Hostname: page-component-76fb5796d-45l2p Total loading time: 0 Render date: 2024-04-25T16:10:13.791Z Has data issue: false hasContentIssue false

Non-alcoholic fatty liver disease: a multi-system disease influenced by ageing and sex, and affected by adipose tissue and intestinal function

Published online by Cambridge University Press:  22 November 2021

Josh Bilson
Affiliation:
Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
Jaswinder K. Sethi
Affiliation:
Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK Institute for Life Sciences, University of Southampton, Southampton, UK
Christopher D. Byrne*
Affiliation:
Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
*
*Corresponding author: Christopher D. Byrne, email C.D.Byrne@soton.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

In recent years, a wealth of factors are associated with increased risk of developing non-alcoholic fatty liver disease (NAFLD) and NAFLD is now thought to increase the risk of multiple extra-hepatic diseases. The aim of this review is first to focus on the role of ageing and sex as key, poorly understood risk factors in the development and progression of NAFLD. Secondly, we aim to discuss the roles of white adipose tissue (WAT) and intestinal dysfunction, as producers of extra-hepatic factors known to further contribute to the pathogenesis of NAFLD. Finally, we aim to summarise the role of NAFLD as a multi-system disease affecting other organ systems beyond the liver. Both increased age and male sex increase the risk of NAFLD and this may be partly driven by alterations in the distribution and function of WAT. Similarly, changes in gut microbiota composition and intestinal function with ageing and chronic overnutrition are likely to contribute to the development of NAFLD both directly (i.e. by affecting hepatic function) and indirectly via exacerbating WAT dysfunction. Consequently, the presence of NAFLD significantly increases the risk of various extra-hepatic diseases including CVD, type 2 diabetes mellitus, chronic kidney disease and certain extra-hepatic cancers. Thus changes in WAT and intestinal function with ageing and chronic overnutrition contribute to the development of NAFLD – a multi-system disease that subsequently contributes to the development of other chronic cardiometabolic diseases.

Type
Conference on ‘Nutrition in a changing world’
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author(s), 2021. Published by Cambridge University Press on behalf of The Nutrition Society

Current estimates indicate that about 30 % of the global adult population are affected by non-alcoholic fatty liver disease (NAFLD) and the increasing prevalence of this disease has occurred in parallel with the global epidemic of obesity and type 2 diabetes mellitus (T2DM)(Reference Targher, Tilg and Byrne1,Reference Sayiner, Koenig and Henry2) . Considered to be the predominant cause of chronic liver disease in many parts of the world, NAFLD represents a spectrum of progressive hepatic disease phenotypes extending from hepatic steatosis to non-alcoholic steatohepatitis (NASH), liver fibrosis and cirrhosis(Reference Targher, Tilg and Byrne1,Reference Mantovani, Scorletti and Mosca3,Reference Paik, Golabi and Younossi4) . Evidence now shows that NAFLD increases the risk of liver-related complications and is also a multi-system disease that increases the risk of CVD and cardiac disease(Reference Byrne and Targher5,Reference Targher, Corey and Byrne6) , chronic kidney disease (CKD)(Reference Byrne and Targher7), T2DM(Reference Morrison, Zaccardi and Khunti8,Reference Mantovani, Byrne and Bonora9) and some extra-hepatic cancers(Reference Targher, Tilg and Byrne1). Therefore, it is no surprise that the presence of NAFLD is strongly associated with an increased risk of all-cause mortality(Reference Mantovani, Scorletti and Mosca3,Reference Tsochatzis and Newsome10) . Indeed, CVD is the main cause of mortality in patients with NAFLD, followed by extra-hepatic cancers and liver-related complications(Reference Tsochatzis and Newsome10). Additionally, recent evidence suggests that there may be an even greater cardiometabolic risk with the more advanced stages of liver disease, such as liver fibrosis, which is also a strong predictor of all-cause and disease-specific mortality(Reference Ekstedt, Hagström and Nasr11Reference Taylor, Taylor and Bayliss13).

In recent years, a wealth of factors have been shown to be associated with an increased risk of developing NAFLD. The aim of this review is first to focus on the role of ageing and sex as key, poorly understood risk factors in the development and progression of NAFLD. Secondly, we will discuss the roles of white adipose tissue (WAT) and intestinal dysfunction, as producers of extra-hepatic factors known to further contribute to the pathogenesis of NAFLD. Finally, we will summarise the role of NAFLD as a multi-system disease affecting other organ systems beyond the liver.

Sex and age as risk factors for non-alcoholic fatty liver disease

The involvement of age and sex in the development of NAFLD has received increased attention in recent years yet the reasons why these are risk factors for NAFLD remain poorly understood. The prevalence of NAFLD is higher in men and is thought to increase into middle age and then decline after the age of 50–60 years(Reference Lonardo, Bellentani and Argo14). In contrast, pre-menopausal women appear to be relatively protected from NAFLD; however, this protective capacity is lost after the fifth decade of life when the prevalence of NAFLD is thought to be similar in both sexes(Reference Lonardo, Bellentani and Argo14,Reference Lonardo, Nascimbeni and Ballestri15) . The incidence of NASH and cirrhosis is also thought to be greater in both men and women who are ≥50 years of age compared to younger age groups(Reference Sayiner, Koenig and Henry2). Recent meta-analysis suggests that whilst pre-menopausal women may have a lower risk of NAFLD, women ≥50 years of age may be at an increased risk of NAFLD progression, compared to men of a similar age(Reference Balakrishnan, Patel and Dunn-Valadez16). Specifically, among older age groups (≥50 years of age), the relative risk of NASH and advanced liver fibrosis was found to be 17 and 56 % higher respectively, in women compared to men(Reference Balakrishnan, Patel and Dunn-Valadez16). Conversely, the risk of NAFLD progression was not significantly different between men and women in populations with an average age of ≤50 years(Reference Balakrishnan, Patel and Dunn-Valadez16). Further work is required to elucidate potential mechanisms underlying the apparent increased risk of NAFLD progression in older women. For example, studies exploring sexual dimorphism in liver metabolism have recently linked hepatic actions of oestrogens to lipid metabolism and female reproductive functions(Reference Maggi and Della Torre17). Whether these or other sexually dimorphic metabolic or endocrine factors are important in NAFLD remains to be investigated(Reference Lefebvre and Staels18).

Advancing age also increases the risk of hepatic and extra-hepatic complications of NAFLD(Reference Lonardo, Bellentani and Argo14). Thus it is expected that older patients with NAFLD will have a higher likelihood of overall and disease-specific mortality(Reference Bertolotti, Lonardo and Mussi19,Reference Ong, Pitts and Younossi20) . Whether the association between NAFLD and all-cause mortality is modified by sex is currently unclear. Previous studies suggest a worse outcome in men(Reference Ong, Pitts and Younossi20,Reference Bedogni, Miglioli and Masutti21) , whilst others have found trends suggesting that NAFLD is associated with an increased risk of all-cause mortality in women but not men(Reference Liu, Zhong and Tan22). Thus, further large prospective cohort studies should explore whether the direction and magnitude of the association between NAFLD and mortality are modified by sex.

White adipose tissue mass and distribution in non-alcoholic fatty liver disease

A wealth of evidence indicates that obesity increases the risk of NAFLD(Reference Younossi23Reference Younossi, Koenig and Abdelatif26). Obesity is defined as excess body fat and results from chronic overnutrition. For adults, it is most frequently classified as a weight for height index or BMI and includes underweight or ‘wasting’ (<18⋅5 kg/m2), overweight (≥25 kg/m2), obesity (≥30 kg/m2) and morbid obesity (≥40 kg/m2)(Reference Kelly, Yang and Chen27). In contrast, waist circumference provides a simpler anthropometric measurement to diagnose central obesity which is an important independent risk factor for NAFLD and an important component of the metabolic syndrome (MetS). As previously described(Reference Alberti, Eckel and Grundy28), MetS is defined as the presence of three or more of the following criteria; increased waist circumference, hypertriglyceridemia, reduced HDL-cholesterol, hypertension and hyperglycaemia. It is worth highlighting that neither BMI nor waist circumference is considered reliable indicators of adiposity per se since they do not provide an assessment of WAT mass nor volume(29). Nonetheless, BMI and waist circumference have proved to be extremely useful measures for population-based studies and firmly established the importance of obesity as a risk factor for NAFLD. Despite this, it is an oversimplification to consider NAFLD solely as a consequence of obesity given the growing evidence indicating that NAFLD can also occur in individuals with a non-obese BMI, or low WAT mass(Reference Ye, Zou and Yeo30,Reference Azzu, Vacca and Virtue31) . It has been proposed that an increase in the accumulation of central WAT and a reduction in the functional capacity of WAT (particularly subcutaneous WAT (SAT)) to store excess energy as TAG are crucial factors that underpin the relationship between obesity, systemic metabolic disease and NAFLD(Reference Azzu, Vacca and Virtue31).

Studies utilising adipose tissue-targeted technologies coupled with histological assessment have suggested that the hypertrophic expansion of adipocytes within visceral WAT (VAT) rather than SAT is particularly associated with NAFLD. After approximately 4 years of follow up, a larger VAT area was found to be associated with a higher risk of incident NAFLD, whereas larger areas of SAT were associated with regression of NAFLD(Reference Kim, Chung and Kwak32). Moreover, several recent studies have demonstrated that increased VAT, as opposed to SAT, increases the risk of, and predicts advanced liver fibrosis in patients with NAFLD(Reference Eguchi, Eguchi and Mizuta33Reference Yu, Kim and Kim35). Similarly, evidence also indicates that VAT accumulation is an independent risk factor for hepatocellular carcinoma (HCC) recurrence in patients with suspected NASH(Reference Ohki, Tateishi and Shiina36). Thus, this evidence supports a fundamental hypothesis that ‘the risk of developing metabolic disease associated with obesity is governed by the regional distribution of WAT within the individual, with the expansion of certain fat depots being more strongly associated with metabolic dysfunction than others’(Reference Jensen37). Collectively, it is likely that the distribution and capacity of SAT to effectively expand and store lipid, rather than the obesity per se, is a pivotal factor in the relationship between increased adiposity and NAFLD risk.

The distribution of WAT is known to differ significantly between sexes, changes with increasing age and has been hypothesised to be partly responsible for the increased prevalence of NAFLD in men and older age groups, particularly post-menopausal women (Fig. 1)(Reference De Carvalho, Justice and Freitas38Reference Karastergiou, Smith and Greenberg40). Whilst the mechanisms regulating the distribution of WAT remain largely elusive, evidence indicates that ageing and male sex are associated with a restricted capacity to effectively expand so-called ‘metabolically protective’ SAT depots(Reference Mancuso and Bouchard41). Whilst pre-menopausal women typically have greater total adiposity, men tend to accumulate greater amounts of VAT with ageing and pre-menopausal women accumulate gluteal femoral SAT which is associated with a lower risk of metabolic disease and NAFLD(Reference Eaton and Sethi42). In both men and women, older age (i.e. post-menopausal women and men >50 years) is associated with a reduction in the capacity of SAT to expand and an increase in VAT(Reference Hughes, Roubenoff and Wood43Reference Kuk, Saunders and Davidson45). The limited capacity of SAT to store TAG in men and with increasing age is likely to re-direct lipid accumulation ectopically in non-adipose tissues, including the liver, leading to lipotoxicity, a chronic local and systemic pro-inflammatory environment and eventually NAFLD development(Reference Godoy-Matos, Silva Júnior and Valerio46). The importance of effective SAT expansion can be seen in individuals with certain genetic or acquired lipodystrophies that are characterised by the complete or partial absence of SAT(Reference Polyzos, Perakakis and Mantzoros47). In spite of their often lean appearance, these individuals appear to exhibit much higher rates of NAFLD/NASH progression and other cardiometabolic complications than would be expected based on their BMI alone(Reference Azzu, Vacca and Virtue31,Reference Polyzos, Perakakis and Mantzoros47) . Given this, it is likely that differences in WAT distribution between sexes and changes occurring with increasing age are both important in the increased risk of NAFLD associated with ageing and with male sex.

Fig. 1. Age-related changes in WAT distribution in men and women are associated with increased risk NAFLD, MetS, T2DM and CVD. Sex and age are key factors that modify the risk of NAFLD and NAFLD progression. NAFLD risk is lower in younger women compared to younger men whereas the risk of NAFLD is similar in older men and women (i.e. post-menopausal). Younger women have an increased capacity to preferentially expand gluteal femoral SAT consequently protecting them from NAFLD. Age-associated changes in WAT leads to the redistribution of WAT which is typically characterised by a marked reduction in SAT and increased central metabolically-unfavourable VAT which may partly explain the increased risk of NAFLD associated with ageing in both men and women. WAT distribution is different between men and women, is heavily influenced by ageing and is strongly associated with NAFLD risk. T2DM, type 2 diabetes; MetS, metabolic syndrome; SAT, subcutaneous adipose tissue; VAT, visceral adipose tissue; NAFLD, non-alcoholic fatty liver disease; WAT, white adipose tissue.

Adipose tissue dysfunction and non-alcoholic fatty liver disease

WAT is composed of mature unilocular adipocyte fraction and a stromal vascular fraction, comprised of numerous cell types such as vascular, mesenchymal and immune cells. At a cellular level, WAT expansion can be mediated by an enlargement of individual adipocytes (hypertrophy), an increase in the number of adipocytes (hyperplasia) or a mixture of both. Adipocyte hypertrophy, rather than hyperplasia, is more closely associated with WAT dysfunction and metabolic disease(Reference Longo, Zatterale and Naderi48). Factors including hypoxia, low-grade chronic inflammation (i.e. metaflammation) and improper extracellular matrix remodelling are thought to limit adipocyte differentiation and the healthy expansion of adipose tissue (hyperplasia)(Reference Sethi and Vidal-Puig49,Reference Cawthorn, Heyd and Hegyi50) . This limit can result in adipocyte hypertrophy, dysfunction, stress and eventually death(Reference Rutkowski, Stern and Scherer51,Reference Halberg, Khan and Trujillo52) . In this context, WAT dysfunction refers to a reduction in the tissues ability to effectively sense and respond to dynamic changes in nutrient availability (i.e. metabolic inflexibility) and can coexist with adipose insulin resistance and metaflammation. Specifically, this dysfunction is thought to affect WAT metabolism and in particular its ability to handle lipids and increase the lipolytic rate of WAT due to a reduction in tissue insulin sensitivity, increasing the flux of non-esterified fatty acids (NEFA) to the liver and consequently increasing the risk of NAFLD(Reference Azzu, Vacca and Virtue31,Reference Blüher53Reference Byrne55) .

Accompanying the changes in the distribution of WAT, ageing is associated with a marked reduction in insulin, lipolytic and NEFA responsiveness in WAT. This metabolic inflexibility may underly the known association between ageing and increased risk of NAFLD(Reference Hughes, Roubenoff and Wood43Reference Kuk, Saunders and Davidson45). The reduction in SAT with ageing in both men and women may in part be driven by a reduction in the adipogenic potential of progenitor cells and the accumulation of senescent adipocytes in aged WAT. Preadipocytes isolated from peripheral SAT in elderly individuals were found to have a reduced rate of replication compared to those isolated from younger individuals(Reference Caso, McNurlan and Mileva56). Additionally, ageing is associated with an accumulation of senescent adipocyte-derived stem cells within SAT which lack the ability to differentiate into adipocytes in response to metabolic stress, consequently affecting the tissue's capacity to store TAG(Reference Schipper, Marra and Zhang57). Through their senescence-associated secretory phenotype, senescent adipocyte progenitor cells within WAT are also likely to contribute to WAT inflammation and subsequent metabolic complications(Reference Palmer and Kirkland58,Reference Xu, Palmer and Ding59) .

In addition to ageing, there are also sexually dimorphic differences in WAT function whereby WAT in females is generally more insulin-sensitive, more lipogenic and less susceptible to inflammation than WAT from males. This phenomenon is also strongly associated with differences in sex hormone concentrations(Reference Grove, Fried and Greenberg60,Reference Macotela, Boucher and Tran61) . Menopause appears to associate with a preferential increase in VAT (rather than SAT) in both obese and non-obese women(Reference Phillips, Jing and Heymsfield62Reference Leeners, Geary and Tobler65), further supporting a role for sex hormones, such as oestrogen, in regulating the beneficial distribution and function of WAT. Circulating concentrations of oestrogen decrease markedly after menopause which is thought to lead to the redistribution of lipids into VAT and the liver which, in combination with overnutrition, increases the risk of VAT accumulation and NAFLD in post-menopausal women(Reference DiStefano66). Pre-clinical studies utilising ovariectomised murine models also support a causative relationship between reduced oestrogen production, increased VAT mass and the development of NASH(Reference Eaton and Sethi42,Reference DiStefano66Reference Rogers, Perfield and Strissel68) . Whilst an in-depth discussion of the role of oestrogen within WAT is beyond the scope of this review (see other relevant reviews(Reference Eaton and Sethi42,Reference Lizcano and Guzmán69,Reference Cooke and Naaz70) ), it is thought that the increased expression of oestrogen receptor α in the gluteal femoral SAT of premenopausal women promotes lipoprotein lipase activity and accumulation of TAG in adipocytes within this depot(Reference Frank, de Souza Santos and Palmer71). Thus, it is likely that differences in WAT function (partly driven by differences in sex hormone concentrations and the expression of functional target receptors) is an important factor underlying the observed differences in NAFLD risk between men and women. Furthermore, changes in WAT with ageing are likely to exacerbate WAT dysfunction associated with a state of chronic energy surplus and are likely to have an important role in the increased risk of NAFLD associated with older age.

Adipokines and non-alcoholic fatty liver disease

WAT is an endocrine tissue capable of secreting a wide range of adipokines which have various roles in the regulation of whole-body energy homeostasis and inter-organ communication(Reference Funcke and Scherer72). The aberrant production of these adipokines has been linked to multiple obesity-related metabolic diseases. Amongst these adipokines, leptin and adiponectin are predominately produced by adipocytes. In addition to its well-established role in regulating appetite and energy homeostasis(Reference Sethi and Vidal-Puig49,Reference Montague, Farooqi and Whitehead73) , leptin exerts a dual action on hepatic function and NAFLD severity. Recent meta-analyses including an analysis of over thirty studies indicated that circulating concentrations of leptin are elevated in patients with NAFLD compared to healthy controls and supports a positive relationship between leptin and NAFLD(Reference Polyzos, Aronis and Kountouras74). As recently highlighted(Reference Polyzos, Kountouras and Mantzoros75,Reference Jiménez-Cortegana, García-Galey and Tami76) , under normoleptinemia conditions, leptin is thought to suppress hepatic glucose production and hepatic lipogenesis thus providing an insulin-sensitising anti-steatotic effect. Conversely, in the context of chronic hyperleptinemia as is common in obesity, a state of leptin resistance can result, which may also contribute to the NASH phenotype. It is suggested that in the liver, high concentrations of leptin can increase the expression of matrix remodelling enzymes via interacting with leptin receptors on Kupffer and sinusoidal endothelial cells, in turn activating hepatic stellate cells, and possibly contributing to liver fibrosis(Reference Polyzos, Kountouras and Zavos77).

Sexual dimorphism has also been reported for leptin expression(Reference Saad, Damani and Gingerich78). Despite their lower risk of NAFLD, circulating concentrations of leptin are higher in pre-menopausal women compared to age-matched men and higher leptin levels are thought to be driven by both greater adiposity and an increased production rate of leptin per unit mass of WAT in women compared to men(Reference Castracane, Kraemer and Franken79). In both men and women, circulating concentrations of leptin are thought to gradually decline with ageing, with reductions being most noticeable in women compared to men whilst appearing to be independent of menopausal status(Reference Castracane, Kraemer and Franken79,Reference Isidori, Strollo and Morè80) . Despite these findings, it is currently unknown whether differences in circulating concentrations of leptin between sexes and age groups have an impact on the risk of NAFLD.

Similar to leptin, a wealth of studies indicate that the circulating concentrations of adiponectin, the most systemically abundant adipokine, are altered in patients with NAFLD (as reviewed in(Reference Boutari and Mantzoros81)). Adiponectin is a hepatoprotective adipokine that has well-established anti-inflammatory(Reference Maeda, Shimomura and Kishida82Reference Tilg and Hotamisligil84) and insulin-sensitising effects(Reference Yamauchi, Kamon and Minokoshi85) both systemically and within the liver. Meta-analysis indicates that adiponectin concentrations are significantly lower in patients with NAFLD compared to healthy controls; furthermore, NASH is associated with lower adiponectin when compared to simple steatosis(Reference Polyzos, Toulis and Goulis86). Conversely, adiponectin concentrations are thought to increase in patients with NAFLD-cirrhosis potentially due to a reduction in the hepatic clearance of adiponectin and/or an increase in its production as a result of the tissue repair process associated with NAFLD-cirrhosis(Reference Sohara, Takagi and Kakizaki87Reference Polyzos, Kountouras and Zavos89). Along with its well-established role in promoting hepatic insulin sensitivity(Reference Buechler, Wanninger and Neumeier90,Reference Polyzos, Kountouras and Zavos91) , evidence indicates that adiponectin also has antifibrogenic effects via inhibiting the proliferation of hepatic stellate cells(Reference Adachi and Brenner92). Whilst the role of adiponectin in ageing remains uncertain, it is thought that circulating concentrations of adiponectin are paradoxically increased in older age and are positively associated with physical disability and mortality in elderly individuals(Reference Kizer, Arnold and Strotmeyer93). Furthermore, some evidence suggests that the association between adiponectin and ageing may be modified by sex(Reference Adamczak, Rzepka and Chudek94). In addition to leptin and adiponectin, a wealth of other studies have demonstrated that numerous other adipokines may be involved in the development and progression of NAFLD (Table 1). It should be noted that there is a substantial amount of conflicting evidence regarding the changes in circulating concentrations of other adipokines in the context of NAFLD and little is known about the potential pathological role of these adipokines in NAFLD (Table 1). Moreover, further studies are required to elucidate whether the effects of age and sex on adipokine production influences NAFLD risk.

Table 1. Changes in circulating concentrations of adipokines and their potential roles in NAFLD

HSC, hepatic stellate cells; NAFLD, non-alcoholic fatty liver disease; RBP-4, retinol binding protein-4.

In contrast to classic adipocyte-derived adipokines leptin and adiponectin, studies investigating changes in circulating concentrations of other adipokines in patients with NAFLD are largely inconsistent. Similarly, whilst the role of leptin and adiponectin in the development and progression of NAFLD remains somewhat debated, there is currently very little known about the potential roles of other adipokines on hepatic function and NAFLD. It should be noted that the expression of many adipokines (e.g. chemerin and RBP-4) is not restricted to WAT and also occurs within other tissues including the liver. Consequently, whilst changes in the secretion of WAT-derived adipokines may contribute to altered circulating concentrations, other sources (particularly hepatic) may also influence circulating concentrations, hepatic function and NAFLD.

WAT dysfunction and changes in adipokine secretion are also strongly associated with increased low-grade chronic inflammation in WAT (metaflammation); characterised by the infiltration of various leucocytes, an increase in the ratio of proinflammatory/anti-inflammatory macrophages and leucocytes and the increased presence of crown-like structures (dying adipocytes surrounded by pro-inflammatory macrophages)(Reference Reilly and Saltiel95). Consequently, metaflammation in WAT (particularly VAT inflammation) is associated with an increased expression of pro-inflammatory cytokines such as IL-6, IL-1β, TNF-α and monocyte chemoattractant protein-1(Reference Mohamed-Ali, Flower and Sethi96Reference Sethi and Hotamisiligil98). Some of these have been shown to contribute to local insulin resistance, elevated fatty acid lipolysis, anti-adipogenesis and pro-inflammatory macrophage infiltration(Reference Sethi and Vidal-Puig49,Reference Cawthorn and Sethi97Reference Ota101) . This in turn can impact both metabolic and endocrine functions of WAT. Evidence from murine diet-induced obesity studies indicates that WAT inflammation and reductions in protective anti-inflammatory lipokines such as palmitoleic acid may be important in the development of NASH(Reference Duval, Thissen and Keshtkar102Reference Souza, Teixeira and Biondo104).

By virtue of its anatomical links, via the portal vein, increased VAT inflammation is of particular importance in NAFLD/NASH since VAT-derived inflammatory cytokines (other adipokines, lipokines and metabolites (e.g. NEFA)) are initially transported to the liver and therefore may exacerbate NAFLD severity. Consequently, this may in turn increase the associated risk of T2DM and CVD(Reference Item and Konrad105,Reference Kabir, Catalano and Ananthnarayan106) . Collectively, findings indicate that changes in the production of adipokines and increased WAT inflammation may contribute to NAFLD via modulating local and hepatic function, inducing insulin resistance and modulating the local and systemic pro-inflammatory conditions. Whether these changes in WAT function contribute to the increased risk of extra-hepatic diseases associated with NAFLD independently requires further investigation.

Intestinal dysfunction, dysbiosis and non-alcoholic fatty liver disease

Emerging evidence now suggests that changes in gut microbiota (GM) (i.e. dysbiosis) and intestinal function may exacerbate WAT dysfunction which may indirectly contribute to metabolic dysfunction and NAFLD(Reference Lundgren and Thaiss107). The gastrointestinal tract is the first point of contact for ingested nutrients where it has an integral role in nutrient breakdown and absorption, regulation of whole-body energy homeostasis and is an important host defence barrier. Occupying the gastrointestinal tract is an extensive number of microorganisms, collectively known as the GM which are thought to modulate local and distal tissue function via a range of complex mechanisms(Reference Valdes, Walter and Segal108,Reference Zmora, Suez and Elinav109) . The microbial organisms occupying the gastrointestinal tract mainly include bacteria, archaea, fungi and viruses (predominantly bacteriophages); however, studies exploring the role of the GM in NAFLD have predominantly focused on bacteria(Reference Hu, Lin and Kong110,Reference Camarillo-Guerrero, Almeida and Rangel-Pineros111) .

A plethora of studies have revealed that GM dysbiosis is associated with and is a contributing factor to NAFLD(Reference Canfora, Meex and Venema112Reference Le Roy, Llopis and Lepage115). The dominating phyla within human GM are Bacteroidetes and Firmicutes with a significant inter-individual variation in the GM at lower taxonomical levels(Reference Bäckhed, Ley and Sonnenburg116,Reference Mouzaki, Comelli and Arendt117) . Previous evidence indicates that the relative abundance of Bacteroidetes is lower in patients with NASH compared to those with hepatic steatosis and healthy controls(Reference Mouzaki, Comelli and Arendt117). More recently, Bacteroides abundance was found to be significantly increased in patients with NASH and the abundance of Ruminococcus was increased in patients with liver fibrosis(Reference Boursier, Mueller and Barret118). As recently reviewed(Reference Aron-Wisnewsky, Vigliotti and Witjes113), this shifting in GM in relation to NAFLD severity is supported by numerous other studies. Indeed, the presence of bacteria belonging to the Proteobacteria phylum was increased significantly in patients with ≥F3 when compared to patients with F0–F2 liver fibrosis (Table 2)(Reference Loomba, Seguritan and Li119). Emerging evidence also supports a strong link between the GM and NAFLD-cirrhosis indicating that the composition of the GM may be a useful tool for the identification and staging of NAFLD. Utilising a unique twin and family study design, one study identified a specific GM signature that had a robust diagnostic accuracy, with an area under the receiver operating characteristic of 0⋅92, for the detection of NAFLD-cirrhosis(Reference Caussy, Tripathi and Humphrey120). Further work demonstrated the robustness and potential universal applicability of this microbiome signature of NAFLD-cirrhosis in two independent cohorts across geographically and culturally distinct populations(Reference Oh, Kim and Caussy121). However, given the impact of host genetics and environmental factors on the composition of GM(Reference Li, Peng and Zhou122), it is unlikely that a single GM signature will be able to distinguish between NAFLD phenotypes at an individual level.

Table 2. Histological definitions of liver fibrosis stages and corresponding liver-biopsy validated liver VCTE cut-off values

VCTE, vibration-controlled transient elastography; kPa, kilopascal; PPV, positive predictive value; NPV, negative predictive value; NASH, non-alcoholic steatohepatitis.

Liver fibrosis stages and corresponding histological definitions are based on the NASH clinical scoring network scoring system(Reference Kleiner, Brunt and Van Natta201). Liver VCTE cut-off values are based on the findings from a recent large validation study(Reference Eddowes, Sasso and Allison202). The liver VCTE threshold of 8⋅2 kPa was found to have a: sensitivity of 0⋅71 (0⋅64–0⋅77), specificity of 0⋅70 (0⋅62–0⋅77), PPV of 0⋅78 (0⋅71–0⋅83) and NPV of 0⋅61 (0⋅54–0⋅69) for the identification of ≥F2 liver fibrosis. For the prediction of ≥F3 liver fibrosis, 9⋅7 kPa was found to have a sensitivity of 0⋅71 (0⋅62–0⋅78), specificity of 0⋅75 (0⋅69–0⋅80), PPV of 0⋅63 (0⋅55–0⋅71) and NPV of 0⋅81 (0⋅74–0⋅85). For the prediction of ≥F4 fibrosis, 13⋅6 kPa was found to have a sensitivity of 0⋅85 (0⋅69–0⋅95), specificity of 0⋅79 (0⋅74–0⋅83), PPV of 0⋅29 (0⋅24–0⋅57) and NPV of 0⋅98 (0⋅95–0⋅99).

Miele et al. were the first to identify that patients with NAFLD generally have increased intestinal permeability and alterations in intestinal tight junction integrity (observed as a reduction in zonula occludens-1 within intestinal crypt cells), compared to healthy subjects(Reference Miele, Valenza and La Torre123). Recent meta-analysis found that 39⋅1 % of NAFLD patients had evidence of increased intestinal permeability compared to 6⋅8 % of healthy controls (OR 5⋅08, 95 % CI 1⋅98, 13⋅05)(Reference Luther, Garber and Khalili124). Furthermore, subgroup analysis indicated that there was a higher incidence of increased intestinal permeability in patients with NASH compared to patients with simple steatosis(Reference Luther, Garber and Khalili124). It is generally well-accepted that the increased intestinal permeability commonly seen in NAFLD facilitates the translocation of GM-derived metabolites and bacterial products (such as lipopolysaccharides (LPS) and ethanol) which may in turn contribute to metaflammation and the pathogenesis of NAFLD(Reference Kolodziejczyk, Zheng and Shibolet125).

In addition to altered GM and intestinal permeability, the abundance of GM-dependent metabolites is thought to be altered in NAFLD, many of which may be detected in stool samples and may offer a tool for the assessment of disease severity. For example, work comparing the abundance of distinct stool metabolites in patients with NAFLD-cirrhosis v. healthy subjects revealed 17 metabolites which, in combination, were able to accurately detect the presence of NAFLD-cirrhosis (AUROC 0⋅91, 95 % CI 0⋅89, 0⋅93)(Reference Oh, Kim and Caussy121). Thus, evidence is accumulating to suggest that accumulation of certain microbial species, changes in intestinal function and increased intestinal permeability are likely to contribute not only to the pathogenesis of NAFLD but also to increased liver disease severity. Further studies are required to elucidate the potential role of non-bacterial species within the GM on the development and progression of NAFLD.

Intestinal dysfunction, dysbiosis and links with white adipose tissue function in non-alcoholic fatty liver disease

Associated with WAT dysfunction are changes in intestinal function and GM dysbiosis, which have also been proposed to be key factors contributing to NAFLD. Receiving about 70 % of its blood supply from intestinal vascularisation, the liver is constantly exposed to the metabolic products, toxins and nutrients produced by the GM(Reference Ridlon, Kang and Hylemon126). It has been suggested that when in a dysbiotic state, GM may contribute to the development and progression of NAFLD via a range of pathways; including changes in dietary energy harvest(Reference Bäckhed, Manchester and Semenkovich127,Reference Bäckhed, Ding and Wang128) , alterations in SCFA production (particularly butyrate)(Reference Rau, Rehman and Dittrich129,Reference Zhou, Pan and Xin130) , increased bacterial LPS translocation(Reference Kolodziejczyk, Zheng and Shibolet125,Reference Cani, Amar and Iglesias131) , alternations in bile acid profiles(Reference Ferslew, Xie and Johnston132) and increased endogenous ethanol production(Reference Zhu, Baker and Gill133). Indeed, the potential effects of these factors on hepatic function and NAFLD have been discussed in various recent reviews(Reference Hu, Lin and Kong110,Reference Canfora, Meex and Venema112Reference Jiang, Zheng and Zhang114,Reference Kolodziejczyk, Zheng and Shibolet125,Reference Grabherr, Grander and Effenberger134) ; furthermore, alterations in appetite-regulating gut hormones are also likely to have an important role in the development and progression of NAFLD, as recently reviewed(Reference Mells and Anania135Reference Koukias, Buzzetti and Tsochatzis137).

Disruptions in intestinal permeability associated with obesity and NAFLD are likely to be accompanied by a reduction in the integrity of intestinal tight junctions(Reference Miele, Valenza and La Torre123,Reference Durkin, Childs and Calder138) . Increased intestinal permeability in the presence of GM dysbiosis is thought to facilitate the translocation of bacterial products including pro-inflammatory endotoxins such as LPS. Circulating concentrations of LPS were found to be significantly higher in patients with NAFLD compared to healthy controls(Reference Harte, da Silva and Creely139,Reference Kitabatake, Tanaka and Fujimori140) and have been shown to be positively associated with the expression of pro-inflammatory genes within both VAT and SAT in individuals with obesity(Reference Clemente-Postigo, Oliva-Olivera and Coin-Aragüez141). This is supported by evidence from pre-clinical murine studies indicating that increased LPS may directly contribute to WAT inflammation and increase the release of WAT-derived pro-inflammatory cytokines(Reference Chang, Sia and Chang142). Accompanying these findings, various other studies have proposed additional mechanisms by which changes in intestinal function and GM dysbiosis may impact NAFLD development both directly and in-directly via detrimentally impacting WAT function (Table 3 and Fig. 2).

Fig. 2. NAFLD is associated with changes in gut microbiota-derived factors that can alter hepatic and WAT function Changes in GM in NAFLD result in alterations in the production of various metabolites/factors that are thought to contribute to NAFLD both directly (i.e. by directly impacting hepatic function) and indirectly through detrimentally influencing WAT function. As highlighted on the left, intestinal eubiosis and healthy gut function (such as that typically found in young individuals) promotes intestinal barrier integrity and homeostasis whilst restricting the production and dissemination of metabolically detrimental factors (such as LPS and endogenous ethanol) into circulation, the liver and WAT. Conversely, as highlighted on the right, intestinal dysbiosis (such as that often associated with older age) leads to alterations in various GM-derived factors/metabolites that impair the function of tight junction-associated proteins located within the intestinal epithelium. Consequently, these changes are thought to contribute to an increased risk of NAFLD both directly (via inducing hepatic mitochondrial function, inflammation and steatosis) and indirectly through detrimentally impacting WAT function (impairing WAT expansion, metabolic flexibility and increasing the production of pro-inflammatory cytokines). The increased production of inflammatory cytokines is thought to lead to a state of chronic low-grade inflammation which is likely to further disrupt the function of tight junction-associated proteins, thus forming a vicious cycle of worsening metabolic dysfunction and NAFLD disease severity. GM, gut microbiota; LPS, lipopolysaccharide; TMAO, trimethylamine N-oxide; NAFLD, non-alcoholic fatty liver disease; WAT, white adipose tissue.

Table 3. Changes in GM-derived factors/metabolites in NAFLD and their proposed effects in WAT and the liver

LPS, lipopolysaccharide; HSCs, hepatic stellate cells; TMAO, trimethylamine N-oxide; NAFLD, non-alcoholic fatty liver disease; WAT, white adipose tissue.

Changes in the production of various GM-derived metabolites/factors may contribute to the development of NAFLD both directly (i.e. via a direct action within the liver) and indirectly via affecting the function of WAT. Whilst evidence of altered circulating concentrations of certain factors (namely LPS, endogenous ethanol and TMAO) is well-reported in patients with NAFLD, changes in circulating concentrations of other factors (particularly SCFA) require further investigation. Furthermore, more research is required to elucidate the potential contribution of endogenously produced ethanol on WAT dysfunction in the context of NAFLD.

Evidence also suggests that the composition of the GM and intestinal function can differ between sexes and such differences may partly explain differences in the risk of metabolic disease between sexes(Reference Kim, Unno and Kim143Reference Sheng, Jena and Liu145). Similarly, changes in GM composition and intestinal function are strongly associated with ageing and are likely to contribute to the increased risk of NAFLD associated with older age both directly and indirectly via exacerbating WAT dysfunction(Reference Rea, O'Sullivan and Shanahan146Reference Nagpal, Mainali and Ahmadi148). Similar to obesity, ageing is also associated with disruptions in intestinal permeability subsequently facilitating the translocation of bacterial products such as LPS which are known to contribute to both hepatic and WAT dysfunction (Fig. 2)(Reference Tran and Greenwood-Van Meerveld149,Reference Man, Bertelli and Rentini150) . Collectively, existing studies demonstrate the existence of a gut-WAT axis which, in addition to the well-established gut-liver axis, may indirectly contribute to NAFLD pathogenesis. Furthermore, differences in GM composition and intestinal function between men and women and with ageing may contribute to both hepatic and WAT dysfunction and subsequently drive the development of NAFLD.

Non-alcoholic fatty liver disease and extra-hepatic complications

Non-alcoholic fatty liver disease, type 2 diabetes mellitus and metabolic syndrome

Type 2 diabetes is both a risk factor for NAFLD and an extra-hepatic complication of NAFLD. The association between T2DM and NAFLD is well-established and T2DM is considered to be one of the most important risk factors for NAFLD. A meta-analysis of twenty-four studies found that the pooled prevalence of NAFLD in patients with T2DM was 59⋅7 % (95 % CI 54⋅3, 64⋅9 %), with the prevalence of NAFLD being slightly higher in men (60⋅1 %, 95 % CI 53⋅6, 66⋅4 %), compared to women (59⋅35 %, 95 % CI 53⋅3, 65⋅3 %)(Reference Dai, Ye and Liu151). Furthermore, the presence of obesity, hypertension and dyslipidaemia, as features of the MetS, were associated with an increased prevalence of NAFLD in patients with T2DM, suggesting that these factors may act with T2DM to further increase the risk of NAFLD(Reference Dai, Ye and Liu151). The presence of T2DM increases the risk of liver fibrosis by approximately 2–6-fold(Reference Targher, Tilg and Byrne1). The mechanism by which T2DM increases the risk of liver fibrosis is uncertain. However, numerous factors have been proposed that could mediate the increase in the risk of liver fibrosis in patients with T2DM and these include insulin resistance, hyperglycaemia, hypoadiponectinemia, mitochondrial dysfunction, increased reactive oxygen species, excess free cholesterol, increased proinflammatory cytokines and endoplasmic reticulum stress(Reference Cariou, Byrne and Loomba152). Recently we have shown in patients with NAFLD that increased circulating concentrations of growth-differentiation factor-15, a stress-inducible cytokine, are independently associated with the presence of ≥F3 and ≥F2 liver fibrosis (Table 2)(Reference Bilson, Scorletti and Bindels153). We also showed in this work that growth-differentiation factor-15 may be an important factor contributing to the increased risk of liver fibrosis associated with T2DM, and that HbA1c levels explained about 30 % of the variance in growth-differentiation factor-15 concentrations(Reference Bilson, Scorletti and Bindels153). However, further work is required to fully elucidate the role of growth-differentiation factor-15 in the development and progression of NAFLD in patients with T2DM.

The estimated global prevalence of NAFLD among patients with T2DM is 55⋅5 % (95 % CI 47⋅3, 63⋅7 %) with prevalence estimates varying between geographical regions(Reference Younossi, Golabi and de Avila154). This study also found that the estimated global prevalence of NASH and advanced fibrosis in patients with T2DM was 37⋅3 % (95 % CI 24⋅7, 50⋅0 %) and 4⋅8 % (95 % CI 0⋅0, 17⋅5 %) respectively(Reference Younossi, Golabi and de Avila154). The presence of T2DM is also an important risk factor for the faster progression of NAFLD towards NASH, cirrhosis or HCC(Reference Targher, Tilg and Byrne1,Reference Targher, Corey and Byrne155,Reference Jarvis, Craig and Barker156) . Patients with NAFLD and coexisting T2DM are thought to have between a 2 and 6-fold increased risk of developing advanced fibrosis compared to patients with only NAFLD(Reference Targher, Tilg and Byrne1). In addition to T2DM, the presence of MetS is also recognised as an important NAFLD risk factor. The presence of MetS in patients with NAFLD but without diabetes is associated with more severe NAFLD compared to patients without MetS(Reference Kanwar, Nelson and Yates157). Furthermore, this study suggested that a higher number of MetS features was associated with a greater probability of NASH, with 70 % of patients diagnosed with NASH having three or more features of MetS. The presence of MetS has also recently been shown to be associated with progression to advanced fibrosis in patients with NAFLD(Reference Kleiner, Brunt and Wilson158). These findings support those of others which also show that NAFLD severity is positively associated with the presence of MetS features, particularly the level of hypertension, hyperglycaemia and hypertriglyceridemia(Reference Hsiao, Kuo and Shin159).

It is important to highlight that the link between T2DM, MetS and NAFLD is complex and bi-directional. Evidence from a recent large meta-analysis of over 500 000 individuals found that NAFLD was associated with an about 2⋅2-fold increased risk of incident diabetes independently of age, sex, adiposity and other common metabolic risk factors(Reference Mantovani, Petracca and Beatrice160). Interestingly, in this study, the risk of incident diabetes was found to increase in relation to the underlying severity of NAFLD with a particularly noticeable increase in risk according to the severity of liver fibrosis (n 5 studies; random-effects HR 3⋅42, 95 % CI 2⋅3, 5⋅1)(Reference Mantovani, Petracca and Beatrice160). These findings support other evidence from meta-analyses and observational studies which demonstrate that individuals with NAFLD had a higher risk for incident T2DM than individuals without NAFLD(Reference Morrison, Zaccardi and Khunti8,Reference Li, Wang and Tang161) . Evidence collated from eight studies with a median follow-up period of 4⋅5 years indicated that NAFLD was associated with an increased risk of incident MetS with a pooled relative risk of 3⋅2 (95 % CI 3⋅1, 3⋅4) when NAFLD was diagnosed via ultrasonography(Reference Ballestri, Zona and Targher162). Collectively, this evidence suggests that a vicious cycle of worsening disease states is likely to exist between T2DM, MetS and NAFLD(Reference Targher, Tilg and Byrne1).

Non-alcoholic fatty liver disease and CVD

Evidence indicates that NAFLD is an important risk factor for various extra-hepatic diseases and the detrimental relationship between T2DM and NAFLD likely exacerbates this risk. Furthermore, given the strong associations with NAFLD and other cardiometabolic risk factors, including central obesity, atherogenic dyslipidaemia and hypertension, it is no surprise that NAFLD is also associated with an increased risk of CVD(Reference Targher, Corey and Byrne6,Reference Targher, Byrne and Tilg163) . Recent evidence suggests that CVD is one of the most important causes of death among people with NAFLD(Reference Paik, Henry and De Avila164), and patients with NAFLD are more likely to experience CVD-related death than a liver-related death(Reference Younossi, Koenig and Abdelatif26,Reference Targher, Byrne and Tilg163,Reference Przybyszewski, Targher and Roden165) . Recent meta-analysis incorporating a total of sixteen observational studies and over 34 000 individuals with a median follow-up of about 7 years concluded that NAFLD conferred an OR of 1⋅6 for fatal and/or non-fatal CVD events (random-effects OR of 1⋅6, 95 % CI 1⋅3, 2⋅1)(Reference Targher, Byrne and Lonardo166). This is consistent with findings from others that suggest that the risk of incident CVD events increases further with greater severity of NAFLD even after adjusting for other established CVD risk factors(Reference Taylor, Taylor and Bayliss13). Emerging data also support the evolving notion that sex is an important modifier of NAFLD outcomes and suggest that the occurrence and prevalence of CVD-related events and mortality are likely to differ between sexes. One study found that in about 108 000 individuals with NAFLD, cardiovascular events were two times higher in women compared to men (OR 2⋅1, 95 % CI 1⋅7, 2⋅7)(Reference Khalid, Dasu and Suga167). Women also had higher cardiovascular mortality with advancing age starting at age 42 years further highlighting the importance of both age and sex as important risk factors for both NAFLD and CVD(Reference Khalid, Dasu and Suga167).

Non-alcoholic fatty liver disease and chronic kidney disease

The risk of CKD is also increased in patients with NAFLD. CKD is a complex, progressive chronic condition that is defined by an abnormality in either the structure and/or function of the kidneys for ≥3 months with serious implications for health(Reference Byrne and Targher7,Reference Stevens and Levin168) . Evidence from three meta-analyses demonstrates a higher incidence of CKD in patients with NAFLD(Reference Musso, Gambino and Tabibian169Reference Mantovani, Petracca and Beatrice171). The first of these studies, which included thirty-three observational (twenty cross-sectional and thirteen longitudinal) studies concluded that NAFLD was associated with a 2-fold increased prevalence of CKD (random-effects OR 2⋅1, 95 % CI 1⋅7, 2⋅7) and that NAFLD was associated with a nearly 80 % increased risk of incident CKD (random-effects HR 1⋅8, 95 % CI 1⋅7, 2⋅0)(Reference Byrne and Targher7,Reference Musso, Gambino and Tabibian169) . Similarly, the second more recent meta-analysis confirmed that NAFLD was associated with an about 40 % increase in the long-term risk of incident CKD (random-effects HR 1⋅4, 95 % CI 1⋅2, 1⋅5)(Reference Mantovani, Zaza and Byrne170). Most recently, findings from a large updated meta-analysis indicate that NAFLD was significantly associated with an about 1⋅45-fold increased long-term risk of incident CKD and this association was independent of age, sex and conventional CKD risk factors(Reference Mantovani, Petracca and Beatrice171). Interestingly, these studies also support an association between increased NAFLD severity (particularly the presence of advanced fibrosis) and increased risk of CKD(Reference Musso, Gambino and Tabibian169Reference Mantovani, Petracca and Beatrice171). Another large database study in Germany also supports a strong link between NAFLD and increased risk of CKD that is independent of age, sex and the presence of additional cardiometabolic risk factors such as diabetes, obesity and hypertension(Reference Kaps, Labenz and Galle172).

Non-alcoholic fatty liver disease and non-hepatic cancers

In addition to increasing the risk of HCC, recent evidence suggests that NAFLD may also increase the risk of various non-hepatic cancers. Findings from a recent large population-based cohort study concluded that, compared to healthy controls, patients with biopsy-confirmed NAFLD had significantly increased overall cancer incidence over a median 13⋅8 years follow-up period (adjusted HR 1⋅3, 95 % CI 1⋅2, 1⋅4)(Reference Simon, Roelstraete and Sharma173). Whilst this increase was mostly driven by a higher HCC incidence, the presence of NAFLD was also associated with modestly increased rates of melanoma, pancreatic and kidney/bladder cancers(Reference Simon, Roelstraete and Sharma173). In support of these findings, a meta-analysis of ten cohort studies (>180 000 individuals, 24⋅8 % with NAFLD) found that NAFLD was significantly associated with a nearly 1⋅5–2-fold increased risk of developing gastrointestinal cancers (oesophagus, stomach, pancreas or colorectal cancers) independently of confounding factors such as age, sex, obesity, diabetes and smoking status(Reference Mantovani, Petracca and Beatrice174). There is currently very limited data on the severity of NAFLD (particularly the severity of liver fibrosis) and the risk of developing extra-hepatic cancers. One recent study found that more severe NAFLD was associated with significantly increased overall mortality with most of the excess mortality observed being driven by extrahepatic cancer and liver cirrhosis(Reference Simon, Roelstraete and Khalili175). Whilst it is reasonable to assume that the risk of developing extra-hepatic cancers is increased in relation to NAFLD severity, further large prospective studies are needed to confirm this link. Such studies should account for the potential modifying effect of important genetic variants, age, sex and obesity along with other NAFLD-associated comorbidities when considering the relationship between NAFLD severity and risk of specific extra-hepatic cancers. This latter consideration is particularly important since it is not yet clear whether NAFLD is associated with an increased risk of certain extra-hepatic cancers simply as a consequence of shared metabolic risk factors or whether NAFLD itself directly contributes to an increased risk of developing extrahepatic cancers(Reference Mantovani, Petracca and Beatrice174).

Conclusions

The risk of developing NAFLD differs between sexes, changes with age and is likely to be modulated by complex interactions between genetic and environmental factors. Differences in WAT mass, its distribution (VAT v. SAT) and functionality (metabolic and endocrine), are likely to be key drivers of hepatic steatosis and NAFLD development. Similarly, differences in the regional distribution and function of WAT between men and women and between age groups are likely to contribute to the increased risk of NAFLD progression associated with sex and age. The development of GM dysbiosis and intestinal dysfunction is likely to contribute to NAFLD both directly and indirectly via the exacerbation of WAT inflammation and dysfunction through a range of GM-derived factors. Collectively, in the presence of chronic nutritional surplus, both WAT and intestinal dysfunction act in a synergistic manner to drive systemic metabolic dysfunction and the development of NAFLD and are further influenced by sex and age. In turn, NAFLD increases the risk of chronic hepatic and extra-hepatic metabolic diseases including T2DM, CVD, CKD, HCC and certain extra-hepatic cancers.

Acknowledgements

The authors would like to thank the National Institute for Health Research Southampton Biomedical Research Centre for their funding and support.

Financial Support

J. B., J. K. S. and C. B. are supported by the National Institution for Health Research through the NIHR Southampton Biomedical Research Centre; J. K. S. is funded by the Wellcome Trust (grant number 206453/Z/17/Z).

Conflict of Interest

None.

Authorship

The authors had sole responsibility for all aspects of preparation of this paper.

References

Targher, G, Tilg, H & Byrne, CD (2021) Non-alcoholic fatty liver disease: a multisystem disease requiring a multidisciplinary and holistic approach. Lancet Gastroenterol Hepatol 6, 578588.CrossRefGoogle ScholarPubMed
Sayiner, M, Koenig, A, Henry, L et al. (2016) Epidemiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis in the United States and the rest of the world. Clin Liver Dis 20, 205214.10.1016/j.cld.2015.10.001CrossRefGoogle ScholarPubMed
Mantovani, A, Scorletti, E, Mosca, A et al. (2020) Complications, morbidity and mortality of nonalcoholic fatty liver disease. Metabolism 111s, 154170.10.1016/j.metabol.2020.154170CrossRefGoogle ScholarPubMed
Paik, JM, Golabi, P, Younossi, Y et al. (2020) Changes in the global burden of chronic liver diseases from 2012 to 2017: the growing impact of NAFLD. Hepatology 72, 16051616.CrossRefGoogle ScholarPubMed
Byrne, CD & Targher, G (2015) NAFLD: a multisystem disease. J Hepatol 62, S47S64.CrossRefGoogle ScholarPubMed
Targher, G, Corey, KE & Byrne, CD (2021) NAFLD, and cardiovascular and cardiac diseases: factors influencing risk, prediction and treatment. Diabetes Metab 47, 101215.10.1016/j.diabet.2020.101215CrossRefGoogle ScholarPubMed
Byrne, CD & Targher, G (2020) NAFLD as a driver of chronic kidney disease. J Hepatol 72, 785801.10.1016/j.jhep.2020.01.013CrossRefGoogle ScholarPubMed
Morrison, AE, Zaccardi, F, Khunti, K et al. (2019) Causality between non-alcoholic fatty liver disease and risk of cardiovascular disease and type 2 diabetes: a meta-analysis with bias analysis. Liver Int 39, 557567.CrossRefGoogle ScholarPubMed
Mantovani, A, Byrne, CD, Bonora, E et al. (2018) Nonalcoholic fatty liver disease and risk of incident type 2 diabetes: a meta-analysis. Diabetes Care 41, 372.CrossRefGoogle ScholarPubMed
Tsochatzis, EA & Newsome, PN (2018) Non-alcoholic fatty liver disease and the interface between primary and secondary care. Lancet Gastroenterol Hepatol 3, 509517.CrossRefGoogle ScholarPubMed
Ekstedt, M, Hagström, H, Nasr, P et al. (2015) Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 61, 15471554.CrossRefGoogle ScholarPubMed
Angulo, P, Kleiner, DE, Dam-Larsen, S et al. (2015) Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 149, 389397.e310.10.1053/j.gastro.2015.04.043CrossRefGoogle ScholarPubMed
Taylor, RS, Taylor, RJ, Bayliss, S et al. (2020) Association between fibrosis stage and outcomes of patients with nonalcoholic fatty liver disease: a systematic review and meta-analysis. Gastroenterology 158, 16111625.e1612.10.1053/j.gastro.2020.01.043CrossRefGoogle ScholarPubMed
Lonardo, A, Bellentani, S, Argo, CK et al. (2015) Epidemiological modifiers of non-alcoholic fatty liver disease: focus on high-risk groups. Dig Liver Dis 47, 9971006.CrossRefGoogle ScholarPubMed
Lonardo, A, Nascimbeni, F, Ballestri, S et al. (2019) Sex differences in nonalcoholic fatty liver disease: state of the art and identification of research gaps. Hepatology 70, 14571469.CrossRefGoogle ScholarPubMed
Balakrishnan, M, Patel, P, Dunn-Valadez, S et al. (2021) Women have a lower risk of nonalcoholic fatty liver disease but a higher risk of progression vs men: a systematic review and meta-analysis. Clin Gastroenterol Hepatol 19, 6171.e15.CrossRefGoogle ScholarPubMed
Maggi, A & Della Torre, S (2018) Sex, metabolism and health. Mol Metab 15, 37.CrossRefGoogle ScholarPubMed
Lefebvre, P & Staels, B (2021) Hepatic sexual dimorphism – implications for non-alcoholic fatty liver disease. Nature Reviews Endocrinology 11, 662670.CrossRefGoogle Scholar
Bertolotti, M, Lonardo, A, Mussi, C et al. (2014) Nonalcoholic fatty liver disease and aging: epidemiology to management. World J Gastroenterol 20, 1418514204.10.3748/wjg.v20.i39.14185CrossRefGoogle Scholar
Ong, JP, Pitts, A & Younossi, ZM (2008) Increased overall mortality and liver-related mortality in non-alcoholic fatty liver disease. J Hepatol 49, 608612.CrossRefGoogle ScholarPubMed
Bedogni, G, Miglioli, L, Masutti, F et al. (2007) Incidence and natural course of fatty liver in the general population: the Dionysos study. Hepatology 46, 13871391.10.1002/hep.21827CrossRefGoogle ScholarPubMed
Liu, Y, Zhong, G-C, Tan, H-Y et al. (2019) Nonalcoholic fatty liver disease and mortality from all causes, cardiovascular disease, and cancer: a meta-analysis. Sci Rep 9, 11124.CrossRefGoogle ScholarPubMed
Younossi, ZM (2019) Non-alcoholic fatty liver disease – a global public health perspective. J Hepatol 70, 531544.10.1016/j.jhep.2018.10.033CrossRefGoogle ScholarPubMed
Fan, J-G, Kim, S-U & Wong, VW-S (2017) New trends on obesity and NAFLD in Asia. J Hepatol 67, 862873.CrossRefGoogle ScholarPubMed
Younossi, Z, Anstee, QM, Marietti, M et al. (2018) Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol 15, 1120.CrossRefGoogle ScholarPubMed
Younossi, ZM, Koenig, AB, Abdelatif, D et al. (2016) Global epidemiology of nonalcoholic fatty liver disease-meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 64, 7384.CrossRefGoogle ScholarPubMed
Kelly, T, Yang, W, Chen, CS et al. (2008) Global burden of obesity in 2005 and projections to 2030. Int J Obes 32, 14311437.10.1038/ijo.2008.102CrossRefGoogle ScholarPubMed
Alberti, KG, Eckel, RH, Grundy, SM et al. (2009) Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 120, 16401645.CrossRefGoogle Scholar
WHO (2000) Obesity: preventing and managing the global epidemic. Report of a WHO consultation. World Health Organization technical report series 894, i-xii, 1253.Google Scholar
Ye, Q, Zou, B, Yeo, YH et al. (2020) Global prevalence, incidence, and outcomes of non-obese or lean non-alcoholic fatty liver disease: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol 8, 739752.CrossRefGoogle Scholar
Azzu, V, Vacca, M, Virtue, S et al. (2020) Adipose tissue-liver cross talk in the control of whole-body metabolism: implications in nonalcoholic fatty liver disease. Gastroenterology 158, 18991912.CrossRefGoogle ScholarPubMed
Kim, D, Chung, GE, Kwak, MS et al. (2016) Body fat distribution and risk of incident and regressed nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 14, 132138.e134.10.1016/j.cgh.2015.07.024CrossRefGoogle ScholarPubMed
Eguchi, Y, Eguchi, T, Mizuta, T et al. (2006) Visceral fat accumulation and insulin resistance are important factors in nonalcoholic fatty liver disease. J Gastroenterol 41, 462469.CrossRefGoogle ScholarPubMed
Motamed, N, Khonsari, MR, Rabiee, B et al. (2017) Discriminatory ability of visceral adiposity index (VAI) in diagnosis of metabolic syndrome: a population based study. Exp Clin Endocrinol Diabetes 125, 202207.Google ScholarPubMed
Yu, SJ, Kim, W, Kim, D et al. (2015) Visceral obesity predicts significant fibrosis in patients with nonalcoholic fatty liver disease. Medicine 94, e2159e2159.10.1097/MD.0000000000002159CrossRefGoogle ScholarPubMed
Ohki, T, Tateishi, R, Shiina, S et al. (2009) Visceral fat accumulation is an independent risk factor for hepatocellular carcinoma recurrence after curative treatment in patients with suspected NASH. Gut 58, 839.10.1136/gut.2008.164053CrossRefGoogle ScholarPubMed
Jensen, MD (2008) Role of body fat distribution and the metabolic complications of obesity. J Clin Endocrinol Metab 93, S57S63.10.1210/jc.2008-1585CrossRefGoogle ScholarPubMed
De Carvalho, FG, Justice, JN, Freitas, ECD et al. (2019) Adipose tissue quality in aging: how structural and functional aspects of adipose tissue impact skeletal muscle quality. Nutrients 11, 2553.10.3390/nu11112553CrossRefGoogle ScholarPubMed
Vlassopoulos, A, Combet, E & Lean, ME (2014) Changing distributions of body size and adiposity with age. Int J Obes 38, 857864.CrossRefGoogle ScholarPubMed
Karastergiou, K, Smith, SR, Greenberg, AS et al. (2012) Sex differences in human adipose tissues – the biology of pear shape. Biol Sex Differ 3, 13.10.1186/2042-6410-3-13CrossRefGoogle ScholarPubMed
Mancuso, P & Bouchard, B (2019) The impact of aging on adipose function and adipokine synthesis. Front Endocrinol 10, 137137.10.3389/fendo.2019.00137CrossRefGoogle ScholarPubMed
Eaton, SA & Sethi, JK (2019) Immunometabolic links between estrogen, adipose tissue and female reproductive metabolism. Biology 8, 1.CrossRefGoogle ScholarPubMed
Hughes, VA, Roubenoff, R, Wood, M et al. (2004) Anthropometric assessment of 10-y changes in body composition in the elderly. Am J Clin Nutr 80, 475482.10.1093/ajcn/80.2.475CrossRefGoogle ScholarPubMed
Tchkonia, T, Morbeck, DE, Von Zglinicki, T et al. (2010) Fat tissue, aging, and cellular senescence. Aging Cell 9, 667684.10.1111/j.1474-9726.2010.00608.xCrossRefGoogle ScholarPubMed
Kuk, JL, Saunders, TJ, Davidson, LE et al. (2009) Age-related changes in total and regional fat distribution. Ageing Res Rev 8, 339348.CrossRefGoogle ScholarPubMed
Godoy-Matos, AF, Silva Júnior, WS & Valerio, CM (2020) NAFLD as a continuum: from obesity to metabolic syndrome and diabetes. Diabetol Metab Syndr 12, 60.10.1186/s13098-020-00570-yCrossRefGoogle ScholarPubMed
Polyzos, SA, Perakakis, N & Mantzoros, CS (2019) Fatty liver in lipodystrophy: a review with a focus on therapeutic perspectives of adiponectin and/or leptin replacement. Metabolism 96, 6682.10.1016/j.metabol.2019.05.001CrossRefGoogle ScholarPubMed
Longo, M, Zatterale, F, Naderi, J et al. (2019) Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int J Mol Sci 20, 2358.10.3390/ijms20092358CrossRefGoogle ScholarPubMed
Sethi, JK & Vidal-Puig, AJ (2007) Thematic review series: adipocyte biology. Adipose tissue function and plasticity orchestrate nutritional adaptation. J Lipid Res 48, 12531262.10.1194/jlr.R700005-JLR200CrossRefGoogle ScholarPubMed
Cawthorn, WP, Heyd, F, Hegyi, K et al. (2007) Tumour necrosis factor-alpha inhibits adipogenesis via a beta-catenin/TCF4(TCF7L2)-dependent pathway. Cell Death Differ 14, 13611373.10.1038/sj.cdd.4402127CrossRefGoogle Scholar
Rutkowski, JM, Stern, JH & Scherer, PE (2015) The cell biology of fat expansion. J Cell Biol 208, 501512.CrossRefGoogle ScholarPubMed
Halberg, N, Khan, T, Trujillo, ME et al. (2009) Hypoxia-inducible factor 1alpha induces fibrosis and insulin resistance in white adipose tissue. Mol Cell Biol 29, 44674483.10.1128/MCB.00192-09CrossRefGoogle ScholarPubMed
Blüher, M (2009) Adipose tissue dysfunction in obesity. Exp Clin Endocrinol Diabetes 117, 241250.CrossRefGoogle ScholarPubMed
Gastaldelli, A (2017) Insulin resistance and reduced metabolic flexibility: cause or consequence of NAFLD? Clin Sci 131, 27012704.CrossRefGoogle ScholarPubMed
Byrne, CD (2013) Ectopic fat, insulin resistance and non-alcoholic fatty liver disease. Proc Nutr Soc 72, 412419.10.1017/S0029665113001249CrossRefGoogle ScholarPubMed
Caso, G, McNurlan, MA, Mileva, I et al. (2013) Peripheral fat loss and decline in adipogenesis in older humans. Metabolism 62, 337340.CrossRefGoogle ScholarPubMed
Schipper, BM, Marra, KG, Zhang, W et al. (2008) Regional anatomic and age effects on cell function of human adipose-derived stem cells. Ann Plast Surg 60, 538544.10.1097/SAP.0b013e3181723bbeCrossRefGoogle ScholarPubMed
Palmer, AK & Kirkland, JL (2016) Aging and adipose tissue: potential interventions for diabetes and regenerative medicine. Exp Gerontol 86, 97105.10.1016/j.exger.2016.02.013CrossRefGoogle ScholarPubMed
Xu, M, Palmer, AK, Ding, H et al. (2015) Targeting senescent cells enhances adipogenesis and metabolic function in old age. eLife 4, e12997.10.7554/eLife.12997CrossRefGoogle ScholarPubMed
Grove, KL, Fried, SK, Greenberg, AS et al. (2010) A microarray analysis of sexual dimorphism of adipose tissues in high-fat-diet-induced obese mice. Int J Obes 34, 9891000.10.1038/ijo.2010.12CrossRefGoogle ScholarPubMed
Macotela, Y, Boucher, J, Tran, TT et al. (2009) Sex and depot differences in adipocyte insulin sensitivity and glucose metabolism. Diabetes 58, 803812.CrossRefGoogle ScholarPubMed
Phillips, GB, Jing, T & Heymsfield, SB (2008) Does insulin resistance, visceral adiposity, or a sex hormone alteration underlie the metabolic syndrome? Studies in women. Metabolism 57, 838844.10.1016/j.metabol.2008.01.029CrossRefGoogle ScholarPubMed
Lovejoy, JC, Champagne, CM, de Jonge, L et al. (2008) Increased visceral fat and decreased energy expenditure during the menopausal transition. Int J Obes 32, 949958.10.1038/ijo.2008.25CrossRefGoogle ScholarPubMed
Abdulnour, J, Doucet, E, Brochu, M et al. (2012) The effect of the menopausal transition on body composition and cardiometabolic risk factors: a Montreal-Ottawa new emerging team group study. Menopause 19, 760767.10.1097/gme.0b013e318240f6f3CrossRefGoogle ScholarPubMed
Leeners, B, Geary, N, Tobler, PN et al. (2017) Ovarian hormones and obesity. Hum Reprod Update 23, 300321.CrossRefGoogle ScholarPubMed
DiStefano, JK (2020) NAFLD and NASH in postmenopausal women: implications for diagnosis and treatment. Endocrinology 161.CrossRefGoogle ScholarPubMed
Babaei, P, Mehdizadeh, R, Ansar, MM et al. (2010) Effects of ovariectomy and estrogen replacement therapy on visceral adipose tissue and serum adiponectin levels in rats. Menopause Int 16, 100104.10.1258/mi.2010.010028CrossRefGoogle ScholarPubMed
Rogers, NH, Perfield, JW III, Strissel, KJ et al. (2009) Reduced energy expenditure and increased inflammation are early events in the development of ovariectomy-induced obesity. Endocrinology 150, 21612168.CrossRefGoogle ScholarPubMed
Lizcano, F & Guzmán, G (2014) Estrogen deficiency and the origin of obesity during menopause. Biomed Res Int 2014, 757461.CrossRefGoogle ScholarPubMed
Cooke, PS & Naaz, A (2004) Role of estrogens in adipocyte development and function. Exp Biol Med 229, 11271135.CrossRefGoogle ScholarPubMed
Frank, AP, de Souza Santos, R, Palmer, BF et al. (2019) Determinants of body fat distribution in humans may provide insight about obesity-related health risks. J Lipid Res 60, 17101719.10.1194/jlr.R086975CrossRefGoogle ScholarPubMed
Funcke, JB & Scherer, PE (2019) Beyond adiponectin and leptin: adipose tissue-derived mediators of inter-organ communication. J Lipid Res 10, 16481684.10.1194/jlr.R094060CrossRefGoogle Scholar
Montague, CT, Farooqi, IS, Whitehead, JP et al. (1997) Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387, 903908.CrossRefGoogle ScholarPubMed
Polyzos, SA, Aronis, KN, Kountouras, J et al. (2016) Circulating leptin in non-alcoholic fatty liver disease: a systematic review and meta-analysis. Diabetologia 59, 3043.10.1007/s00125-015-3769-3CrossRefGoogle ScholarPubMed
Polyzos, SA, Kountouras, J & Mantzoros, CS (2015) Leptin in nonalcoholic fatty liver disease: a narrative review. Metabolism 64, 6078.10.1016/j.metabol.2014.10.012CrossRefGoogle ScholarPubMed
Jiménez-Cortegana, C, García-Galey, A, Tami, M et al. (2021) Role of leptin in non-alcoholic fatty liver disease. Biomedicines 9, 762.10.3390/biomedicines9070762CrossRefGoogle ScholarPubMed
Polyzos, SA, Kountouras, J, Zavos, C et al. (2011) The potential adverse role of leptin resistance in nonalcoholic fatty liver disease: a hypothesis based on critical review of the literature. J Clin Gastroenterol 45, 5054.CrossRefGoogle ScholarPubMed
Saad, MF, Damani, S, Gingerich, RL et al. (1997) Sexual dimorphism in plasma leptin concentration. J Clin Endocrinol Metab 82, 579584.Google ScholarPubMed
Castracane, VD, Kraemer, RR, Franken, MA et al. (1998) Serum leptin concentration in women: effect of age, obesity, and estrogen administration. Fertil Steril 70, 472477.CrossRefGoogle ScholarPubMed
Isidori, AM, Strollo, F, Morè, M et al. (2000) Leptin and aging: correlation with endocrine changes in male and female healthy adult populations of different body weights. J Clin Endocrinol Metab 85, 19541962.10.1210/jcem.85.5.6572CrossRefGoogle ScholarPubMed
Boutari, C & Mantzoros, CS (2020) Adiponectin and leptin in the diagnosis and therapy of NAFLD. Metabolism 103, 154028.CrossRefGoogle ScholarPubMed
Maeda, N, Shimomura, I, Kishida, K et al. (2002) Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nat Med 8, 731737.10.1038/nm724CrossRefGoogle ScholarPubMed
Ouchi, N, Kihara, S, Arita, Y et al. (1999) Novel modulator for endothelial adhesion molecules: adipocyte-derived plasma protein adiponectin. Circulation 100, 24732476.CrossRefGoogle ScholarPubMed
Tilg, H & Hotamisligil, GS (2006) Nonalcoholic fatty liver disease: cytokine-adipokine interplay and regulation of insulin resistance. Gastroenterology 131, 934945.CrossRefGoogle ScholarPubMed
Yamauchi, T, Kamon, J, Minokoshi, Y et al. (2002) Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med 8, 12881295.10.1038/nm788CrossRefGoogle ScholarPubMed
Polyzos, SA, Toulis, KA, Goulis, DG et al. (2011) Serum total adiponectin in nonalcoholic fatty liver disease: a systematic review and meta-analysis. Metabolism 60, 313326.CrossRefGoogle ScholarPubMed
Sohara, N, Takagi, H, Kakizaki, S et al. (2005) Elevated plasma adiponectin concentrations in patients with liver cirrhosis correlate with plasma insulin levels. Liver Intr 25, 2832.10.1111/j.1478-3231.2004.0986.xCrossRefGoogle ScholarPubMed
Tietge, UJ, Böker, KH, Manns, MP et al. (2004) Elevated circulating adiponectin levels in liver cirrhosis are associated with reduced liver function and altered hepatic hemodynamics. Am. J Physiol Endocrinol Metab 287, E82E89.10.1152/ajpendo.00494.2003CrossRefGoogle ScholarPubMed
Polyzos, SA, Kountouras, J & Zavos, C (2010) Nonlinear distribution of adiponectin in patients with nonalcoholic fatty liver disease limits its use in linear regression analysis. J Clin Gastroenterol 44, 229230; author reply 230–221.10.1097/MCG.0b013e3181b5ce68CrossRefGoogle ScholarPubMed
Buechler, C, Wanninger, J & Neumeier, M (2011) Adiponectin, a key adipokine in obesity related liver diseases. World J Gastroenterol 17, 28012811.Google ScholarPubMed
Polyzos, SA, Kountouras, J, Zavos, C et al. (2010) The role of adiponectin in the pathogenesis and treatment of non-alcoholic fatty liver disease. Diabetes, Obes Metab 12, 365383.10.1111/j.1463-1326.2009.01176.xCrossRefGoogle ScholarPubMed
Adachi, M & Brenner, DA (2008) High molecular weight adiponectin inhibits proliferation of hepatic stellate cells via activation of adenosine monophosphate-activated protein kinase. Hepatology 47, 677685.CrossRefGoogle ScholarPubMed
Kizer, JR, Arnold, AM, Strotmeyer, ES et al. (2010) Change in circulating adiponectin in advanced old age: determinants and impact on physical function and mortality. The cardiovascular health study all stars study. J Gerontol A, Biol Sci Med Sci 65, 12081214.10.1093/gerona/glq122CrossRefGoogle ScholarPubMed
Adamczak, M, Rzepka, E, Chudek, J et al. (2005) Ageing and plasma adiponectin concentration in apparently healthy males and females. Clin Endocrinol 62, 114118.10.1111/j.1365-2265.2004.02182.xCrossRefGoogle ScholarPubMed
Reilly, SM & Saltiel, AR (2017) Adapting to obesity with adipose tissue inflammation. Nat Rev Endocrinol 13, 633643.CrossRefGoogle ScholarPubMed
Mohamed-Ali, V, Flower, L, Sethi, J et al. (2001) beta-Adrenergic regulation of IL-6 release from adipose tissue: in vivo and in vitro studies. J Clin Endocrinol Metab 86, 58645869.Google ScholarPubMed
Cawthorn, WP & Sethi, JK (2008) TNF-alpha and adipocyte biology. FEBS Lett 582, 117131.CrossRefGoogle ScholarPubMed
Sethi, JK & Hotamisiligil, GS (2021) Metabolic messengers: tumour necrosis factor. Nat Metab 3, 13021312.CrossRefGoogle ScholarPubMed
Hotamisligil, GS, Arner, P, Caro, JF et al. (1995) Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest 95, 24092415.10.1172/JCI117936CrossRefGoogle ScholarPubMed
Hotamisligil, G, Shargill, N & Spiegelman, B (1993) Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Sci 259, 8791.10.1126/science.7678183CrossRefGoogle ScholarPubMed
Ota, T (2013) Chemokine systems link obesity to insulin resistance. Diabetes Metab J 37, 165172.10.4093/dmj.2013.37.3.165CrossRefGoogle ScholarPubMed
Duval, C, Thissen, U, Keshtkar, S et al. (2010) Adipose tissue dysfunction signals progression of hepatic steatosis towards nonalcoholic steatohepatitis in C57Bl/6 mice. Diabetes 59, 31813191.CrossRefGoogle ScholarPubMed
Cao, H, Gerhold, K, Mayers, JR et al. (2008) Identification of a lipokine, a lipid hormone linking adipose tissue to systemic metabolism. Cell 134, 933944.CrossRefGoogle ScholarPubMed
Souza, CO, Teixeira, AAS, Biondo, LA et al. (2020) Palmitoleic acid reduces high fat diet-induced liver inflammation by promoting PPAR-γ-independent M2a polarization of myeloid cells. Biochim Biophys Acta Mol Cell Biol Lipids 1865, 158776.CrossRefGoogle ScholarPubMed
Item, F & Konrad, D (2012) Visceral fat and metabolic inflammation: the portal theory revisited. Obes Rev 13(Suppl. 2), 3039.CrossRefGoogle ScholarPubMed
Kabir, M, Catalano, KJ, Ananthnarayan, S et al. (2005) Molecular evidence supporting the portal theory: a causative link between visceral adiposity and hepatic insulin resistance. Am J Physiol Endocrinol Metab 288, E454E461.10.1152/ajpendo.00203.2004CrossRefGoogle ScholarPubMed
Lundgren, P & Thaiss, CA (2020) The microbiome-adipose tissue axis in systemic metabolism. Am J Physiol-Gastrointest Liver Physiol 318, G717G724.CrossRefGoogle ScholarPubMed
Valdes, AM, Walter, J, Segal, E et al. (2018) Role of the gut microbiota in nutrition and health. Br Med J 361, k2179.10.1136/bmj.k2179CrossRefGoogle ScholarPubMed
Zmora, N, Suez, J & Elinav, E (2019) You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepat 16, 3556.CrossRefGoogle ScholarPubMed
Hu, H, Lin, A, Kong, M et al. (2020) Intestinal microbiome and NAFLD: molecular insights and therapeutic perspectives. J Gastroenterol 55, 142158.CrossRefGoogle ScholarPubMed
Camarillo-Guerrero, LF, Almeida, A, Rangel-Pineros, G et al. (2021) Massive expansion of human gut bacteriophage diversity. Cell 184, 10981109.e1099.CrossRefGoogle ScholarPubMed
Canfora, EE, Meex, RCR, Venema, K et al. (2019) Gut microbial metabolites in obesity, NAFLD and T2DM. Nat Rev Endocrinol 15, 261273.10.1038/s41574-019-0156-zCrossRefGoogle ScholarPubMed
Aron-Wisnewsky, J, Vigliotti, C, Witjes, J et al. (2020) Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepat 17, 279297.CrossRefGoogle ScholarPubMed
Jiang, X, Zheng, J, Zhang, S et al. (2020) Advances in the involvement of gut microbiota in pathophysiology of NAFLD. Front Med 7.10.3389/fmed.2020.00361CrossRefGoogle ScholarPubMed
Le Roy, T, Llopis, M, Lepage, P et al. (2013) Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut 62, 17871794.CrossRefGoogle ScholarPubMed
Bäckhed, F, Ley, RE, Sonnenburg, JL et al. (2005) Host-bacterial mutualism in the human intestine. Science 307, 19151920.CrossRefGoogle ScholarPubMed
Mouzaki, M, Comelli, EM, Arendt, BM et al. (2013) Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 58, 120127.CrossRefGoogle ScholarPubMed
Boursier, J, Mueller, O, Barret, M et al. (2016) The severity of nonalcoholic fatty liver disease is associated with gut dysbiosis and shift in the metabolic function of the gut microbiota. Hepatology 63, 764775.10.1002/hep.28356CrossRefGoogle ScholarPubMed
Loomba, R, Seguritan, V, Li, W et al. (2017) Gut microbiome-based metagenomic signature for non-invasive detection of advanced fibrosis in human nonalcoholic fatty liver disease. Cell Metab 25, 10541062.e1055.CrossRefGoogle ScholarPubMed
Caussy, C, Tripathi, A, Humphrey, G et al. (2019) A gut microbiome signature for cirrhosis due to nonalcoholic fatty liver disease. Nat Commun 10, 1406.CrossRefGoogle ScholarPubMed
Oh, TG, Kim, SM, Caussy, C et al. (2020) A universal gut-microbiome-derived signature predicts cirrhosis. Cell Metab 32, 878888.10.1016/j.cmet.2020.06.005CrossRefGoogle ScholarPubMed
Li, K, Peng, W, Zhou, Y et al. (2020) Host genetic and environmental factors shape the composition and function of gut microbiota in populations living at high altitude. Biomed Res Int 2020, 1482109.Google ScholarPubMed
Miele, L, Valenza, V, La Torre, G et al. (2009) Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 49, 18771887.10.1002/hep.22848CrossRefGoogle ScholarPubMed
Luther, J, Garber, JJ, Khalili, H et al. (2015) Hepatic injury in nonalcoholic steatohepatitis contributes to altered intestinal permeability. Cell Mol Gastroenterol Hepatol 1, 222232.10.1016/j.jcmgh.2015.01.001CrossRefGoogle ScholarPubMed
Kolodziejczyk, AA, Zheng, D, Shibolet, O et al. (2019) The role of the microbiome in NAFLD and NASH. EMBO Mol Med 11, e9302.CrossRefGoogle ScholarPubMed
Ridlon, JM, Kang, DJ, Hylemon, PB et al. (2014) Bile acids and the gut microbiome. Curr Opin Gastroenterol 30, 332338.10.1097/MOG.0000000000000057CrossRefGoogle ScholarPubMed
Bäckhed, F, Manchester, JK, Semenkovich, CF et al. (2007) Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci USA 104, 979984.CrossRefGoogle ScholarPubMed
Bäckhed, F, Ding, H, Wang, T et al. (2004) The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA 101, 1571815723.10.1073/pnas.0407076101CrossRefGoogle ScholarPubMed
Rau, M, Rehman, A, Dittrich, M et al. (2018) Fecal SCFAs and SCFA-producing bacteria in gut microbiome of human NAFLD as a putative link to systemic T-cell activation and advanced disease. United European Gastroenterol J 6, 14961507.CrossRefGoogle ScholarPubMed
Zhou, D, Pan, Q, Xin, F-Z et al. (2017) Sodium butyrate attenuates high-fat diet-induced steatohepatitis in mice by improving gut microbiota and gastrointestinal barrier. World J Gastroenterol 23, 6075.CrossRefGoogle ScholarPubMed
Cani, PD, Amar, J, Iglesias, MA et al. (2007) Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56, 17611772.10.2337/db06-1491CrossRefGoogle ScholarPubMed
Ferslew, BC, Xie, G, Johnston, CK et al. (2015) Altered bile acid metabolome in patients with nonalcoholic steatohepatitis. Dig Dis Sci 60, 33183328.10.1007/s10620-015-3776-8CrossRefGoogle ScholarPubMed
Zhu, L, Baker, SS, Gill, C et al. (2013) Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 57, 601609.CrossRefGoogle ScholarPubMed
Grabherr, F, Grander, C, Effenberger, M et al. (2019) Gut dysfunction and non-alcoholic fatty liver disease. Front Endocrinol 10, 611611.CrossRefGoogle ScholarPubMed
Mells, JE & Anania, FA (2013) The role of gastrointestinal hormones in hepatic lipid metabolism. Semin Liver Dis 33, 343357.CrossRefGoogle ScholarPubMed
Mok, JKW, Makaronidis, JM & Batterham, RL (2019) The role of gut hormones in obesity. Curr Opin Endocr Metab Res 4, 413.10.1016/j.coemr.2018.09.005CrossRefGoogle Scholar
Koukias, N, Buzzetti, E & Tsochatzis, EA (2017) Intestinal hormones, gut microbiota and non-alcoholic fatty liver disease. Minerva Endocrinol 42, 184194.CrossRefGoogle ScholarPubMed
Durkin, LA, Childs, CE & Calder, PC (2021) Omega-3 polyunsaturated fatty acids and the intestinal epithelium – a review. Foods 10, 1.10.3390/foods10010199CrossRefGoogle ScholarPubMed
Harte, AL, da Silva, NF, Creely, SJ et al. (2010) Elevated endotoxin levels in non-alcoholic fatty liver disease. J Inflamm 7, 1515.CrossRefGoogle ScholarPubMed
Kitabatake, H, Tanaka, N, Fujimori, N et al. (2017) Association between endotoxemia and histological features of nonalcoholic fatty liver disease. World J Gastroenterol 23, 712722.CrossRefGoogle ScholarPubMed
Clemente-Postigo, M, Oliva-Olivera, W, Coin-Aragüez, L et al. (2019) Metabolic endotoxemia promotes adipose dysfunction and inflammation in human obesity. Am J Physiol-Endocrinol Metab 316, E319E332.CrossRefGoogle ScholarPubMed
Chang, C-C, Sia, K-C, Chang, J-F et al. (2019) Lipopolysaccharide promoted proliferation and adipogenesis of preadipocytes through JAK/STAT and AMPK-regulated cPLA2 expression. Int J Med Sci 16, 167179.CrossRefGoogle ScholarPubMed
Kim, YS, Unno, T, Kim, BY et al. (2020) Sex differences in gut microbiota. World J Mens Health 38, 4860.CrossRefGoogle ScholarPubMed
Valeri, F & Endres, K (2021) How biological sex of the host shapes its gut microbiota. Front Neuroendocrinol 61, 100912.CrossRefGoogle ScholarPubMed
Sheng, L, Jena, PK, Liu, HX et al. (2017) Gender differences in bile acids and microbiota in relationship with gender dissimilarity in steatosis induced by diet and FXR inactivation. Sci Rep 7, 1748.CrossRefGoogle ScholarPubMed
Rea, MC, O'Sullivan, O, Shanahan, F et al. (2012) Clostridium difficile carriage in elderly subjects and associated changes in the intestinal microbiota. J Clin Microbiol 50, 867875.CrossRefGoogle ScholarPubMed
O'Toole, PW & Jeffery, IB (2015) Gut microbiota and aging. Science 350, 12141215.10.1126/science.aac8469CrossRefGoogle ScholarPubMed
Nagpal, R, Mainali, R, Ahmadi, S et al. (2018) Gut microbiome and aging: physiological and mechanistic insights. Nutr Healthy Aging 4, 267285.CrossRefGoogle ScholarPubMed
Tran, L & Greenwood-Van Meerveld, B (2013) Age-associated remodeling of the intestinal epithelial barrier. J Gerontol A 68, 10451056.CrossRefGoogle ScholarPubMed
Man, AL, Bertelli, E, Rentini, S et al. (2015) Age-associated modifications of intestinal permeability and innate immunity in human small intestine. Clin Sci 129, 515527.CrossRefGoogle ScholarPubMed
Dai, W, Ye, L, Liu, A et al. (2017) Prevalence of nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus: a meta-analysis. Medicine 96, e8179e8179.CrossRefGoogle ScholarPubMed
Cariou, B, Byrne, CD, Loomba, R et al. (2021) Nonalcoholic fatty liver disease as a metabolic disease in humans: a literature review. Diabetes Obes Metab 23, 10691083.10.1111/dom.14322CrossRefGoogle ScholarPubMed
Bilson, J, Scorletti, E, Bindels, LB et al. (2021) Growth differentiation factor-15 and the association between type 2 diabetes and liver fibrosis in NAFLD. Nutr Diabetes 11.10.1038/s41387-021-00170-3CrossRefGoogle ScholarPubMed
Younossi, ZM, Golabi, P, de Avila, L et al. (2019) The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: a systematic review and meta-analysis. J Hepatol 71, 793801.CrossRefGoogle ScholarPubMed
Targher, G, Corey, KE, Byrne, CD et al. (2021) The complex link between NAFLD and type 2 diabetes mellitus – mechanisms and treatments. Nat Rev Gastroenterol Hepat 18, 599612.CrossRefGoogle ScholarPubMed
Jarvis, H, Craig, D, Barker, R et al. (2020) Metabolic risk factors and incident advanced liver disease in non-alcoholic fatty liver disease (NAFLD): a systematic review and meta-analysis of population-based observational studies. PLoS Med 17, e1003100.CrossRefGoogle ScholarPubMed
Kanwar, P, Nelson, JE, Yates, K et al. (2016) Association between metabolic syndrome and liver histology among NAFLD patients without diabetes. BMJ Open Gastroenterol 3, e000114.CrossRefGoogle ScholarPubMed
Kleiner, DE, Brunt, EM, Wilson, LA et al. (2019) Association of histologic disease activity with progression of nonalcoholic fatty liver disease. JAMA Network Open 2, e1912565.CrossRefGoogle ScholarPubMed
Hsiao, PJ, Kuo, KK, Shin, SJ et al. (2007) Significant correlations between severe fatty liver and risk factors for metabolic syndrome. J Gastroenterol Hepatol 22, 21182123.CrossRefGoogle ScholarPubMed
Mantovani, A, Petracca, G, Beatrice, G et al. (2020) Nonalcoholic fatty liver disease and risk of incident diabetes mellitus: an updated meta-analysis of 501 022 adult individuals. Gut 70, 962969.CrossRefGoogle ScholarPubMed
Li, Y, Wang, J, Tang, Y et al. (2017) Bidirectional association between nonalcoholic fatty liver disease and type 2 diabetes in Chinese population: evidence from the Dongfeng-Tongji cohort study. PLoS ONE 12, e0174291.CrossRefGoogle ScholarPubMed
Ballestri, S, Zona, S, Targher, G et al. (2016) Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome. Evidence from a systematic review and meta-analysis. J Gastroenterol Hepatol 31, 936944.CrossRefGoogle ScholarPubMed
Targher, G, Byrne, CD & Tilg, H (2020) NAFLD and increased risk of cardiovascular disease: clinical associations, pathophysiological mechanisms and pharmacological implications. Gut 69, 16911705.CrossRefGoogle ScholarPubMed
Paik, JM, Henry, L, De Avila, L et al. (2019) Mortality related to nonalcoholic fatty liver disease is increasing in the United States. Hepatol Commun 3, 14591471.CrossRefGoogle ScholarPubMed
Przybyszewski, EM, Targher, G, Roden, M et al. (2021) Nonalcoholic fatty liver disease and cardiovascular disease. Clin Liver Dis 17, 1922.10.1002/cld.1017CrossRefGoogle ScholarPubMed
Targher, G, Byrne, CD, Lonardo, A et al. (2016) Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: a meta-analysis. J Hepatol 65, 589600.CrossRefGoogle ScholarPubMed
Khalid, YS, Dasu, NR, Suga, H et al. (2020) Increased cardiovascular events and mortality in females with NAFLD: a meta-analysis. Am J Cardiovasc Dis 10, 258271.Google ScholarPubMed
Stevens, PE & Levin, A (2013) Evaluation and management of chronic kidney disease: synopsis of the kidney disease: improving global outcomes 2012 clinical practice guideline. Ann Intern Med 158, 825830.CrossRefGoogle ScholarPubMed
Musso, G, Gambino, R, Tabibian, JH et al. (2014) Association of non-alcoholic fatty liver disease with chronic kidney disease: a systematic review and meta-analysis. PLoS Med 11, e1001680.CrossRefGoogle ScholarPubMed
Mantovani, A, Zaza, G, Byrne, CD et al. (2018) Nonalcoholic fatty liver disease increases risk of incident chronic kidney disease: a systematic review and meta-analysis. Metabolism 79, 6476.10.1016/j.metabol.2017.11.003CrossRefGoogle ScholarPubMed
Mantovani, A, Petracca, G, Beatrice, G et al. (2020) Non-alcoholic fatty liver disease and risk of incident chronic kidney disease: an updated meta-analysis. Gut 71, 156162.CrossRefGoogle ScholarPubMed
Kaps, L, Labenz, C, Galle, PR et al. (2020) Non-alcoholic fatty liver disease increases the risk of incident chronic kidney disease. United European Gastroenterol J 8, 942948.CrossRefGoogle ScholarPubMed
Simon, TG, Roelstraete, B, Sharma, R et al. (2021) Cancer risk in patients with biopsy-confirmed nonalcoholic fatty liver disease: a population-based cohort study. Hepatology 74, 24102423.10.1002/hep.31845CrossRefGoogle ScholarPubMed
Mantovani, A, Petracca, G, Beatrice, G et al. (2021) Non-alcoholic fatty liver disease and increased risk of incident extrahepatic cancers: a meta-analysis of observational cohort studies. Gut, Epub ahead of print.Google ScholarPubMed
Simon, TG, Roelstraete, B, Khalili, H et al. (2021) Mortality in biopsy-confirmed nonalcoholic fatty liver disease: results from a nationwide cohort. Gut 70, 1375.10.1136/gutjnl-2020-322786CrossRefGoogle ScholarPubMed
Cao, Q, Mak, KM & Lieber, CS (2007) Leptin represses matrix metalloproteinase-1 gene expression in LX2 human hepatic stellate cells. J Hepatol 46, 124133.CrossRefGoogle ScholarPubMed
Cao, Q, Mak, KM, Ren, C et al. (2004) Leptin stimulates tissue inhibitor of metalloproteinase-1 in human hepatic stellate cells: respective roles of the JAK/STAT and JAK-mediated H2O2-dependant MAPK pathways. J Biol Chem 279, 42924304.10.1074/jbc.M308351200CrossRefGoogle ScholarPubMed
Polyzos, SA, Kountouras, J, Zavos, C et al. (2010) The role of adiponectin in the pathogenesis and treatment of non-alcoholic fatty liver disease. Diabetes Obes Metab 12, 365383.10.1111/j.1463-1326.2009.01176.xCrossRefGoogle ScholarPubMed
Baranova, A, Gowder, SJ, Schlauch, K et al. (2006) Gene expression of leptin, resistin, and adiponectin in the white adipose tissue of obese patients with non-alcoholic fatty liver disease and insulin resistance. Obes Surg 16, 11181125.10.1381/096089206778392149CrossRefGoogle ScholarPubMed
Musso, G, Gambino, R, Durazzo, M et al. (2005) Adipokines in NASH: postprandial lipid metabolism as a link between adiponectin and liver disease. Hepatology 42, 11751183.CrossRefGoogle ScholarPubMed
Wong, VW, Hui, AY, Tsang, SW et al. (2006) Metabolic and adipokine profile of Chinese patients with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 4, 11541161.CrossRefGoogle ScholarPubMed
Ajmera, V, Perito, ER, Bass, NM et al. (2017) Novel plasma biomarkers associated with liver disease severity in adults with nonalcoholic fatty liver disease. Hepatology 65, 6577.CrossRefGoogle ScholarPubMed
Jamali, R, Razavizade, M, Arj, A et al. (2016) Serum adipokines might predict liver histology findings in non-alcoholic fatty liver disease. World J Gastroenterol 22, 50965103.CrossRefGoogle ScholarPubMed
Dendi, VSR, Aloor, S, Runkana, A et al. (2015) Elevated serum resistin in non-alcoholic fatty liver disease and steatohepatitis: a meta-analysis: 2149. Am Coll Gastroenterol 110, S896.10.14309/00000434-201510001-02149CrossRefGoogle Scholar
Silswal, N, Singh, AK, Aruna, B et al. (2005) Human resistin stimulates the pro-inflammatory cytokines TNF-alpha and IL-12 in macrophages by NF-kappaB-dependent pathway. Biochem Biophys Res Commun 334, 10921101.CrossRefGoogle ScholarPubMed
Chen, X, Shen, T, Li, Q et al. (2017) Retinol binding protein-4 levels and non-alcoholic fatty liver disease: a community-based cross-sectional study. Sci Rep 7, 45100.CrossRefGoogle ScholarPubMed
Wang, X, Chen, X, Zhang, H et al. (2020) Circulating retinol-binding protein 4 is associated with the development and regression of non-alcoholic fatty liver disease. Diabetes Metab 46, 119128.CrossRefGoogle ScholarPubMed
Liu, Y, Mu, D, Chen, H et al. (2016) Retinol-binding protein 4 induces hepatic mitochondrial dysfunction and promotes hepatic steatosis. J Clin Endocrinol Metab 101, 43384348.CrossRefGoogle ScholarPubMed
Xia, M, Liu, Y, Guo, H et al. (2013) Retinol binding protein 4 stimulates hepatic sterol regulatory element-binding protein 1 and increases lipogenesis through the peroxisome proliferator-activated receptor-γ coactivator 1β-dependent pathway. Hepatology 58, 564575.10.1002/hep.26227CrossRefGoogle ScholarPubMed
Zhang, J, Li, K, Pan, L et al. (2021) Association of circulating adipsin with nonalcoholic fatty liver disease in obese adults: a cross-sectional study. BMC Gastroenterol 21, 131.CrossRefGoogle ScholarPubMed
Yilmaz, Y, Yonal, O, Kurt, R et al. (2011) Serum levels of omentin, chemerin and adipsin in patients with biopsy-proven nonalcoholic fatty liver disease. Scand J Gastroenterol 46, 9197.CrossRefGoogle ScholarPubMed
Qiu, Y, Wang, SF, Yu, C et al. (2019) Association of circulating adipsin, visfatin, and adiponectin with nonalcoholic fatty liver disease in adults: a case-control study. Ann Nutr Metab 74, 4452.CrossRefGoogle ScholarPubMed
Lo, JC, Ljubicic, S, Leibiger, B et al. (2014) Adipsin is an adipokine that improves β cell function in diabetes. Cell 158, 4153.CrossRefGoogle ScholarPubMed
Kukla, M, Zwirska-Korczala, K, Hartleb, M et al. (2010) Serum chemerin and vaspin in non-alcoholic fatty liver disease. Scand J Gastroenterol 45, 235242.CrossRefGoogle ScholarPubMed
Bekaert, M, Verhelst, X, Geerts, A et al. (2016) Association of recently described adipokines with liver histology in biopsy-proven non-alcoholic fatty liver disease: a systematic review. Obes Rev 17, 6880.CrossRefGoogle ScholarPubMed
Cash, JL, Hart, R, Russ, A et al. (2008) Synthetic chemerin-derived peptides suppress inflammation through ChemR23. J Exp Med 205, 767775.CrossRefGoogle ScholarPubMed
Aktas, B, Yilmaz, Y, Eren, F et al. (2011) Serum levels of vaspin, obestatin, and apelin-36 in patients with nonalcoholic fatty liver disease. Metabolism 60, 544549.CrossRefGoogle ScholarPubMed
Montazerifar, F, Bakhshipour, AR, Karajibani, M et al. (2017) Serum omentin-1, vaspin, and apelin levels and central obesity in patients with nonalcoholic fatty liver disease. J Res Med Sci 22, 70.Google ScholarPubMed
Wang, Y, Song, J, Bian, H et al. (2019) Apelin promotes hepatic fibrosis through ERK signaling in LX-2 cells. Mol Cell Biochem 460, 205215.10.1007/s11010-019-03581-0CrossRefGoogle ScholarPubMed
Lv, S-Y, Cui, B, Chen, W-D et al. (2017) Apelin/APJ system: a key therapeutic target for liver disease. Oncotarget 8, 112145112151.CrossRefGoogle ScholarPubMed
Kleiner, DE, Brunt, EM, Van Natta, M et al. (2005) Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 41, 13131321.CrossRefGoogle ScholarPubMed
Eddowes, PJ, Sasso, M, Allison, M et al. (2019) Accuracy of FibroScan controlled attenuation parameter and liver stiffness measurement in assessing steatosis and fibrosis in patients with nonalcoholic fatty liver disease. Gastroenterology 156, 17171730.10.1053/j.gastro.2019.01.042CrossRefGoogle ScholarPubMed
Carpino, G, Del Ben, M, Pastori, D et al. (2020) Increased liver localization of lipopolysaccharides in human and experimental NAFLD. Hepatology 72, 470485.10.1002/hep.31056CrossRefGoogle ScholarPubMed
Song, MJ, Kim, KH, Yoon, JM et al. (2006) Activation of toll-like receptor 4 is associated with insulin resistance in adipocytes. Biochem Biophys Res Commun 346, 739745.10.1016/j.bbrc.2006.05.170CrossRefGoogle ScholarPubMed
Mehta, NN, McGillicuddy, FC, Anderson, PD et al. (2010) Experimental endotoxemia induces adipose inflammation and insulin resistance in humans. Diabetes 59, 172.CrossRefGoogle ScholarPubMed
Douhara, A, Moriya, K, Yoshiji, H et al. (2015) Reduction of endotoxin attenuates liver fibrosis through suppression of hepatic stellate cell activation and remission of intestinal permeability in a rat non-alcoholic steatohepatitis model. Mol Med Rep 11, 16931700.CrossRefGoogle Scholar
Volynets, V, Küper, MA, Strahl, S et al. (2012) Nutrition, intestinal permeability, and blood ethanol levels are altered in patients with nonalcoholic fatty liver disease (NAFLD). Dig Dis Sci 57, 19321941.CrossRefGoogle Scholar
Kema, VH, Mojerla, NR, Khan, I et al. (2015) Effect of alcohol on adipose tissue: a review on ethanol mediated adipose tissue injury. Adipocyte 4, 225231.CrossRefGoogle ScholarPubMed
Chen, X, Zhang, Z, Li, H et al. (2020) Endogenous ethanol produced by intestinal bacteria induces mitochondrial dysfunction in non-alcoholic fatty liver disease. J Gastroenterol Hepatol 35, 20092019.CrossRefGoogle ScholarPubMed
Chen, J & Vitetta, L (2020) Gut Microbiota metabolites in NAFLD pathogenesis and therapeutic implications. Int J Mol Sci 21, 5214.CrossRefGoogle ScholarPubMed
Wang, X, He, G, Peng, Y et al. (2015) Sodium butyrate alleviates adipocyte inflammation by inhibiting NLRP3 pathway. Sci Rep 5, 12676.CrossRefGoogle ScholarPubMed
Ohira, H, Fujioka, Y, Katagiri, C et al. (2013) Butyrate attenuates inflammation and lipolysis generated by the interaction of adipocytes and macrophages. J Atheroscler Thromb 20, 425442.CrossRefGoogle ScholarPubMed
Mollica, MP, Mattace Raso, G, Cavaliere, G et al. (2017) Butyrate regulates liver mitochondrial function, efficiency, and dynamics in insulin-resistant obese mice. Diabetes 66, 14051418.CrossRefGoogle ScholarPubMed
Zhou, D, Chen, Y-W, Zhao, Z-H et al. (2018) Sodium butyrate reduces high-fat diet-induced non-alcoholic steatohepatitis through upregulation of hepatic GLP-1R expression. Exp Mol Med 50, 112.Google ScholarPubMed
Chen, Y-M, Liu, Y, Zhou, R-F et al. (2016) Associations of gut-flora-dependent metabolite trimethylamine-N-oxide, betaine and choline with non-alcoholic fatty liver disease in adults. Sci Rep 6, 19076.CrossRefGoogle ScholarPubMed
León-Mimila, P, Villamil-Ramírez, H, Li, XS et al. (2021) Trimethylamine N-oxide levels are associated with NASH in obese subjects with type 2 diabetes. Diabetes Metab 47, 101183.CrossRefGoogle ScholarPubMed
Gao, X, Liu, X, Xu, J et al. (2014) Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet. J Biosci Bioeng 118, 476481.10.1016/j.jbiosc.2014.03.001CrossRefGoogle ScholarPubMed
Gao, X, Xu, J, Jiang, C et al. (2015) Fish oil ameliorates trimethylamine N-oxide-exacerbated glucose intolerance in high-fat diet-fed mice. Food Funct 6, 11171125.CrossRefGoogle ScholarPubMed
Tan, X, Liu, Y, Long, J et al. (2019) Trimethylamine N-oxide aggravates liver steatosis through modulation of bile acid metabolism and inhibition of farnesoid X receptor signaling in nonalcoholic fatty liver disease. Mol Nutr Food Res 63, e1900257.CrossRefGoogle ScholarPubMed
Ma, L, Li, H, Hu, J et al. (2020) Indole alleviates diet-induced hepatic steatosis and inflammation in a manner involving myeloid cell 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3. Hepatology 72, 11911203.CrossRefGoogle Scholar
Virtue, AT, McCright, SJ, Wright, JM et al. (2019) The gut microbiota regulates white adipose tissue inflammation and obesity via a family of microRNAs. Sci Transl Med 11, eaav1892.CrossRefGoogle Scholar
Figure 0

Fig. 1. Age-related changes in WAT distribution in men and women are associated with increased risk NAFLD, MetS, T2DM and CVD. Sex and age are key factors that modify the risk of NAFLD and NAFLD progression. NAFLD risk is lower in younger women compared to younger men whereas the risk of NAFLD is similar in older men and women (i.e. post-menopausal). Younger women have an increased capacity to preferentially expand gluteal femoral SAT consequently protecting them from NAFLD. Age-associated changes in WAT leads to the redistribution of WAT which is typically characterised by a marked reduction in SAT and increased central metabolically-unfavourable VAT which may partly explain the increased risk of NAFLD associated with ageing in both men and women. WAT distribution is different between men and women, is heavily influenced by ageing and is strongly associated with NAFLD risk. T2DM, type 2 diabetes; MetS, metabolic syndrome; SAT, subcutaneous adipose tissue; VAT, visceral adipose tissue; NAFLD, non-alcoholic fatty liver disease; WAT, white adipose tissue.

Figure 1

Table 1. Changes in circulating concentrations of adipokines and their potential roles in NAFLD

Figure 2

Table 2. Histological definitions of liver fibrosis stages and corresponding liver-biopsy validated liver VCTE cut-off values

Figure 3

Fig. 2. NAFLD is associated with changes in gut microbiota-derived factors that can alter hepatic and WAT function Changes in GM in NAFLD result in alterations in the production of various metabolites/factors that are thought to contribute to NAFLD both directly (i.e. by directly impacting hepatic function) and indirectly through detrimentally influencing WAT function. As highlighted on the left, intestinal eubiosis and healthy gut function (such as that typically found in young individuals) promotes intestinal barrier integrity and homeostasis whilst restricting the production and dissemination of metabolically detrimental factors (such as LPS and endogenous ethanol) into circulation, the liver and WAT. Conversely, as highlighted on the right, intestinal dysbiosis (such as that often associated with older age) leads to alterations in various GM-derived factors/metabolites that impair the function of tight junction-associated proteins located within the intestinal epithelium. Consequently, these changes are thought to contribute to an increased risk of NAFLD both directly (via inducing hepatic mitochondrial function, inflammation and steatosis) and indirectly through detrimentally impacting WAT function (impairing WAT expansion, metabolic flexibility and increasing the production of pro-inflammatory cytokines). The increased production of inflammatory cytokines is thought to lead to a state of chronic low-grade inflammation which is likely to further disrupt the function of tight junction-associated proteins, thus forming a vicious cycle of worsening metabolic dysfunction and NAFLD disease severity. GM, gut microbiota; LPS, lipopolysaccharide; TMAO, trimethylamine N-oxide; NAFLD, non-alcoholic fatty liver disease; WAT, white adipose tissue.

Figure 4

Table 3. Changes in GM-derived factors/metabolites in NAFLD and their proposed effects in WAT and the liver