study), this is precisely the group likely to be considered for MASLD screening. In contrast, the Rotterdam study did not have pre-specified exclusion criteria [186], and given the similar findings of all of these major studies, it is highly likely that MASLD does not lead to an excess of premature mortality in older adults. This has been further evaluated using genetic markers of MASLD rather than composite scores or radiology, with similar findings [187].

6. MASLD Treatments in Older Persons

6.1. Treatment Overview

A wide range of treatments are described in the literature and are well summarised in the recent North American [91] and European guidelines [17] as well as a recent review article focusing on liver-directed therapies [188]. While historically MASLD treatment has focused on (a) lifestyle interventions as well as (b) the management of known co-morbidities, there is now a therapy specifically licensed for the treatment of MASH—resmetirom [189]—with several more in the pipeline [188]. However, there are specific nuances to consider regarding both lifestyle and adjunctive pharmacotherapy, as well as the prescription of MASLD-specific medications in older persons, due to differences in ageing physiology, the risks of polypharmacy, and change in competing risks over time as the population ages. As such, here we describe key considerations around guidelines and recommendations that tend to be based on data from middle-aged persons and highlight opportunities for future research.

6.2. Lifestyle Therapies: Weight Loss and Diet

Weight loss features prominently in North American and European MASLD guidelines, with a target of 3–5+% to reduce steatosis and 7–10+% to improve MASH and/or fibrosis stage [17,91], particularly in those who are overweight/obese. These recommendations are primarily supported by studies in middle-aged adults. Multiple iterations of calorie-restricted diets have been studied in MASLD, with some benefits identified in a recent systemic review and meta-analysis (although none had a mean participant age over 55 years) [190]. Whilst these weight loss targets improve not only steatosis itself and also concomitant metabolic dysfunction (such as insulin resistance), they are challenging to achieve and maintain, with weight regain occurring slowly in both MASLD-focused studies [191,192] and in general weight loss studies [193]. Additionally, not all patients benefit from significant weight loss. In particular, in so-called ‘lean’ MASLD subjects, the target weight loss is capped at 3–5% [17], while in those with a background history of disordered eating patterns, implementation of weight loss measures requires a careful and balanced evaluation of the risks and benefits as well as additional psychological support [194]. Importantly, for the older population, there are also data that suggested they may be at increased risk of mortality as a result of significant weight loss [195], although the implications for this in the MASLD-group specifically are unknown.
An additional strategy compared to weight loss alone is to optimise overall diet quality, as well as macronutrient and micronutrient composition, for those with MASLD. One option is to institute a Mediterranean Diet pattern of eating, with a focus on high intakes of polyphenol-rich olive oil, vegetables, fruits, nuts/seeds, legumes, and seafood. It also focuses on a reduction in added sugar, refined carbohydrates, saturated fat, red and processed meats, and ultra-processed foods [196,197]. The Mediterranean Diet was shown in a 2021 meta-analysis to be associated with reduced odds of having MASLD [198] (however, it included only one study with a mean participant age over 65 years), which has also been shown in a more recent study in older persons [199]. Importantly, its improvements in MASLD are at least partly independent of overall weight loss [200]. These results have been confirmed in prospective trials specifically in MASLD, demonstrating benefits to the Mediterranean Diet alone [201] as well as a polyphenol enriched Mediterranean Diet [202], and higher adherence to the Mediterranean Diet has also been shown to improve the HSI (a surrogate for hepatic steatosis) in the PREDIMED study [203]. Other dietary options have been explored, including time restricted eating [192,204] and a low-carbohydrate, high-fat diet [205], although there is significantly less research on these strategies compared with the Mediterranean Diet. Conversely, there is a relationship between higher intakes of ultra-processed foods (UPFs) and an increased prevalence of MASLD in older persons [199]. It remains uncertain if this is related to co-morbid frailty [206] rather than a true causal link. Importantly, while UPFs are positively associated with MASLD, it has not yet been shown that reducing their intake would improve MASLD incidence or outcomes. In the context of older persons, UPF reduction should be undertaken cautiously, to ensure appropriate non-UPF substitution.
This nuance is especially relevant in older persons. There are significant additional co-morbidities in older persons living with MASLD compared to middle-aged people. As such, a primary focus on weight loss through diet may carry greater risk in this cohort, and while focusing instead on improving dietary patterns (e.g., shifting towards a more Mediterranean Diet pattern) may be a reasonable alternative goal, both are difficult to implement in practice for several reasons. Firstly, older adults with MASLD are more likely to be pre-frail or frail [124] as well as to develop persistent physical disability [138], and significant weight loss is an independent risk for the development of frailty [207,208]. Still, it should be noted that there is evidence to suggest that some obese older adults may functionally benefit from weight loss, particularly when combined with exercise [209]. Secondly, the Mediterranean Diet—beyond its utility for MASLD—is associated with a reduced risk of frailty in older persons [206,210,211]. Additionally, it is associated with reduced all-cause mortality, CVD, and improved quality of life [212,213,214,215]. Thirdly, food insecurity is common in older adults [216] and is associated with reduced adherence to a Mediterranean Dietary pattern of eating [217] as well as both MASLD and advanced hepatic fibrosis in the middle-aged [218]. Finally, multiple potential barriers are often noted in the implementation of dietary and lifestyle change, including psychosocial barriers [219], functional/motivational/financial issues [220,221,222], and diet palatability [223]. Understanding the optimal dietary therapy (ies) for older persons with MASLD requires further study.
Recommendations for Future Research
  • Longitudinally assess the impact of (a) weight loss and (b) Mediterranean Diet adherence on older persons with MASLD in terms of both hepatic steatosis as well as functional outcomes
  • Longitudinally assess the impact of the above on liver-related and extra-hepatic outcomes in older adults with MASLD
  • Assess the barriers and facilitators to dietary modification in older persons with MASLD

6.3. Lifestyle Therapies: Physical Activity and Exercise

Similar to the recommendations for weight loss and dietary modification, both North American and European guidelines focus on exercise as a key tenet in the management of MASLD [17,91]. Sedentary behaviour is a known risk factor for MASLD [101,224,225], and conversely, higher levels of non-exercise physical activity (daily activities that involve movement without planning or strict control of exertion) is associated with both a lower risk for MASLD [225] as well as for both all-cause mortality and CVD in the general population [226,227]. Physical activity in MASLD specifically is also known to reduce all-cause mortality [228]. Recommended exercise targets for MASLD are >150 min/week of moderate intensity or >75 min/week of vigorous intensity exercise [17]. Once more, however, this is based on heterogeneous data, with a paucity of studies in older persons [229,230,231,232]. This lack of relevant data is akin to the lack of evidence underpinning the overall healthy physical activity guidelines for adults aged over 65 years in both the USA and Australia [233,234].
Importantly, physical activity is associated with a reduced risk of frailty [235], symptoms of frailty [236], and nursing home placement in older adults [237]. Whilst there are limited data on the impact of a physical activity intervention in older persons with MASLD, it is likely that—given the relationship between MASLD, frailty, sarcopenia, and myosteatosis [124,238]—a targeted intervention may improve not only hepatic steatosis but also the known associated physical dysfunction. This is particularly important in the setting of weight loss. If reducing bodyweight is considered an appropriate therapeutic goal for an overweight older person, the institution of calorie/kilojoule restriction in combination with resistance training has been shown to markedly attenuate weight loss-induced lean mass loss [239]. Older-person specific interventions that recognise and attempt to ameliorate the known barriers to exercise with ageing [114,222] may improve both liver and non-liver related outcomes as well as improve health span.
Recommendations for Future Research
  • Assess the type, level and duration of exercise required to ameliorate poorer outcomes in older persons with MASLD, with a particular focus on frailty, persistent physical disability, and quality of life

6.4. Directed Pharmacotherapies

6.4.1. Incretin Mimetics

In addition to the lifestyle recommendations of diet and exercise, adjunctive weight loss pharmacotherapy is becoming increasingly common in the treatment of overweight/obesity [240]—with an associated decrease in the need for bariatric surgery [241]. Despite the lack of a specific glucagon-like peptide (GLP)-1 target in hepatocytes, GLP-1 agonists may have additional liver-specific benefits via improvement in insulin resistance, managing co-morbid T2DM, and improving lipotoxicity [242,243]. These actions are particularly important in MASLD, both because of the known link between T2DM and an increased risk of progressive liver disease [14,244], as well as the known challenges of sustaining weight loss through diet and exercise alone [193,245]. A recent study compared lifestyle alone against GLP-1 agonist therapy in MASLD and showed comparable reductions in liver fat content, although improvements in glucose homeostasis were only seen in the GLP-1 agonist group [246].
Given this, it is no surprise that promising data are emerging to showcase the benefits from incretin monotherapy or combination therapy (a GLP-1 agonist combined with another incretin or glucagon) prescription in middle-aged adults with MASLD on a range of liver-related parameters including histology, transaminases, and MRI-quantified liver fat content [247,248,249,250,251]. Recently, the final data from the ESSENCE randomised controlled trial of 2.4 mg semaglutide versus placebo in MASH showed marked improvements in both steatohepatitis and liver fibrosis compared with placebo [252]. Additionally, observational data suggest a reduction in liver-related outcomes and reduced mortality in individuals with cirrhosis and T2DM treated with GLP-1 agonists (although not necessarily only due to MASLD) [253,254]. While earlier studies focused solely on those with compensated liver disease, a recent small pilot trial of survodutide (a GLP-1/glucagon dual-agonist) was safe and efficacious in decompensated cirrhosis [255]. Additionally, recent Taiwanese data show that there is a reduced risk of progression to cirrhosis amongst patients with T2DM compared to those using long-acting insulin [256] amongst middle-aged adults. Further data on the current role of GLP-1 receptor agonists can be found in a review article in Gut [257].
There are less data on the safety and efficacy of these agents in older adults with MASLD, although multiple studies have investigated their use in older adults in other conditions. A recent systematic review showed that GLP-1 agonists led to a reduced risk of MACE in adults ≥65 years [258], which is of particular interest given the previously described link between MASLD and MACE [128,129]. However, as previously mentioned, weight loss in older adults may present a double-edged sword, as it may be associated with an increased risk of frailty [207], or in the obese, may be protective [209]. This is especially concerning in older persons with MASLD, given the previously identified links with frailty and physical disability [124,138]. The addition of exercise training to incretin therapies to mitigate lean mass loss is an as-yet poorly studied area in the general literature [259], but may be important in the safe utilisation of these therapies in older persons to minimise potential harms. To our knowledge, there is no specific study investigating physical function outcomes in older persons with MASLD treated with incretin mimetics, nor are there data to guide decision making around BMI strata that may most benefit from such therapies in older persons. Additionally, while therapy cessation is known to lead to weight regain, there are less data on this in older persons [260], especially if the distribution of that gain is disproportionately adipose tissue, or whether there is a return of lean mass.
Recommendations for Future Research
  • Evaluate the safety of incretin mimetics in older adults with MASLD in terms of body composition as well as changes associated with weight regain with medication cessation
  • Evaluate whether incretin mimetics improve (a) liver-related and (b) non-liver related cardiometabolic outcomes in older adults with MASLD
  • Evaluate whether incretin mimetics improve physical function in older adults within various BMI strata
  • Describe changes in dietary intake in older adults prescribed incretin mimetics

6.4.2. Thyroid Hormone Receptor-β Agonists

In 2024, the Federal Drug Administration (FDA) in the USA approved resmetirom [189] as the first MASH-specific agent licensed for use, due to its Phase III data showing improvements in both steatohepatitis and fibrosis in at-risk disease [261]. Previous data had established a relationship between altered thyroid hormone metabolism and MASLD-related inflammation and fibrosis [262]. Resmetirom is an orally active liver-specific compound that preferentially binds to thyroid hormone receptor beta (THR-β), the predominant receptor isoform in hepatocytes. This reduces T4 without impacting thyroid stimulating hormone (TSH) or the more biologically active triiodothyronine (T3) [262]. Taken together, these actions improve lipid metabolism, mitochondrial β-oxidation, reduce VLDL and triglycerides, and reduce liver stiffness and fat content, all with relatively minor deleterious impacts on other tissues, such as the heart and bone density [261,262,263]. In addition to these key cardiometabolic benefits, secondary outcomes suggest that resmetirom improves health-related quality of life in MASLD [264]. Even more potent THR-β agents are in development and may have improved efficacy over resmetirom, although these trials are ongoing [262]. It is uncertain whether resmetirom or other THR-β agents will have any role in MASLD-related cirrhosis.
Again, there are no data on the use of resmetirom in older persons. This is relevant given the links between ageing and thyroid dysfunction generally and the data on the impact of thyroid status on cognitive function [265], as well as the increased resistance to thyroid hormones in older persons [266]. Resmetirom can cause gastrointestinal side-effects (including diarrhoea and nausea) [261], and currently the impact of this on dietary patterns and overall nutritional intake is unknown. Pharmacologically, resmetirom is hepatically metabolised as a CYP2C8 substrate, which is the same cytochrome P450 enzyme that metabolises a number of commonly prescribed medications, including certain non-steroidal anti-inflammatory drugs, some statins, and amiodarone, as well as being inhibited by clopidogrel and gemfibrozil [267,268,269]. Older persons are more likely to be prescribed multiple agents [270], and the impact of polypharmacy on the efficacy of resmetirom and other agents remains understudied.
Recommendations for Future Research
  • Evaluate whether resmetirom has a differential impact on thyroid function parameters in older persons with MASLD
  • Evaluate whether the gastrointestinal side-effects of resmetirom lead to an undesirable dietary shift in older persons with MASLD
  • Evaluate the potential polypharmacy and co-prescribing risks to older persons with MASLD being offered Resmetirom

6.4.3. Fibroblast Growth Factor-21

While resmetirom was the first FDA licensed therapy for MASH, other anti-fibrotic agents are currently undergoing Phase II trials. Fibroblast Growth Factor 21 (FGF21) is associated with obesity, dyslipidaemia, pericardial fat, and insulin resistance, with a half-life of approximately 0.5–2 h [271,272]. Pegozafermin is a long-acting recombinant FGF21 analogue with a half-life of 55–100 h that has been shown to improve insulin sensitivity and decrease de novo lipogenesis, hypertriglyceridaemia [273], hepatic fibrosis and MASH [274]. One trial showed up to 27% improvement in fibrosis stage over 24 weeks (compared to 7% for placebo) in non-cirrhotic middle-aged adults. While these data need to be confirmed to be sustained over a longer duration and evaluated in larger studies, pegozafermin is a promising future option for at-risk MASH.
Efruxifermin, another FGF21 analogue, has been studied in MASH with fibrosis stage 2 or 3 (but not cirrhosis) with similarly promising results, including improvements in fibrosis stage (without worsening of MASH) as well as improvements in non-invasive markers of liver injury (such as transaminase levels and ELF score) in the HARMONY study [275]. Preliminary data presented in abstract form of up to 96 weeks of use confirmed ongoing improvements in both steatohepatitis and fibrosis, including in those participants who did not respond in the initial 24-week period [276]. Another Phase II study has demonstrated safety with signals of efficacy in patients with compensated cirrhosis, expanding the potential population that may benefit from efruxifermin [277]. While early published data from the SYMMETRY study showed no improvement in fibrosis stage at 36 weeks, 50 mg efruxifermin appeared to reduce fibrosis in subjects with cirrhosis at 96 weeks [278]—further work evaluating longer-term use of FGF21 analogues is expected.
Like resmetirom, there are GI related side-effects from FGF-21 analogues, though, once more, these are inadequately studied in older adults [274,275,277]. Importantly for older patients, there have also been safety concerns raised regarding bone health, with accelerated bone loss in some (but not all) of the studies to date [188,278].
Recommendations for Future Research
  • Evaluate the efficacy of FGF21 agonists in older persons with MASLD
  • Determine whether accelerated bone loss in those with and without pre-existing osteopenia or osteoporosis occurs with FGF21 agonist use in older persons
  • Evaluate the impact of gastrointestinal side-effects on the dietary patterns of older persons with MASLD

6.4.4. Co-Prescriptions and Combinations

Given the potential synergies between different therapeutic options, early work has begun to investigate the potential combinations of therapies with differing mechanisms of action. One recent study showed an additive benefit of efruxifermin to GLP-1 agonism in terms of hepatic fat fraction reduction and lipid parameters compared to GLP-1 monotherapy in participants with T2DM [279], and combination therapies have been recently editorialised as the “next frontier in MASH” [280]. While relatively few participants in the landmark resmetirom MAESTRO-NASH study were taking GLP-1 agonists at baseline (and thus co-prescription is difficult to endorse), the AASLD has recently noted that individual clinical judgement can be applied when considering the use of both therapies concurrently [269].
Recommendations for Future Research
  • Evaluate whether co-therapy is synergistically beneficial for fibrosis reduction as well as metabolic risk management in (a) middle-aged persons and (b) older adults specifically

6.5. Pharmacotherapies: Co-Morbidity Management

It is well known that participants with MASLD are more likely to have, and to develop, T2DM [96,281,282], dyslipidaemia [105], and renal impairment [283,284,285]. Further, these diseases are more common in older adults than in the general population, despite the need to re-evaluate ‘normal’ cut-offs with aging [286,287,288,289]. Some of the therapies used to manage these conditions may have indirect benefits in MASLD, and while they are not licensed for liver-specific indications, their continued prescription may have ancillary benefits.
Historically, there has been caution around the use of statins in individuals with MASLD or other chronic liver diseases, due to their perceived risk of precipitating acute liver injury [290]. However, this has been largely debunked, with numerous contemporary studies supporting the initiation of statins in those with various liver diseases or elevated transaminase levels [291,292]. In addition to their role in the management of dyslipidaemia and the primary and secondary prevention of CVD, statins may reduce the risk of both MASLD development, as well as protect against fibrosis in those with existing MASLD [293]. Additionally, they may reduce portal pressure in cirrhosis and reduce the risk of hepatocellular carcinoma development [294,295]. Nevertheless, statin use in older persons can be complicated by concerns around increasing the risk of T2DM development [296] and potential inefficacy or reduced efficacy when used for primary prevention of CVD [297,298]. While statin therapy may have benefits in persons with MASLD, their relative risks and benefits in older persons warrants further study; STAREE (Statins in Reducing Events in the Elderly) trial results are keenly awaited [299].
Similarly, from a cardiovascular and preventive health perspective, aspirin for use as primary prevention remains a contentious issue. The landmark ASPREE study showed no net benefit of aspirin in older adults for MACE, disability-free survival, or all-cause mortality [300,301,302] (although it did show benefit in reducing the incidence of T2DM [303]), and a post-hoc analysis showed no benefit on mortality or MACE in older persons with MASLD [128]. However, existing data do show potential benefits of aspirin in terms of reducing MASH and rates of fibrosis progression [304,305], as well as a marked reduction of hepatocellular carcinoma rates in mixed-aetiology chronic liver disease [306] and MASLD [307]. Given the risks of excess bleeding with aspirin use in older adults [301,308], careful patient selection is important if considering aspirin for chemoprevention or treatment for MASLD and MASLD-specific complications in older persons.
In terms of the management of co-morbid T2DM, sodium-glucose co-transporter 2 inhibitors (SGLT2i) are commonly used to not only improve glycaemic control but to reduce the risk of incident kidney disease and manage co-morbid heart failure [309,310]. These agents may also confer benefits for MASLD [311], including on histologic endpoints [312]. When considering their role in preventing outcomes for patients with coexistent T2DM and MASLD, recent data are supportive—a Japanese study showed that SGLT2i were superior to DPP4 inhibitors for the reducing the development of varices and malignancy [313], and a Korean study has shown that SGLT2i therapy is associated with both higher rates of MASLD regression and lower rates of adverse liver outcomes than other oral antidiabetic therapies [314]. Importantly, SGLT2i are considered safe in older adults [315,316], and thus the existing data support consideration of therapy with SGLT2i for older adults with co-morbid MASLD and T2DM, especially if GLP-1 agonists are not tolerated or available. A key caveat is the diuretic and blood pressure lowering effects of SGLT2i, with a potential for increased risk of falls [317,318]. Frailty assessment (given its relative frequency in MASLD [124]) may assist in identifying subgroups of older persons most likely to benefit with lower risk.
Recommendations for Future Research
  • Determine whether any subgroup(s) of older participants, particularly in relation to frailty and disability status, may particularly benefit from the potential MASLD reduction associated with aspirin and/or statin therapy
  • Determine whether older persons with combination T2DM and MASLD have an outsized benefit from SGLT2 inhibition compared to other therapies, and whether stratifying by frailty identifies a subgroup more likely to benefit

7. Economic Impacts

MASLD, as a major clinical concern and public health burden, represents a significant cost to the health system. A study evaluating the economic impact of MASLD in the US and parts of Europe (Germany, France, Italy, and the United Kingdom) found projected annual costs to be as high as USD 292.19 billion and EUR 227.84, respectively [319]. A more recent study in Italy highlighted excess potential direct costs of EUR 1.77 billion compared to those without MASLD, and up to EUR 7.94 billion including modelled flow-on effects (including increased rates of cancer and T2DM but excluding quality of life impacts) [320]. Importantly, it is likely that those economic costs are most significant in the older adult population; in the US, the per-patient costs were calculated to be highest in the ≥65 year old group [319].
In the context of the developing literature highlighted in this review, it is also important to note that many of these models do not necessarily directly account for the impacts of frailty or physical disability. In the general ageing literature, it is well recognised that frailty is costly [321,322,323]. However, the interplay between MASLD and ageing and the consequent potential increase in older persons with frailty, and at risk of persistent physical disability, is understudied from a health economic perspective. Consequently, the costs identified in MASLD modelling studies could be significantly underestimated in relation to older adults.
Similarly, when considering pharmacoeconomic modelling for the benefits of potential MASLD therapies, studies often do not consider the impact on older persons specifically [324], and we do not yet know the impact of modern pharmacotherapeutic options on frailty or the progression to disability in older persons with MASLD. As highlighted in Section 6, future work to better understand the risks and benefits of the novel therapies for MASLD in older persons will also help guide more accurate health economic modelling.
Recommendations for Future Research
  • Determine the health economic burden of MASLD in older adults including both direct and indirect costs
  • Determine whether modern pharmacotherapies are cost-effective for treating MASLD in older persons

8. Conclusions

MASLD is a commonly identified condition in older persons and, like in the middle-aged, is strongly associated with multiple other cardiometabolic conditions. However, the treatment paradigm may differ significantly. A greater focus on frailty and functional assessment, appropriate selection of adjunctive therapies for co-morbid conditions, and consideration of polypharmacy and the competing risks inherent to ageing are important and understudied areas; focusing solely on weight loss may not yield the benefits we and our patients seek. When counselling older persons with MASLD in the clinic, highlighting these links, and gaps in the literature, will be important, and shifting the focus away from mortality towards improving health span will likely realign expectations for both clinicians and patients. Further research into understanding the impact of MASLD on older persons and how best to care for them is crucial, as the epidemiology and risks differ compared to younger and middle-aged persons, while, at the same time, the population continues to age rapidly.

Author Contributions

Conceptualisaion, D.C.-C. and S.K.R.; methodology, D.C.-C., writing—original draft preparation, D.C.-C., P.W.L. and S.K.R.; writing—review and editing, D.C.-C., W.W.K., A.M., P.W.L., J.A.F., K.V., J.S.L., A.D.H., J.R., J.J.M., A.J.O., R.L.W. and S.K.R.; visualization, D.C.-C.; supervision, S.K.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Addison, T. Observations on fatty degeneration of the liver. Guys Hosp. Rep. 1836, 1, 485. [Google Scholar]
  2. Schaffner, F.; Thaler, H. Nonalcoholic fatty liver disease. Prog. Liver Dis. 1986, 8, 283–298. [Google Scholar] [PubMed]
  3. Ludwig, J.; Viggiano, T.R.; Mcgill, D.B.; Oh, B. Nonalcoholic steatohepatitis: Mayo Clinic experiences with a hitherto unnamed disease. Mayo Clin. Proc. 1980, 55, 434–438. [Google Scholar] [CrossRef] [PubMed]
  4. Marchesini, G.; Brizi, M.; Morselli-Labate, A.M.; Bianchi, G.; Bugianesi, E.; McCullough, A.J.; Forlani, G.; Melchionda, N. Association of nonalcoholic fatty liver disease with insulin resistance. Am. J. Med. 1999, 107, 450–455. [Google Scholar] [CrossRef]
  5. Boutari, C.; Mantzoros, C.S. A 2022 update on the epidemiology of obesity and a call to action: As its twin COVID-19 pandemic appears to be receding, the obesity and dysmetabolism pandemic continues to rage on. Metabolism 2022, 133, 155217. [Google Scholar] [CrossRef]
  6. 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. [Google Scholar] [CrossRef]
  7. Younossi, Z.M.; Kalligeros, M.; Henry, L. Epidemiology of Metabolic Dysfunction-Associated Steatotic Liver Disease. Clin. Mol. Hepatol. 2024, 31, S32–S50. [Google Scholar] [CrossRef]
  8. Cortez-Pinto, H.; Camilo, M.E.; Baptista, A.; De Oliveira, A.G.; De Moura, M.C. Non-alcoholic fatty liver: Another feature of the metabolic syndrome? Clin. Nutr. 1999, 18, 353–358. [Google Scholar] [CrossRef]
  9. Alberti, K.G.; Zimmet, P.; Shaw, J. Metabolic syndrome--a new world-wide definition. A Consensus Statement from the International Diabetes Federation. Diabet. Med. 2006, 23, 469–480. [Google Scholar] [CrossRef]
  10. Rinella, M.E.; Lazarus, J.V.; Ratziu, V.; Francque, S.M.; Sanyal, A.J.; Kanwal, F.; Romero, D.; Abdelmalek, M.F.; Anstee, Q.M.; Arab, J.P.; et al. A multi-society Delphi consensus statement on new fatty liver disease nomenclature. J. Hepatol. 2023, 78, 1966–1986. [Google Scholar] [CrossRef]
  11. Hagstrom, H.; Vessby, J.; Ekstedt, M.; Shang, Y. 99% of patients with NAFLD meet MASLD criteria and natural history is therefore identical. J. Hepatol. 2024, 80, e76–e77. [Google Scholar] [CrossRef]
  12. Song, S.J.; Che-To Lai, J.; Lai-Hung Wong, G.; Wai-Sun Wong, V.; Cheuk-Fung Yip, T. Can we use old NAFLD data under the new MASLD definition? J. Hepatol. 2024, 80, e54–e56. [Google Scholar] [CrossRef] [PubMed]
  13. Riley, D.R.; Hydes, T.; Hernadez, G.; Zhao, S.S.; Alam, U.; Cuthbertson, D.J. The synergistic impact of type 2 diabetes and MASLD on cardiovascular, liver, diabetes-related and cancer outcomes. Liver Int. 2024, 44, 2538–2550. [Google Scholar] [CrossRef] [PubMed]
  14. O’Beirne, J.; Skoien, R.; Leggett, B.A.; Hartel, G.F.; Gordon, L.G.; Powell, E.E.; Valery, P.C. Diabetes mellitus and the progression of non-alcoholic fatty liver disease to decompensated cirrhosis: A retrospective cohort study. Med. J. Aust. 2023, 219, 358–365. [Google Scholar] [CrossRef] [PubMed]
  15. 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. [Google Scholar] [CrossRef]
  16. Deprince, A.; Haas, J.T.; Staels, B. Dysregulated lipid metabolism links NAFLD to cardiovascular disease. Mol. Metab. 2020, 42, 101092. [Google Scholar] [CrossRef]
  17. European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). J. Hepatol. 2024, 81, 492–542. [Google Scholar] [CrossRef]
  18. 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. [Google Scholar] [CrossRef]
  19. Younossi, Z.M.; Stepanova, M.; Younossi, Y.; Golabi, P.; Mishra, A.; Rafiq, N.; Henry, L. Epidemiology of chronic liver diseases in the USA in the past three decades. Gut 2020, 69, 564–568. [Google Scholar] [CrossRef]
  20. Younossi, Z.M.; Stepanova, M.; Ong, J.; Trimble, G.; AlQahtani, S.; Younossi, I.; Ahmed, A.; Racila, A.; Henry, L. Nonalcoholic Steatohepatitis Is the Most Rapidly Increasing Indication for Liver Transplantation in the United States. Clin. Gastroenterol. Hepatol. 2021, 19, 580–589 e5. [Google Scholar] [CrossRef]
  21. Adams, L.A.; Roberts, S.K.; Strasser, S.I.; Mahady, S.E.; Powell, E.; Estes, C.; Razavi, H.; George, J. Nonalcoholic fatty liver disease burden: Australia, 2019–2030. J. Gastroenterol. Hepatol. 2020, 35, 1628–1635. [Google Scholar] [CrossRef] [PubMed]
  22. Le, P.; Tatar, M.; Dasarathy, S.; Alkhouri, N.; Herman, W.H.; Taksler, G.B.; Deshpande, A.; Ye, W.; Adekunle, O.A.; McCullough, A.; et al. Estimated Burden of Metabolic Dysfunction-Associated Steatotic Liver Disease in US Adults, 2020 to 2050. JAMA Netw. Open 2025, 8, e2454707. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, D.; Kim, W.R.; Kim, H.J.; Therneau, T.M. Association between noninvasive fibrosis markers and mortality among adults with nonalcoholic fatty liver disease in the United States. Hepatology 2013, 57, 1357–1365. [Google Scholar] [CrossRef] [PubMed]
  24. Singh, S.; Allen, A.M.; Wang, Z.; Prokop, L.J.; Murad, M.H.; Loomba, R. Fibrosis progression in nonalcoholic fatty liver vs. nonalcoholic steatohepatitis: A systematic review and meta-analysis of paired-biopsy studies. Clin. Gastroenterol. Hepatol. 2015, 13, 643–654.e9. [Google Scholar] [CrossRef]
  25. Ekstedt, M.; Hagstrom, H.; Nasr, P.; Fredrikson, M.; Stal, P.; Kechagias, S.; Hultcrantz, R. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 2015, 61, 1547–1554. [Google Scholar] [CrossRef]
  26. Taylor, R.S.; Taylor, R.J.; Bayliss, S.; Hagstrom, H.; Nasr, P.; Schattenberg, J.M.; Ishigami, M.; Toyoda, H.; Wai-Sun Wong, V.; Peleg, N.; et al. Association Between Fibrosis Stage and Outcomes of Patients with Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Gastroenterology 2020, 158, 1611–1625.e12. [Google Scholar] [CrossRef]
  27. Lazarus, J.V.; Mark, H.E.; Villota-Rivas, M.; Palayew, A.; Carrieri, P.; Colombo, M.; Ekstedt, M.; Esmat, G.; George, J.; Marchesini, G.; et al. The global NAFLD policy review and preparedness index: Are countries ready to address this silent public health challenge? J. Hepatol. 2022, 76, 771–780. [Google Scholar] [CrossRef]
  28. United Nations, Department of Economic and Social Affairs, Population Division. World Population Ageing 2019 Highlights; UN: New York, NY, USA, 2019.
  29. Mathers, C.D.; Stevens, G.A.; Boerma, T.; White, R.A.; Tobias, M.I. Causes of international increases in older age life expectancy. Lancet 2015, 385, 540–548. [Google Scholar] [CrossRef]
  30. Rahman, M.M.; Rana, R.; Khanam, R. Determinants of life expectancy in most polluted countries: Exploring the effect of environmental degradation. PLoS ONE 2022, 17, e0262802. [Google Scholar] [CrossRef]
  31. World Health Organization. Burden of Disease Attributable to Unsafe Drinking-Water, Sanitation and Hygiene, 2019 Update; World Health Organization: Geneva, Switzerland, 2023.
  32. Hao, L.; Xu, X.; Dupre, M.E.; Guo, A.; Zhang, X.; Qiu, L.; Zhao, Y.; Gu, D. Adequate access to healthcare and added life expectancy among older adults in China. BMC Geriatr. 2020, 20, 129. [Google Scholar] [CrossRef]
  33. Marques, S.; Mariano, J.; Mendonca, J.; De Tavernier, W.; Hess, M.; Naegele, L.; Peixeiro, F.; Martins, D. Determinants of Ageism against Older Adults: A Systematic Review. Int. J. Environ. Res. Public Health 2020, 17, 2560. [Google Scholar] [CrossRef] [PubMed]
  34. Florisson, S.; Aagesen, E.K.; Bertelsen, A.S.; Nielsen, L.P.; Rosholm, J.U. Are older adults insufficiently included in clinical trials?—An umbrella review. Basic Clin. Pharmacol. Toxicol. 2021, 128, 213–223. [Google Scholar] [CrossRef]
  35. Markham, S.C.; McNab, J.; O’Loughlin, K.; Clemson, L. International responses addressing the under-representation of older people in clinical research. Australas. J. Ageing 2023, 42, 762–768. [Google Scholar] [CrossRef] [PubMed]
  36. Tian, Y.E.; Cropley, V.; Maier, A.B.; Lautenschlager, N.T.; Breakspear, M.; Zalesky, A. Heterogeneous aging across multiple organ systems and prediction of chronic disease and mortality. Nat. Med. 2023, 29, 1221–1231. [Google Scholar] [CrossRef]
  37. Allaire, M.; Gilgenkrantz, H. The aged liver: Beyond cellular senescence. Clin. Res. Hepatol. Gastroenterol. 2020, 44, 6–11. [Google Scholar] [CrossRef]
  38. Wynne, H.A.; Cope, L.H.; Mutch, E.; Rawlins, M.D.; Woodhouse, K.W.; James, O.F. The effect of age upon liver volume and apparent liver blood flow in healthy man. Hepatology 1989, 9, 297–301. [Google Scholar] [CrossRef]
  39. Boland, B.S.; Dong, M.H.; Bettencourt, R.; Barrett-Connor, E.; Loomba, R. Association of serum bilirubin with aging and mortality. J. Clin. Exp. Hepatol. 2014, 4, 1–7. [Google Scholar] [CrossRef] [PubMed]
  40. Clayton-Chubb, D.; Majeed, A.; Roberts, S.K.; Schneider, H.G.; Commins, I.; Fitzpatrick, J.; Woods, R.L.; Ryan, J.; Hussain, S.M.; Tan, N.; et al. Serum Transaminases And Older Adults: Distribution And Associations with All-Cause Mortality. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glae203. [Google Scholar] [CrossRef]
  41. Munoz-Espin, D.; Serrano, M. Cellular senescence: From physiology to pathology. Nat. Rev. Mol. Cell Biol. 2014, 15, 482–496. [Google Scholar] [CrossRef]
  42. Wakabayashi, H.; Nishiyama, Y.; Ushiyama, T.; Maeba, T.; Maeta, H. Evaluation of the effect of age on functioning hepatocyte mass and liver blood flow using liver scintigraphy in preoperative estimations for surgical patients: Comparison with CT volumetry. J. Surg. Res. 2002, 106, 246–253. [Google Scholar] [CrossRef]
  43. Belahnech, Y.; Rodenas-Alesina, E.; Munoz, M.A.; Verdu-Rotellar, J.M.; Sao-Aviles, A.; Urio-Garmendia, G.; Osorio, D.; Salas, K.; Pantoja, E.; Ribera, A.; et al. Systematic Coronary Risk Evaluation 2 for Older Persons (SCORE2-OP): 10 years risk validation, clinical utility and potential improvement. Eur. J. Prev. Cardiol. 2024, 32, 527–536. [Google Scholar] [CrossRef] [PubMed]
  44. Kouvari, M.; Valenzuela-Vallejo, L.; Guatibonza-Garcia, V.; Polyzos, S.A.; Deng, Y.; Kokkorakis, M.; Agraz, M.; Mylonakis, S.C.; Katsarou, A.; Verrastro, O.; et al. Liver biopsy-based validation, confirmation and comparison of the diagnostic performance of established and novel non-invasive steatotic liver disease indexes: Results from a large multi-center study. Metabolism 2023, 147, 155666. [Google Scholar] [CrossRef] [PubMed]
  45. Ratziu, V.; Charlotte, F.; Heurtier, A.; Gombert, S.; Giral, P.; Bruckert, E.; Grimaldi, A.; Capron, F.; Poynard, T.; Group, L.S. Sampling variability of liver biopsy in nonalcoholic fatty liver disease. Gastroenterology 2005, 128, 1898–1906. [Google Scholar] [CrossRef] [PubMed]
  46. Ooi, G.J.; Clouston, A.; Johari, Y.; Kemp, W.W.; Roberts, S.K.; Brown, W.A.; Burton, P.R. Evaluation of the histological variability of core and wedge biopsies in nonalcoholic fatty liver disease in bariatric surgical patients. Surg. Endosc. 2021, 35, 1210–1218. [Google Scholar] [CrossRef]
  47. Lund, J.; Saunders, C.L.; Edwards, D.; Mant, J. Anticoagulation trends in adults aged 65 years and over with atrial fibrillation: A cohort study. Open Heart 2021, e001737. [Google Scholar] [CrossRef]
  48. Navaratnarajah, A.; Jackson, S.H.D. The physiology of ageing. Medicine 2017, 45, 6–10. [Google Scholar] [CrossRef]
  49. Hatheway, O.L.; Mitnitski, A.; Rockwood, K. Frailty affects the initial treatment response and time to recovery of mobility in acutely ill older adults admitted to hospital. Age Ageing 2017, 46, 920–925. [Google Scholar] [CrossRef]
  50. Schwenzer, N.F.; Springer, F.; Schraml, C.; Stefan, N.; Machann, J.; Schick, F. Non-invasive assessment and quantification of liver steatosis by ultrasound, computed tomography and magnetic resonance. J. Hepatol. 2009, 51, 433–445. [Google Scholar] [CrossRef]
  51. Hernaez, R.; Lazo, M.; Bonekamp, S.; Kamel, I.; Brancati, F.L.; Guallar, E.; Clark, J.M. Diagnostic accuracy and reliability of ultrasonography for the detection of fatty liver: A meta-analysis. Hepatology 2011, 54, 1082–1090. [Google Scholar] [CrossRef]
  52. Lee, S.S.; Park, S.H. Radiologic evaluation of nonalcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 7392–7402. [Google Scholar] [CrossRef]
  53. Gu, J.; Liu, S.; Du, S.; Zhang, Q.; Xiao, J.; Dong, Q.; Xin, Y. Diagnostic value of MRI-PDFF for hepatic steatosis in patients with non-alcoholic fatty liver disease: A meta-analysis. Eur. Radiol. 2019, 29, 3564–3573. [Google Scholar] [CrossRef] [PubMed]
  54. Karlas, T.; Petroff, D.; Sasso, M.; Fan, J.G.; Mi, Y.Q.; de Ledinghen, V.; Kumar, M.; Lupsor-Platon, M.; Han, K.H.; Cardoso, A.C.; et al. Individual patient data meta-analysis of controlled attenuation parameter (CAP) technology for assessing steatosis. J. Hepatol. 2017, 66, 1022–1030. [Google Scholar] [CrossRef]
  55. Ferraioli, G.; Barr, R.G.; Berzigotti, A.; Sporea, I.; Wong, V.W.; Reiberger, T.; Karlas, T.; Thiele, M.; Cardoso, A.C.; Ayonrinde, O.T.; et al. WFUMB Guidelines/Guidance on Liver Multiparametric Ultrasound. Part 2: Guidance on Liver Fat Quantification. Ultrasound Med. Biol. 2024, 50, 1088–1098. [Google Scholar] [CrossRef] [PubMed]
  56. Bedogni, G.; Bellentani, S.; Miglioli, L.; Masutti, F.; Passalacqua, M.; Castiglione, A.; Tiribelli, C. The Fatty Liver Index: A simple and accurate predictor of hepatic steatosis in the general population. BMC Gastroenterol. 2006, 6, 33. [Google Scholar] [CrossRef] [PubMed]
  57. Long, M.T.; Pedley, A.; Colantonio, L.D.; Massaro, J.M.; Hoffmann, U.; Muntner, P.; Fox, C.S. Development and Validation of the Framingham Steatosis Index to Identify Persons with Hepatic Steatosis. Clin. Gastroenterol. Hepatol. 2016, 14, 1172–1180.e2. [Google Scholar] [CrossRef]
  58. McHenry, S.; Park, Y.; Browning, J.D.; Sayuk, G.; Davidson, N.O. Dallas Steatosis Index Identifies Patients with Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2020, 18, 2073–2080.e7. [Google Scholar] [CrossRef]
  59. Lee, J.H.; Kim, D.; Kim, H.J.; Lee, C.H.; Yang, J.I.; Kim, W.; Kim, Y.J.; Yoon, J.H.; Cho, S.H.; Sung, M.W.; et al. Hepatic steatosis index: A simple screening tool reflecting nonalcoholic fatty liver disease. Dig. Liver Dis. 2010, 42, 503–508. [Google Scholar] [CrossRef]
  60. Amato, M.C.; Giordano, C.; Galia, M.; Criscimanna, A.; Vitabile, S.; Midiri, M.; Galluzzo, A.; AlkaMeSy Study, G. Visceral Adiposity Index: A reliable indicator of visceral fat function associated with cardiometabolic risk. Diabetes Care 2010, 33, 920–922. [Google Scholar] [CrossRef]
  61. Wang, J.; Xu, C.; Xun, Y.; Lu, Z.; Shi, J.; Yu, C.; Li, Y. ZJU index: A novel model for predicting nonalcoholic fatty liver disease in a Chinese population. Sci. Rep. 2015, 5, 16494. [Google Scholar] [CrossRef]
  62. Koehler, E.M.; Schouten, J.N.; Hansen, B.E.; Hofman, A.; Stricker, B.H.; Janssen, H.L. External validation of the fatty liver index for identifying nonalcoholic fatty liver disease in a population-based study. Clin. Gastroenterol. Hepatol. 2013, 11, 1201–1204. [Google Scholar] [CrossRef]
  63. Clayton-Chubb, D.; Commins, I.; Roberts, S.K.; Majeed, A.; Woods, R.L.; Ryan, J.; Schneider, H.G.; Lubel, J.S.; Hodge, A.D.; McNeil, J.J.; et al. Scores to predict steatotic liver disease—Correlates and outcomes in older adults. NPJ Gut Liver 2025, 2, 9. [Google Scholar] [CrossRef]
  64. MAFLD Consensus Statement Working Group. Recommendations for the Assessment of Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD) in Primary Care: A Consensus Statement; Gastroenterological Society of Australia: Melbourne, Australia, 2024. Available online: https://www.gesa.org.au/public/13/files/Education%20%26%20Resources/Clinical%20Practice%20Resources/MAFLD/MAFLD%20consensus%20statement%202024.pdf (accessed on 1 June 2025).
  65. Winter, J.E.; MacInnis, R.J.; Wattanapenpaiboon, N.; Nowson, C.A. BMI and all-cause mortality in older adults: A meta-analysis. Am. J. Clin. Nutr. 2014, 99, 875–890. [Google Scholar] [CrossRef] [PubMed]
  66. Deurenberg, P.; Yap, M.; van Staveren, W.A. Body mass index and percent body fat: A meta analysis among different ethnic groups. Int. J. Obes. Relat. Metab. Disord. 1998, 22, 1164–1171. [Google Scholar] [CrossRef]
  67. Caleyachetty, R.; Barber, T.M.; Mohammed, N.I.; Cappuccio, F.P.; Hardy, R.; Mathur, R.; Banerjee, A.; Gill, P. Ethnicity-specific BMI cutoffs for obesity based on type 2 diabetes risk in England: A population-based cohort study. Lancet Diabetes Endocrinol. 2021, 9, 419–426. [Google Scholar] [CrossRef]
  68. Dong, M.H.; Bettencourt, R.; Brenner, D.A.; Barrett-Connor, E.; Loomba, R. Serum levels of alanine aminotransferase decrease with age in longitudinal analysis. Clin. Gastroenterol. Hepatol. 2012, 10, 285–290.e1. [Google Scholar] [CrossRef] [PubMed]
  69. Xiao, G.; Zhu, S.; Xiao, X.; Yan, L.; Yang, J.; Wu, G. Comparison of laboratory tests, ultrasound, or magnetic resonance elastography to detect fibrosis in patients with nonalcoholic fatty liver disease: A meta-analysis. Hepatology 2017, 66, 1486–1501. [Google Scholar] [CrossRef] [PubMed]
  70. Selvaraj, E.A.; Mozes, F.E.; Jayaswal, A.N.A.; Zafarmand, M.H.; Vali, Y.; Lee, J.A.; Levick, C.K.; Young, L.A.J.; Palaniyappan, N.; Liu, C.H.; et al. Diagnostic accuracy of elastography and magnetic resonance imaging in patients with NAFLD: A systematic review and meta-analysis. J. Hepatol. 2021, 75, 770–785. [Google Scholar] [CrossRef]
  71. Eddowes, P.J.; Sasso, M.; Allison, M.; Tsochatzis, E.; Anstee, Q.M.; Sheridan, D.; Guha, I.N.; Cobbold, J.F.; Deeks, J.J.; Paradis, V.; et al. Accuracy of FibroScan Controlled Attenuation Parameter and Liver Stiffness Measurement in Assessing Steatosis and Fibrosis in Patients with Nonalcoholic Fatty Liver Disease. Gastroenterology 2019, 156, 1717–1730. [Google Scholar] [CrossRef]
  72. Mozes, F.E.; Lee, J.A.; Selvaraj, E.A.; Jayaswal, A.N.A.; Trauner, M.; Boursier, J.; Fournier, C.; Staufer, K.; Stauber, R.E.; Bugianesi, E.; et al. Diagnostic accuracy of non-invasive tests for advanced fibrosis in patients with NAFLD: An individual patient data meta-analysis. Gut 2021, 71, 1006–1019. [Google Scholar] [CrossRef]
  73. Boursier, J.; Hagstrom, H.; Ekstedt, M.; Moreau, C.; Bonacci, M.; Cure, S.; Ampuero, J.; Nasr, P.; Tallab, L.; Canivet, C.M.; et al. Non-invasive tests accurately stratify patients with NAFLD based on their risk of liver-related events. J. Hepatol. 2022, 76, 1013–1020. [Google Scholar] [CrossRef]
  74. Vallet-Pichard, A.; Mallet, V.; Nalpas, B.; Verkarre, V.; Nalpas, A.; Dhalluin-Venier, V.; Fontaine, H.; Pol, S. FIB-4: An inexpensive and accurate marker of fibrosis in HCV infection. comparison with liver biopsy and fibrotest. Hepatology 2007, 46, 32–36. [Google Scholar] [CrossRef] [PubMed]
  75. Sun, W.; Cui, H.; Li, N.; Wei, Y.; Lai, S.; Yang, Y.; Yin, X.; Chen, D.F. Comparison of FIB-4 index, NAFLD fibrosis score and BARD score for prediction of advanced fibrosis in adult patients with non-alcoholic fatty liver disease: A meta-analysis study. Hepatol. Res. 2016, 46, 862–870. [Google Scholar] [CrossRef]
  76. Forlano, R.; Stanic, T.; Jayawardana, S.; Mullish, B.H.; Yee, M.; Mossialos, E.; Goldin, R.D.; Petta, S.; Tsochatzis, E.; Thursz, M.R.; et al. Derivation and validation of the BIMAST score for predicting the presence of fibrosis due to Metabolic dysfunction-associated steatotic liver disease among diabetic patients in the community. PLoS ONE 2024, 19, e0307500. [Google Scholar] [CrossRef]
  77. van Kleef, L.A.; Francque, S.M.; Prieto-Ortiz, J.E.; Sonneveld, M.J.; Sanchez-Luque, C.B.; Prieto-Ortiz, R.G.; Kwanten, W.J.; Vonghia, L.; Verrijken, A.; De Block, C.; et al. Metabolic Dysfunction-Associated Fibrosis 5 (MAF-5) Score Predicts Liver Fibrosis Risk and Outcome in the General Population with Metabolic Dysfunction. Gastroenterology 2024, 167, 357–367.e9. [Google Scholar] [CrossRef]
  78. Sripongpun, P.; Kim, W.R.; Mannalithara, A.; Charu, V.; Vidovszky, A.; Asch, S.; Desai, M.; Kim, S.H.; Kwong, A.J. The steatosis-associated fibrosis estimator (SAFE) score: A tool to detect low-risk NAFLD in primary care. Hepatology 2023, 77, 256–267. [Google Scholar] [CrossRef] [PubMed]
  79. Jung, J.; Loomba, R.R.; Imajo, K.; Madamba, E.; Gandhi, S.; Bettencourt, R.; Singh, S.; Hernandez, C.; Valasek, M.A.; Behling, C.; et al. MRE combined with FIB-4 (MEFIB) index in detection of candidates for pharmacological treatment of NASH-related fibrosis. Gut 2021, 70, 1946–1953. [Google Scholar] [CrossRef] [PubMed]
  80. Younossi, Z.M.; Felix, S.; Jeffers, T.; Younossi, E.; Nader, F.; Pham, H.; Afendy, A.; Cable, R.; Racila, A.; Younoszai, Z.; et al. Performance of the Enhanced Liver Fibrosis Test to Estimate Advanced Fibrosis Among Patients with Nonalcoholic Fatty Liver Disease. JAMA Netw. Open 2021, 4, e2123923. [Google Scholar] [CrossRef]
  81. McPherson, S.; Hardy, T.; Dufour, J.F.; Petta, S.; Romero-Gomez, M.; Allison, M.; Oliveira, C.P.; Francque, S.; Van Gaal, L.; Schattenberg, J.M.; et al. Age as a Confounding Factor for the Accurate Non-Invasive Diagnosis of Advanced NAFLD Fibrosis. Am. J. Gastroenterol. 2017, 112, 740–751. [Google Scholar] [CrossRef]
  82. Morimoto, K.; Takeuchi, Y.; Takaki, A.; Wada, N.; Oyama, A.; Adachi, T.; Onishi, H.; Shiraha, H.; Okada, H. Disease Progression-Related Markers for Aged Non-Alcoholic Fatty Liver Disease Patients. Acta Med. Okayama 2023, 77, 377–385. [Google Scholar] [CrossRef]
  83. van Kleef, L.A.; de Knegt, R.J.; Ayada, I.; Pan, Q.; Brouwer, W.P. The Steatosis-associated fibrosis estimator (SAFE) score: Validation in the general US population. Hepatol. Commun. 2023, 7, e0075. [Google Scholar] [CrossRef]
  84. Pitisuttithum, P.; Chan, W.K.; Piyachaturawat, P.; Imajo, K.; Nakajima, A.; Seki, Y.; Kasama, K.; Kakizaki, S.; Fan, J.G.; Song, M.J.; et al. Predictors of advanced fibrosis in elderly patients with biopsy-confirmed nonalcoholic fatty liver disease: The GOASIA study. BMC Gastroenterol. 2020, 20, 88. [Google Scholar] [CrossRef] [PubMed]
  85. Tamaki, N.; Takaura, K.; Higuchi, M.; Yasui, Y.; Itakura, J.; Tsuchiya, K.; Nakanishi, H.; Izumi, N.; Kurosaki, M. Enhanced Liver Fibrosis Score for Diagnosing Liver Fibrosis in Chronic Hepatitis. Diagnostics 2024, 14, 1317. [Google Scholar] [CrossRef] [PubMed]
  86. Dellavance, A.; Fernandes, F.; Shimabokuro, N.; Latini, F.; Baldo, D.; Barreto, J.A.; Marvulle, V.; Andrade, L.E.; Ferraz, M.L. Enhanced liver fibrosis (ELF) score: Analytical performance and distribution range in a large cohort of blood donors. Clin. Chim. Acta 2016, 461, 151–155. [Google Scholar] [CrossRef]
  87. Lichtinghagen, R.; Pietsch, D.; Bantel, H.; Manns, M.P.; Brand, K.; Bahr, M.J. The Enhanced Liver Fibrosis (ELF) score: Normal values, influence factors and proposed cut-off values. J. Hepatol. 2013, 59, 236–242. [Google Scholar] [CrossRef] [PubMed]
  88. Perier, C.; Granouillet, R.; Chamson, A.; Gonthier, R.; Frey, J. Nutritional markers, acute phase reactants and tissue inhibitor of matrix metalloproteinase 1 in elderly patients with pressure sores. Gerontology 2002, 48, 298–301. [Google Scholar] [CrossRef]
  89. Pettersson-Pablo, P.; Samyn, D.; Wasim, J.; Vink, M. Reference interval for type III procollagen (PIIINP) using the Advia centaur PIIINP assay in adults and elderly. Scand. J. Clin. Lab. Investig. 2021, 81, 649–652. [Google Scholar] [CrossRef]
  90. Balduini, C.L.; Noris, P. Platelet count and aging. Haematologica 2014, 99, 953–955. [Google Scholar] [CrossRef]
  91. Rinella, M.E.; Neuschwander-Tetri, B.A.; Siddiqui, M.S.; Abdelmalek, M.F.; Caldwell, S.; Barb, D.; Kleiner, D.E.; Loomba, R. AASLD Practice Guidance on the clinical assessment and management of nonalcoholic fatty liver disease. Hepatology 2023, 77, 1797–1835. [Google Scholar] [CrossRef]
  92. Sookoian, S.; Pirola, C.J. Genetic predisposition in nonalcoholic fatty liver disease. Clin. Mol. Hepatol. 2017, 23, 1–12. [Google Scholar] [CrossRef]
  93. Driessen, S.; Francque, S.M.; Anker, S.D.; Castro Cabezas, M.; Grobbee, D.E.; Tushuizen, M.E.; Holleboom, A.G. Metabolic dysfunction-associated steatotic liver disease and the heart. Hepatology 2023. [Google Scholar] [CrossRef]
  94. Mantovani, A.; Petracca, G.; Beatrice, G.; Csermely, A.; Tilg, H.; Byrne, C.D.; Targher, G. Non-alcoholic fatty liver disease and increased risk of incident extrahepatic cancers: A meta-analysis of observational cohort studies. Gut 2022, 71, 778–788. [Google Scholar] [CrossRef] [PubMed]
  95. Caussy, C.; Aubin, A.; Loomba, R. The Relationship Between Type 2 Diabetes, NAFLD, and Cardiovascular Risk. Curr. Diab. Rep. 2021, 21, 15. [Google Scholar] [CrossRef]
  96. Mantovani, A.; Petracca, G.; Beatrice, G.; Tilg, H.; Byrne, C.D.; Targher, G. Non-alcoholic fatty liver disease and risk of incident diabetes mellitus: An updated meta-analysis of 501 022 adult individuals. Gut 2021, 70, 962–969. [Google Scholar] [CrossRef] [PubMed]
  97. Zelber-Sagi, S.; Nitzan-Kaluski, D.; Goldsmith, R.; Webb, M.; Blendis, L.; Halpern, Z.; Oren, R. Long term nutritional intake and the risk for non-alcoholic fatty liver disease (NAFLD): A population based study. J. Hepatol. 2007, 47, 711–717. [Google Scholar] [CrossRef]
  98. Wehmeyer, M.H.; Zyriax, B.C.; Jagemann, B.; Roth, E.; Windler, E.; Schulze Zur Wiesch, J.; Lohse, A.W.; Kluwe, J. Nonalcoholic fatty liver disease is associated with excessive calorie intake rather than a distinctive dietary pattern. Medicine 2016, 95, e3887. [Google Scholar] [CrossRef] [PubMed]
  99. Zelber-Sagi, S.; Ivancovsky-Wajcman, D.; Fliss Isakov, N.; Webb, M.; Orenstein, D.; Shibolet, O.; Kariv, R. High red and processed meat consumption is associated with non-alcoholic fatty liver disease and insulin resistance. J. Hepatol. 2018, 68, 1239–1246. [Google Scholar] [CrossRef]
  100. Chen, H.; Wang, J.; Li, Z.; Lam, C.W.K.; Xiao, Y.; Wu, Q.; Zhang, W. Consumption of Sugar-Sweetened Beverages Has a Dose-Dependent Effect on the Risk of Non-Alcoholic Fatty Liver Disease: An Updated Systematic Review and Dose-Response Meta-Analysis. Int. J. Environ. Res. Public Health 2019, 16, 2192. [Google Scholar] [CrossRef]
  101. Zelber-Sagi, S.; Nitzan-Kaluski, D.; Goldsmith, R.; Webb, M.; Zvibel, I.; Goldiner, I.; Blendis, L.; Halpern, Z.; Oren, R. Role of leisure-time physical activity in nonalcoholic fatty liver disease: A population-based study. Hepatology 2008, 48, 1791–1798. [Google Scholar] [CrossRef]
  102. Gerber, L.; Otgonsuren, M.; Mishra, A.; Escheik, C.; Birerdinc, A.; Stepanova, M.; Younossi, Z.M. Non-alcoholic fatty liver disease (NAFLD) is associated with low level of physical activity: A population-based study. Aliment. Pharmacol. Ther. 2012, 36, 772–781. [Google Scholar] [CrossRef]
  103. Bowden Davies, K.A.; Sprung, V.S.; Norman, J.A.; Thompson, A.; Mitchell, K.L.; Harrold, J.O.A.; Finlayson, G.; Gibbons, C.; Wilding, J.P.H.; Kemp, G.J.; et al. Physical Activity and Sedentary Time: Association with Metabolic Health and Liver Fat. Med. Sci. Sports Exerc. 2019, 51, 1169–1177. [Google Scholar] [CrossRef]
  104. Lonardo, A.; Ballestri, S.; Marchesini, G.; Angulo, P.; Loria, P. Nonalcoholic fatty liver disease: A precursor of the metabolic syndrome. Dig. Liver Dis. 2015, 47, 181–190. [Google Scholar] [CrossRef] [PubMed]
  105. 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. [Google Scholar] [CrossRef]
  106. Fabbrini, E.; Sullivan, S.; Klein, S. Obesity and nonalcoholic fatty liver disease: Biochemical, metabolic, and clinical implications. Hepatology 2010, 51, 679–689. [Google Scholar] [CrossRef] [PubMed]
  107. Cusi, K. Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: Pathophysiology and clinical implications. Gastroenterology 2012, 142, 711–725.e6. [Google Scholar] [CrossRef]
  108. He, X.; Zhang, S.; Bai, Q.; Pan, M.; Jiang, Y.; Liu, W.; Li, W.; Gong, Y.; Li, X. Air pollution exposure and prevalence of non-alcoholic fatty liver disease and related cirrhosis: A systematic review and meta-analysis. Ecotoxicol. Environ. Saf. 2025, 289, 117469. [Google Scholar] [CrossRef]
  109. Ju, S.Y.; Lee, J.Y.; Kim, D.H. Association of metabolic syndrome and its components with all-cause and cardiovascular mortality in the elderly: A meta-analysis of prospective cohort studies. Medicine 2017, 96, e8491. [Google Scholar] [CrossRef] [PubMed]
  110. Sun, F.; Norman, I.J.; While, A.E. Physical activity in older people: A systematic review. BMC Public Health 2013, 13, 449. [Google Scholar] [CrossRef]
  111. Long, T.; Zhang, K.; Chen, Y.; Wu, C. Trends in Diet Quality Among Older US Adults From 2001 to 2018. JAMA Netw. Open 2022, 5, e221880. [Google Scholar] [CrossRef] [PubMed]
  112. Govindaraju, T.; Owen, A.J.; McCaffrey, T.A. Past, present and future influences of diet among older adults—A scoping review. Ageing Res. Rev. 2022, 77, 101600. [Google Scholar] [CrossRef]
  113. Crombie, I.K.; Irvine, L.; Williams, B.; McGinnis, A.R.; Slane, P.W.; Alder, E.M.; McMurdo, M.E. Why older people do not participate in leisure time physical activity: A survey of activity levels, beliefs and deterrents. Age Ageing 2004, 33, 287–292. [Google Scholar] [CrossRef]
  114. Costello, E.; Kafchinski, M.; Vrazel, J.; Sullivan, P. Motivators, barriers, and beliefs regarding physical activity in an older adult population. J. Geriatr. Phys. Ther. 2011, 34, 138–147. [Google Scholar] [CrossRef]
  115. Rai, R.; Jongenelis, M.I.; Jackson, B.; Newton, R.U.; Pettigrew, S. Factors influencing physical activity participation among older people with low activity levels. Ageing Soc. 2019, 40, 2593–2613. [Google Scholar] [CrossRef]
  116. Marmot, M.; Bell, R. Social determinants and non-communicable diseases: Time for integrated action. BMJ 2019, 364, l251. [Google Scholar] [CrossRef] [PubMed]
  117. McNamara, C.L.; Balaj, M.; Thomson, K.H.; Eikemo, T.A.; Solheim, E.F.; Bambra, C. The socioeconomic distribution of non-communicable diseases in Europe: Findings from the European Social Survey (2014) special module on the social determinants of health. Eur. J. Public Health 2017, 27 (Suppl. S1), 22–26. [Google Scholar] [CrossRef] [PubMed]
  118. Walker, R.J.; Strom Williams, J.; Egede, L.E. Influence of Race, Ethnicity and Social Determinants of Health on Diabetes Outcomes. Am. J. Med. Sci. 2016, 351, 366–373. [Google Scholar] [CrossRef] [PubMed]
  119. Gracey, M.; King, M. Indigenous health part 1: Determinants and disease patterns. Lancet 2009, 374, 65–75. [Google Scholar] [CrossRef]
  120. Smith, K.B.; Humphreys, J.S.; Wilson, M.G. Addressing the health disadvantage of rural populations: How does epidemiological evidence inform rural health policies and research? Aust. J. Rural. Health 2008, 16, 56–66. [Google Scholar] [CrossRef]
  121. Flavel, J.; Kedzior, S.G.; Isaac, V.; Cameron, D.; Baum, F. Regional health inequalities in Australia and social determinants of health: Analysis of trends and distribution by remoteness. Rural. Remote Health 2024, 24, 7726. [Google Scholar] [CrossRef]
  122. Czapla, B.C.; Dalvi, A.; Hu, J.; Moran, I.J.; Wijarnpreecha, K.; Chen, V.L. Physical activity, diet, and social determinants of health associate with health related quality of life and fibrosis in MASLD. Sci. Rep. 2025, 15, 7976. [Google Scholar] [CrossRef]
  123. Rich, N.E.; Oji, S.; Mufti, A.R.; Browning, J.D.; Parikh, N.D.; Odewole, M.; Mayo, H.; Singal, A.G. Racial and Ethnic Disparities in Nonalcoholic Fatty Liver Disease Prevalence, Severity, and Outcomes in the United States: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2018, 16, 198–210.e2. [Google Scholar] [CrossRef]
  124. Clayton-Chubb, D.; Kemp, W.W.; Majeed, A.; Lubel, J.S.; Woods, R.L.; Tran, C.; Ryan, J.; Hodge, A.; Schneider, H.G.; McNeil, J.J.; et al. Metabolic dysfunction-associated steatotic liver disease in older adults is associated with frailty and social disadvantage. Liver Int. 2024, 44, 39–51. [Google Scholar] [CrossRef] [PubMed]
  125. Kim, R.G.; Ballantyne, A.; Conroy, M.B.; Price, J.C.; Inadomi, J.M. Screening for social determinants of health among populations at risk for MASLD: A scoping review. Front. Public Health 2024, 12, 1332870. [Google Scholar] [CrossRef]
  126. Mantovani, A.; Csermely, A.; Petracca, G.; Beatrice, G.; Corey, K.E.; Simon, T.G.; Byrne, C.D.; Targher, G. Non-alcoholic fatty liver disease and risk of fatal and non-fatal cardiovascular events: An updated systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2021, 6, 903–913. [Google Scholar] [CrossRef]
  127. Duell, P.B.; Welty, F.K.; Miller, M.; Chait, A.; Hammond, G.; Ahmad, Z.; Cohen, D.E.; Horton, J.D.; Pressman, G.S.; Toth, P.P.; et al. Nonalcoholic Fatty Liver Disease and Cardiovascular Risk: A Scientific Statement From the American Heart Association. Arterioscler. Thromb. Vasc. Biol. 2022, 42, e168–e185. [Google Scholar] [CrossRef] [PubMed]
  128. Clayton-Chubb, D.; Roberts, S.K.; Majeed, A.; Woods, R.L.; Tonkin, A.M.; Nelson, M.R.; Chan, A.T.; Ryan, J.; Tran, C.; Hodge, A.; et al. Associations between MASLD, atrial fibrillation, cardiovascular events, mortality and aspirin use in older adults. Geroscience 2024, 47, 1303–1318. [Google Scholar] [CrossRef] [PubMed]
  129. Neumann, J.T.; Thao, L.T.P.; Callander, E.; Chowdhury, E.; Williamson, J.D.; Nelson, M.R.; Donnan, G.; Woods, R.L.; Reid, C.M.; Poppe, K.K.; et al. Cardiovascular risk prediction in healthy older people. Geroscience 2022, 44, 403–413. [Google Scholar] [CrossRef]
  130. Liu, Y.; Zhong, G.C.; Tan, H.Y.; Hao, F.B.; Hu, J.J. Nonalcoholic fatty liver disease and mortality from all causes, cardiovascular disease, and cancer: A meta-analysis. Sci. Rep. 2019, 9, 11124. [Google Scholar] [CrossRef]
  131. Simons, N.; Isaacs, A.; Koek, G.H.; Kuč, S.; Schaper, N.C.; Brouwers, M. PNPLA3, TM6SF2, and MBOAT7 Genotypes and Coronary Artery Disease. Gastroenterology 2017, 152, 912–913. [Google Scholar] [CrossRef]
  132. van Kleef, L.A.; Lu, Z.; Ikram, M.A.; de Groot, N.M.S.; Kavousi, M.; de Knegt, R.J. Liver stiffness not fatty liver disease is associated with atrial fibrillation: The Rotterdam study. J. Hepatol. 2022, 77, 931–938. [Google Scholar] [CrossRef]
  133. Mantovani, A.; Dauriz, M.; Sandri, D.; Bonapace, S.; Zoppini, G.; Tilg, H.; Byrne, C.D.; Targher, G. Association between non-alcoholic fatty liver disease and risk of atrial fibrillation in adult individuals: An updated meta-analysis. Liver Int. 2019, 39, 758–769. [Google Scholar] [CrossRef]
  134. Simon, T.G.; Roelstraete, B.; Khalili, H.; Hagstrom, H.; Ludvigsson, J.F. Mortality in biopsy-confirmed nonalcoholic fatty liver disease: Results from a nationwide cohort. Gut 2021, 70, 1375–1382. [Google Scholar] [CrossRef]
  135. Arase, Y.; Kobayashi, M.; Suzuki, F.; Suzuki, Y.; Kawamura, Y.; Akuta, N.; Imai, N.; Kobayashi, M.; Sezaki, H.; Matsumoto, N.; et al. Difference in malignancies of chronic liver disease due to non-alcoholic fatty liver disease or hepatitis C in Japanese elderly patients. Hepatol. Res. 2012, 42, 264–272. [Google Scholar] [CrossRef]
  136. Shaukat, A.; Kahi, C.J.; Burke, C.A.; Rabeneck, L.; Sauer, B.G.; Rex, D.K. ACG Clinical Guidelines: Colorectal Cancer Screening 2021. Am. J. Gastroenterol. 2021, 116, 458–479. [Google Scholar] [CrossRef] [PubMed]
  137. Issaka, R.B.; Chan, A.T.; Gupta, S. AGA Clinical Practice Update on Risk Stratification for Colorectal Cancer Screening and Post-Polypectomy Surveillance: Expert Review. Gastroenterology 2023, 165, 1280–1291. [Google Scholar] [CrossRef] [PubMed]
  138. Clayton-Chubb, D.; Kemp, W.W.; Majeed, A.; Woods, R.L.; Ryan, J.; Murray, A.M.; Chong, T.T.J.; Lubel, J.S.; Tran, C.; Hodge, A.D.; et al. Late-Life Metabolic Dysfunction-Associated Steatotic Liver Disease and its Association with Physical Disability and Dementia. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glae011. [Google Scholar] [CrossRef]
  139. Oduwole, O.; Ding, C.; Bitar, N.; Nair, D.; Salter, S.; Silverman, M.; Allen, R.; Ng Fat, L.; Tsochatzis, E.; Bell, S.; et al. Steatotic liver disease is a marker of multimorbidity, not underlying cirrhosis, in older adults. NPJ Gut Liver 2025, 2, 10. [Google Scholar] [CrossRef] [PubMed]
  140. Tursi, A.; Scarpignato, C.; Strate, L.L.; Lanas, A.; Kruis, W.; Lahat, A.; Danese, S. Colonic diverticular disease. Nat. Rev. Dis. Primers 2020, 6, 20. [Google Scholar] [CrossRef]
  141. Everhart, J.E.; Ruhl, C.E. Burden of digestive diseases in the United States part II: Lower gastrointestinal diseases. Gastroenterology 2009, 136, 741–754. [Google Scholar] [CrossRef]
  142. Milovanovic, T.; Pantic, I.; Dragasevic, S.; Lugonja, S.; Dumic, I.; Rajilic-Stojanovic, M. The Interrelationship Among Non-Alcoholic Fatty Liver Disease, Colonic Diverticulosis and Metabolic Syndrome. J. Gastrointestin. Liver Dis. 2021, 30, 274–282. [Google Scholar] [CrossRef]
  143. Pantic, I.; Lugonja, S.; Rajovic, N.; Dumic, I.; Milovanovic, T. Colonic Diverticulosis and Non-Alcoholic Fatty Liver Disease: Is There a Connection? Medicina 2021, 58, 38. [Google Scholar] [CrossRef]
  144. Nagata, N.; Sakamoto, K.; Arai, T.; Niikura, R.; Shimbo, T.; Shinozaki, M.; Aoki, T.; Sekine, K.; Okubo, H.; Watanabe, K.; et al. Visceral Abdominal Obesity Measured by Computed Tomography is Associated with Increased Risk of Colonic Diverticulosis. J. Clin. Gastroenterol. 2015, 49, 816–822. [Google Scholar] [CrossRef] [PubMed]
  145. Kim, D.; Chung, G.E.; Kwak, M.S.; Kim, Y.J.; Yoon, J.H. Effect of longitudinal changes of body fat on the incidence and regression of nonalcoholic fatty liver disease. Dig. Liver Dis. 2018, 50, 389–395. [Google Scholar] [CrossRef] [PubMed]
  146. Clayton-Chubb, D.; Kemp, W.; Majeed, A.; Lubel, J.S.; Hodge, A.; Roberts, S.K. Understanding NAFLD: From Case Identification to Interventions, Outcomes, and Future Perspectives. Nutrients 2023, 15, 687. [Google Scholar] [CrossRef]
  147. Aron-Wisnewsky, J.; Vigliotti, C.; Witjes, J.; Le, P.; Holleboom, A.G.; Verheij, J.; Nieuwdorp, M.; Clement, K. Gut microbiota and human NAFLD: Disentangling microbial signatures from metabolic disorders. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 279–297. [Google Scholar] [CrossRef] [PubMed]
  148. Gupta, H.; Min, B.H.; Ganesan, R.; Gebru, Y.A.; Sharma, S.P.; Park, E.; Won, S.M.; Jeong, J.J.; Lee, S.B.; Cha, M.G.; et al. Gut Microbiome in Non-Alcoholic Fatty Liver Disease: From Mechanisms to Therapeutic Role. Biomedicines 2022, 10, 550. [Google Scholar] [CrossRef]
  149. Kline, K.A.; Bowdish, D.M. Infection in an aging population. Curr. Opin. Microbiol. 2016, 29, 63–67. [Google Scholar] [CrossRef]
  150. Dorrington, M.G.; Bowdish, D.M. Immunosenescence and novel vaccination strategies for the elderly. Front. Immunol. 2013, 4, 171. [Google Scholar] [CrossRef]
  151. Aberle, J.H.; Stiasny, K.; Kundi, M.; Heinz, F.X. Mechanistic insights into the impairment of memory B cells and antibody production in the elderly. Age 2013, 35, 371–381. [Google Scholar] [CrossRef]
  152. Mittelbrunn, M.; Kroemer, G. Hallmarks of T cell aging. Nat. Immunol. 2021, 22, 687–698. [Google Scholar] [CrossRef]
  153. Mantovani, A.; Morandin, R.; Fiorio, V.; Lando, M.G.; Gaviraghi, A.; Motta, L.; Gobbi, F.; Tilg, H.; Byrne, C.D.; Targher, G. Association between MASLD and increased risk of serious bacterial infections requiring hospital admission: A meta-analysis. Liver Int. 2025, 45, e16101. [Google Scholar] [CrossRef]
  154. Ebrahimi, F.; Simon, T.G.; Hagstrom, H.; Soderling, J.; Wester, A.; Roelstraete, B.; Ludvigsson, J.F. Risk of Severe Infection in Patients with Biopsy-proven Nonalcoholic Fatty Liver Disease—A Population-based Cohort Study. Clin. Gastroenterol. Hepatol. 2023, 21, 3346–3355.e19. [Google Scholar] [CrossRef] [PubMed]
  155. Krznaric, J.; Vince, A. The Role of Non-Alcoholic Fatty Liver Disease in Infections. Life 2022, 12, 2052. [Google Scholar] [CrossRef] [PubMed]
  156. Benede-Ubieto, R.; Cubero, F.J.; Nevzorova, Y.A. Breaking the barriers: The role of gut homeostasis in Metabolic-Associated Steatotic Liver Disease (MASLD). Gut Microbes 2024, 16, 2331460. [Google Scholar] [CrossRef]
  157. Andersen, C.J.; Murphy, K.E.; Fernandez, M.L. Impact of Obesity and Metabolic Syndrome on Immunity. Adv. Nutr. 2016, 7, 66–75. [Google Scholar] [CrossRef]
  158. Ma, C.; Kesarwala, A.H.; Eggert, T.; Medina-Echeverz, J.; Kleiner, D.E.; Jin, P.; Stroncek, D.F.; Terabe, M.; Kapoor, V.; ElGindi, M.; et al. NAFLD causes selective CD4+ T lymphocyte loss and promotes hepatocarcinogenesis. Nature 2016, 531, 253–257. [Google Scholar] [CrossRef]
  159. Tilg, H.; Adolph, T.E.; Dudek, M.; Knolle, P. Non-alcoholic fatty liver disease: The interplay between metabolism, microbes and immunity. Nat. Metab. 2021, 3, 1596–1607. [Google Scholar] [CrossRef]
  160. Sim, B.C.; Kang, Y.E.; You, S.K.; Lee, S.E.; Nga, H.T.; Lee, H.Y.; Nguyen, T.L.; Moon, J.S.; Tian, J.; Jang, H.J.; et al. Hepatic T-cell senescence and exhaustion are implicated in the progression of fatty liver disease in patients with type 2 diabetes and mouse model with nonalcoholic steatohepatitis. Cell Death Dis. 2023, 14, 618. [Google Scholar] [CrossRef] [PubMed]
  161. Spinelli, R.; Baboota, R.K.; Gogg, S.; Beguinot, F.; Bluher, M.; Nerstedt, A.; Smith, U. Increased cell senescence in human metabolic disorders. J. Clin. Investig. 2023, e169922. [Google Scholar] [CrossRef] [PubMed]
  162. Ebadi, M.; Burra, P.; Zanetto, A.; Montano-Loza, A.J. Current treatment strategies and future possibilities for sarcopenia in cirrhosis. J. Hepatol. 2023, 78, 889–892. [Google Scholar] [CrossRef]
  163. Sinclair, M.; Poltavskiy, E.; Dodge, J.L.; Lai, J.C. Frailty is independently associated with increased hospitalisation days in patients on the liver transplant waitlist. World J. Gastroenterol. 2017, 23, 899–905. [Google Scholar] [CrossRef]
  164. Lai, J.C.; Dodge, J.L.; Kappus, M.R.; Dunn, M.A.; Volk, M.L.; Duarte-Rojo, A.; Ganger, D.R.; Rahimi, R.S.; McCulloch, C.E.; Haugen, C.E.; et al. Changes in frailty are associated with waitlist mortality in patients with cirrhosis. J. Hepatol. 2020, 73, 575–581. [Google Scholar] [CrossRef] [PubMed]
  165. Zhong, Q.; Zhou, R.; Huang, Y.N.; Huang, R.D.; Li, F.R.; Chen, H.W.; Wei, Y.F.; Liu, K.; Cao, B.F.; Liao, K.Y.; et al. Frailty and risk of metabolic dysfunction-associated steatotic liver disease and other chronic liver diseases. J. Hepatol. 2025, 82, 427–437. [Google Scholar] [CrossRef] [PubMed]
  166. Khanmohammadi, S.; Masrour, M.; Fallahtafti, P.; Habibzadeh, A.; Schuermans, A.; Kuchay, M.S. The relationship between nonalcoholic fatty liver disease and frailty: A systematic review and meta-analysis. Diabetes Metab. Syndr. 2025, 19, 103187. [Google Scholar] [CrossRef]
  167. Naimimohasses, S.; O’Gorman, P.; McCormick, E.; Ferguson, D.; Monaghan, A.; McGrath, M.; Robinson, M.W.; Gormley, J.; Norris, S. Prevalence of frailty in patients with non-cirrhotic non-alcoholic fatty liver disease. BMJ Open Gastroenterol. 2022, 9, e000861. [Google Scholar] [CrossRef]
  168. Geng, C.; Gao, P.; Tang, Y. Associations of Metabolic Dysfunction-Related Fatty Liver Disease and Dementia risk: A Prospective Study Based on the UK Biobank. Arch. Gerontol. Geriatr. 2025, 135, 105845. [Google Scholar] [CrossRef]
  169. George, E.S.; Sood, S.; Daly, R.M.; Tan, S.Y. Is there an association between non-alcoholic fatty liver disease and cognitive function? A systematic review. BMC Geriatr. 2022, 22, 47. [Google Scholar] [CrossRef] [PubMed]
  170. Weinstein, G.; Zelber-Sagi, S.; Preis, S.R.; Beiser, A.S.; DeCarli, C.; Speliotes, E.K.; Satizabal, C.L.; Vasan, R.S.; Seshadri, S. Association of Nonalcoholic Fatty Liver Disease with Lower Brain Volume in Healthy Middle-aged Adults in the Framingham Study. JAMA Neurol. 2018, 75, 97–104. [Google Scholar] [CrossRef]
  171. Shang, Y.; Widman, L.; Hagstrom, H. Nonalcoholic Fatty Liver Disease and Risk of Dementia: A Population-Based Cohort Study. Neurology 2022, 99, e574–e582. [Google Scholar] [CrossRef]
  172. Huang, H.; Liu, Z.; Xie, J.; Xu, C. NAFLD does not increase the risk of incident dementia: A prospective study and meta-analysis. J. Psychiatr. Res. 2023, 161, 435–440. [Google Scholar] [CrossRef]
  173. Xiao, T.; van Kleef, L.A.; Ikram, M.K.; de Knegt, R.J.; Ikram, M.A. Association of Nonalcoholic Fatty Liver Disease and Fibrosis with Incident Dementia and Cognition: The Rotterdam Study. Neurology 2022, 99, e565–e573. [Google Scholar] [CrossRef]
  174. Johnson, D.K.; Wilkins, C.H.; Morris, J.C. Accelerated weight loss may precede diagnosis in Alzheimer disease. Arch. Neurol. 2006, 63, 1312–1317. [Google Scholar] [CrossRef]
  175. Kivimaki, M.; Luukkonen, R.; Batty, G.D.; Ferrie, J.E.; Pentti, J.; Nyberg, S.T.; Shipley, M.J.; Alfredsson, L.; Fransson, E.I.; Goldberg, M.; et al. Body mass index and risk of dementia: Analysis of individual-level data from 1.3 million individuals. Alzheimers Dement. 2018, 14, 601–609. [Google Scholar] [CrossRef] [PubMed]
  176. Watson, R.; Sanson-Fisher, R.; Bryant, J.; Mansfield, E. Dementia is the second most feared condition among Australian health service consumers: Results of a cross-sectional survey. BMC Public Health 2023, 23, 876. [Google Scholar] [CrossRef] [PubMed]
  177. Tang, W.; Kannaley, K.; Friedman, D.B.; Edwards, V.J.; Wilcox, S.; Levkoff, S.E.; Hunter, R.H.; Irmiter, C.; Belza, B. Concern about developing Alzheimer’s disease or dementia and intention to be screened: An analysis of national survey data. Arch. Gerontol. Geriatr. 2017, 71, 43–49. [Google Scholar] [CrossRef] [PubMed]
  178. Parker, M.; Barlow, S.; Hoe, J.; Aitken, L. Persistent barriers and facilitators to seeking help for a dementia diagnosis: A systematic review of 30 years of the perspectives of carers and people with dementia. Int. Psychogeriatr. 2020, 32, 611–634. [Google Scholar] [CrossRef]
  179. Milne, A. Dementia screening and early diagnosis: The case for and against. Health Risk Soc. 2010, 12, 65–76. [Google Scholar] [CrossRef]
  180. Martin, S.; Kelly, S.; Khan, A.; Cullum, S.; Dening, T.; Rait, G.; Fox, C.; Katona, C.; Cosco, T.; Brayne, C.; et al. Attitudes and preferences towards screening for dementia: A systematic review of the literature. BMC Geriatr. 2015, 15, 66. [Google Scholar] [CrossRef]
  181. Hou, X.H.; Feng, L.; Zhang, C.; Cao, X.P.; Tan, L.; Yu, J.T. Models for predicting risk of dementia: A systematic review. J. Neurol. Neurosurg. Psychiatry 2019, 90, 373–379. [Google Scholar] [CrossRef]
  182. Dunn, W.; Xu, R.; Wingard, D.L.; Rogers, C.; Angulo, P.; Younossi, Z.M.; Schwimmer, J.B. Suspected nonalcoholic fatty liver disease and mortality risk in a population-based cohort study. Am. J. Gastroenterol. 2008, 103, 2263–2271. [Google Scholar] [CrossRef]
  183. 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. [Google Scholar] [CrossRef]
  184. van Kleef, L.A.; Sonneveld, M.J.; Kavousi, M.; Ikram, M.A.; de Man, R.A.; de Knegt, R.J. Fatty liver disease is not associated with increased mortality in the elderly: A prospective cohort study. Hepatology 2023, 77, 585–593. [Google Scholar] [CrossRef] [PubMed]
  185. Golabi, P.; Paik, J.; Reddy, R.; Bugianesi, E.; Trimble, G.; Younossi, Z.M. Prevalence and long-term outcomes of non-alcoholic fatty liver disease among elderly individuals from the United States. BMC Gastroenterol. 2019, 19, 56. [Google Scholar] [CrossRef]
  186. Ikram, M.A.; Kieboom, B.C.T.; Brouwer, W.P.; Brusselle, G.; Chaker, L.; Ghanbari, M.; Goedegebure, A.; Ikram, M.K.; Kavousi, M.; de Knegt, R.J.; et al. The Rotterdam Study. Design update and major findings between 2020 and 2024. Eur. J. Epidemiol. 2024, 39, 183–206. [Google Scholar] [CrossRef] [PubMed]
  187. Gellert-Kristensen, H.; Tybjaerg-Hansen, A.; Nordestgaard, B.G.; Ghouse, J.; Fuchs, A.; Kuhl, J.T.; Sigvardsen, P.E.; Kofoed, K.F.; Stender, S. Genetic risk of fatty liver disease and mortality in the general population: A Mendelian randomization study. Liver Int. 2023, 43, 1955–1965. [Google Scholar] [CrossRef]
  188. Kim, H.Y.; Rinella, M.E. Emerging Therapies and Real-World Application of Metabolic Dysfunction-Associated Steatotic Liver Disease Treatment. Clin. Mol. Hepatol. 2025. [Google Scholar] [CrossRef]
  189. Keam, S.J. Resmetirom: First Approval. Drugs 2024, 84, 729–735. [Google Scholar] [CrossRef] [PubMed]
  190. Dobbie, L.J.; Burgess, J.; Hamid, A.; Nevitt, S.J.; Hydes, T.J.; Alam, U.; Cuthbertson, D.J. Effect of a Low-Calorie Dietary Intervention on Liver Health and Body Weight in Adults with Metabolic-Dysfunction Associated Steatotic Liver Disease (MASLD) and Overweight/Obesity: A Systematic Review and Meta-Analysis. Nutrients 2024, 16, 1030. [Google Scholar] [CrossRef]
  191. Marin-Alejandre, B.A.; Cantero, I.; Perez-Diaz-Del-Campo, N.; Monreal, J.I.; Elorz, M.; Herrero, J.I.; Benito-Boillos, A.; Quiroga, J.; Martinez-Echeverria, A.; Uriz-Otano, J.I.; et al. Effects of two personalized dietary strategies during a 2-year intervention in subjects with nonalcoholic fatty liver disease: A randomized trial. Liver Int. 2021, 41, 1532–1544. [Google Scholar] [CrossRef]
  192. Wei, X.; Lin, B.; Huang, Y.; Yang, S.; Huang, C.; Shi, L.; Liu, D.; Zhang, P.; Lin, J.; Xu, B.; et al. Effects of Time-Restricted Eating on Nonalcoholic Fatty Liver Disease: The TREATY-FLD Randomized Clinical Trial. JAMA Netw. Open 2023, 6, e233513. [Google Scholar] [CrossRef]
  193. Machado, A.M.; Guimaraes, N.S.; Bocardi, V.B.; da Silva, T.P.R.; Carmo, A.S.D.; Menezes, M.C.; Duarte, C.K. Understanding weight regain after a nutritional weight loss intervention: Systematic review and meta-analysis. Clin. Nutr. ESPEN 2022, 49, 138–153. [Google Scholar] [CrossRef]
  194. Brodosi, L.; Stecchi, M.; Marchignoli, F.; Lucia, E.; Magnani, L.; Guarneri, V.; Petroni, M.L.; Marchesini, G.; Pironi, L. Risk of binge eating disorder in patients with metabolic dysfunction-associated steatotic liver disease. Eat. Weight. Disord. 2023, 28, 100. [Google Scholar] [CrossRef] [PubMed]
  195. Hussain, S.M.; Newman, A.B.; Beilin, L.J.; Tonkin, A.M.; Woods, R.L.; Neumann, J.T.; Nelson, M.; Carr, P.R.; Reid, C.M.; Owen, A.; et al. Associations of Change in Body Size with All-Cause and Cause-Specific Mortality Among Healthy Older Adults. JAMA Netw. Open 2023, 6, e237482. [Google Scholar] [CrossRef] [PubMed]
  196. Keys, A.; Keys, M. How to Eat Well and Stay Well the Mediterranean Way; Doubleday: New York, NY, USA, 1975. [Google Scholar]
  197. Giuffre, D.; Giuffre, A.M. Mediterranean diet and health in the elderly. AIMS Public Health 2023, 10, 568–576. [Google Scholar] [CrossRef]
  198. Hassani Zadeh, S.; Mansoori, A.; Hosseinzadeh, M. Relationship between dietary patterns and non-alcoholic fatty liver disease: A systematic review and meta-analysis. J. Gastroenterol. Hepatol. 2021, 36, 1470–1478. [Google Scholar] [CrossRef]
  199. Commins, I.; Clayton-Chubb, D.; Fitzpatrick, J.A.; George, E.S.; Schneider, H.G.; Phyo, A.Z.Z.; Majeed, A.; Janko, N.; Vaughan, N.; Woods, R.L.; et al. Associations Between MASLD, Ultra-Processed Food and a Mediterranean Dietary Pattern in Older Adults. Nutrients 2025, 17, 1415. [Google Scholar] [CrossRef]
  200. Gepner, Y.; Shelef, I.; Komy, O.; Cohen, N.; Schwarzfuchs, D.; Bril, N.; Rein, M.; Serfaty, D.; Kenigsbuch, S.; Zelicha, H.; et al. The beneficial effects of Mediterranean diet over low-fat diet may be mediated by decreasing hepatic fat content. J. Hepatol. 2019, 71, 379–388. [Google Scholar] [CrossRef]
  201. Properzi, C.; O’Sullivan, T.A.; Sherriff, J.L.; Ching, H.L.; Jeffrey, G.P.; Buckley, R.F.; Tibballs, J.; MacQuillan, G.C.; Garas, G.; Adams, L.A. Ad Libitum Mediterranean and Low-Fat Diets both Significantly Reduce Hepatic Steatosis: A Randomized Controlled Trial. Hepatology 2018, 68, 1741–1754. [Google Scholar] [CrossRef]
  202. Yaskolka Meir, A.; Rinott, E.; Tsaban, G.; Zelicha, H.; Kaplan, A.; Rosen, P.; Shelef, I.; Youngster, I.; Shalev, A.; Bluher, M.; et al. Effect of green-Mediterranean diet on intrahepatic fat: The DIRECT PLUS randomised controlled trial. Gut 2021, 70, 2085–2095. [Google Scholar] [CrossRef]
  203. Cueto-Galán, R.; Fontalba-Navas, A.; Gutiérrez-Bedmar, M.; Ruiz-Canela, M.; Martínez-González, M.A.; Alves, L.; Babio, N.; Fitó, M.; Ros, E.; Fiol, M.; et al. Adherence to the Mediterranean diet to prevent or delay hepatic steatosis: A longitudinal analysis within the PREDIMED study. Front. Nutr. 2025, 12, 1518082. [Google Scholar] [CrossRef] [PubMed]
  204. Oh, J.H.; Yoon, E.L.; Park, H.; Lee, S.; Jo, A.J.; Cho, S.; Kwon, E.; Nah, E.H.; Lee, J.H.; Park, J.H.; et al. Efficacy and safety of time-restricted eating in metabolic dysfunction-associated steatotic liver disease. J. Hepatol. 2025. [Google Scholar] [CrossRef]
  205. Holmer, M.; Lindqvist, C.; Petersson, S.; Moshtaghi-Svensson, J.; Tillander, V.; Brismar, T.B.; Hagstrom, H.; Stal, P. Treatment of NAFLD with intermittent calorie restriction or low-carb high-fat diet—A randomised controlled trial. JHEP Rep. 2021, 3, 100256. [Google Scholar] [CrossRef] [PubMed]
  206. Clayton-Chubb, D.; Vaughan, N.V.; George, E.S.; Chan, A.T.; Roberts, S.K.; Ryan, J.; Phyo, A.Z.Z.; McNeil, J.J.; Beilin, L.J.; Tran, C.; et al. Mediterranean Diet and Ultra-Processed Food Intake in Older Australian Adults-Associations with Frailty and Cardiometabolic Conditions. Nutrients 2024, 16, 2978. [Google Scholar] [CrossRef]
  207. Crow, R.S.; Petersen, C.L.; Cook, S.B.; Stevens, C.J.; Titus, A.J.; Mackenzie, T.A.; Batsis, J.A. Reported Weight Change in Older Adults and Presence of Frailty. J. Frailty Aging 2020, 9, 74–81. [Google Scholar] [CrossRef] [PubMed]
  208. Fougère, B.; Morley, J.E. Editorial: Weight Loss is a Major Cause of Frailty. J. Nutr. Health Aging 2017, 21, 933–935. [Google Scholar] [CrossRef] [PubMed]
  209. Villareal, D.T.; Chode, S.; Parimi, N.; Sinacore, D.R.; Hilton, T.; Armamento-Villareal, R.; Napoli, N.; Qualls, C.; Shah, K. Weight loss, exercise, or both and physical function in obese older adults. N. Engl. J. Med. 2011, 364, 1218–1229. [Google Scholar] [CrossRef]
  210. Leon-Munoz, L.M.; Guallar-Castillon, P.; Lopez-Garcia, E.; Rodriguez-Artalejo, F. Mediterranean diet and risk of frailty in community-dwelling older adults. J. Am. Med. Dir. Assoc. 2014, 15, 899–903. [Google Scholar] [CrossRef]
  211. Talegawkar, S.A.; Bandinelli, S.; Bandeen-Roche, K.; Chen, P.; Milaneschi, Y.; Tanaka, T.; Semba, R.D.; Guralnik, J.M.; Ferrucci, L. A higher adherence to a Mediterranean-style diet is inversely associated with the development of frailty in community-dwelling elderly men and women. J. Nutr. 2012, 142, 2161–2166. [Google Scholar] [CrossRef]
  212. Eleftheriou, D.; Benetou, V.; Trichopoulou, A.; La Vecchia, C.; Bamia, C. Mediterranean diet and its components in relation to all-cause mortality: Meta-analysis. Br. J. Nutr. 2018, 120, 1081–1097. [Google Scholar] [CrossRef]
  213. Govindaraju, T.; Sahle, B.W.; McCaffrey, T.A.; McNeil, J.J.; Owen, A.J. Dietary Patterns and Quality of Life in Older Adults: A Systematic Review. Nutrients 2018, 10, 971. [Google Scholar] [CrossRef]
  214. Rosato, V.; Temple, N.J.; La Vecchia, C.; Castellan, G.; Tavani, A.; Guercio, V. Mediterranean diet and cardiovascular disease: A systematic review and meta-analysis of observational studies. Eur. J. Nutr. 2019, 58, 173–191. [Google Scholar] [CrossRef]
  215. Martinez-Gonzalez, M.A.; Gea, A.; Ruiz-Canela, M. The Mediterranean Diet and Cardiovascular Health. Circ. Res. 2019, 124, 779–798. [Google Scholar] [CrossRef]
  216. Pooler, J.A.; Hartline-Grafton, H.; DeBor, M.; Sudore, R.L.; Seligman, H.K. Food Insecurity: A Key Social Determinant of Health for Older Adults. J. Am. Geriatr. Soc. 2019, 67, 421–424. [Google Scholar] [CrossRef]
  217. Leung, C.W.; Wolfson, J.A. Food Insecurity Among Older Adults: 10-Year National Trends and Associations with Diet Quality. J. Am. Geriatr. Soc. 2021, 69, 964–971. [Google Scholar] [CrossRef] [PubMed]
  218. Golovaty, I.; Tien, P.C.; Price, J.C.; Sheira, L.; Seligman, H.; Weiser, S.D. Food Insecurity May Be an Independent Risk Factor Associated with Nonalcoholic Fatty Liver Disease among Low-Income Adults in the United States. J. Nutr. 2020, 150, 91–98. [Google Scholar] [CrossRef]
  219. Walker-Clarke, A.; Walasek, L.; Meyer, C. Psychosocial factors influencing the eating behaviours of older adults: A systematic review. Ageing Res. Rev. 2022, 77, 101597. [Google Scholar] [CrossRef] [PubMed]
  220. Tsofliou, F.; Vlachos, D.; Hughes, C.; Appleton, K.M. Barriers and Facilitators Associated with the Adoption of and Adherence to a Mediterranean Style Diet in Adults: A Systematic Review of Published Observational and Qualitative Studies. Nutrients 2022, 14, 4314. [Google Scholar] [CrossRef] [PubMed]
  221. van Erck, D.; Dolman, C.D.; Henriques, J.P.; Schoufour, J.D.; Delewi, R.; Scholte Op Reimer, W.J.M.; Snaterse, M. Exploring barriers and facilitators of behavioural changes in dietary intake and physical activity: A qualitative study in older adults undergoing transcatheter aortic valve implantation. Eur. Geriatr. Med. 2023, 14, 503–510. [Google Scholar] [CrossRef]
  222. Gadowski, A.M.; Curtis, A.J.; Heritier, S.; Zoungas, S.; Owen, A.J. Barriers to healthy lifestyle behaviours and adherence to physical activity and dietary guidelines by older Australians: A descriptive analysis. Hum. Nutr. Metab. 2021, 25, 200127. [Google Scholar] [CrossRef]
  223. Sam-Yellowe, T.Y. Nutritional Barriers to the Adherence to the Mediterranean Diet in Non-Mediterranean Populations. Foods 2024, 13, 1750. [Google Scholar] [CrossRef]
  224. Kim, D.; Vazquez-Montesino, L.M.; Li, A.A.; Cholankeril, G.; Ahmed, A. Inadequate Physical Activity and Sedentary Behavior Are Independent Predictors of Nonalcoholic Fatty Liver Disease. Hepatology 2020, 72, 1556–1568. [Google Scholar] [CrossRef]
  225. Hallsworth, K.; Thoma, C.; Moore, S.; Ploetz, T.; Anstee, Q.M.; Taylor, R.; Day, C.P.; Trenell, M.I. Non-alcoholic fatty liver disease is associated with higher levels of objectively measured sedentary behaviour and lower levels of physical activity than matched healthy controls. Frontline Gastroenterol. 2015, 6, 44–51. [Google Scholar] [CrossRef] [PubMed]
  226. Ekblom-Bak, E.; Ekblom, B.; Vikstrom, M.; de Faire, U.; Hellenius, M.L. The importance of non-exercise physical activity for cardiovascular health and longevity. Br. J. Sports Med. 2014, 48, 233–238. [Google Scholar] [CrossRef]
  227. Hamer, M.; de Oliveira, C.; Demakakos, P. Non-exercise physical activity and survival: English longitudinal study of ageing. Am. J. Prev. Med. 2014, 47, 452–460. [Google Scholar] [CrossRef]
  228. Kim, D.; Murag, S.; Cholankeril, G.; Cheung, A.; Harrison, S.A.; Younossi, Z.M.; Ahmed, A. Physical Activity, Measured Objectively, Is Associated with Lower Mortality in Patients with Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2021, 19, 1240–1247.e5. [Google Scholar] [CrossRef] [PubMed]
  229. Sabag, A.; Barr, L.; Armour, M.; Armstrong, A.; Baker, C.J.; Twigg, S.M.; Chang, D.; Hackett, D.A.; Keating, S.E.; George, J.; et al. The Effect of High-intensity Interval Training vs. Moderate-intensity Continuous Training on Liver Fat: A Systematic Review and Meta-Analysis. J. Clin. Endocrinol. Metab. 2022, 107, 862–881. [Google Scholar] [CrossRef]
  230. Kwak, M.S.; Kim, D.; Chung, G.E.; Kim, W.; Kim, J.S. The preventive effect of sustained physical activity on incident nonalcoholic fatty liver disease. Liver Int. 2017, 37, 919–926. [Google Scholar] [CrossRef]
  231. 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. [Google Scholar] [CrossRef] [PubMed]
  232. Babu, A.F.; Csader, S.; Lok, J.; Gomez-Gallego, C.; Hanhineva, K.; El-Nezami, H.; Schwab, U. Positive Effects of Exercise Intervention without Weight Loss and Dietary Changes in NAFLD-Related Clinical Parameters: A Systematic Review and Meta-Analysis. Nutrients 2021, 13, 3135. [Google Scholar] [CrossRef]
  233. Piercy, K.L.; Troiano, R.P.; Ballard, R.M.; Carlson, S.A.; Fulton, J.E.; Galuska, D.A.; George, S.M.; Olson, R.D. The Physical Activity Guidelines for Americans. JAMA 2018, 320, 2020–2028. [Google Scholar] [CrossRef]
  234. Australian Government Department of Health and Aged Care. Physical Activity and Exercise Guidelines for All Australians. Available online: https://www.health.gov.au/topics/physical-activity-and-exercise/physical-activity-and-exercise-guidelines-for-all-australians (accessed on 28 April 2025).
  235. Racey, M.; Ali, M.U.; Sherifali, D.; Fitzpatrick-Lewis, D.; Lewis, R.; Jovkovic, M.; Bouchard, D.R.; Giguère, A.; Holroyd-Leduc, J.; Tang, A.; et al. Effectiveness of physical activity interventions in older adults with frailty or prefrailty: A systematic review and meta-analysis. CMAJ Open 2021, 9, E728–E743. [Google Scholar] [CrossRef]
  236. Nagai, K.; Miyamato, T.; Okamae, A.; Tamaki, A.; Fujioka, H.; Wada, Y.; Uchiyama, Y.; Shinmura, K.; Domen, K. Physical activity combined with resistance training reduces symptoms of frailty in older adults: A randomized controlled trial. Arch. Gerontol. Geriatr. 2018, 76, 41–47. [Google Scholar] [CrossRef]
  237. Gibson, A.A.; Gale, J.; Stamatakis, E.; Lindley, R.I.; Fontana, L.; Cistulli, P.A.; Nassar, N. Impact of lifestyle risk factors on admission to nursing home care: A cohort study of 127 108 people aged 60 years and over. J. Epidemiol. Community Health 2023, 77, 744–751. [Google Scholar] [CrossRef] [PubMed]
  238. Henin, G.; Loumaye, A.; Leclercq, I.A.; Lanthier, N. Myosteatosis: Diagnosis, pathophysiology and consequences in metabolic dysfunction-associated steatotic liver disease. JHEP Rep. 2024, 6, 100963. [Google Scholar] [CrossRef] [PubMed]
  239. Sardeli, A.V.; Komatsu, T.R.; Mori, M.A.; Gaspari, A.F.; Chacon-Mikahil, M.P.T. Resistance Training Prevents Muscle Loss Induced by Caloric Restriction in Obese Elderly Individuals: A Systematic Review and Meta-Analysis. Nutrients 2018, 10, 423. [Google Scholar] [CrossRef]
  240. Watanabe, J.H.; Kwon, J.; Nan, B.; Reikes, A. Trends in glucagon-like peptide 1 receptor agonist use, 2014 to 2022. J. Am. Pharm. Assoc. 2024, 64, 133–138. [Google Scholar] [CrossRef]
  241. Lin, K.; Mehrotra, A.; Tsai, T.C. Metabolic Bariatric Surgery in the Era of GLP-1 Receptor Agonists for Obesity Management. JAMA Netw. Open 2024, 7, e2441380. [Google Scholar] [CrossRef] [PubMed]
  242. Abushamat, L.A.; Shah, P.A.; Eckel, R.H.; Harrison, S.A.; Barb, D. The Emerging Role of Glucagon-Like Peptide-1 Receptor Agonists for the Treatment of Metabolic Dysfunction-Associated Steatohepatitis. Clin. Gastroenterol. Hepatol. 2024, 22, 1565–1574. [Google Scholar] [CrossRef]
  243. Pyke, C.; Heller, R.S.; Kirk, R.K.; Orskov, C.; Reedtz-Runge, S.; Kaastrup, P.; Hvelplund, A.; Bardram, L.; Calatayud, D.; Knudsen, L.B. GLP-1 receptor localization in monkey and human tissue: Novel distribution revealed with extensively validated monoclonal antibody. Endocrinology 2014, 155, 1280–1290. [Google Scholar] [CrossRef]
  244. Huang, D.Q.; Noureddin, N.; Ajmera, V.; Amangurbanova, M.; Bettencourt, R.; Truong, E.; Gidener, T.; Siddiqi, H.; Majzoub, A.M.; Nayfeh, T.; et al. Type 2 diabetes, hepatic decompensation, and hepatocellular carcinoma in patients with non-alcoholic fatty liver disease: An individual participant-level data meta-analysis. Lancet Gastroenterol. Hepatol. 2023, 8, 829–836. [Google Scholar] [CrossRef]
  245. Anderson, J.W.; Konz, E.C.; Frederich, R.C.; Wood, C.L. Long-term weight-loss maintenance: A meta-analysis of US studies. Am. J. Clin. Nutr. 2001, 74, 579–584. [Google Scholar] [CrossRef]
  246. Moolla, A.; Poolman, T.; Othonos, N.; Dong, J.; Smith, K.; Cornfield, T.; White, S.; Ray, D.W.; Mouchti, S.; Mózes, F.E.; et al. Randomised trial comparing weight loss through lifestyle and GLP-1 receptor agonist therapy in people with MASLD. JHEP Rep. 2025, 7, 101363. [Google Scholar] [CrossRef] [PubMed]
  247. Newsome, P.N.; Buchholtz, K.; Cusi, K.; Linder, M.; Okanoue, T.; Ratziu, V.; Sanyal, A.J.; Sejling, A.S.; Harrison, S.A.; Investigators, N.N. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2021, 384, 1113–1124. [Google Scholar] [CrossRef] [PubMed]
  248. Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; LEAN trial team; Abouda, G.; et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): A multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet 2016, 387, 679–690. [Google Scholar] [CrossRef]
  249. Nahra, R.; Wang, T.; Gadde, K.M.; Oscarsson, J.; Stumvoll, M.; Jermutus, L.; Hirshberg, B.; Ambery, P. Effects of Cotadutide on Metabolic and Hepatic Parameters in Adults with Overweight or Obesity and Type 2 Diabetes: A 54-Week Randomized Phase 2b Study. Diabetes Care 2021, 44, 1433–1442. [Google Scholar] [CrossRef] [PubMed]
  250. Romero-Gomez, M.; Lawitz, E.; Shankar, R.R.; Chaudhri, E.; Liu, J.; Lam, R.L.H.; Kaufman, K.D.; Engel, S.S.; Group, M.-P.S. A phase IIa active-comparator-controlled study to evaluate the efficacy and safety of efinopegdutide in patients with non-alcoholic fatty liver disease. J. Hepatol. 2023, 79, 888–897. [Google Scholar] [CrossRef]
  251. Harrison, S.A.; Browne, S.K.; Suschak, J.J.; Tomah, S.; Gutierrez, J.A.; Yang, J.; Roberts, M.S.; Harris, M.S. Effect of pemvidutide, a GLP-1/glucagon dual receptor agonist, on MASLD: A randomized, double-blind, placebo-controlled study. J. Hepatol. 2025, 82, 7–17. [Google Scholar] [CrossRef]
  252. Sanyal, A.J.; Newsome, P.N.; Kliers, I.; Ostergaard, L.H.; Long, M.T.; Kjaer, M.S.; Cali, A.M.G.; Bugianesi, E.; Rinella, M.E.; Roden, M.; et al. Phase 3 Trial of Semaglutide in Metabolic Dysfunction-Associated Steatohepatitis. N. Engl. J. Med. 2025, 392, 2089–2099. [Google Scholar] [CrossRef]
  253. Simon, T.G.; Patorno, E.; Schneeweiss, S. Glucagon-Like Peptide-1 Receptor Agonists and Hepatic Decompensation Events in Patients with Cirrhosis and Diabetes. Clin. Gastroenterol. Hepatol. 2022, 20, 1382–1393.e19. [Google Scholar] [CrossRef]
  254. Yen, F.S.; Hou, M.C.; Cheng-Chung Wei, J.; Shih, Y.H.; Hsu, C.Y.; Hsu, C.C.; Hwu, C.M. Glucagon-like Peptide-1 Receptor Agonist Use in Patients with Liver Cirrhosis and Type 2 Diabetes. Clin. Gastroenterol. Hepatol. 2024, 22, 1255–1264.e18. [Google Scholar] [CrossRef]
  255. Lawitz, E.J.; Fraessdorf, M.; Neff, G.W.; Schattenberg, J.M.; Noureddin, M.; Alkhouri, N.; Schmid, B.; Andrews, C.P.; Takacs, I.; Hussain, S.A.; et al. Efficacy, tolerability and pharmacokinetics of survodutide, a glucagon/glucagon-like peptide-1 receptor dual agonist, in cirrhosis. J. Hepatol. 2024, 81, 837–846. [Google Scholar] [CrossRef]
  256. Yang, C.T.; Yao, W.Y.; Yang, C.Y.; Peng, Z.Y.; Ou, H.T.; Kuo, S. Lower risks of cirrhosis and hepatocellular carcinoma with GLP-1RAs in type 2 diabetes: A nationwide cohort study using target trial emulation framework. J. Intern. Med. 2024, 295, 357–368. [Google Scholar] [CrossRef]
  257. Targher, G.; Mantovani, A.; Byrne, C.D.; Tilg, H. Recent advances in incretin-based therapy for MASLD: From single to dual or triple incretin receptor agonists. Gut 2025, 74, 487–497. [Google Scholar] [CrossRef]
  258. Karagiannis, T.; Tsapas, A.; Athanasiadou, E.; Avgerinos, I.; Liakos, A.; Matthews, D.R.; Bekiari, E. GLP-1 receptor agonists and SGLT2 inhibitors for older people with type 2 diabetes: A systematic review and meta-analysis. Diabetes Res. Clin. Pract. 2021, 174, 108737. [Google Scholar] [CrossRef] [PubMed]
  259. Locatelli, J.C.; Costa, J.G.; Haynes, A.; Naylor, L.H.; Fegan, P.G.; Yeap, B.B.; Green, D.J. Incretin-Based Weight Loss Pharmacotherapy: Can Resistance Exercise Optimize Changes in Body Composition? Diabetes Care 2024, 47, 1718–1730. [Google Scholar] [CrossRef]
  260. Berg, S.; Stickle, H.; Rose, S.J.; Nemec, E.C. Discontinuing glucagon-like peptide-1 receptor agonists and body habitus: A systematic review and meta-analysis. Obes. Rev. 2025, e13929. [Google Scholar] [CrossRef] [PubMed]
  261. Harrison, S.A.; Bedossa, P.; Guy, C.D.; Schattenberg, J.M.; Loomba, R.; Taub, R.; Labriola, D.; Moussa, S.E.; Neff, G.W.; Rinella, M.E.; et al. A Phase 3, Randomized, Controlled Trial of Resmetirom in NASH with Liver Fibrosis. N. Engl. J. Med. 2024, 390, 497–509. [Google Scholar] [CrossRef] [PubMed]
  262. Kokkorakis, M.; Boutari, C.; Hill, M.A.; Kotsis, V.; Loomba, R.; Sanyal, A.J.; Mantzoros, C.S. Resmetirom, the first approved drug for the management of metabolic dysfunction-associated steatohepatitis: Trials, opportunities, and challenges. Metabolism 2024, 154, 155835. [Google Scholar] [CrossRef]
  263. Suvarna, R.; Shetty, S.; Pappachan, J.M. Efficacy and safety of Resmetirom, a selective thyroid hormone receptor-beta agonist, in the treatment of metabolic dysfunction-associated steatotic liver disease (MASLD): A systematic review and meta-analysis. Sci. Rep. 2024, 14, 19790. [Google Scholar] [CrossRef]
  264. Younossi, Z.M.; Stepanova, M.; Taub, R.A.; Barbone, J.M.; Harrison, S.A. Hepatic Fat Reduction Due to Resmetirom in Patients with Nonalcoholic Steatohepatitis Is Associated with Improvement of Quality of Life. Clin. Gastroenterol. Hepatol. 2022, 20, 1354–1361.e7. [Google Scholar] [CrossRef]
  265. Eslami-Amirabadi, M.; Sajjadi, S.A. The relation between thyroid dysregulation and impaired cognition/behaviour: An integrative review. J. Neuroendocrinol. 2021, 33, e12948. [Google Scholar] [CrossRef]
  266. Mooradian, A.D. Age-related thyroid hormone resistance: A friend or foe. Endocr. Metab. Sci. 2023, 11, 100132. [Google Scholar] [CrossRef]
  267. Daily, E.B.; Aquilante, C.L. Cytochrome P450 2C8 pharmacogenetics: A review of clinical studies. Pharmacogenomics 2009, 10, 1489–1510. [Google Scholar] [CrossRef]
  268. Itkonen, M.K.; Tornio, A.; Neuvonen, M.; Neuvonen, P.J.; Niemi, M.; Backman, J.T. Clopidogrel and Gemfibrozil Strongly Inhibit the CYP2C8-Dependent Formation of 3-Hydroxydesloratadine and Increase Desloratadine Exposure In Humans. Drug Metab. Dispos. 2019, 47, 377–385. [Google Scholar] [CrossRef] [PubMed]
  269. Chen, V.L.; Morgan, T.R.; Rotman, Y.; Patton, H.M.; Cusi, K.; Kanwal, F.; Kim, W.R. Resmetirom therapy for metabolic dysfunction-associated steatotic liver disease: October 2024 updates to AASLD Practice Guidance. Hepatology 2025, 81, 312–320. [Google Scholar] [CrossRef] [PubMed]
  270. Delara, M.; Murray, L.; Jafari, B.; Bahji, A.; Goodarzi, Z.; Kirkham, J.; Chowdhury, M.; Seitz, D.P. Prevalence and factors associated with polypharmacy: A systematic review and Meta-analysis. BMC Geriatr. 2022, 22, 601. [Google Scholar] [CrossRef]
  271. Lee, Y.; Lim, S.; Hong, E.S.; Kim, J.H.; Moon, M.K.; Chun, E.J.; Choi, S.I.; Kim, Y.B.; Park, Y.J.; Park, K.S.; et al. Serum FGF21 concentration is associated with hypertriglyceridaemia, hyperinsulinaemia and pericardial fat accumulation, independently of obesity, but not with current coronary artery status. Clin. Endocrinol. 2014, 80, 57–64. [Google Scholar] [CrossRef]
  272. Weng, Y.; Ishino, T.; Sievers, A.; Talukdar, S.; Chabot, J.R.; Tam, A.; Duan, W.; Kerns, K.; Sousa, E.; He, T.; et al. Glyco-engineered Long Acting FGF21 Variant with Optimal Pharmaceutical and Pharmacokinetic Properties to Enable Weekly to Twice Monthly Subcutaneous Dosing. Sci. Rep. 2018, 8, 4241. [Google Scholar] [CrossRef]
  273. Bailey, N.N.; Peterson, S.J.; Parikh, M.A.; Jackson, K.A.; Frishman, W.H. Pegozafermin Is a Potential Master Therapeutic Regulator in Metabolic Disorders: A Review. Cardiol. Rev. 2023. [Google Scholar] [CrossRef]
  274. Loomba, R.; Sanyal, A.J.; Kowdley, K.V.; Bhatt, D.L.; Alkhouri, N.; Frias, J.P.; Bedossa, P.; Harrison, S.A.; Lazas, D.; Barish, R.; et al. Randomized, Controlled Trial of the FGF21 Analogue Pegozafermin in NASH. N. Engl. J. Med. 2023, 389, 998–1008. [Google Scholar] [CrossRef]
  275. Harrison, S.A.; Frias, J.P.; Neff, G.; Abrams, G.A.; Lucas, K.J.; Sanchez, W.; Gogia, S.; Sheikh, M.Y.; Behling, C.; Bedossa, P.; et al. Safety and efficacy of once-weekly efruxifermin versus placebo in non-alcoholic steatohepatitis (HARMONY): A multicentre, randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Gastroenterol. Hepatol. 2023, 8, 1080–1093. [Google Scholar] [CrossRef]
  276. Harrison, S.A.; Frias, J.P.; Neff, G.; Abrams, G.; Lucas, K.J.; Sanchez, W.; Gogia, S.; Sheikh, M.Y.; Behling, C.; Bedossa, P. LBO-002 Efruxifermin significantly reduced liver fibrosis in MASH patients with F2-F3 fibrosis, with sustained improvement in liver injury and resolution of steatohepatitis over 96 weeks (HARMONY phase 2b study). J. Hepatol. 2024, 80, S8. [Google Scholar] [CrossRef]
  277. Harrison, S.A.; Ruane, P.J.; Freilich, B.; Neff, G.; Patil, R.; Behling, C.; Hu, C.; Shringarpure, R.; de Temple, B.; Fong, E.; et al. A randomized, double-blind, placebo-controlled phase IIa trial of efruxifermin for patients with compensated NASH cirrhosis. JHEP Rep. 2023, 5, 100563. [Google Scholar] [CrossRef]
  278. Noureddin, M.; Rinella, M.E.; Chalasani, N.P.; Neff, G.W.; Lucas, K.J.; Rodriguez, M.E.; Rudraraju, M.; Patil, R.; Behling, C.; Burch, M.; et al. Efruxifermin in Compensated Liver Cirrhosis Caused by MASH. N. Engl. J. Med. 2025, 392, 2413–2424. [Google Scholar] [CrossRef] [PubMed]
  279. Harrison, S.A.; Frias, J.P.; Lucas, K.J.; Reiss, G.; Neff, G.; Bollepalli, S.; Su, Y.; Chan, D.; Tillman, E.J.; Moulton, A.; et al. Safety and Efficacy of Efruxifermin in Combination with a GLP-1 Receptor Agonist in Patients with NASH/MASH and Type 2 Diabetes in a Randomized Phase 2 Study. Clin. Gastroenterol. Hepatol. 2025, 23, 103–113. [Google Scholar] [CrossRef]
  280. Tsochatzis, E.A.; Noureddin, M. Combination Treatment in MASLD: The Next Frontier. Clin. Gastroenterol. Hepatol. 2025, 23, 31–32. [Google Scholar] [CrossRef] [PubMed]
  281. Valenti, L.; Bugianesi, E.; Pajvani, U.; Targher, G. Nonalcoholic fatty liver disease: Cause or consequence of type 2 diabetes? Liver Int. 2016, 36, 1563–1579. [Google Scholar] [CrossRef] [PubMed]
  282. Dai, W.; Ye, L.; Liu, A.; Wen, S.W.; Deng, J.; Wu, X.; Lai, Z. Prevalence of nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus: A meta-analysis. Medicine 2017, 96, e8179. [Google Scholar] [CrossRef] [PubMed]
  283. Marcuccilli, M.; Chonchol, M. NAFLD and Chronic Kidney Disease. Int. J. Mol. Sci. 2016, 17, 562. [Google Scholar] [CrossRef]
  284. Byrne, C.D.; Targher, G. NAFLD as a driver of chronic kidney disease. J. Hepatol. 2020, 72, 785–801. [Google Scholar] [CrossRef]
  285. Mantovani, A.; Petracca, G.; Beatrice, G.; Csermely, A.; Lonardo, A.; Schattenberg, J.M.; Tilg, H.; Byrne, C.D.; Targher, G. Non-alcoholic fatty liver disease and risk of incident chronic kidney disease: An updated meta-analysis. Gut 2022, 71, 156–162. [Google Scholar] [CrossRef]
  286. Rodgers, J.L.; Jones, J.; Bolleddu, S.I.; Vanthenapalli, S.; Rodgers, L.E.; Shah, K.; Karia, K.; Panguluri, S.K. Cardiovascular Risks Associated with Gender and Aging. J. Cardiovasc. Dev. Dis. 2019, 6, 19. [Google Scholar] [CrossRef] [PubMed]
  287. Denic, A.; Glassock, R.J.; Rule, A.D. Structural and Functional Changes with the Aging Kidney. Adv. Chronic Kidney Dis. 2016, 23, 19–28. [Google Scholar] [CrossRef]
  288. Kovesdy, C.P. Epidemiology of chronic kidney disease: An update 2022. Kidney Int. Suppl. 2022, 12, 7–11. [Google Scholar] [CrossRef]
  289. Zheng, Y.; Ley, S.H.; Hu, F.B. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat. Rev. Endocrinol. 2018, 14, 88–98. [Google Scholar] [CrossRef]
  290. Del Ben, M.; Baratta, F.; Polimeni, L.; Pastori, D.; Loffredo, L.; Averna, M.; Violi, F.; Angelico, F. Under-prescription of statins in patients with non-alcoholic fatty liver disease. Nutr. Metab. Cardiovasc. Dis. 2017, 27, 161–167. [Google Scholar] [CrossRef]
  291. Tziomalos, K.; Athyros, V.G.; Paschos, P.; Karagiannis, A. Nonalcoholic fatty liver disease and statins. Metabolism 2015, 64, 1215–1223. [Google Scholar] [CrossRef]
  292. Pastori, D.; Pani, A.; Di Rocco, A.; Menichelli, D.; Gazzaniga, G.; Farcomeni, A.; D’Erasmo, L.; Angelico, F.; Del Ben, M.; Baratta, F. Statin liver safety in non-alcoholic fatty liver disease: A systematic review and metanalysis. Br. J. Clin. Pharmacol. 2022, 88, 441–451. [Google Scholar] [CrossRef] [PubMed]
  293. Lee, J.I.; Lee, H.W.; Lee, K.S.; Lee, H.S.; Park, J.Y. Effects of Statin Use on the Development and Progression of Nonalcoholic Fatty Liver Disease: A Nationwide Nested Case-Control Study. Am. J. Gastroenterol. 2021, 116, 116–124. [Google Scholar] [CrossRef] [PubMed]
  294. Bosch, J.; Gracia-Sancho, J.; Abraldes, J.G. Cirrhosis as new indication for statins. Gut 2020, 69, 953–962. [Google Scholar] [CrossRef]
  295. Huang, D.Q.; El-Serag, H.B.; Loomba, R. Global epidemiology of NAFLD-related HCC: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 223–238. [Google Scholar] [CrossRef]
  296. Laakso, M.; Fernandes Silva, L. Statins and risk of type 2 diabetes: Mechanism and clinical implications. Front. Endocrinol. 2023, 14, 1239335. [Google Scholar] [CrossRef] [PubMed]
  297. Ramos, R.; Comas-Cufi, M.; Marti-Lluch, R.; Ballo, E.; Ponjoan, A.; Alves-Cabratosa, L.; Blanch, J.; Marrugat, J.; Elosua, R.; Grau, M.; et al. Statins for primary prevention of cardiovascular events and mortality in old and very old adults with and without type 2 diabetes: Retrospective cohort study. BMJ 2018, 362, k3359. [Google Scholar] [CrossRef]
  298. Cholesterol Treatment Trialists, C. Efficacy and safety of statin therapy in older people: A meta-analysis of individual participant data from 28 randomised controlled trials. Lancet 2019, 393, 407–415. [Google Scholar] [CrossRef]
  299. Zoungas, S.; Moran, C.; Curtis, A.J.; Spark, S.; Flanagan, Z.; Beilin, L.; Chong, T.T.; Cloud, G.C.; Hopper, I.; Kost, A.; et al. Baseline Characteristics of Participants in STAREE: A Randomized Trial for Primary Prevention of Cardiovascular Disease Events and Prolongation of Disability-Free Survival in Older People. J. Am. Heart Assoc. 2024, 13, e036357. [Google Scholar] [CrossRef] [PubMed]
  300. McNeil, J.J.; Nelson, M.R.; Woods, R.L.; Lockery, J.E.; Wolfe, R.; Reid, C.M.; Kirpach, B.; Shah, R.C.; Ives, D.G.; Storey, E.; et al. Effect of Aspirin on All-Cause Mortality in the Healthy Elderly. N. Engl. J. Med. 2018, 379, 1519–1528. [Google Scholar] [CrossRef]
  301. McNeil, J.J.; Wolfe, R.; Woods, R.L.; Tonkin, A.M.; Donnan, G.A.; Nelson, M.R.; Reid, C.M.; Lockery, J.E.; Kirpach, B.; Storey, E.; et al. Effect of Aspirin on Cardiovascular Events and Bleeding in the Healthy Elderly. N. Engl. J. Med. 2018, 379, 1509–1518. [Google Scholar] [CrossRef] [PubMed]
  302. McNeil, J.J.; Woods, R.L.; Nelson, M.R.; Reid, C.M.; Kirpach, B.; Wolfe, R.; Storey, E.; Shah, R.C.; Lockery, J.E.; Tonkin, A.M.; et al. Effect of Aspirin on Disability-free Survival in the Healthy Elderly. N. Engl. J. Med. 2018, 379, 1499–1508. [Google Scholar] [CrossRef] [PubMed]
  303. Zoungas, S.; Zhou, Z.; Owen, A.J.; Curtis, A.J.; Espinoza, S.E.; Ernst, M.E.; Woods, R.L.; Orchard, S.G.; McNeil, J.J.; Murray, A.M.; et al. Daily low-dose aspirin and incident type 2 diabetes in community-dwelling healthy older adults: A post-hoc analysis of efficacy and safety in the ASPREE randomised placebo-controlled trial. Lancet Diabetes Endocrinol. 2024, 12, 98–106. [Google Scholar] [CrossRef]
  304. Simon, T.G.; Henson, J.; Osganian, S.; Masia, R.; Chan, A.T.; Chung, R.T.; Corey, K.E. Daily Aspirin Use Associated with Reduced Risk For Fibrosis Progression in Patients with Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2019, 17, 2776–2784.e4. [Google Scholar] [CrossRef]
  305. Simon, T.G.; Wilechansky, R.M.; Stoyanova, S.; Grossman, A.; Dichtel, L.E.; Lauer, G.M.; Miller, K.K.; Hoshida, Y.; Corey, K.E.; Loomba, R.; et al. Aspirin for Metabolic Dysfunction-Associated Steatotic Liver Disease without Cirrhosis: A Randomized Clinical Trial. JAMA 2024, 331, 920–929. [Google Scholar] [CrossRef]
  306. Memel, Z.N.; Arvind, A.; Moninuola, O.; Philpotts, L.; Chung, R.T.; Corey, K.E.; Simon, T.G. Aspirin Use Is Associated with a Reduced Incidence of Hepatocellular Carcinoma: A Systematic Review and Meta-analysis. Hepatol. Commun. 2021, 5, 133–143. [Google Scholar] [CrossRef] [PubMed]
  307. Lee, T.Y.; Hsu, Y.C.; Ho, H.J.; Lin, J.T.; Chen, Y.J.; Wu, C.Y. Daily aspirin associated with a reduced risk of hepatocellular carcinoma in patients with non-alcoholic fatty liver disease: A population-based cohort study. EClinicalMedicine 2023, 61, 102065. [Google Scholar] [CrossRef] [PubMed]
  308. Mahady, S.E.; Margolis, K.L.; Chan, A.; Polekhina, G.; Woods, R.L.; Wolfe, R.; Nelson, M.R.; Lockery, J.E.; Wood, E.M.; Reid, C.; et al. Major GI bleeding in older persons using aspirin: Incidence and risk factors in the ASPREE randomised controlled trial. Gut 2021, 70, 717–724. [Google Scholar] [CrossRef]
  309. Bailey, C.J.; Day, C.; Bellary, S. Renal Protection with SGLT2 Inhibitors: Effects in Acute and Chronic Kidney Disease. Curr. Diab. Rep. 2022, 22, 39–52. [Google Scholar] [CrossRef] [PubMed]
  310. Vaduganathan, M.; Docherty, K.F.; Claggett, B.L.; Jhund, P.S.; de Boer, R.A.; Hernandez, A.F.; Inzucchi, S.E.; Kosiborod, M.N.; Lam, C.S.P.; Martinez, F.; et al. SGLT-2 inhibitors in patients with heart failure: A comprehensive meta-analysis of five randomised controlled trials. Lancet 2022, 400, 757–767. [Google Scholar] [CrossRef]
  311. 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. [Google Scholar] [CrossRef]
  312. Takeshita, Y.; Honda, M.; Harada, K.; Kita, Y.; Takata, N.; Tsujiguchi, H.; Tanaka, T.; Goto, H.; Nakano, Y.; Iida, N.; et al. Comparison of Tofogliflozin and Glimepiride Effects on Nonalcoholic Fatty Liver Disease in Participants with Type 2 Diabetes: A Randomized, 48-Week, Open-Label, Active-Controlled Trial. Diabetes Care 2022, 45, 2064–2075. [Google Scholar] [CrossRef]
  313. Kawaguchi, T.; Fujishima, Y.; Wakasugi, D.; Io, F.; Sato, Y.; Uchida, S.; Kitajima, Y. Effects of SGLT2 inhibitors on the onset of esophageal varices and extrahepatic cancer in type 2 diabetic patients with suspected MASLD: A nationwide database study in Japan. J. Gastroenterol. 2024, 59, 1120–1132. [Google Scholar] [CrossRef]
  314. Jang, H.; Kim, Y.; Lee, D.H.; Joo, S.K.; Koo, B.K.; Lim, S.; Lee, W.; Kim, W. Outcomes of Various Classes of Oral Antidiabetic Drugs on Nonalcoholic Fatty Liver Disease. JAMA Intern. Med. 2024, 184, 375–383. [Google Scholar] [CrossRef]
  315. Lunati, M.E.; Cimino, V.; Gandolfi, A.; Trevisan, M.; Montefusco, L.; Pastore, I.; Pace, C.; Betella, N.; Favacchio, G.; Bulgheroni, M.; et al. SGLT2-inhibitors are effective and safe in the elderly: The SOLD study. Pharmacol. Res. 2022, 183, 106396. [Google Scholar] [CrossRef]
  316. Scheen, A.J.; Bonnet, F. Efficacy and safety profile of SGLT2 inhibitors in the elderly: How is the benefit/risk balance? Diabetes Metab. 2023, 49, 101419. [Google Scholar] [CrossRef]
  317. van Poelgeest, E.P.; Handoko, M.L.; Muller, M.; van der Velde, N.; Task, E.; Finish group on Fall-risk-increasing, d. Diuretics, SGLT2 inhibitors and falls in older heart failure patients: To prescribe or to deprescribe? A clinical review. Eur. Geriatr. Med. 2023, 14, 659–674. [Google Scholar] [CrossRef]
  318. Suzuki, Y.; Suzuki, H.; Maruo, K.; Matsuda, T.; Murayama, Y.; Sugano, Y.; Osaki, Y.; Iwasaki, H.; Sekiya, M.; Hada, Y.; et al. Longitudinal association of SGLT2 inhibitors and GLP-1RAs on falls in persons with type 2 diabetes. Sci. Rep. 2025, 15, 9178. [Google Scholar] [CrossRef]
  319. Younossi, Z.M.; Blissett, D.; Blissett, R.; Henry, L.; Stepanova, M.; Younossi, Y.; Racila, A.; Hunt, S.; Beckerman, R. The economic and clinical burden of nonalcoholic fatty liver disease in the United States and Europe. Hepatology 2016, 64, 1577–1586. [Google Scholar] [CrossRef]
  320. Torre, E.; Di Matteo, S.; Martinotti, C.; Bruno, G.M.; Goglia, U.; Testino, G.; Rebora, A.; Bottaro, L.C.; Colombo, G.L. Economic Impact of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) in Italy. Analysis and Perspectives. Clinicoecon Outcomes Res. 2024, 16, 773–784. [Google Scholar] [CrossRef] [PubMed]
  321. Alkhodary, A.; Aljunid, S.M.; Ismail, A.; Nur, A.; Shahar, S. The economic burden of frailty among elderly people: A review of the current literature. Malays. J. Public Health Med. 2020, 20, 224–232. [Google Scholar] [CrossRef]
  322. Bock, J.O.; Konig, H.H.; Brenner, H.; Haefeli, W.E.; Quinzler, R.; Matschinger, H.; Saum, K.U.; Schottker, B.; Heider, D. Associations of frailty with health care costs--results of the ESTHER cohort study. BMC Health Serv. Res. 2016, 16, 128. [Google Scholar] [CrossRef] [PubMed]
  323. Alvarez-Bustos, A.; Rodriguez-Sanchez, B.; Carnicero-Carreno, J.A.; Sepulveda-Loyola, W.; Garcia-Garcia, F.J.; Rodriguez-Manas, L. Healthcare cost expenditures associated to frailty and sarcopenia. BMC Geriatr. 2022, 22, 747. [Google Scholar] [CrossRef]
  324. Wallace, C.; Gamkrelidze, I.; Estes, C.; Razavi, H.; Sanyal, A.J. Modeling the health and economic impact of pharmacologic therapies for MASLD in the United States. J. Hepatol. 2025, 83, 21–30. [Google Scholar] [CrossRef]
Table 1. Components of selected steatosis indices. Adapted from Clayton-Chubb et al. [63].
Table 1. Components of selected steatosis indices. Adapted from Clayton-Chubb et al. [63].
Score ComponentsFLIFSIDSIHSIZJUVAILAP
BMI (kg/m2)
Abdominal circumference (cm)
GGT (U/L)
Triglycerides (Yes/No)
ALT (U/L)
AST (U/L)
Age (years)
Sex (Male/Female)
Hypertension (Yes/No)
Diabetes (Yes/No)
Serum glucose
Ethnicity
HDL cholesterol
BMI: Body Mass Index; GGT: gamma glutamyltransferase; ALT: alanine aminotransferase; AST: aspartate aminotransferase; FLI: Fatty Liver Index; FSI: Framingham Steatosis Index; DSI: Dallas Steatosis Index; HSI: Hepatic Steatosis Index; ZJU: ZJU Index; VAI: Visceral Adiposity Index; LAP: Lipid Accumulation Product.
Table 2. Components of selected liver fibrosis scores.
Table 2. Components of selected liver fibrosis scores.
Score ComponentsFIB-4BARDAPRINAFLD Fibrosis ScoreBIMASTSAFEMAF-5ELF
Age
ALT (U/L)
AST (U/L)
Platelets
BMI
Waist Circumference
Diabetes (Yes/No)
Impaired Fasting Glucose (Yes/No)
Albumin
Globulins
Tissue inhibitor of metalloproteinases 1 (TIMP-1)
Amino-terminal propeptide of type III procollagen (PIIINP)
Hyaluronic acid (HA)
BMI: Body Mass Index; ALT: alanine aminotransferase; AST: aspartate aminotransferase; FIB-4: Fibrosis-4; BARD: BMI, AST/ALT Ratio, Diabetes; APRI: AST to Platelet Ratio Index; BIMAST: BMI and AST; SAFE: Steatosis-Associated Fibrosis Estimator; MAF-5: Metabolic Dysfunction-Associated Fibrosis-5; ELF: Enhanced Liver Fibrosis.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Clayton-Chubb, D.; Kemp, W.W.; Majeed, A.; Lange, P.W.; Fitzpatrick, J.A.; Vaz, K.; Lubel, J.S.; Hodge, A.D.; Ryan, J.; McNeil, J.J.; et al. Steatotic Liver Disease in Older Adults: Clinical Implications and Unmet Needs. Nutrients 2025, 17, 2189. https://doi.org/10.3390/nu17132189

AMA Style

Clayton-Chubb D, Kemp WW, Majeed A, Lange PW, Fitzpatrick JA, Vaz K, Lubel JS, Hodge AD, Ryan J, McNeil JJ, et al. Steatotic Liver Disease in Older Adults: Clinical Implications and Unmet Needs. Nutrients. 2025; 17(13):2189. https://doi.org/10.3390/nu17132189

Chicago/Turabian Style

Clayton-Chubb, Daniel, William W. Kemp, Ammar Majeed, Peter W. Lange, Jessica A. Fitzpatrick, Karl Vaz, John S. Lubel, Alexander D. Hodge, Joanne Ryan, John J. McNeil, and et al. 2025. "Steatotic Liver Disease in Older Adults: Clinical Implications and Unmet Needs" Nutrients 17, no. 13: 2189. https://doi.org/10.3390/nu17132189

APA Style

Clayton-Chubb, D., Kemp, W. W., Majeed, A., Lange, P. W., Fitzpatrick, J. A., Vaz, K., Lubel, J. S., Hodge, A. D., Ryan, J., McNeil, J. J., Owen, A. J., Woods, R. L., & Roberts, S. K. (2025). Steatotic Liver Disease in Older Adults: Clinical Implications and Unmet Needs. Nutrients, 17(13), 2189. https://doi.org/10.3390/nu17132189

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop