Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Molecular Aspects of Medicine


journal homepage: www.elsevier.com/locate/mam

Omega-3 fatty acids and non-alcoholic fatty liver disease: Evidence of


efficacy and mechanism of action
Eleonora Scorlettia,b,∗, Christopher D. Byrnea,b
a
Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
b
National Institute for Health Research, Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation
Trust, Southampton, UK

A R T I C LE I N FO A B S T R A C T

Keywords: For many years it has been known that high doses of long chain omega-3 fatty acids are beneficial in the
Non-alcoholic fatty liver disease treatment of hypertriglyceridaemia. Over the last three decades, there has also been a wealth of in vitro and in
NAFLD vivo data that has accumulated to suggest that long chain omega-3 fatty acid treatment might be beneficial to
Fatty liver decrease liver triacylglycerol. Several biological mechanisms have been identified that support this hypothesis;
Liver fibrosis
notably, it has been shown that long chain omega-3 fatty acids have a beneficial effect: a) on bioactive meta-
Omega-3 fatty acids
bolites involved in inflammatory pathways, and b) on alteration of nuclear transcription factor activities such as
n-3 fatty acids
Docosahexanoic acid peroxisome proliferator-activated receptors (PPARs), sterol regulatory element-binding protein 1c (SREBP-1c)
Eicosapentanoic acid and carbohydrate-responsive element-binding protein (ChREBP), involved in inflammatory pathways and liver
Nutrition lipid metabolism. Since the pathogenesis of non alcoholic fatty liver disease (NAFLD) begins with the accu-
mulation of liver lipid and progresses with inflammation and then several years later with development of
fibrosis; it has been thought in patients with NAFLD omega-3 fatty acid treatment would be beneficial in treating
liver lipid and possibly also in ameliorating inflammation. Meta-analyses (of predominantly dietary studies and
small trials) have tended to support the assertion that omega-3 fatty acids are beneficial in decreasing liver lipid,
but recent randomised controlled trials have produced conflicting data. These trials have suggested that omega-3
fatty acid might be beneficial in decreasing liver triglyceride (docosahexanoic acid also possibly being more
effective than eicosapentanoic acid) but not in decreasing other features of steatohepatitis (or liver fibrosis). The
purpose of this review is to discuss recent evidence regarding biological mechanisms by which long chain
omega-3 fatty acids might act to ameliorate liver disease in NAFLD; to consider the recent evidence from ran-
domised trials in both adults and children with NAFLD; and finally to discuss key ‘known unknowns’ that need to
be considered, before planning future studies that are focussed on testing the effects of omega-3 fatty acid
treatment in patients with NAFLD.

1. Introduction to cirrhosis and liver failure (Calzadilla Bertot and Adams, 2016).
NAFLD has become one of the most common causes of chronic liver
Non-alcoholic fatty liver disease (NAFLD) is a pathologic condition disease and liver related mortality worldwide, and is now becoming a
defined by the presence of triglycerides (TG) deposition in the liver major reason for liver transplantation (Rinella, 2015; Sherif et al.,
greater than 5% of the total liver weight (Chalasani et al., 2012; 2016). According to the World Gastroenterology Organization's global
Calzadilla Bertot and Adams, 2016; de Alwis and Day, 2008; Angulo, guidelines (http://www.worldgastroenterology.org/UserFiles/file/
2002). The term NAFLD encompasses a spectrum of liver diseases guidelines/nafld-nash-english-2012.pdf) approximately 10–20% of
where the first stage is characterized by simple steatosis with liver fat people with NAFLD progress to NASH.
accumulation in the hepatocytes (Angulo, 2002; Buzzetti et al., 2016; Although NAFLD also occurs in normal weight people, the bur-
Byrne, 2010; Angulo et al., 2007). The second stage is non-alcoholic geoning epidemic of overweight, obesity and type 2 diabetes is also
steatohepatitis (NASH) characterized by hepatocyte injury due to in- contributing to a marked increase in the burden of chronic disease
flammation, ballooning and possible collagen deposition. NASH is a caused by NAFLD. Whilst national mortality data show that the ma-
progressive form of fatty liver that can worsen over time and may lead jority of liver disease deaths were previously attributed to alcoholic


Corresponding author. Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK.
E-mail address: e.scorletti@soton.ac.uk (E. Scorletti).

https://doi.org/10.1016/j.mam.2018.03.001
Received 14 January 2018; Received in revised form 7 March 2018; Accepted 9 March 2018
0098-2997/ Crown Copyright © 2018 Published by Elsevier Ltd. All rights reserved.

Please cite this article as: Scorletti, E., Molecular Aspects of Medicine (2018), https://doi.org/10.1016/j.mam.2018.03.001
E. Scorletti, C.D. Byrne Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

cirrhosis, there is emerging evidence of the importance of non-alcoholic With regard to EPA and DHA, an adequate intake ranges between 0.25
fatty liver as a risk factor for severe chronic liver disease (Dulai et al., and 2 g/day (corresponding to two fish potions per week, with at least
2017; Whalley et al., 2007). The prevalence of disease progression from one oily fishmeal consumed) (Smit et al., 2009). With a healthy diet, the
NAFLD to NASH is approximately 10–20% in the general population; physiological ratio between omega-6 and omega-3 fatty acids should be
however, the prevalence increases up to 37% in the presence of obesity 4:1. Whereas, in the Western diet, the consumption of linoleic acid for
(Calzadilla Bertot and Adams, 2016). Furthermore, it is now clear that the omega-6 series is 5–20-fold higher than the consumption of α-li-
NAFLD is also a risk factor for type 2 diabetes and cardiovascular dis- nolenic acid. In the presence of a physiological equilibrium, desaturases
ease (Byrne and Targher, 2015) and therefore NAFLD has an important and elongases enzymes exhibit greater affinity to metabolize omega-3
adverse impact not only on hepatology and gastroenterology services fatty acids (Smit et al., 2009). Interestingly, several studies described
but also diabetes, cardiology and cardiac surgery services within the the associations between SNPs of the fatty acid desaturases (FADS1 and
National Health Service. The prognosis of NAFLD is hard to establish FADS2) gene cluster and members of the elongation-of-very-long-chain-
due to the heterogeneity of the condition and to the fact that most fatty-acids (ELOVL) gene family with plasma levels of AA, EPA, DPA
studies are small with relatively short follow-up (Targher and Byrne, and DHA (Lemaitre et al., 2011; Tanaka et al., 2009; Rzehak et al.,
2013). The pathophysiology of NAFLD is complex and involves several 2009; Malerba et al., 2008; Cormier et al., 2014). Hepatic Elovl5, Fads1
metabolic and genetic aspects. The multifactorial mechanisms re- & Fads3 expression are suppressed by increases in dietary omega-3 fatty
sponsible for the development and progression of NAFLD include ge- acids (Wang et al., 2006). Since these enzymes are involved in both
netic polymorphisms (Scorletti et al., 2015) and metabolic factors omega-3 and omega-6 fatty acid synthesis, dietary omega-3 fatty acids
(Levene and Goldin, 2012), such as sedentary lifestyle, increased intake will increase tissue omega-3 and lower tissue omega-6 fatty acid
of energy-rich foods (Veena et al., 2014), malnutrition due to an im- downstream products. FADS1 and FADS2 are genes encoding for key
balanced intake of nutrients (e.g. high fat, high carbohydrates and high enzymes in the omega-3 and omega-6 fatty acid series, the Δ-5 and Δ-6
protein diet (Palmer et al., 2012), low fibre intake (Musso et al., 2003), desaturase respectively. Elongases are enzymes encoded by genes
high fructose intake (Softic et al., 2016)), altered gut microbiota com- within the ELOVL family and are responsible for catalysing the elon-
position (dysbiosis) (Leung et al., 2016; Henao-Mejia et al., 2013; Yu gation of the aliphatic chain of carbons leading to the formation of long-
et al., 2016), obesity (Veena et al., 2014; Scorletti et al., 2011). Several chain omega-3 polyunsaturated fatty acids (Cormier et al., 2014). In-
research studies have attempted to test various treatments in NAFLD: terestingly, FADS and ELOVL polymorphisms are associated with re-
however, these studies have produced controversial results with limited duced Δ-5 and Δ-6 desaturase activity and accumulation of desaturase
success and serious safety concerns about long-term therapy (Ratziu substrates and a reduction of desaturase products (Schaeffer et al.,
et al., 2008, 2010; Sanyal et al., 2010; Caldwell, 2017; Dyson and Day, 2006; Lattka et al., 2010). Omega-3 fatty acid supplementation can
2014). Currently, lifestyle changes may ameliorate steatosis, but improve Δ-5 and Δ-6 desaturase activity through a gene-treatment in-
sometimes weight loss and its maintenance are difficult to achieve (St teraction. Cormier et al. showed that 6 weeks supplementation with 2 g
George et al., 2009; Bellentani et al., 2008). Typically, within a Wes- of EPA plus 1 g of DHA daily in 210 healthy people increased Δ-5 de-
ternized diet, omega-6 fatty acid consumption is markedly greater than saturase activity and decreased Δ-6 desaturase activity increasing
omega-3 fatty acid. The potential consequences of an increased ratio of omega-3 and omega-6 fatty acid plasma levels (Cormier et al., 2014).
omega-6 to omega-3 fatty acid consumption are increased production of
pro-inflammatory arachidonic acid-derived eicosanoids and impaired 3. Pathogenesis of NAFLD and treatment with omega-3 fatty acids
regulation of hepatic and adipose function, predisposing to NAFLD
(Scorletti and Byrne, 2013). Several studies have shown that a diet with 3.1. Obesity, insulin resistance and adipose tissue dysfunction
an inadequate intake of “omega-3 essential fatty acids” is associated
with metabolic syndrome (Mirmiran et al., 2012), cardiovascular dis- Overweight and obesity are major risk factors for several chronic
ease (Simopoulos, 2008), dyslipidaemia and fatty liver disease diseases such as type 2 diabetes, metabolic syndrome and NAFLD. In
(Pachikian et al., 2008; Marsman et al., 2011). particular, excessive intake of fat and carbohydrates can affect lipid
metabolism. Dietary lipids are essential macronutrient in the human
2. Omega-3 fatty acids diet because provide energy for the body. However, there are different
types of dietary fat, some of them are potentially harmful (such as sa-
Omega-3 polyunsaturated fatty acids are long-chain fatty acids turated fat and trans-fat) some other types of fat are beneficial for
characterized by the presence of a double bond (C]C) at the third health (such as monounsaturated fatty acids, polyunsaturated fatty
carbon atom of the hydrocarboxylic chain counting form the methyl acids and omega-3 fatty acids (Scorletti and Byrne, 2013; Jump et al.,
end (Calder, 2017). α-linolenic acid is the simplest fatty acid with an 2015)). For example, a low intake of omega-3 fatty acids as well as a
18-carbon hydrocarboxylic chain and three double bonds. α-linolenic high omega-6/omega-3 fatty acid ratio is associated with a risk to de-
acid is one of the two “essential fatty acids” (the other is linoleic acid velop NAFLD (Jump et al., 2015; Toshimitsu et al., 2007). In the pre-
from the omega-6 series), namely they cannot be synthesised by ani- sence of obesity there is an excessive accumulation of fat, in form of
mals, including humans; therefore, these fatty acids need to be obtained triglycerides, in the adipose tissue, causing expansion of the visceral
through diet. Both α-linolenic acid and linoleic acid are synthesized in and peripheral adipose depots. After a meal, in a physiological condi-
plants and consequently are found in numerous seeds, nuts, and seed tion, the production of insulin by pancreatic β-cells suppresses hepatic
oils such as linseeds (flaxseeds), and their oil, soybean oil, rapeseed oil, glucose output via inhibition of gluconeogenesis and glycogenolysis;
walnuts and sunflower oil (Calder and Yaqoob, 2009). Typically, lin- and increases hepatic glycogen synthesis. In the adipose tissue, insulin
seeds contain 45–55% of fatty acids as α-linolenic acid whereas sun- inhibits lipolysis through suppression of hormone-sensitive lipase and
flower oil is highly rich in linoleic acid. Eicosapentaenoic acid [EPA; upregulates lipogenesis (Bugianesi et al., 2005). The adipose tissue is an
20:5(ω-3)], docosapentaenoic acid [DPA; 22:5(ω-3)], and docosahex- endocrine organ secreting adipokines (such as leptin, adiponectin, re-
aenoic acid [DHA; 22:6(ω-3)] are functionally the most important very sistin, apelin, and visfatin; chemokines such as monocyte chemotactic
long chain highly unsaturated omega-3 fatty acids (Fig. 1). Although protein (MCP)-1 and IL-8; other proinflammatory cytokines such as IL-
these very long chain fatty acids are found in a variety of foods, fish 6, IL-1, angiotensin-II, and TNF-α; and antiinflammatory cytokines such
(especially oily fish) and other seafood are the richest sources of EPA, as IL-10) involved in the regulation of energy balance, glucose home-
DPA and DHA. α -linolenic acid is the precursor of the long chain ostasis, inflammation and immune function (Kalupahana et al., 2011).
omega-3 fatty acid series and the recommended daily intake is 1.6 g/ The overload of triglycerides in the adipose tissue results in an increase
day for men and 1.1 g/day for women (≥0.5% total fat) (Calder, 2017). in adipocyte size accompanied by adipocytes hypertrophy, activation of

2
E. Scorletti, C.D. Byrne Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

Fig. 1. Linoleic acid and α-linoleic acid are converted to polyunsaturated fatty acids by enzymatic elongase and desaturase enzymes. Essential fatty acids are found in seeds and vegetable
oils; whereas polyunsaturated fatty acids are found in fish, fish oil, algae, meat, and eggs.

the death receptor and mitochondrial pathways of adipocyte apoptosis,


macrophage recruitment and activation, and dysregulation of adipokine
secretory patterns (Ibrahim et al., 2011; Feldstein, 2010). The im-
balance between secretions of pro-compared to anti-inflammatory
adipokines causes an inflammatory state of subcutaneous and visceral
adipose tissue. This condition of adipose tissue overload and in-
flammation leads to ectopic deposition of fat in the liver (NAFLD),
metabolic inflammation and insulin resistance. Inflamed adipocytes fail
to suppress intracellular production of reactive oxygen species, with
consequent upregulation of nuclear factor-κB (NF-κB) causing adipo-
cyte secretion of adipokines, furthering inflammation. Omega-3 fatty
acids have anti-inflammatory effects by regulating NF-κB subunit
abundance (Siriwardhana et al., 2012). In a state of insulin resistance,
there is an impairment in insulin-mediated suppression of hepatic
glucose production and inhibition of lipolysis. This leads to hypergly- Fig. 2. Mechanisms potentially responsible for a beneficial effect of omega-3 fatty acid
treatment in NAFLD.
caemia and increases plasma levels of non-esterified fatty acids (NEFAs)
(Neuschwander-Tetri, 2010). The rate of NEFA entering the circulation
is a reflection of the balance between lipolysis of triglycerides in the SREBP-1: SREBP-1a, SREBP-1c, and SREBP-2. SREBP-1a is expressed
adipose tissue and uptake of post prandial NEFA in the adipocyte for re- only at low levels in the liver, SREBP-1c is the predominant isoform in
esterification into triglycerides and storage. In the presence of insulin adult liver. Both SREBP-1a, SREBP-1c activate genes required for fatty
resistance, adipose tissue lipolysis is inhibited as insulin fail to suppress acid synthesis and is involved in the regulation of enzymes that catalyse
hormone-sensitive lipase. In addition, fatty acid esterification is di- lipogenesis, such as acetyl-CoA carboxylase (ACC), fatty acid synthase
minished as this mechanism is dependent on the supply of glycerol-3- (FAS), and TG synthesis. SREBP-2 is involved in cellular cholesterol
phosphate derived from insulin-mediated glucose uptake and glycolysis homeostasis activating the LDL receptor gene and various genes re-
in the adipocyte. The anatomic connection between adipose tissue and quired for cholesterol synthesis (Horton et al., 2002). ChREBP is a
liver through the vasculature, facilitate the venous effluent of visceral transcriptional regulator expressed in the liver that activates glycolytic
fat into the portal vein resulting in NEFA flux to the liver (Bugianesi and lipogenic gene expressions for DNL in response to high glucose.
et al., 2005). Omega-3 fatty acids have beneficial effects in regulating Omega-3 fatty acids regulate ChREBP activity by controlling the cel-
hepatic lipid metabolism, adipose tissue function, and inflammation lular abundance of max-like factor X (MLX), the ChREBP heterodimer
reducing hepatic TG accumulation (Scorletti and Byrne, 2013; partner that is required for ChREBP/MLX to bind DNA and activate
Todorcevic and Hodson, 2015; Nobili et al., 2016) (Fig. 2). glycolytic and lipogenic gene expressions (Xu et al., 2006).
A chronic stimulation of DNL (and reduced fatty acid oxidation) in
the liver can in turn enhance intracellular availability of triglyceride,
3.2. De novo lipogenesis
promoting fatty liver (Lambert et al., 2014). Although in a physiological
condition, the contribution of DNL to VLDL-TG synthesis is only around
The excess intake of dietary carbohydrates together with insulin
5%, this percentage increase to 20–30% in the presence of high car-
resistance promotes hepatic de novo (de novo lipogenesis: DNL) synth-
bohydrate diet (Paglialunga and Dehn, 2016). Omega-3 fatty acids can
esis of free fatty acids form acetyl-coenzyme A (CoA). In the liver, DNL
suppress DNL in two ways: by reducing the activities of the lipogenic
can be increased by activation of transcription factors such as sterol
enzymes acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS)
regulatory element-binding protein-1 (SREBP-1), carbohydrate re-
and by suppressing the nuclear abundance of SREBP1c. Omega-3 fatty
sponse element-binding protein (ChREBP). There are three isoforms of

3
E. Scorletti, C.D. Byrne Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

acids accelerate SREBP1c proteasome-mediated degradation with little Sabatti et al., 2009; Kathiresan et al., 2009; Nakayama et al., 2010).
effect on SREBP1c precursors (Botolin et al., 2006). (Scorletti and Currently, it is unclear whether treatment with omega-3 fatty acids can
Byrne, 2013; Gnoni and Giudetti, 2016) (Fig. 2). affect Δ-5 and Δ-6 desaturase enzyme activities in people with NAFLD
Omega-3 fatty acids increase hepatic fatty acid oxidation and reduce through a gene-DHA + EPA interaction. Interestingly, FADS and
triglycerides (TG) synthesis by inhibiting the expression of SREBP-1c ELOVL polymorphisms are associated with reduced Δ-5 and Δ-6 desa-
and ChREBP activity (nuclear transcription factors that stimulate he- turase activity and accumulation of desaturase substrates and a re-
patic de novo lipogenesis). In adipose tissue, omega-3 fatty acids de- duction of desaturase products (Schaeffer et al., 2006; Lattka et al.,
crease fatty acid and adipokine release and have a potential anti-in- 2010). Omega-3 fatty acid supplementation can improve Δ-5 and Δ-6
flammatory effect by inhibiting macrophage recruitment and desaturase activity through a gene-treatment interaction. Cormier et al.
activation. Omega-3 PUFA treatment also increases abundance of bu- showed that 6 weeks supplementation with 2 g of EPA plus 1 g of DHA
tyrate-producing bacterial species in the intestine reducing lipopoly- daily in 210 healthy people increased Δ-5 desaturase activity and de-
saccharide production. creased Δ-6 desaturase activity increasing omega-3 and decreasing
omega-6 fatty acid plasma levels (Cormier et al., 2014).
3.3. Genetic polymorphisms
3.5. Gut microbiota dysbiosis
3.3.1. Influence of PNPLA3 genotype on NAFLD pathogenesis and
progression The development and consequences of NAFLD involve not only al-
There is evidence showing that the genetic variation in patatin-like tered liver function, but also dysfunction of key extra-hepatic tissues,
phospholipase domain-containing protein-3 (PNPLA3-I148M) influ- such as intestine, with the production of endotoxin and bacterial pro-
ences severity of liver disease, and serum TG concentrations in NAFLD. ducts derived from the gut microbiota. Imbalances in the gut micro-
PNPLA3 encodes a 481-amino acid membrane protein, also called adi- biota (dysbiosis) can lead to metabolic endotoxemia, obesity, insulin
ponutrin, localised in the endoplasmic reticulum and at the surface of resistance and inflammation, all factors implicated in NAFLD. In the
lipid droplets. Adiponutrin promotes either triacylglycerol hydrolase or presence of dysbiosis, there is an increased production of endotoxins
acylglycerol transacetylase activity in the liver. Exome wide association from the Gram-negative bacteria that can damage the intestinal barrier.
study identified a single nucleotide polymorphism (SNP) rs738409 in These endotoxins are then released in the blood stream causing a sub-
exon3 of the PNPLA3 gene, encoding for the isoleucine to methionine clinical elevation in circulating levels of lipopolysaccharide (Cani et al.,
substitution at position 148 (I148M). The loss-of-function 148 M var- 2007). Although this is a very new area of research, there is evidence
iant is characterised by dysfunctional PNPLA3 (adiponutrin) protein that omega-3 PUFA supplementation increases abundance of butyrate-
that accumulates on the surface of lipid droplets (Pingitore et al., 2014). producing bacteria which decrease production of endotoxins (lipopo-
This variant is associated to a loss of lipolytic activity and impairment lysaccharide) and contribute to gut health (Costantini et al., 2017;
of liver lipid catabolism, with consequent lipid droplets remodelling, Watson et al., 2017). Moreover, Rajkumar et al. showed that the
and impairment of VLDL secretions. This would favour hepatocellular combination of probiotic (Bifidobacteria, Lactobacilli, and Streptococcus
accumulation of triglycerides with consequent impairment of mobili- thermophilus) and omega-3 fatty acids supplement (180 mg EPA and
zation of fatty acids from the hepatocytes. Romeo et al. first showed 120 mg of DHA) was more effective in improving lipid profile, insulin
that this PNPLA3-I148M variation was strongly associated with NAFLD sensitivity and inflammatory biomarkers in overweight healthy adults,
(Romeo et al., 2008). than probiotic alone (Fig. 2).

3.4. Fads 4. Omega-3 fatty acid treatment for NAFLD

Fatty acid desaturase 1 and fatty acid desaturase 2 (FADS1 and Several studies have attempted to test the effects of long chain
FADS2) genes encode for Δ-5 and Δ-6 desaturases enzymes. This are key omega-3 fatty acid treatment in NAFLD. However, these studies have
enzymes mainly expressed in the liver and are responsible for catalysing produced controversial results with limited success and serious safety
the formation of double bonds at the Δ-5 and Δ-6 positions in long chain concerns about long-term therapy (Ratziu et al., 2008, 2010; Sanyal
polyunsaturated fatty acids, respectively. Both genes, FADS1 and et al., 2010; Caldwell, 2017; Dyson and Day, 2014; Jump et al., 2018).
FADS2, are oriented head-to-head and localized in a cluster on chro- Lifestyle changes (exercise and diet) (Glass et al., 2015; Sofi and Casini,
mosome 11 (11q12–13.1). Elongases are enzymes responsible for cat- 2014) may ameliorate steatosis, but sometimes weight loss and its
alysing the elongation of the aliphatic chain of carbons adding two maintenance are difficult to achieve (St George et al., 2009; Bellentani
carbon units to the carboxylic end of a fatty acid chain, leading to the et al., 2008). Studies that have attempted to treat NAFLD by targeting
formation of long-chain omega-3 and omega-6 polyunsaturated fatty specific pathways in the pathogenesis of NAFLD have to date met with
acids (Leonard et al., 2004). Elongases are encoded by members of the limited success. The use of medications for glucose control and insulin
elongation-of-very-long-chain-fatty-acids (ELOVL) gene family located resistance (metformin, glitazones (Ratziu et al., 2008; Ratziu et al.,
on chromosome 6 (Alsaleh et al., 2014). Several genome-wide asso- 2010) or GLP1 agonist) (Lomonaco et al., 2012), lipid metabolism
ciation (GWA) studies have shown that single nucleotide polymorph- (PPAR and FXR agonists (Musso et al., 2010; Nakade et al., 2017), and
isms (SNPs) in both FADS1-FADS2 gene clusters and ELOVL gene family oxidative stress (vitamin E) (Sanyal et al., 2010) produced variable
were strongly associated with: higher EPA and lower DHA proportions results. These relatively small trials have also generated controversy,
(Lemaitre et al., 2011; Al-Hilal et al., 2013), NAFLD (Wang et al., not least because any positive effects of treatment are limited by side-
2015), metabolic syndrome, (Truong et al., 2009) and dyslipidaemia effects and concerns about long term safety of glitazones (Murphy and
(Lemaitre et al., 2011; Tanaka et al., 2009; Rzehak et al., 2009; Malerba Rodgers, 2007) and the high dose of vitamin E required (Dietrich et al.,
et al., 2008; Cormier et al., 2014; Al-Hilal et al., 2013). These studies 2009). Initial clinical trials testing the effects of omega-3 fatty acid
have also shown that low Δ5-desaturase enzyme activity was associated treatment in NAFLD differed markedly in four cardinal areas: 1) dura-
with accumulation of desaturase substrates and low desaturase pro- tion of the treatment, (Calzadilla Bertot and Adams, 2016) composition
ducts (Schaeffer et al., 2006; Lattka et al., 2010). Other investigations of the omega-3 fatty acid treatment, (de Alwis and Day, 2008) dosage of
have found that the concurrence of low Δ-5 desaturase activity and high omega-3 fatty acids, and (Angulo, 2002) testing for both adherence to
Δ6-desaturase activity was associated with dyslipidaemia, suggesting the omega-3 intervention and for contamination with omega-3 fatty
that heritable differences in omega-3 and omega-6 fatty acids meta- acids obtained from other readily available sources (Table 1).
bolism also influenced plasma lipid profiles (Aulchenko et al., 2009; These differences in the design of the clinical trial has added to the

4
Table 1
Studies investigating the effect of omega-3 fatty acids in patients with NAFLD.

Authors, year Study design Intervention Population Outcome measurements Results Comments

Hatzitolios et al. Interventional Fish oil (15 ml daily; 64 Patients with non-alcoholic fatty liver disease Liver function, lipid levels and Ultrasonography showed No control group.
(2004) DHA = 1.58 g/d and EPA associated with hyperlipidaemia liver ultrasonography resolution of fatty liver in 35% of A significant decrease (13%)
E. Scorletti, C.D. Byrne

(Hatzitolios 2.25 g/d for 24 weeks); Fish oil: Group A (n = 23) NAFLD with predominant patients in Group A, 61% in Group in BMI was found only in the
et al., 2004) atorvastatin (20 mg/daily); hypertriglyceridemia; Atorvastatin: Group B B, and in 86% in Group C Orlistat group.
orlistat (120 mg thrice daily) (n = 28): NAFLD with predominant (p < 0.001, Group C vs. A).
for 6 months hypercholesterolemia; Orlistat: Group C (n = 21):
overweight patients with NAFLD
Capanni et al. Open-label Oral administration 56 patients with NAFLD (42 subjects receiving Liver function, omega-6/ Improvement in AST (P = 0.003), Absence of
(2006) of omega-3 PUFA, 1-g therapy; 14 controls) omega-3 ratio, liver US and ALT (P = 0.002), GGT (P = 0.03), blinding and
(Capanni et al., capsule/day (EPA liver perfusion by DPI and TG (P = 0.02) randomization
2006) 375 mg/d and DHA
625 mg/d)
for 12 months
Spadaro et al. Randomized; AHA diet + 2 g/day 36 patients with Liver fat assessed by Reduction in ALT (P < 0.01), TG Lack of placebo, and the non
(2008) open-label n-3 PUFA (fish oil) vs AHA diet NAFLD (18 subjects receiving therapy + AHA diet; abdominal US, ALT, AST, TNF- (P < 0.01), serum TNF-α blinding of
(Spadaro et al., for 6 months 18 controls AHA diet alone) α serum levels, and HOMA (P < 0.05) and HOMA (IR) participants and investigators;
2008) (P < 0.05) the amount of EPA and DHA in
fish oil was not reported.
Zhu et al. (2008) Randomized Seal oils 2 g/three times a day 134 patients with NAFLD Liver function test fatty liver Decrease in ALT, TG, LDL The amount of EPA and DHA
(Zhu et al., Placebo-control plus caloric restriction determined by ultrasound assessed by US (P < 0.05); complete fatty liver in fish oil and the composition
2008) to 25-30 kcal/d) (66 subjects receiving regression (P = 0.004) of placebo was not reported.
vs placebo Therapy; 68 receiving placebo)
for 6 months
Vega et al. (2008) Open label with Fish oil (9 g/d, EPA 17 patients with previous elevated liver fat on MRS Liver fat content assessed by Improvement of plasma Causes of liver
(Vega et al., washout period 4.63 g/d and DHA (17patientis: 4 weeks on placebo oil followed by 8 MRS; liver enzymes, TG and triglyceride level by disease other than NAFLD

5
2008) between placebo and 2.15 g/d) vs weeks on fish oil treatment) adiponectin levels (P < 0.03), VLDL + IDL were not excluded and
treatment Placebo (P < 0.03), ApoB (P < 0.03) alcohol intake was not
capsules consisted of and liver fat content. reported; short duration
6.5 g/d of vaccenic acid, 0.9 g/d
of linoleic acid and 1.1 g/d of
palmitic acid
for 2 months
Tanaka et al. (2008) Interventional Purified EPA ethyl ester (2.7 g/ 23 patients with biopsy proven NASH Improvement in steatosis, Improvement of steatosis, fibrosis No control group; only 7
(Tanaka et al., d) fibrosis and ballooning; and ballooning in 6 out of 7 patients consented to undergo
2008) for 12 months improvement of liver enzymes. subjects who underwent second post-treatment liver biopsy.
liver biopsy.
Cussons et al. Double blind, Fish oil (4 g/d: 1.08 g/d EPA 25 patients with polycystic ovary syndrome and Liver fat measured by MRS Improvement of liver fat In the fish oil supplement,
(2009) (Cussons crossover study with and 2.24 g/d DHA); Placebo NAFLD determined by percentage on MRS after fish oil DHA concentration was higher
et al., 2009) 4 weeks wash out olive oil (4 g/d) MRS treatment (14.8 fish oil vs 18.2% than EPA. This may have
for 2 months placebo). contributed to the
(8 weeks of fish oil or olive oil; improvement of liver fat
wash out period 4 weeks) percentage.
Nobili et al. (2011) Randomized DHA (250 and 60 children with biopsy proven NAFLD. Primary: change in liver fat Improvement of liver fat detected
(Nobili et al., 500 mg/day); Placebo: germ oil DHA 250 mg/d (n = 20), DHA 500 mg/d (n = 20) or content as detected by US; by US - DHA 250 mg vs placebo
2011) for 6 months Placebo (n = 20) secondary: changes in ALT, TG (p < 0.001) and DHA 500 mg vs
and BMI placebo (p = 0.01)
Sanyal et al. (2014) Double-blind Purified EPA ethyl ester 243 patients with biopsy proven NASH. Improvement of the NAS score No significant effect in hepatic fat
(Sanyal et al., randomised (1.8 mg/day, and 2.7 g/day); EPA 1.8 g/d (n = 82) by 2 points or more with and enzymes, insulin resistance
2014) Placebo-controlled Placebo (composition EPA 2.7 g/d (n = 86) contribution form more than 1 and
not reported) Placebo (n = 75) parameter and no worsening inflammatory markers
for 12 months of fibrosis
Scorletti et al. Double-blind Purified EPA-EE (1.84 g/day) Patients with NAFLD Reduction of liver fat Erythrocyte DHA but not These results supported the
(2014) randomised and confirmed by biopsy, measured by magnetic DHA + EPA enrichment was evidence that DHA might be
Placebo-controlled MRS, computed resonance spectroscopy (MRI)
(continued on next page)
Molecular Aspects of Medicine xxx (xxxx) xxx–xxx
Table 1 (continued)

Authors, year Study design Intervention Population Outcome measurements Results Comments

(Scorletti et al., DHA-EE (1.52 g/day) tomography or and improvement of two associated with a reduction in more effective than EPA in
2014a) For 15–18 months ultrasound fibrosis scores liver fat. decreasing liver fat.
DHA + EPA (n = 51)
E. Scorletti, C.D. Byrne

Placebo (n = 52)
Pacifico et al. Double-blinded, 250 mg/day algal oil (39% DHA Children with NAFLD Change in hepatic fat fraction DHA supplementation reduced
(2015) (Pacifico parallel-group, ≈97.5 mg/day). Low-caloric diagnosed by MRI as estimated by MRI hepatic fat by 53.4% and the
et al., 2015) randomized, placebo diet DHA + diet (n = 25) hepatic fat fraction from 14% to
controlled (25–30 kcal/kg/day) Placebo + diet (n = 26) 6.5% assessed by MRI.
and daily exercise
(60 min/day, 5 times/week)
For 6 months
Dasarathy et al. Prospective, EPA (2.16 g/d) and DHA 37 patients with well controlled diabetes and Improvement of ≥2 points in EPA/DHA supplement did not No information regarding
(2015) randomized, double (1.44 g/d); Placebo: corn oil biopsy proven NASH the NAS determined by liver provide beneficial effect over erythrocytes enrichment after
(Dasarathy blind placebo- For 12 months within 6 months prior to the start of study. biopsy placebo in NASH patients with treatment. No information
et al., 2015) controlled study EPA/DHA (n = 18) diabetes regarding compliance with
Placebo (n = 19) supplements, diet or life style.
Argo et al. (2015) Double-blind, EPA (1.05 g/day) and DHA 34 patients with NASH Decrease of at least two points Significantly decreased liver fat
(Argo et al., randomized (0.75 g/day), aerobic exercise at diagnosed by biopsy in the NAS score (p = 0.0009) and markers of liver
2015) placebo-controlled least 150 min/week and EPA + DHA + diet + exercise (n = 17) injury but not NAS score.
decrease energy intake Placebo + diet + exercise (n = 17)
by 500–1000 calories/day
for 12 months
Boyraz et al. (2015) Double-blinded, Omega-3 fatty acids 1 g/d Obese adolescents with NAFLD Improvement in liver The combination of omega-3 fatty No mention was made
(Boyraz et al., randomized, placebo (containing 720 mg omega-3; Omega-3 fatty acids (n = 56) functions, liver brightness and acids and lifestyle change showed about the placebo
2015) controlled 380 mg EPA and 200 mg DHA) Placebo (n = 52) insulin resistance a greater improvement in hepatic composition.
plus a calorie restriction diet fat compared with placebo. The composition of omega-3

6
with fatty acid supplement was
25-30 kcal/kg/day and available on supplement's
Physical activity (one hour, manufacture website, no
three times per week). information was mentioned in
Placebo (no composition the article
available)
For 12 months
Janczyk et al. Randomized, double- Omega-3 fatty acids (DHA and 64 Overweight/obese children with NAFLD. Decreased ALT Omega-3 fatty acids did not Liver steatosis was a secondary
(2015) (Janczyk blind, placebo EPA 3:2 proportion); Omega-3 fatty acids (n = 30) activity by = 0.3 times the decrease serum outcome, omega-3 fatty acids
et al., 2015) controlled Treatment was adjusted for Placebo (N = 34) upper limit of normal ALT did not affect liver steatosis on
weight: < 40 kg: ultrasound
DHA = 0.27 g/d and
EPA = 0.18 g/d; 40–60 kg:
DHA = 0.53 g/d and
EPA = 0.36 g/d;
> 60 kg: DHA = 0.8 g/d and
EPA = 0.53 g/d;
Placebo: sunflower oil
For 6 months
Li et al. (2015) (Li Prospective, 50 ml omega-3 fatty acids (1:1 78 patients with biopsy proven NASH. Improvement of NASH Improvements in liver histology No placebo group.
et al., 2015) randomized, ratio Omega-3 fatty acid (n = 39) and metabolic profile compared to BMI reduction in the treatment
controlled EPA:DHA) added into Control (normal saline) (n = 39) control group. group
unblinded daily diet.
Nogueira et al. Double-blind, 0.945 g n-3 per day 50 patients with NAFLD Effect of treatment in patients No changes in the treatment or There was an increase in
(2016) randomized and (605 mg ALA, 143 mg EPA and diagnosed by liver with biopsy proven NASH placebo group were observed plasma omega-3
(Nogueira et al., placebo-controlled 177 mg DHA) biopsy fatty acids in both placebo and
2016) For 6 months Treatment (n = 27) treatment group
Placebo (n = 23)
Molecular Aspects of Medicine xxx (xxxx) xxx–xxx
E. Scorletti, C.D. Byrne Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

confusion about the efficacy of omega-3 fatty acid treatment in NAFLD Welch et al., 2010; Mori and Woodman, 2006; Mori et al., 2000;
(Jump et al., 2018; Parker et al., 2012; de Castro and Calder, 2018). Woodman et al., 2003; Mesa et al., 2004; Leigh-Firbank et al., 2002;
Hatzitolios et al., studied the effect of different lipid lowering treat- Baker et al., 2016). Trials testing omega-3 fatty acid interventions have
ments in patients with NAFLD and hyperlipidaemia for 6 months predominantly focussed on testing the effects of DHA. The effects of
(Hatzitolios et al., 2004). The authors compared the effects of fish oil high dose purified DHA only has been tested in children by Nobili et al.
with those of atorvastatin and orlistat on liver fat. At the end of the (2014). These authors studied the effect of 18 months treatment with
study, there was a general improvement in liver fat assessed by ultra- DHA in children with biopsy proven NAFLD. At the end of the study
sound with 35% improvement in patients receiving fish oil, 61% im- there was an improvement in hepatic steatosis ballooning, inflamma-
provement in patients receiving atorvastatin and 86% improvement in tion but there was no beneficial effect on fibrosis. However, it was not
patients receiving orlistat. However, there was a significant decrease considered ethical to subject the children in the placebo-arm of the trial
(13%) in BMI in the orlistat group resulting in a significant reduction in to an end of study liver biopsy. The combination of EPA + DHA has
liver fat in the orlistat group compared with the fish oil group been tested in several clinical trials. Dasarathy et al. tested the effect of
(p < 0.001 orlistat vs. fish oil) (Hatzitolios et al., 2004). Capanni, Sofi EPA + DHA on patients with diabetes and biopsy proven NASH for 12
and Spadaro suggested a beneficial effect of omega-3 fatty acid treat- months (Dasarathy et al., 2015). At the end of the study, there was no
ment on liver enzymes after 6–12 months intervention (Capanni et al., beneficial effect over placebo in NASH patients with diabetes. The re-
2006; Spadaro et al., 2008; He et al., 2016; Sofi et al., 2010). The sults of the biopsy showed a greater improvement in the placebo group
composition and dosage of omega-3 fatty acids used in a clinical trial is compared to the EPA + DHA group. Interestingly, there was no in-
also potentially very important, as EPA and DHA are not absorbed and formation regarding compliance with supplements, diet or life style
metabolised in the same way. For example, omega-3 fatty acids used in (Dasarathy et al., 2015). In another study, Li et al. evaluated the effect
clinical trials have been fish oil, seal oil, or purified EPA, DHA or of omega-3 fatty acid on NASH diagnosed by liver biopsy (Li et al.,
EPA + DHA. EPA and DHA are metabolised differently and they may 2015). Patients were randomised to receive 50 ml of omega-3 fatty
not have equivalent effects on the liver in NAFLD. EPA supplements acids (1:1 ratio EPA:DHA) added into daily diet or to receive normal
increases blood levels of EPA and DPA, but not DHA; whereas, DHA saline for 6 months. Liver biopsies were undertaken at the beginning
supplements increase blood levels of DHA, DPA and EPA. EPA and DHA and at the end of the study. After 6 months of treatment, the
may not have equivalent effects on oxidative stress (Depner et al., EPA + DHA group showed improvement in steatosis grade, necro-in-
2013), inflammation (Allaire et al., 2016) or fibrosis (Lytle et al., 2015). flammatory grade, fibrosis stage and ballooning score compared to
The range of effects of omega-3 fatty acids on NAFLD and/or NASH may control group. However, both groups showed an increase in physical
depend on the composition and purity of omega-3 fatty acids used in activity and a decrease in BMI at the end of the study (Li et al., 2015). In
the study and on the severity of liver disease. Previous studies used overweight and obese children with NAFLD, Janczyk et al. tested the
mixtures of EPA and DHA with different degree of purification. Two effect of a weight-adjusted dosage of EPA + DHA (Janczyk et al.,
studies tested the effect of purified EPA, one in 2008 (Tanaka et al., 2015). The primary outcome was an improvement of ALT activity
2008) and one in 2014 (Sanyal et al., 2014). The first study investigated by = 0.3 times the upper limit of normal, whereas improvement in liver
the effect of 2.7 g/d of EPA for 12 months on markers of NAFLD and steatosis was a secondary outcome. After 6 months there was a decrease
NASH in 23 patients with liver biopsy proven NASH. However, the end in liver function tests and liver steatosis in both placebo and
of study biopsy was performed in only 7 subjects (Tanaka et al., 2008). EPA + DHA groups. There was no significant reduction of ALT or liver
At the end of the study, the ultrasound showed an improvement in liver steatosis measured by ultrasound in the EPA + DHA group compared to
steatosis in 12 patients and the liver biopsy showed a decrease in placebo (Janczyk et al., 2015). Boyraz et al. studied obese adolescents
steatosis (29%), fibrosis (59%), lobular inflammation (48%), ballooning with NAFLD in a double-blinded, randomized, placebo-controlled trial
(44%) and NAS (39%) (Tanaka et al., 2008). After this small study, (Boyraz et al., 2015) and the intervention was: 1 g/d of omega-3 fatty
Sanyal et al. showed no improvement of NAS score after supple- acids (380 mg of EPA and 200 mg of DHA) (as per the supplement's
mentation with purified EPA in patients with NASH (Sanyal et al., manufacturer website) or placebo (composition not available in the
2014). On the contrary, Pacifico and Nobili showed that DHA was ef- published paper). All adolescents received lifestyle advice with a calorie
fective in reducing liver fat and markers of liver fibrosis (Pacifico et al., restricted diet (25–30 kcal/kg/d) for weight loss and physical activity
2015; Nobili et al., 2013). Cusson et al. showed an improvement in MRS (one hour, three times per week). After 12 months of treatment, there
liver fat percentage after fish oil treatment (14.8 fish oil vs 18.2% was a decrease in weight and hepatic steatosis measured by ultrasound
placebo). Interestingly, the fish oil supplement contained higher con- in both groups. The improvements were more pronounced in the PUFA
centration of DHA (2.24 g/d) than EPA (1.08 g/d) and this may have group, showing an additional effect of omega-3 fatty acid supple-
contributed to the improvement of liver fat percentage (Cussons et al., mentation in adolescents that underwent a lifestyle change (Boyraz
2009). These opposing results may be explained not only by the com- et al., 2015).
position of omega-3 fatty acid used in the trial, but also by the severity Only one clinical trial the WELCOME study (Wessex Evaluation of
of liver disease in patients recruited to the trials. Omega-3 fatty acids fatty Liver and Cardiovascular markers in NAFLD with OMacor
maybe effective in the early stages of NAFLD, and further studies are thErapy, undertaken by the authors) has assessed erythrocyte EPA and
needed to test the specific effects of DHA on NASH/fibrosis. To date, DHA enrichment to assess compliance during the study (Scorletti et al.,
only one clinical trial has tested the effect of purified EPA in NASH. The 2014a, 2014b, 2015). Using erythrocyte DHA percentage enrichment or
primary outcomes were either a NAS≤3 without worsening of fibrosis erythrocyte EPA percentage enrichment, we were able to test the spe-
on a drop in NAS by two or more points with no worsening of fibrosis cific contribution of each omega-3 fatty acid. Thus, it was possible to
(Sanyal et al., 2014). The authors reported no beneficial effect on the test associations between percentage DHA enrichment (or percentage
histologic features of NASH or on serum triglyceride levels. The very EPA enrichment), and changes in liver fat percentage measured by
modest effect of EPA on triglyceride levels supports the possibility that magnetic resonance spectroscopy (Scorletti et al., 2014a). In this study,
the dosage of EPA may have been too low, as we might have expected a we showed an independent association between a decrease in liver fat
triglyceride-lowering effective even though patients did not have hy- percentage and erythrocyte DHA enrichment (but not with erythrocyte
pertriglyceridaemia. In addition, supplementation with EPA may sup- EPA enrichment).
presses the conversion of EPA to DHA (Itakura et al., 2011). There is
also evidence that EPA and DHA metabolism may be different in men
and women. For example, in men, the conversion of EPA to DHA
is < 1%, whereas in women is up to 9% (Burdge and Calder, 2006;

7
E. Scorletti, C.D. Byrne Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

5. Areas of uncertainty and unanswered questions for future 5.3. Composition and chemical purity of omega-3 fatty acid treatment
research
All clinical trials used different composition of omega-3 fatty acid
We consider there are several poorly answered questions in con- treatment, some studies used fish oil, others used DHA or EPA or a
sidering whether omega-3 fatty acids have efficacy in ameliorating liver combination of DHA and EPA. One study used the highest licence dose
disease in NAFLD. We have listed some of the important areas of un- of omega-3 fatty acids, 380 mg DHA and 460 mg EPA per g of oil(94).
certainty that we consider still need to be resolved in the planning of Notably, contrary to fish oil preparations used in other studies no lipid-
future studies testing the effects of omega-3 fatty acids on the different soluble vitamins A and D were present (Parker et al., 2012). This
components of liver disease in NAFLD. combination was chosen because of the different characteristics of DHA
and EPA and specifically because they produce different lipid media-
tors: EPA-derived eicosanoids and DHA-derived resolvins and pro-
5.1. Duration of treatment with omega-3 fatty acids tectins. EPA-derived eicosanoids have an anti-inflammatory effect.;
DHA-derived resolvins and protectins have a role in the resolution of
Omega-3 fatty acid interventions of variable durations have been inflammation and have thus been described as ‘‘specialized pro-resol-
tested in different studies (Parker et al., 2012). The duration of the ving lipid mediators’’ (Calder, 2006). The key function of these med-
WELCOME study intervention (minimum 15 months, maximum 18 iators is to reduce liver macrophage infiltration, and induce a specific
months) was informed by a meta-analysis by He et al. showing the ef- hepatic miRNA signature, in order to reduce inflammatory adipokine
fectiveness of ≥12 months treatment with omega-3 fatty acids on AST, expression (Nobili et al., 2016; Rius et al., 2014). Moreover, DHA also
TG and liver fat and studies from Capanni, Sofi and Spadaro showing a inhibits the secretion of apoB-100 by promoting its autophagic de-
beneficial effect of omega-3 fatty acids on liver fat and liver enzymes gradation causing a reduction in VLDL synthesis in the liver (Scorletti
after 6–12 months treatment (Capanni et al., 2006; Spadaro et al., 2008; and Byrne, 2013). With regard to the chemical purity of the treatment,
He et al., 2016; Sofi et al., 2010). Whether it is necessary to intervene only one study selected the most highly purified DHA and EPA available
for > 12 months with omega-3 fatty acids to decrease liver triglyceride on the market(120). This decision was made to avoid contamination
seems unlikely as other intervention that decrease liver lipid (such as with polluting particles and reduce toxicity due to other compounds
weight loss) are known to decrease liver triglyceride concentrations that might be present within the fish oil preparation. This was again a
over much shorter periods of time. However, it seems likely that a substantial departure from previous studies. It is noteworthy that many
longer period of intervention would be needed to have an impact on of the trials testing the effects of different omega-3 fatty acid/fish oil
NASH. Most of the biopsy-based end point trials that have tested the preparations have used various different preparations containing long
effects of other agents (or drugs) on histological end points (focussed on chain omega-3 fatty acids. It is plausible that not only the dose of
improving NAS by = 2 points), have intervened for ∼ 2 years. Such a specific fatty acids, and their variable tissue enrichment, but also the
timescale has been based on an assumption that the longer duration of purity of the preparations and their contaminants, may also influence
intervention is needed to improve features of steatohepatitis such as whether these agents affect liver disease in NAFLD.
inflammation and ballooning of hepatocytes (and to have a potential
impact on fibrosis). Therefore, most of the studies to date that have 5.4. Methods used to assess severity of liver disease in NAFLD and
tested the effects of omega-3 fatty acids on liver disease in NAFLD have heterogeneity of participants recruited within the spectrum of NAFLD
been of too short a duration to be certain of whether there might be any
benefit or not on NASH. Different methods can be used to assess the severity of liver disease.
Four studies measured liver fat with B-mode ultrasound imaging and
liver echotexture was scored on a four-grade scale by comparing it with
5.2. Appropriate dosage of omega-3 fatty acid the right kidney cortical echogenicity (Capanni et al., 2006; Spadaro
et al., 2008; Zhu et al., 2008; Nobili et al., 2011). Although the ultra-
The dosage of 4 g of DHA + EPA daily was selected for the sound scan is a cheap procedure, it is operator dependant and therefore
WELCOME study for two reasons. sometime could be difficult to identify different grade of echogenicity
First, in light of the proven effectiveness of this dose to decrease to quantify liver fat. Two studies used magnetic resonance spectroscopy
serum triglyceride levels in patients with hypertriglyceridemia (Kris- (MRS) to quantify liver fat percentage (Vega et al., 2008; Pacifico et al.,
Etherton et al., 2003). Secondly, it was hypothesised that this dosage 2015) and one study (Scorletti et al., 2014a) used a synergistic com-
would raise the erythrocyte level of EPA and DHA within phospholipids bination of MRS and non-invasive markers of liver function, and NAFLD
above the pre-specified level of 0.7% and 2% respectively in the disease severity. The combination of MRS and markers of liver function
phospholipid fraction of red blood cell membrane preparations. This was a unique feature and a substantial departure at the time from the
level had been previously deemed to be the minimum increase for traditional approach of carrying out assessment of liver disease solely
improving the DHA + EPA sum after treatment, to a value thought to through liver biopsy. Multiple reasons are in support of the choice to
result in decreased risk of CVD (Harris and Von Schacky, 2004). use MRS over liver biopsy. First, liver biopsy is invasive, expensive, and
In the WELCOME study, we were concerned that there might be subject to sampling variability (Bedossa et al., 2003; Colloredo et al.,
variable tissue enrichment with omega-3 fatty acids, despite good 2003; Ratziu et al., 2005; Vuppalanchi et al., 2009; Kleiner and
compliance from the participants, and consequently we tested enrich- Bedossa, 2015), and many investigators currently consider it a high-risk
ment of red blood cell membrane phospholipid (as a validated proxy for procedure that is unacceptable as a research test for monitoring NAFLD.
liver enrichment with DHA and EPA). To our surprise we found very Also, liver biopsy evaluates only a tiny portion (0.05 cm3) of the liver
variable tissue enrichment despite us using the highest licensed dose of (800–1000 cm3), and NAFLD is often a patchy disease. Secondly, MRS is
DHA + EPA (as Omacor or Lovaza). Participants in the trial were also currently considered the non-invasive gold-standard technique for as-
questioned about their compliance and virtually all assured us that they sessing liver fat percentage and has excellent reproducibility and sen-
had been compliant with taking their allocated medication. We also sitivity (Szczepaniak et al., 2005; Machann et al., 2006) with a coeffi-
counted returned unused capsules from participants at 6 monthly in- cient of variance of only 8%, and liver fat signals of only 0.2% are
tervals during the trial to monitor compliance. Since an explanation for clearly evident above the noise level (Machann et al., 2006). It is also
variable tissue enrichment with DHA and EPA is uncertain, it is possible plausible that omega-3 fatty acid treatment only affects liver fat
that liver enrichment is sub-optimal in some individuals leading to little without affecting inflammation or fibrosis in NAFLD. Interestingly, it
response to therapy. has also been shown recently by Argo et al. (2015), that omega-3 fatty

8
E. Scorletti, C.D. Byrne Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

acid treatment caused a significant reduction in liver fat on the paired oxidants such as vitamin E (Sanyal et al., 2010), have produced variable
analysis of MRI image-assisted lipid morphometry, regardless of weight results with ∼50% of patients with NASH, not responding to treatment
loss or gain. In this study, Argo et al. tested the effects of 3 g of fish oil/ and to date it is uncertain why liver disease in some patients responds
day for 1 year in 41 participants with non-cirrhotic NASH and in this to therapy and in other patients, there is no improvement in liver dis-
study each 1000 mg capsule contained 70% total N-3 as triglyceride: ease. With no easy way to identify non responders, there is a reluctance
35% EPA, 25% DHA, 10% other omega-3's, and a scant amount of amongst many clinicians to use agents that have potential side effects
lemon oil. and at the same time might not ameliorate the liver disease. There is
some support for the hypothesis that omega-3 fatty acids treatment
5.5. Effect of genotype to modulate the influence of omega-3 fatty acid might have a beneficial effect on NAFLD, and at the same time produce
treatment minimal side effects (Scorletti and Byrne, 2013). As discussed above
several biological mechanisms have been identified that support this
Scorletti et al. showed that there was a beneficial effect of achieving hypothesis; notably, it has been shown that these omega-3 fatty acids
high levels of DHA tissue enrichment. Specifically, they showed that have a beneficial effect on bioactive metabolites, alteration of tran-
high levels of erythrocyte DHA enrichment (≥2%), was effective for scription factor activity such as peroxisome proliferator-activated re-
reducing liver fat. Patients with high liver fat percentage obtained the ceptors (PPARs), sterol regulatory element-binding protein 1c (SREBP-
most benefit from achieving good DHA enrichment (≥2%): a 6% en- 1c) and carbohydrate-responsive element-binding protein (ChREBP)
richment in DHA resulted in a (6 × 3.3%) = ∼20% decrease in liver fat (Scorletti and Byrne, 2013). There is evidence showing that EPA has
percentage. These results were consistent with previous published lit- modest effects on fatty acid oxidation and triglyceride catabolism by
erature (Nobili et al., 2011, 2013; Pacifico et al., 2015; de Castro and binding to and activating PPAR (α, β, and g) (Xu et al., 1999; Pawar and
Calder, 2018; Della Corte et al., 2016; Yu et al., 2017; Kelley, 2016). Jump, 2003; Tripathy et al., 2014). As far as we know, to date there are
Nobili et al. conducted two clinical trials testing the effect of DHA no clinical studies testing the effect of long term treatment with omega-
supplementation in children with NAFLD. The authors showed specific 3 fatty acids in humans. Nonetheless, there are divergent opinions re-
beneficial effects of DHA on liver biopsy with improvement on hepatic garding the beneficial effect of omega-3 fatty acid treatment on liver fat
steatosis, ballooning, and inflammation NAS, but DHA was ineffective accumulation. In an animal model, supplemental feeding with ALA or
on fibrosis (Nobili et al., 2011, 2013). Pacifico and colleagues, showed EPA and DHA decreased 3-hydroxy-3-methylglutaryl (HMG)-CoA re-
that after 6 months of DHA supplementation in children with NAFLD, ductase activity and increased biliary secretion causing more oxidative
MRS liver fat was reduced by 53.4% (95% CI, 33.4–73.4; p = 0.04) in stress, and consequently more liver damage (Chih-Cheng Chen et al.,
the DHA group. Interestingly, there is evidence showing that PNPLA3- 2013). It is possible that hydroxyl (OH) radical attack on the highly
148 MM genotype adversely affected response to DHA + EPA treat- unsaturated omega-3 fatty acids creates lipid peroxyl radicals and then
ment in NAFLD (Scorletti et al., 2015). Although, the numbers of sub- lipid peroxides which would be the obvious route to creating oxidative
jects studied in the trial with PNPLA3-148 MM genotype was small liver damage initiated by high tissue levels of omega-3 fatty acids. An in
(∼13%), there was a suggestion that subjects with this genotype had vitro study testing the effects of various concentrations and durations of
lower levels of DHA tissue enrichment only (with no effect on EPA incubation of saturated (palmitate), mono-unsaturated fatty acids
enrichment), and also no decrease in liver fat, with omega-3 fatty acid (oleate) and omega-3 fatty acids (eicopentanoate) on triglyceride and
treatment. apo-B metabolism in HepG2 cells showed over 25 years ago that in
PNPLA3 is a multifunctional enzyme with both triacylglycerol lipase contrast to palmitate and oleate, 250 micromolar eicosapentanoate
and acylglycerol O-acyltransferase activity that participates in tria- adversely affected cell viability after only 72 h incubation. Whether
cylglycerol hydrolysis. Whereas, the isoleucine to methionine sub- such high concentrations of serum EPA could ever be reached with high
stitution leads to a loss of lipolytic activity leading to an impairment of dose EPA treatment is uncertain but this evidence suggests that very
lipid catabolism, lipid droplets remodelling, and impairment of VLDL high supplementation levels over months or years of exposure could
secretions, increasing liver fat accumulation and affecting DHA meta- have a deleterious impact on a damaged liver (NAFLD) in vivo (Byrne
bolism. PNPLA3-I148M variant is attached on the surface of lipid dro- and Hales, 1992). In contrast, EPA and DHA supplementation has been
plets reducing TG breakdown leading to lipid retention in the hepato- shown to attenuate a Western diet-mediated induction of hepatic in-
cyte lipid droplet slowing down the conversion of ethyl ester to TGs. flammation and oxidative stress; and in particular, DHA and not EPA
Thus, it is plausible that ethyl esters of omega-3 fatty acid preparations attenuated hepatic fibrosis (Jump et al., 2015, 2016). Thus, although it
maybe less effective in subjects with NAFLD, who have this particular is not proven, this body of evidence suggests it is important to err on the
genotype. side of caution, as further human studies are needed to show that long
term high dose supplementation with omega-3 fatty acids is safe.
5.6. The present state of recommendations and possible future evolution of A recent systematic review and meta-analysis has shown a benefit of
guidelines omega-3 fatty acid treatment on liver fat in NAFLD (Parker et al.,
2012), emphasising that omega-3 fatty acid treatment might confer a
At present, the NICE NAFLD guidelines (ng49) for England and benefit early in the course of the liver disease in NAFLD. We suggest this
Wales do not recommend omega-3 fatty acids to adults with NAFLD body of evidence should not be diminished by the results of a recent
because there is insufficient evidence of their beneficial effect. To date, trial in which treatment involving high doses of EPA failed to show an
these guidelines, the joint European Association for the Study of improvement in NAFLD histological score in patients with more ad-
Diabetes, European Association for the Study of Liver Disease and the vanced disease who had NASH at recruitment (Sanyal et al., 2014). As
European Association for the Study of Obesity (EASL-EASD-EASO, there is considerable evidence that EPA and DHA have different bio-
2016) and the US Guidelines (Chalasani et al., 2012) recommend pio- logical effects and also different metabolism in men and women (e.g. in
glitazone or vitamin E for adults with NASH, whether they have dia- men, the conversion of EPA to DHA is < 1%, whereas in women is up to
betes or not. In the NICE Guidelines these treatments are advocated for 9%) (Mori and Woodman, 2006; Mori et al., 2000; Woodman et al.,
use only in secondary and tertiary care settings because of their side- 2003; Mesa et al., 2004; Leigh-Firbank et al., 2002; Baker et al., 2016) it
effects and concerns about long term safety (Murphy and Rodgers, is possibly not surprising that Sanyal et al. did not find that EPA
2007; Dietrich et al., 2009). treatment had a beneficial effect in NASH.
However, there is increasing evidence that the paradigm on which
these guidelines were originally based is shifting. The results of trials
testing treatment with thiazolidinediones (Ratziu et al., 2008) and anti-

9
E. Scorletti, C.D. Byrne Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

6. Conclusions 362.
Angulo, P., 2002. Nonalcoholic fatty liver disease. N. Engl. J. Med. 346 (16), 1221–1231.
Angulo, P., Hui, J.M., Marchesini, G., Bugianesi, E., George, J., Farrell, G.C., et al., 2007.
There is increasing support for the hypothesis that omega-3 fatty The NAFLD fibrosis score: a noninvasive system that identifies liver fibrosis in pa-
acids treatment might have a beneficial effect on liver disease in NAFLD tients with NAFLD. Hepatology 45 (4), 846–854.
and cause minimal side effects (Scorletti and Byrne, 2013). First, sev- Argo, C.K., Patrie, J.T., Lackner, C., Henry, T.D., de Lange, E.E., Weltman, A.L., et al.,
2015. Effects of n-3 fish oil on metabolic and histological parameters in NASH: a
eral biological mechanisms have been identified that support this hy- double-blind, randomized, placebo-controlled trial. J. Hepatol. 62 (1), 190–197.
pothesis; notably, it has been shown that these omega-3 fatty acids have Aulchenko, Y.S., Ripatti, S., Lindqvist, I., Boomsma, D., Heid, I.M., Pramstaller, P.P.,
a beneficial effect on bioactive metabolites, alteration of transcription et al., 2009. Loci influencing lipid levels and coronary heart disease risk in 16
European population cohorts. Nat. Genet. 41 (1), 47–55.
factor activity such as peroxisome proliferator-activated receptors Baker, E.J., Miles, E.A., Burdge, G.C., Yaqoob, P., Calder, P.C., 2016. Metabolism and
(PPARs), sterol regulatory element-binding protein 1c (SREBP-1c) and functional effects of plant-derived omega-3 fatty acids in humans. Prog. Lipid Res. 64,
carbohydrate-responsive element-binding protein (ChREBP) (Scorletti 30–56.
Bedossa, P., Dargere, D., Paradis, V., 2003. Sampling variability of liver fibrosis in chronic
and Byrne, 2013). Additionally, as discussed above there is some evi-
hepatitis C. Hepatology 38 (6), 1449–1457.
dence of benefit from omega-3 fatty acid treatment from randomised Bellentani, S., Dalle Grave, R., Suppini, A., Marchesini, G., 2008. Behavior therapy for
trials in patients with NAFLD. However, as discussed above, we con- nonalcoholic fatty liver disease: the need for a multidisciplinary approach.
sider that certain factors (‘known unknowns’) need to be considered Hepatology 47 (2), 746–754.
Botolin, D., Wang, Y., Christian, B., Jump, D.B., 2006. Docosahexaneoic acid (22:6,n-3)
further, before planning future studies that are focussed on in- regulates rat hepatocyte SREBP-1 nuclear abundance by Erk- and 26S proteasome-
vestigating the effects of omega-3 fatty acids in NAFLD. Moreover, the dependent pathways. J. Lipid Res. 47 (1), 181–192.
mechanistic basis for omega-3 fatty acid action is based on studies in Boyraz, M., Pirgon, O., Dundar, B., Cekmez, F., Hatipoglu, N., 2015. Long-term treatment
with n-3 polyunsaturated fatty acids as a monotherapy in children with nonalcoholic
rodents and cells. These studies have established that omega-3 fatty fatty liver disease. J. Clin. Res. Pediatr. Endocrinol. 7 (2), 121–127.
acids are pleiotropic regulators of multiple pathways, many of which Bugianesi, E., McCullough, A.J., Marchesini, G., 2005. Insulin resistance: a metabolic
are involved in the onset and progression of NAFLD. Whether these pathway to chronic liver disease. Hepatology 42 (5), 987–1000.
Burdge, G.C., Calder, P.C., 2006. Dietary alpha-linolenic acid and health-related out-
same mechanisms are operative in humans is less clear. Further human comes: a metabolic perspective. Nutr. Res. Rev. 19 (1), 26–52.
studies are needed to better understand the beneficial effect of omega-3 Buzzetti, E., Pinzani, M., Tsochatzis, E.A., 2016 Aug. The multiple-hit pathogenesis of
fatty acids, specifically the different effect of EPA and DHA, on NAFLD. non-alcoholic fatty liver disease (NAFLD). Metabolism 65 (8), 1038–1048. http://dx.
doi.org/10.1016/j.metabol.2015.12.012. Epub 2016 Jan 4.
Byrne, C.D., 2010. Fatty liver: role of inflammation and fatty acid nutrition. Prostagl.
Acknowledgements Leukot. Essent. Fat. Acids 82 (4–6), 265–271.
Byrne, C.D.W.T., Hales, C.N., 1992. Control of Hep G2-cell triacylglycerol and apolipo-
protein B synthesis and secretion by polyunsaturated non-esterified fatty acids and
ES and CDB are supported in part by the Southampton National
insulin. Biochem. J. 288 (Pt 1), 101–107.
Institute for Health Research Biomedical Research. ES is supported by Byrne, C.D., Targher, G., 2015. NAFLD: a multisystem disease. J. Hepatol. 62 (1 Suppl. l),
the Parnell Diabetes Trust. S47–S64.
Calder, P.C., 2006. n-3 polyunsaturated fatty acids, inflammation, and inflammatory
diseases. Am. J. Clin. Nutr. 83 (6 Suppl. l) 1505s–19s.
Financial support Calder, P.C., 2017. Very long-chain n-3 fatty acids and human health: fact, fiction and the
future. Proc. Nutr. Soc. 1–21.
None for this work. Calder, P.C., Yaqoob, P., 2009. Understanding omega-3 polyunsaturated fatty acids. PGM
(Postgrad. Med.) 121 (6), 148–157.
Caldwell, S., 2017. NASH Therapy: omega 3 supplementation, vitamin E, insulin sensi-
Conflicts of interest tizers and statin drugs. Clin. Mol. Hepatol. 23 (2), 103–108.
Calzadilla Bertot, L., Adams, L.A., 2016. The natural course of non-alcoholic fatty liver
disease. Int. J. Mol. Sci. 17 (5).
CDB was Principal Investigator for the Wessex Evaluation of fatty Cani, P.D., Amar, J., Iglesias, M.A., Poggi, M., Knauf, C., Bastelica, D., et al., 2007.
Liver and Cardiovascular markers in NAFLD (non alcoholic fatty liver Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56 (7),
disease) with OMacor thErapy) WELCOME Study. The WELCOME study 1761–1772.
Capanni, M., Calella, F., Biagini, M.R., Genise, S., Raimondi, L., Bedogni, G., et al., 2006.
was a randomised placebo-controlled trial in patients with NAFLD that
Prolonged n-3 polyunsaturated fatty acid supplementation ameliorates hepatic stea-
tested the effects of high dose purified n-3 long chain fatty acids tosis in patients with non-alcoholic fatty liver disease: a pilot study. Aliment
(Omacor/Lovaza Abbott/Pronova) 4 grames o.d. on a range of liver and Pharmacol. Therapeut. 23 (8), 1143–1151.
Chalasani, N., Younossi, Z., Lavine, J.E., Diehl, A.M., Brunt, E.M., Cusi, K., et al., 2012.
cardio-metabolic outcomes. The trial was funded independently of the
The diagnosis and management of non-alcoholic fatty liver disease: practice guideline
drug manufacturers/suppliers whose only input was to provide the by the American gastroenterological association, American association for the study
active compound and matched placebo at no cost. There was no of liver diseases, and American college of gastroenterology. Gastroenterology 142 (7),
pharmaceutical input into the study design, conduct of the trial, ana- 1592–1609.
Chih-Cheng Chen, C.-Y.H., Chaung, Hsio-Chi, Tain, You-Lin, Hsieh, Chih-Sung, Kuo, Fang-
lyses of the data, or writing and submission, of the papers resulting Ying, Yang, Chun-Yu, Huang, Li-Tung, 2013. Fish Omega-3 fatty acids induce liver
from this trial. The trial had core funding from NIHR and also attracted fibrosis in the treatment of bile duct-ligated rats. Dig. Dis. Sci. 58, 440–447.
extra grant support from Diabetes UK, the NHS and the University of Colloredo, G., Guido, M., Sonzogni, A., Leandro, G., 2003. Impact of liver biopsy size on
histological evaluation of chronic viral hepatitis: the smaller the sample, the milder
Pavia (Italy), and was registered with the Diabetes and Hepatology the disease. J. Hepatol. 39 (2), 239–244.
networks. The trial was adopted by the NIHR Portfolio. (www. Cormier, H., Rudkowska, I., Lemieux, S., Couture, P., Julien, P., Vohl, M.C., 2014. Effects
clinicaltrials.gov registration number NCT00760513). of FADS and ELOVL polymorphisms on indexes of desaturase and elongase activities:
results from a pre-post fish oil supplementation. Genes & nutrition 9 (6), 437.
Costantini, L., Molinari, R., Farinon, B., Merendino, N., 2017. Impact of Omega-3 fatty
References acids on the gut microbiota. Int. J. Mol. Sci. 18 (12).
Cussons, A.J., Watts, G.F., Mori, T.A., Stuckey, B.G., 2009. Omega-3 fatty acid supple-
mentation decreases liver fat content in polycystic ovary syndrome: a randomized
Al-Hilal, M., Alsaleh, A., Maniou, Z., Lewis, F.J., Hall, W.L., Sanders, T.A., et al., 2013.
controlled trial employing proton magnetic resonance spectroscopy. J. Clin.
Genetic variation at the FADS1-FADS2 gene locus influences delta-5 desaturase ac-
Endocrinol. Metabol. 94 (10), 3842–3848.
tivity and LC-PUFA proportions after fish oil supplement. J. Lipid Res. 54 (2),
Dasarathy, S., Dasarathy, J., Khiyami, A., Yerian, L., Hawkins, C., Sargent, R., et al., 2015.
542–551.
Double-blind randomized placebo-controlled clinical trial of omega 3 fatty acids for
Allaire, J., Couture, P., Leclerc, M., Charest, A., Marin, J., Lepine, M.C., et al., 2016. A
the treatment of diabetic patients with nonalcoholic steatohepatitis. J. Clin.
randomized, crossover, head-to-head comparison of eicosapentaenoic acid and doc-
Gastroenterol. 49 (2), 137–144.
osahexaenoic acid supplementation to reduce inflammation markers in men and
EASL-EASD-EASO, 2016. Clinical Practice Guidelines for the management of non-alco-
women: the comparing EPA to DHA (ComparED) study. Am. J. Clin. Nutr. 104 (2),
holic fatty liver disease. Diabetologia 59 (6), 1121–1140.
280–287.
de Alwis, N.M., Day, C.P., 2008. Non-alcoholic fatty liver disease: the mist gradually
Alsaleh, A., Maniou, Z., Lewis, F.J., Hall, W.L., Sanders, T.A., O'Dell, S.D., 2014. ELOVL2
clears. J. Hepatol. 48 (Suppl. 1), S104–S112.
gene polymorphisms are associated with increases in plasma eicosapentaenoic and
de Castro, G.S., Calder, P.C., 2018 Feb. Non-alcoholic fatty liver disease and its treatment
docosahexaenoic acid proportions after fish oil supplement. Genes & nutrition 9 (1),
with n-3 polyunsaturated fatty acids. Clin. Nutr. 37 (1), 37–55. http://dx.doi.org/10.

10
E. Scorletti, C.D. Byrne Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

1016/j.clnu.2017.01.006. Epub 2017 Jan 19. Genet. 7 (7) e1002193.


Della Corte, C., Carpino, G., De Vito, R., De Stefanis, C., Alisi, A., Cianfarani, S., et al., Leonard, A.E., Pereira, S.L., Sprecher, H., Huang, Y.S., 2004. Elongation of long-chain
2016. Docosahexanoic acid plus vitamin D treatment improves features of NAFLD in fatty acids. Prog. Lipid Res. 43 (1), 36–54.
children with serum vitamin D deficiency: results from a single centre trial. PLoS One Leung, C., Rivera, L., Furness, J.B., Angus, P.W., 2016. The role of the gut microbiota in
11 (12) e0168216. NAFLD. Nat. Rev. Gastroenterol. Hepatol. 13 (7), 412–425.
Depner, C.M., Philbrick, K.A., Jump, D.B., 2013. Docosahexaenoic acid attenuates hepatic Levene, A.P., Goldin, R.D., 2012. The epidemiology, pathogenesis and histopathology of
inflammation, oxidative stress, and fibrosis without decreasing hepatosteatosis in a fatty liver disease. Histopathology 61 (2), 141–152.
Ldlr(-/-) mouse model of western diet-induced nonalcoholic steatohepatitis. J. Nutr. Li, Y.H., Yang, L.H., Sha, K.H., Liu, T.G., Zhang, L.G., Liu, X.X., 2015. Efficacy of poly-
143 (3), 315–323. unsaturated fatty acid therapy on patients with nonalcoholic steatohepatitis. World J.
Dietrich, M., Jacques, P.F., Pencina, M.J., Lanier, K., Keyes, M.J., Kaur, G., et al., 2009. Gastroenterol. 21 (22), 7008–7013.
Vitamin E supplement use and the incidence of cardiovascular disease and all-cause Lomonaco, R., Ortiz-Lopez, C., Orsak, B., Webb, A., Hardies, J., Darland, C., et al., 2012.
mortality in the Framingham Heart Study: does the underlying health status play a Effect of adipose tissue insulin resistance on metabolic parameters and liver histology
role? Atherosclerosis 205 (2), 549–553. in obese patients with nonalcoholic fatty liver disease. Hepatology 55 (5),
Dulai, P.S., Singh, S., Patel, J., Soni, M., Prokop, L.J., Younossi, Z., et al., 2017 May. 1389–1397.
Increased risk of mortality by fibrosis stage in non-alcoholic fatty liver disease: sys- Lytle, K.A., Depner, C.M., Wong, C.P., Jump, D.B., 2015. Docosahexaenoic acid attenuates
tematic Review and Meta-analysis. Hepatology 65 (5), 1557–1565. http://dx.doi. Western diet-induced hepatic fibrosis in Ldlr-/- mice by targeting the TGFbeta-Smad3
org/10.1002/hep.29085. Epub 2017 Mar 31. pathway. J. Lipid Res. 56 (10), 1936–1946.
Dyson, J., Day, C., 2014. Treatment of non-alcoholic fatty liver disease. Dig. Dis. 32 (5), Machann, J., Thamer, C., Schnoedt, B., Stefan, N., Haring, H.U., Claussen, C.D., et al.,
597–604. 2006. Hepatic lipid accumulation in healthy subjects: a comparative study using
Feldstein, A.E., 2010. Novel insights into the pathophysiology of nonalcoholic fatty liver spectral fat-selective MRI and volume-localized 1H-MR spectroscopy. Magn. Reson.
disease. Semin. Liver Dis. 30 (4), 391–401. Med. 55 (4), 913–917.
Glass, L.M., Dickson, R.C., Anderson, J.C., Suriawinata, A.A., Putra, J., Berk, B.S., et al., Malerba, G., Schaeffer, L., Xumerle, L., Klopp, N., Trabetti, E., Biscuola, M., et al., 2008.
2015. Total body weight loss of > /= 10 % is associated with improved hepatic SNPs of the FADS gene cluster are associated with polyunsaturated fatty acids in a
fibrosis in patients with nonalcoholic steatohepatitis. Dig. Dis. Sci. 60 (4), cohort of patients with cardiovascular disease. Lipids 43 (4), 289–299.
1024–1030. Marsman, H.A., Heger, M., Kloek, J.J., Nienhuis, S.L., van Werven, J.R., Nederveen, A.J.,
Gnoni, A., Giudetti, A.M., 2016. Dietary long-chain unsaturated fatty acids acutely and et al., 2011. Reversal of hepatic steatosis by omega-3 fatty acids measured non-in-
differently reduce the activities of lipogenic enzymes and of citrate Carrier in rat vasively by (1) H-magnetic resonance spectroscopy in a rat model. J. Gastroenterol.
liver. J. Physiol. Biochem. 72 (3), 485–494. Hepatol. 26 (2), 356–363.
Harris, W.S., Von Schacky, C., 2004. The Omega-3 Index: a new risk factor for death from Mesa, M.D., Buckley, R., Minihane, A.M., Yaqoob, P., 2004. Effects of oils rich in eico-
coronary heart disease? Prev. Med. 39 (1), 212–220. sapentaenoic and docosahexaenoic acids on the oxidizability and thrombogenicity of
Hatzitolios, A., Savopoulos, C., Lazaraki, G., Sidiropoulos, I., Haritanti, P., Lefkopoulos, low-density lipoprotein. Atherosclerosis 175 (2), 333–343.
A., et al., 2004. Efficacy of omega-3 fatty acids, atorvastatin and orlistat in non- Mirmiran, P., Hosseinpour-Niazi, S., Naderi, Z., Bahadoran, Z., Sadeghi, M., Azizi, F.,
alcoholic fatty liver disease with dyslipidemia. Indian J. Gastroenterol. : official 2012. Association between interaction and ratio of omega-3 and omega-6 poly-
journal of the Indian Society of Gastroenterology 23 (4), 131–134. unsaturated fatty acid and the metabolic syndrome in adults. Nutrition 28 (9),
He, X.X., Wu, X.L., Chen, R.P., Chen, C., Liu, X.G., Wu, B.J., et al., 2016. Effectiveness of 856–863.
Omega-3 polyunsaturated fatty acids in non-alcoholic fatty liver disease: a meta- Mori, T.A., Woodman, R.J., 2006. The independent effects of eicosapentaenoic acid and
analysis of randomized controlled trials. PLoS One 11 (10) e0162368. docosahexaenoic acid on cardiovascular risk factors in humans. Curr. Opin. Clin.
Henao-Mejia, J., Elinav, E., Thaiss, C.A., Licona-Limon, P., Flavell, R.A., 2013. Role of the Nutr. Metab. Care 9 (2), 95–104.
intestinal microbiome in liver disease. J. Autoimmun. 46, 66–73. Mori, T.A., Burke, V., Puddey, I.B., Watts, G.F., O'Neal, D.N., Best, J.D., et al., 2000.
Horton, J.D., Goldstein, J.L., Brown, M.S., 2002. SREBPs: activators of the complete Purified eicosapentaenoic and docosahexaenoic acids have differential effects on
program of cholesterol and fatty acid synthesis in the liver. J. Clin. Invest. 109 (9), serum lipids and lipoproteins, LDL particle size, glucose, and insulin in mildly hy-
1125–1131. perlipidemic men. Am. J. Clin. Nutr. 71 (5), 1085–1094.
Ibrahim, S.H., Kohli, R., Gores, G.J., 2011. Mechanisms of lipotoxicity in NAFLD and Murphy, C.E., Rodgers, P.T., 2007. Effects of thiazolidinediones on bone loss and fracture.
clinical implications. J. Pediatr. Gastroenterol. Nutr. 53 (2), 131–140. Ann. Pharmacother. 41 (12), 2014–2018.
Itakura, H., Yokoyama, M., Matsuzaki, M., Saito, Y., Origasa, H., Ishikawa, Y., et al., 2011. Musso, G., Gambino, R., De Michieli, F., Cassader, M., Rizzetto, M., Durazzo, M., et al.,
Relationships between plasma fatty acid composition and coronary artery disease. J. 2003. Dietary habits and their relations to insulin resistance and postprandial lipemia
Atherosclerosis Thromb. 18 (2), 99–107. in nonalcoholic steatohepatitis. Hepatology 37 (4), 909–916.
Janczyk, W., Lebensztejn, D., Wierzbicka-Rucinska, A., Mazur, A., Neuhoff-Murawska, J., Musso, G., Gambino, R., Cassader, M., Pagano, G., 2010. A meta-analysis of randomized
Matusik, P., et al., 2015. Omega-3 Fatty acids therapy in children with nonalcoholic trials for the treatment of nonalcoholic fatty liver disease. Hepatology 52 (1), 79–104.
Fatty liver disease: a randomized controlled trial. J. Pediatr. 166 (6), 1358–1363 Nakade, Y., Murotani, K., Inoue, T., Kobayashi, Y., Yamamoto, T., Ishii, N., et al., 2017
e1-3. Dec. Ezetimibe for the treatment of non-alcoholic fatty liver disease: a meta-analysis.
Jump, D.B., Depner, C.M., Tripathy, S., Lytle, K.A., 2015. Potential for dietary omega-3 Hepatol. Res. 47 (13), 1417–1428. http://dx.doi.org/10.1111/hepr.12887. Epub
fatty acids to prevent nonalcoholic fatty liver disease and reduce the risk of primary 2017 Apr 1.
liver cancer. Advances in nutrition (Bethesda, Md) 6 (6), 694–702. Nakayama, K., Bayasgalan, T., Tazoe, F., Yanagisawa, Y., Gotoh, T., Yamanaka, K., et al.,
Jump, D.B., Depner, C.M., Tripathy, S., Lytle, K.A., 2016. Impact of dietary fat on the 2010. A single nucleotide polymorphism in the FADS1/FADS2 gene is associated with
development of non-alcoholic fatty liver disease in Ldlr-/- mice. Proc. Nutr. Soc. 75 plasma lipid profiles in two genetically similar Asian ethnic groups with distinctive
(1), 1–9. differences in lifestyle. Hum. Genet. 127 (6), 685–690.
Jump, D.B., Lytle, K.A., Depner, C.M., Tripathy, S., 2018. Omega-3 polyunsaturated fatty Neuschwander-Tetri, B.A., 2010. Hepatic lipotoxicity and the pathogenesis of nonalco-
acids as a treatment strategy for nonalcoholic fatty liver disease. Pharmacol. holic steatohepatitis: the central role of nontriglyceride fatty acid metabolites.
Therapeut. 181, 108–125. Hepatology 52 (2), 774–788.
Kalupahana, N.S., Claycombe, K.J., Moustaid-Moussa, N., 2011. (n-3) Fatty acids alleviate Nobili, V., Bedogni, G., Alisi, A., Pietrobattista, A., Rise, P., Galli, C., et al., 2011.
adipose tissue inflammation and insulin resistance: mechanistic insights. Advances in Docosahexaenoic acid supplementation decreases liver fat content in children with
nutrition (Bethesda, Md) 2 (4), 304–316. non-alcoholic fatty liver disease: double-blind randomised controlled clinical trial.
Kathiresan, S., Willer, C.J., Peloso, G.M., Demissie, S., Musunuru, K., Schadt, E.E., et al., Arch. Dis. Child. 96 (4), 350–353.
2009. Common variants at 30 loci contribute to polygenic dyslipidemia. Nat. Genet. Nobili, V., Alisi, A., Della Corte, C., Rise, P., Galli, C., Agostoni, C., et al., 2013.
41 (1), 56–65. Docosahexaenoic acid for the treatment of fatty liver: randomised controlled trial in
Kelley, N.S., 2016. Treatment of nonalcoholic fatty liver disease with long-chain n-3 children. Nutrition, metabolism, and cardiovascular diseases. Nutr. Metabol.
polyunsaturated fatty acids in humans. Metab. Syndrome Relat. Disord. 14 (9), Cardiovasc. Dis. 23 (11), 1066–1070.
417–430. Nobili, V., Carpino, G., Alisi, A., De Vito, R., Franchitto, A., Alpini, G., et al., 2014. Role of
Kleiner, D.E., Bedossa, P., 2015. Liver histology and clinical trials for nonalcoholic docosahexaenoic acid treatment in improving liver histology in pediatric nonalco-
steatohepatitis-perspectives from 2 pathologists. Gastroenterology 149 (6), holic fatty liver disease. PLoS One 9 (2) e88005.
1305–1308. Nobili, V., Alisi, A., Musso, G., Scorletti, E., Calder, P.C., Byrne, C.D., 2016. Omega-3 fatty
Kris-Etherton, P.M., Harris, W.S., Appel, L.J., 2003. Omega-3 fatty acids and cardiovas- acids: mechanisms of benefit and therapeutic effects in pediatric and adult NAFLD.
cular disease: new recommendations from the American heart association. Crit. Rev. Clin. Lab Sci. 53 (2), 106–120.
Arterioscler. Thromb. Vasc. Biol. 23 (2), 151–152. Nogueira, M.A., Oliveira, C.P., Ferreira Alves, V.A., Stefano, J.T., Rodrigues, L.S.,
Lambert, J.E., Ramos-Roman, M.A., Browning, J.D., Parks, E.J., 2014. Increased de novo Torrinhas, R.S., et al., 2016. Omega-3 polyunsaturated fatty acids in treating non-
lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver alcoholic steatohepatitis: a randomized, double-blind, placebo-controlled trial. Clin.
disease. Gastroenterology 146 (3), 726–735. Nutr. (Edinb.) 35 (3), 578–586.
Lattka, E., Illig, T., Heinrich, J., Koletzko, B., 2010. Do FADS genotypes enhance our Pachikian, B.D., Neyrinck, A.M., Cani, P.D., Portois, L., Deldicque, L., De Backer, F.C.,
knowledge about fatty acid related phenotypes? Clin. Nutr. (Edinb.) 29 (3), 277–287. et al., 2008. Hepatic steatosis in n-3 fatty acid depleted mice: focus on metabolic
Leigh-Firbank, E.C., Minihane, A.M., Leake, D.S., Wright, J.W., Murphy, M.C., Griffin, alterations related to tissue fatty acid composition. BMC Physiol. 8, 21.
B.A., et al., 2002. Eicosapentaenoic acid and docosahexaenoic acid from fish oils: Pacifico, L., Bonci, E., Di Martino, M., Versacci, P., Andreoli, G., Silvestri, L.M., et al.,
differential associations with lipid responses. Br. J. Nutr. 87 (5), 435–445. 2015. A double-blind, placebo-controlled randomized trial to evaluate the efficacy of
Lemaitre, R.N., Tanaka, T., Tang, W., Manichaikul, A., Foy, M., Kabagambe, E.K., et al., docosahexaenoic acid supplementation on hepatic fat and associated cardiovascular
2011. Genetic loci associated with plasma phospholipid n-3 fatty acids: a meta- risk factors in overweight children with nonalcoholic fatty liver disease. Nutr.
analysis of genome-wide association studies from the CHARGE Consortium. PLoS Metabol. Cardiovasc. Dis. NMCD 25 (8), 734–741.

11
E. Scorletti, C.D. Byrne Molecular Aspects of Medicine xxx (xxxx) xxx–xxx

Paglialunga, S., Dehn, C.A., 2016. Clinical assessment of hepatic de novo lipogenesis in Softic, S., Cohen, D.E., Kahn, C.R., 2016. Role of dietary fructose and hepatic de novo
non-alcoholic fatty liver disease. Lipids Health Dis. 15 (1), 159. lipogenesis in fatty liver disease. Dig. Dis. Sci. 61 (5), 1282–1293.
Palmer, C.N., Maglio, C., Pirazzi, C., Burza, M.A., Adiels, M., Burch, L., et al., 2012. Spadaro, L., Magliocco, O., Spampinato, D., Piro, S., Oliveri, C., Alagona, C., et al., 2008.
Paradoxical lower serum triglyceride levels and higher type 2 diabetes mellitus sus- Effects of n-3 polyunsaturated fatty acids in subjects with nonalcoholic fatty liver
ceptibility in obese individuals with the PNPLA3 148M variant. PLoS One 7 (6) disease. Digest. Liver Disease Official journal of the Italian Society of
e39362. Gastroenterology and the Italian Association for the Study of the Liver 40 (3),
Parker, H.M., Johnson, N.A., Burdon, C.A., Cohn, J.S., O'Connor, H.T., George, J., 2012. 194–199.
Omega-3 supplementation and non-alcoholic fatty liver disease: a systematic review St George, A., Bauman, A., Johnston, A., Farrell, G., Chey, T., George, J., 2009.
and meta-analysis. J. Hepatol. 56 (4), 944–951. Independent effects of physical activity in patients with nonalcoholic fatty liver
Pawar, A., Jump, D.B., 2003. Unsaturated fatty acid regulation of peroxisome pro- disease. Hepatology 50 (1), 68–76.
liferator-activated receptor alpha activity in rat primary hepatocytes. J. Biol. Chem. Szczepaniak, L.S., Nurenberg, P., Leonard, D., Browning, J.D., Reingold, J.S., Grundy, S.,
278 (38), 35931–35939. et al., 2005. Magnetic resonance spectroscopy to measure hepatic triglyceride con-
Pingitore, P., Pirazzi, C., Mancina, R.M., Motta, B.M., Indiveri, C., Pujia, A., et al., 2014. tent: prevalence of hepatic steatosis in the general population. Am. J. Physiol.
Recombinant PNPLA3 protein shows triglyceride hydrolase activity and its I148M Endocrinol. Metabol. 288 (2), E462–E468.
mutation results in loss of function. Biochim. Biophys. Acta 1841 (4), 574–580. Tanaka, N., Sano, K., Horiuchi, A., Tanaka, E., Kiyosawa, K., Aoyama, T., 2008. Highly
Ratziu, V., Charlotte, F., Heurtier, A., Gombert, S., Giral, P., Bruckert, E., et al., 2005. purified eicosapentaenoic acid treatment improves nonalcoholic steatohepatitis. J.
Sampling variability of liver biopsy in nonalcoholic fatty liver disease. Clin. Gastroenterol. 42 (4), 413–418.
Gastroenterology 128 (7), 1898–1906. Tanaka, T., Shen, J., Abecasis, G.R., Kisialiou, A., Ordovas, J.M., Guralnik, J.M., et al.,
Ratziu, V., Giral, P., Jacqueminet, S., Charlotte, F., Hartemann-Heurtier, A., Serfaty, L., 2009. Genome-wide association study of plasma polyunsaturated fatty acids in the
et al., 2008. Rosiglitazone for nonalcoholic steatohepatitis: one-year results of the InCHIANTI Study. PLoS Genet. 5 (1) e1000338.
randomized placebo-controlled fatty liver improvement with rosiglitazone therapy Targher, G., Byrne, C.D., 2013. Clinical Review: nonalcoholic fatty liver disease: a novel
(FLIRT) trial. Gastroenterology 135 (1), 100–110. cardiometabolic risk factor for type 2 diabetes and its complications. J. Clin.
Ratziu, V., Charlotte, F., Bernhardt, C., Giral, P., Halbron, M., Lenaour, G., et al., 2010. Endocrinol. Metabol. 98 (2), 483–495.
Long-term efficacy of rosiglitazone in nonalcoholic steatohepatitis: results of the fatty Todorcevic, M., Hodson, L., 2015. The effect of marine derived n-3 fatty acids on adipose
liver improvement by rosiglitazone therapy (FLIRT 2) extension trial. Hepatology 51 tissue metabolism and function. J. Clin. Med. 5 (1).
(2), 445–453. Toshimitsu, K., Matsuura, B., Ohkubo, I., Niiya, T., Furukawa, S., Hiasa, Y., et al., 2007.
Rinella, M.E., 2015. Nonalcoholic fatty liver disease: a systematic review. Jama 313 (22), Dietary habits and nutrient intake in non-alcoholic steatohepatitis. Nutrition 23 (1),
2263–2273. 46–52.
Rius, B., Titos, E., Moran-Salvador, E., Lopez-Vicario, C., Garcia-Alonso, V., Gonzalez- Tripathy, S., Lytle, K.A., Stevens, R.D., Bain, J.R., Newgard, C.B., Greenberg, A.S., et al.,
Periz, A., et al., 2014. Resolvin D1 primes the resolution process initiated by calorie 2014. Fatty acid elongase-5 (Elovl5) regulates hepatic triglyceride catabolism in
restriction in obesity-induced steatohepatitis. FASEB J. Official publication of the obese C57BL/6J mice. J. Lipid Res. 55 (7), 1448–1464.
Federation of American Societies for Experimental Biology 28 (2), 836–848. Truong, H., DiBello, J.R., Ruiz-Narvaez, E., Kraft, P., Campos, H., Baylin, A., 2009. Does
Romeo, S., Kozlitina, J., Xing, C., Pertsemlidis, A., Cox, D., Pennacchio, L.A., et al., 2008. genetic variation in the Delta6-desaturase promoter modify the association between
Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. alpha-linolenic acid and the prevalence of metabolic syndrome? Am. J. Clin. Nutr. 89
Nat. Genet. 40 (12), 1461–1465. (3), 920–925.
Rzehak, P., Heinrich, J., Klopp, N., Schaeffer, L., Hoff, S., Wolfram, G., et al., 2009. Veena, J., Muragundla, A., Sidgiddi, S., Subramaniam, S., 2014. Non-alcoholic fatty liver
Evidence for an association between genetic variants of the fatty acid desaturase 1 disease: need for a balanced nutritional source. Br. J. Nutr. 112 (11), 1858–1872.
fatty acid desaturase 2 ( FADS1 FADS2) gene cluster and the fatty acid composition of Vega, G.L., Chandalia, M., Szczepaniak, L.S., Grundy, S.M., 2008. Effects of N-3 fatty acids
erythrocyte membranes. Br. J. Nutr. 101 (1), 20–26. on hepatic triglyceride content in humans. Journal of investigative medicine. the
Sabatti, C., Service, S.K., Hartikainen, A.L., Pouta, A., Ripatti, S., Brodsky, J., et al., 2009. official publication of the American Federation for Clinical Research 56 (5), 780–785.
Genome-wide association analysis of metabolic traits in a birth cohort from a founder Vuppalanchi, R., Unalp, A., Van Natta, M.L., Cummings, O.W., Sandrasegaran, K.E.,
population. Nat. Genet. 41 (1), 35–46. Hameed, T., et al., 2009. Effects of liver biopsy sample length and number of readings
Sanyal, A.J., Chalasani, N., Kowdley, K.V., McCullough, A., Diehl, A.M., Bass, N.M., et al., on sampling variability in nonalcoholic Fatty liver disease. Clin. Gastroenterol.
2010. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N. Engl. J. Hepatol. The Official Clinical Practice Journal of the American Gastroenterological
Med. 362 (18), 1675–1685. Association 7 (4), 481–486.
Sanyal, A.J., Abdelmalek, M.F., Suzuki, A., Cummings, O.W., Chojkier, M., 2014. No Wang, Y., Botolin, D., Xu, J., Christian, B., Mitchell, E., Jayaprakasam, B., et al., 2006.
significant effects of ethyl-eicosapentanoic acid on histologic features of nonalcoholic Regulation of hepatic fatty acid elongase and desaturase expression in diabetes and
steatohepatitis in a phase 2 trial. Gastroenterology 147 (2), 377–384 e1. obesity. J. Lipid Res. 47 (9), 2028–2041.
Schaeffer, L., Gohlke, H., Muller, M., Heid, I.M., Palmer, L.J., Kompauer, I., et al., 2006. Wang, L., Athinarayanan, S., Jiang, G., Chalasani, N., Zhang, M., Liu, W., 2015. Fatty acid
Common genetic variants of the FADS1 FADS2 gene cluster and their reconstructed desaturase 1 gene polymorphisms control human hepatic lipid composition.
haplotypes are associated with the fatty acid composition in phospholipids. Hum. Hepatology 61 (1), 119–128.
Mol. Genet. 15 (11), 1745–1756. Watson, H., Mitra, S., Croden, F.C., Taylor, M., Wood, H.M., Perry, S.L., et al., 2017 Sep
Scorletti, E., Byrne, C.D., 2013. Omega-3 fatty acids, hepatic lipid metabolism, and 26. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supple-
nonalcoholic fatty liver disease. Annu. Rev. Nutr. 33, 231–248. ments on the human intestinal microbiota. Gut. http://dx.doi.org/10.1136/gutjnl-
Scorletti, E., Calder, P.C., Byrne, C.D., 2011. Non-alcoholic fatty liver disease and car- 2017-314968. pii: gutjnl-2017-314968, [Epub ahead of print].
diovascular risk: metabolic aspects and novel treatments. Endocrine 40 (3), 332–343. Welch, A.A., Shakya-Shrestha, S., Lentjes, M.A., Wareham, N.J., Khaw, K.T., 2010.
Scorletti, E., Bhatia, L., McCormick, K.G., Clough, G.F., Nash, K., Hodson, L., et al., 2014a. Dietary intake and status of n-3 polyunsaturated fatty acids in a population of fish-
Effects of purified eicosapentaenoic and docosahexaenoic acids in nonalcoholic fatty eating and non-fish-eating meat-eaters, vegetarians, and vegans and the product-
liver disease: results from the Welcome* study. Hepatology 60 (4), 1211–1221. precursor ratio [corrected] of alpha-linolenic acid to long-chain n-3 polyunsaturated
Scorletti, E., Bhatia, L., McCormick, K.G., Clough, G.F., Nash, K., Calder, P.C., et al., fatty acids: results from the EPIC-Norfolk cohort. Am. J. Clin. Nutr. 92 (5),
2014b. Design and rationale of the WELCOME trial: a randomised, placebo controlled 1040–1051.
study to test the efficacy of purified long chainomega-3 fatty acid treatment in non- Whalley, S., Puvanachandra, P., Desai, A., Kennedy, H., 2007. Hepatology outpatient
alcoholic fatty liver disease [corrected]. Contemp. Clin. Trials 37 (2), 301–311. service provision in secondary care: a study of liver disease incidence and resource
Scorletti, E., West, A.L., Bhatia, L., Hoile, S.P., McCormick, K.G., Burdge, G.C., et al., costs. Clin. Med. 7 (2), 119–124.
2015. Treating liver fat and serum triglyceride levels in NAFLD, effects of PNPLA3 Woodman, R.J., Mori, T.A., Burke, V., Puddey, I.B., Barden, A., Watts, G.F., et al., 2003.
and TM6SF2 genotypes: results from the WELCOME trial. J. Hepatol. 63 (6), Effects of purified eicosapentaenoic acid and docosahexaenoic acid on platelet, fi-
1476–1483. brinolytic and vascular function in hypertensive type 2 diabetic patients.
Sherif, Z.A., Saeed, A., Ghavimi, S., Nouraie, S.M., Laiyemo, A.O., Brim, H., et al., 2016. Atherosclerosis 166 (1), 85–93.
Global epidemiology of nonalcoholic fatty liver disease and perspectives on US Xu, H.E., Lambert, M.H., Montana, V.G., Parks, D.J., Blanchard, S.G., Brown, P.J., et al.,
minority populations. Dig. Dis. Sci. 61 (5), 1214–1225. 1999. Molecular recognition of fatty acids by peroxisome proliferator-activated re-
Simopoulos, A.P., 2008. The importance of the omega-6/omega-3 fatty acid ratio in ceptors. Mol. Cell 3 (3), 397–403.
cardiovascular disease and other chronic diseases. Exp. Biol. Med. 233 (6), 674–688. Xu, J., Christian, B., Jump, D.B., 2006. Regulation of rat hepatic L-pyruvate kinase pro-
Siriwardhana, N., Kalupahana, N.S., Fletcher, S., Xin, W., Claycombe, K.J., Quignard- moter composition and activity by glucose, n-3 polyunsaturated fatty acids, and
Boulange, A., et al., 2012. n-3 and n-6 polyunsaturated fatty acids differentially peroxisome proliferator-activated receptor-alpha agonist. J. Biol. Chem. 281 (27),
regulate adipose angiotensinogen and other inflammatory adipokines in part via NF- 18351–18362.
kappaB-dependent mechanisms. J. Nutr. Biochem. 23 (12), 1661–1667. Yu, J., Marsh, S., Hu, J., Feng, W., Wu, C., 2016. The pathogenesis of nonalcoholic fatty
Smit, L.A., Mozaffarian, D., Willett, W., 2009. Review of fat and fatty acid requirements liver disease: interplay between diet, gut microbiota, and genetic background.
and criteria for developing dietary guidelines. Ann. Nutr. Metabol. 55 (1–3), 44–55. Gastroenterol. Res. Pract. 2016 2862173.
Sofi, F., Casini, A., 2014. Mediterranean diet and non-alcoholic fatty liver disease: new Yu, L., Yuan, M., Wang, L., 2017. The effect of omega-3 unsaturated fatty acids on non-
therapeutic option around the corner? World J. Gastroenterol. 20 (23), 7339–7346. alcoholic fatty liver disease: a systematic review and meta-analysis of RCTs. Pakistan
Sofi, F., Giangrandi, I., Cesari, F., Corsani, I., Abbate, R., Gensini, G.F., et al., 2010. Effects J. Med. Sci. 33 (4), 1022–1028.
of a 1-year dietary intervention with n-3 polyunsaturated fatty acid-enriched olive oil Zhu, F.S., Liu, S., Chen, X.M., Huang, Z.G., Zhang, D.W., 2008. Effects of n-3 poly-
on non-alcoholic fatty liver disease patients: a preliminary study. Int. J. Food Sci. unsaturated fatty acids from seal oils on nonalcoholic fatty liver disease associated
Nutr. 61 (8), 792–802. with hyperlipidemia. World J. Gastroenterol. 14 (41), 6395–6400.

12

You might also like