Download as pdf or txt
Download as pdf or txt
You are on page 1of 27

REVIEW

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
The Liver as an Endocrine Organ—Linking NAFLD
and Insulin Resistance
Matthew J. Watt,1 Paula M. Miotto,1 William De Nardo,1 and Magdalene K. Montgomery1

1
Department of Physiology, University of Melbourne, Melbourne, Victoria 3010, Australia
ORCiD numbers: 0000-0003-4064-4188 (M. J. Watt).

ABSTRACT The liver is a dynamic organ that plays critical roles in many physiological processes, including the regulation of systemic
glucose and lipid metabolism. Dysfunctional hepatic lipid metabolism is a cause of nonalcoholic fatty liver disease (NAFLD), the most
common chronic liver disorder worldwide, and is closely associated with insulin resistance and type 2 diabetes. Through the use of
advanced mass spectrometry “omics” approaches and detailed experimentation in cells, mice, and humans, we now understand that
the liver secretes a wide array of proteins, metabolites, and noncoding RNAs (miRNAs) and that many of these secreted factors exert
powerful effects on metabolic processes both in the liver and in peripheral tissues. In this review, we summarize the rapidly evolving
field of “hepatokine” biology with a particular focus on delineating previously unappreciated communication between the liver and
other tissues in the body. We describe the NAFLD-induced changes in secretion of liver proteins, lipids, other metabolites, and miRNAs,
and how these molecules alter metabolism in liver, muscle, adipose tissue, and pancreas to induce insulin resistance. We also synthesize
the limited information that indicates that extracellular vesicles, and in particular exosomes, may be an important mechanism for
intertissue communication in normal physiology and in promoting metabolic dysregulation in NAFLD. (Endocrine Reviews 40:
1367 – 1393, 2019)

N onalcoholic fatty liver disease (NAFLD) is


defined by the accumulation of fat in the
liver, in the absence of excessive alcohol con-
threatening conditions such as cirrhosis, hepato-
cellular carcinoma, and terminal liver failure. Ap-
proximately % of adults in industrialized countries
sumption and other causes of hepatic steatosis, and have NAFLD, and the global epidemic of obesity is
encompasses a spectrum of conditions. Hepatic driving a dramatic increase of NAFLD that is fore-
steatosis is also known as nonalcoholic fatty liver casted to result in increased clinical and economic
(NAFL) and is clinically characterized by the burden (, ).
presence of visible lipid droplets containing tri- Hepatic steatosis/NAFL or early stage NAFLD
glycerides in .% of hepatocytes when thin sec- is often described as a “benign condition” in the
tions are assessed by light microscopy (, ), or a context of liver disease; however, the effects of
threshold of ..% when using proton magnetic steatosis extend beyond the liver. There are strong
resonance spectroscopy (). Liver lipid levels are epidemiological links between NAFLD and type 
regulated by the interplay between the delivery of diabetes, and steatosis is strongly associated with
lipids to the liver and their hepatic uptake, syn- insulin resistance in the liver, and also in pe-
thesis, oxidation, and secretion within very low– ripheral tissues such as skeletal muscle and adi-
density lipoproteins (VLDLs). Alterations in the pose tissue (–). A major focus of current ISSN Print: 0163-769X
equilibrium of one or more of these processes can research is understanding the pathogenic mech- ISSN Online: 1945-7189
promote hepatic steatosis (). NAFL can further anisms linking these comorbidities, and in this Printed: in USA
Copyright © 2019
progress to nonalcoholic steatohepatitis (NASH), review, we focus on the impact of NAFLD on
Endocrine Society
which is defined by hepatocyte ballooning, necrosis altering the endocrine function of the liver, and Received: 24 January 2019
near steatotic hepatocytes, and mild inflammation, how the secretion of proteins, metabolites, and Accepted: 4 April 2019
with or without different stages of fibrosis (). nucleic acids contributes to the pathophysiology First Published Online:
Further progression of NASH can lead to life- of insulin resistance. 25 April 2019

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1367


REVIEW

ESSENTIAL POINTS

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
· Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation in hepatocytes and is the most
common chronic liver disorder worldwide, affecting ~25% to 30% of adults in industrialized countries
· NAFLD is strongly associated with metabolic comorbidities, including obesity, type 2 diabetes, and dyslipidemia
· The liver secretes proteins, metabolites, and noncoding RNA that act as autocrine/paracrine and endocrine factors to
influence metabolism in other tissues
· “Hepatokines” exert pleiotropic effects on lipid and glucose metabolism, as well as insulin action, and their secretion is
impacted by NAFLD
· Liver-derived lipids and metabolites can serve as signaling molecules to regulate insulin action and other metabolic
processes
· Liver-derived miRNAs may regulate glycemic control in NAFLD; however, definitive evidence is lacking

NAFLD and Diabetes widely held idea that hepatic steatosis is a benign
condition, it is now clear that steatosis is closely linked
Prevalence to impaired insulin action and type  diabetes, and it is
Concurrent with the increased global prevalence of an early predictor of metabolic disorders, particularly
obesity (, ), NAFLD has emerged as the most in the normal-weight population (, ). Moreover,
common chronic liver disorder worldwide, affecting hepatic steatosis precedes the development of skeletal
~% to % of adults in industrialized countries, with muscle lipid accumulation, macrophage-related in-
~% of these NAFLD cases classified as NASH (, ). flammation, hepatic, skeletal muscle, and adipose tissue
Importantly, NAFLD prevalence is forecasted to in- insulin resistance, and whole-body hyperglycemia and
crease by % in the next  years, from . to . hyperinsulinemia (–) in mice fed a high-fat diet.
million individuals worldwide, with coincident in- Taken together, these observations are consistent with
creases in NASH and liver-related deaths (). Liver- the notion that changes occurring in the fatty liver alter
related deaths are likely due to the development of paracrine and endocrine functions to cause insulin
fibrosis (), which occurs in ~% of NAFLD patients resistance in key glucoregulatory tissues.
() and is a major clinical concern. Although hepatic steatosis is closely associated with
systemic insulin resistance, it is noteworthy that in-
NAFLD and insulin resistance
sulin resistance also predicts the development of
NAFLD is strongly associated with metabolic
NAFLD. This results primarily from an impaired
comorbidities, including obesity, type  diabetes, and
ability of insulin to suppress adipose tissue lipolysis,
dyslipidemia. Steatosis prevalence is increased in co-
leading to increased delivery of free fatty acids to the
horts with obesity undergoing bariatric surgery, ranging
liver (, ), and from increased de novo lipogenesis
from % to % (–), with NASH identified in %
to % (–). Type  diabetes mellitus is closely () that results from stimulation of lipogenic enzymes
associated with NAFLD, with more than three-fourths via sterol receptor–binding protein c (SREBP-c),
of type  diabetes patients reportedly having NAFLD even in an insulin-resistant state (). There is also
(–). evidence that triglyceride synthesis is increased through
Hepatic steatosis is epidemiologically associated the Kennedy pathway () and that b-oxidation of
with insulin resistance (, –). Results from small fatty acids is decreased in insulin resistance, although
cross-sectional studies using gold-standard measures of the latter is controversial (–).
insulin action have consistently shown that hepatic Although the association between NAFLD and
steatosis, independent of adiposity, is associated with insulin resistance in generally clear in most patients,
impaired insulin action in liver, skeletal muscle, and this does not always hold true for specific geneti-
adipose tissue in both lean individuals and nondiabetic cally determined forms of fatty liver. For example,
individuals with obesity (–). Moreover, relatively a frequent sequence variation (IM) in patatin-
small increases in liver fat are associated with hepatic like phospholipase domain–containing protein 
and skeletal muscle insulin resistance, and further ac- (PNPLA) is strongly associated with fatty liver disease
cumulation of liver fat beyond this relatively low in the absence of insulin resistance or dyslipidemia
threshold (~.% for liver insulin resistance and ~% for (–), and similar dissociations are reported in
muscle insulin resistance) is not associated with more individuals with a single nucleotide polymorphism for
severe insulin resistance (). Thus, in contrast to the acyl-coenzyme A (CoA):diacylglycerol acyltransferase

1368 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

() and the Lys allele in transmembrane  su- secretory vesicles or secretory granules for delivery of
perfamily  (TMSF) (). In fact, the TMSF ac- transmembrane proteins to the plasma membrane and

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
tually protects from cardiovascular disease, despite soluble proteins to the extracellular space. Proteins
high liver fat content and high prevalence for pro- secreted in this manner contain a signal peptide on the
gression to NASH and cirrhosis (). These observa- N terminus and most often contain posttranslational
tions highlight the need to understand the mechanistic modifications that occur at the ER and Golgi (i.e.,
bases of NAFLD for predicting the development of glycosylation) ().
comorbidities and applicability of specific therapeutic
interventions. Nonclassical protein secretion
Proteins that do not contain an N terminus signal
peptide can be secreted as cargo contained within ex-
Liver Endocrine Function tracellular vesicles. There are three main types of ex-
tracellular vesicles that differ in terms of size, type of
Protein secretion—the role of “hepatokines” biogenesis, and composition. Exosomes are the smallest
The role of liver as a major secretory organ has long vesicle, ranging from ~ to  nm in diameter, and are
been appreciated, particularly with respect to its roles in formed within multivesicular bodies that fuse to the
regulating coagulation and hemostasis, but only recently plasma membrane and are released into the circulation
has the potential magnitude for protein secretion be- as exosomes. Microvesicles such as ectosomes and
come apparent. Mass spectrometry–based quantitative endosomes range from  to  nm and are formed
proteomics of human liver has quantified ~, by direct budding from the plasma membrane, whereas
proteins (), which parallels observations in mice (). apoptotic bodies range from  to  nm and are
Given that ~% of the transcripts in liver encode secreted as a byproduct of cell death (). These vesicles
secreted proteins (), there is clearly a large scope for contain protein, lipid, and nucleic acid cargo that
significant and varied protein secretion from the liver. somewhat reflects their cell of origin, and the compo-
The liver is composed of parenchymal cells, in- nents of extracellular vesicles can be rapidly altered in
cluding hepatocytes and bile duct cells, which occupy response to metabolic challenges (), highlighting their
~% of the liver volume, and nonparenchymal cells, potential role in regulating intertissue communication
such as sinusoidal endothelial cells, Kupffer cells, and and metabolism.
hepatic stellate cells. Although there are some differ-
ences in the proteins expressed in these different cell Hepatokines, NAFLD, and insulin resistance
types, the vast majority of liver proteins are expressed in Studies examining hepatocytes isolated from healthy
all liver cell types, and the hepatocyte proteome con- and steatotic mouse livers have used quantitative
stitutes the vast majority of the total liver proteome, proteomics to show that liver steatosis alters hep-
indicating that hepatocytes are quantitively the most atokine secretion and that the protein signals origi-
important cell type for liver protein secretion (). nating from the steatotic liver alter fatty acid
Several anatomical, structural, and functional fea- metabolism and induce inflammation and insulin
tures support the notion that the liver is an important resistance in other cell types. This section outlines the
organ for intertissue communication. The liver is large classically secreted hepatokines involved in regulating
(~. kg) and receives ~% of the cardiac output, lipid metabolism and insulin action, and it describes
providing a substantial volume of blood and thereby the effect of NAFLD in these relationships [see Table 
secreted factors for redistribution to other tissues. In this and Fig.  for hepatokine changes in the presence of
context, the liver has unique architecture and blood flow NAFLD (–)]. We close this section with a brief
regulation, whereby hepatocytes and nonparenchymal description of the literature pertaining to liver exo-
cells secrete products into the liver sinusoids, which flow some proteins and metabolism.
via the central veins to the inferior vena cava and
eventually to the heart for redistribution to peripheral Activin E
tissues. The extensive vascular network, and particularly Activin E is a member of the TGFb family and is
the “open pore” sinusoids that are located between encoded by the inhibin bE gene (). Activin E is a
hepatocyte planes, also supports the likelihood that newly identified hepatokine () that is elevated in
hepatokines are prominent in paracrine and autocrine liver and serum in humans with obesity () and
regulation of hepatocyte function. NAFLD (). Mice that overexpress activin E gain less
fat and have improved glucose tolerance when com-
Classical protein secretion pared with wild-type mice fed a high-fat diet. This
Classical or conventional secretion involves the appears to be mediated by an increase in uncoupled
transport of newly synthesized proteins through respiration as evidenced by increased expression of
the organelles of the secretory pathway, including thermogenic proteins in adipose tissue and higher core
the endoplasmic reticulum (ER), the ER exit sites, the temperature in activin E overexpressing mice (), as
Golgi, and eventually to the plasma membrane via well as an inability of activin E knockout mice to

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1369


REVIEW

Table 1. Hepatokine Links With NAFLD and Insulin Resistance

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
Molecular
Gene (Mouse / Mass Expression Contribution to Glucose Tolerance/Insulin
Hepatokine Human) (kDa) in NAFLD Contribution to NAFLD Resistance

Activin E Inhbe/INHBE ~22a Increased Reduces steatosis (50) Activin E overexpression prevents diet-induced glucose
intolerance in rodents (51), unknown in humans

Adropin Enho/ENHO 5 Decreased Suppresses lipogenesis (52) Improves insulin sensitivity (52); stimulates insulin
signaling in skeletal muscle (53)

ANGPTL4 Angptl4/ANGPTL4 45 Increased Promotes hepatic lipid accumulation Controversial findings: (i) improves insulin sensitivity
(54–56) (55, 56) by decreasing hepatic glucose output (55);
(ii) causes insulin resistance in liver, skeletal muscle,
and adipose (57, 58)

DPP4 Dpp4/DPP4 ~30 Increased Increases liver CD36 (59), likely to Inhibits incretin levels and impairs insulin secretion
increase lipid storage (60)

Ectodysplasin EDA ~46 Increased Unknown Induces insulin resistance in skeletal muscle via JNK
activation (61)

Fetuin A Ashg/ASHG ~67 Increased Unknown Promotes insulin resistance in liver via ER stress and
JNK activation (62); inhibits insulin receptor in
skeletal muscle (63); ligand for TLR4, which
promotes lipid-mediated insulin resistance in
adipose tissue (64)

Fetuin B Fetub/FETUB ~60 Increased Unknown Promotes insulin resistance in myocytes/hepatocytes


(65); impairs whole-body glucose tolerance (65)

FGF21 Fgf21/FGF21 ~23 Increased Increases hepatic fat oxidation and decreases Improves insulin sensitivity, decreases diacylglycerol
lipids (66); decreases adipose tissue lipolysis, (68); promotes insulin secretion of pancreatic
reducing lipid availability to the liver (67) b-cells (69)

Follistatin Fst/FST 38 Increased Promotes IL-1b production, may promote Unknown


fibrosis development (70)

HFREP1 Fgl1/FGL1 36 Increased Promotes NAFLD (71); increases lipogenesis Causes insulin resistance in skeletal muscle via JNK
through ERK1/2 activation (72) activation (72)

HMGB1 Hmgb1/HMGB1 30 Increased Unresolved; blocking HMGB1 protects Unknown; however, HMGB1 activates TLR4 and
against NAFLD (73, 74) causes inflammation in hepatocytes that could
impair insulin sensitivity (75)

Inhibin bE Inbe 39 Increased Unknown Unknown


with
obesity

LECT2 Lect2/LECT2 16 Increased Unknown Promotes insulin resistance in skeletal muscle via JNK
activation (76); impairs insulin signaling via serine/
threonine phosphorylation of IRS1 (77)

PEDF Serpinf1/SERPINF1 50 Increased Interacts with ATGL to increase lipolysis; Promotes insulin resistance in skeletal muscle via JNK
ablation promotes steatosis (78, 79). activation (80).

RBP4 Rbp4/RBP4 21 Increased Unknown Equivocal. Overexpression in liver does not alter
glucose homeostasis or insulin sensitivity (81);
whole-body overexpression causes insulin resistance
by activating JNK and TLR4 (82)

SeP Selenop/SELENOP ~60 Increased Unknown Causes insulin resistance in skeletal muscle and liver
(83);inhibits the insulin receptor (83); impairs insulin
secretion from pancreatic b-cells (84)

SHBG Shbg/SHBG 95 Decreased Suppresses lipogenesis in liver, exacerbates Unknown


steatosis (85)

TSK Tsku/TSKU ~40 Increased Promotes steatosis and NASH (86) Associated with whole-body insulin resistance (86)

Abbreviation: ATGL, adipose triglyceride lipase.


aMolecular weight is predicted and requires form validation.

1370 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

maintain body temperature during cold exposure (). glucose tolerance, and lower fatty acid oxidation ().
Additionally, administration of recombinant activin E Although little is known regarding the direct effects of

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
to mice with activin E deficiency (via global knockout adropin on metabolism, acute IP injection of adropin
of follistatin-like ) resulted in hepatic steatosis, increases insulin signaling through protein kinase B
suggesting a role in regulating lipid metabolism (). (AKT) and AKT substrate of  kDa (AS)
The expression pattern in obesity is interesting, and phosphorylation, resulting in greater glucose trans-
potentially unexpected, as on the one hand plasma porter type  (GLUT) translocation to the sarco-
activin E levels are increased in individuals with lemma in skeletal muscle (). Consistent with these
obesity, but on the other hand mice with increased effects, acute administration of recombinant adropin
circulating activin E levels are resistant to weight gain. improves glucose homeostasis in insulin-resistant
Further studies are required to assess how activin E diet–induced obese mice, and it is associated with
drives uncoupled respiration and why this mechanism lower expression of lipogenesis-associated genes, in-
does not prevent weight gain in individuals with cluding sterol CoA desaturase  and fatty acid syn-
obesity. thase, in the liver and adipose tissue (). Others have
shown that adropin increases glucose uptake and
Adropin oxidation via activation of pyruvate dehydrogenase,
Adropin is encoded by the ENHO gene, and ex- the rate-limiting enzyme for mitochondrial pyruvate
pression is decreased in response to elevated hepatic transport, and is associated with downregulation of
lipid availability (). Serum adropin levels are lower in carnitine palmitoyl-transferase I (CPT) activity and
humans with obesity () and in patients with type  other proteins involved in lipid metabolism, including
diabetes (), and although there is no reported link the fatty acid transporter CD () (Table ). Hence,
between serum adropin and NAFLD in humans, high- high adropin expression and secretion appear to
fat feeding reduces ENHO expression in parallel with improve insulin sensitivity and carbohydrate and lipid
the development of hepatic steatosis in mice (). In metabolism while suppressing hepatic steatosis. Taken
line with these observations, hepatic steatosis is ex- together, these studies suggest that restoring adropin
acerbated in adropin-null mice, and this is accom- levels within the liver and/or blood of patients with
panied by impaired glucose tolerance and insulin obesity could be a therapeutic approach for NAFLD
sensitivity at the whole-body level () and, perhaps and insulin resistance.
paradoxically, increased hepatic and whole-body fatty
acid oxidation (). In agreement, hepatic steatosis Angiopoietin-like protein 4
resulting from high-fat feeding is attenuated in mice Angiopoietin-like protein  (ANGPTL) is secreted by
with adropin overexpression, and this occurs in par- liver and adipose tissue () and plays important roles
allel with enhanced whole-body insulin sensitivity, in regulating lipid metabolism. ANGPTL increases

Figure 1. General metabolic processes affected by hepatokines in NAFLD. Liver secretion of various proteins are altered with
NAFLD, and selected proteins can influence insulin responsiveness. In adipose and skeletal muscle, many hepatokines affect
pathways involved in inflammation, lipogenesis/lipolysis, and fatty acid oxidation, which can promote insulin resistance.
Additionally, some hepatokines influence insulin secretion by the pancreas, which can independently affect peripheral tissue
glucose uptake and metabolism.

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1371


REVIEW

adipocyte lipolysis through a process that is dependent studies, liver-specific overexpression of DPP impairs
on its C-terminal fibrinogen-like domain (), and it whole-body glucose tolerance in high-fat–fed mice,

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
suppresses lipoprotein lipase activity via its N-terminal effects that are linked to reduced circulating GLP- ().
coiled-coil domain (). This increases plasma free fatty DPP is likely to directly affect metabolism in peripheral
acids and triglycerides, which is associated with ectopic tissues, as treatment of primary hepatocytes, adipocytes,
lipid accumulation in liver and skeletal muscle (, , and skeletal myotubes with recombinant DPP impairs
) and is highlighted by dyslipidemia in mice with insulin sensitivity (, ). However, a receptor for
liver-specific overexpression of Angptl (). Consistent DPP has not yet been identified. DPP also drives liver
with these functions, individuals heterozygous or ho- steatosis, most likely by increasing fatty acid uptake and
mozygous for the loss-of-function Angptl variant EK storage in hepatocytes (). In agreement with these
have significantly lower fasting plasma triglyceride levels observations, genetic ablation () or administration of
() whereas Angptl2/2 mice have increased VLDL oral DPP inhibitors such as vildagliptin () or
clearance (). Free fatty acids upregulate Angptl sitagliptin () improves both hepatic steatosis and
expression via peroxisome proliferator-activated re- glucose tolerance, further highlighting the systemic and
ceptor (PPAR)a activation in the liver and PPARg in autocrine/paracrine actions of DPP.
adipose tissue (), demonstrating the likelihood of a
feed-forward loop whereby lipid oversupply drives Ectodysplasin A
Angptl expression and high ANGPTL levels further Ectodysplasin A is a newly discovered hepatokine that
drive dyslipidemia. Such regulation would be clinically is associated with obesity and insulin resistance ().
unfavorable for PPAR agonists, which are used as Liver and serum ectodysplasin A levels are increased
antidiabetic and lipid-lowering agents. Insulin sup- in high-fat–fed mice and mice with genetic obesity
presses hepatic Angptl expression; however, this (i.e., leptin receptor–deficient db/db mice), and liver
mechanism is likely to be impaired in insulin-resistant ectodysplasin A mRNA levels increase with steatosis
states (). severity and correlate with a reduction in whole-body
The role of ANGPTL on insulin action and insulin action in humans (). In parallel with these
glycemic control is controversial. Although mice with findings, treatment of CC skeletal myotubes or
ANGPTL overexpression have severe hepatic stea- whole-body overexpression of ectodysplasin A in mice
tosis, they exhibit improvements in hepatic and sys- induces muscle insulin resistance in association with
temic insulin sensitivity (, ). In agreement, ANGPTL activation of c-Jun N-terminal kinase (JNK), and these
increases insulin-mediated inhibition of gluconeogene- effects were reversed upon partial silencing of ecto-
sis and decreases hepatic glucose production in primary dysplasin A in mice () (Fig. ). Notably, ectodys-
hepatocytes, and in humans ANGPTL levels correlate plasin A did not affect hepatic insulin action or other
positively with insulin sensitivity (). In contrast, parameters of energy homeostasis, pointing to a direct
reducing plasma ANGPTL via genetic deletion in liver-to-muscle crosstalk. Additional work is required
mice reduces blood lipids, reduces ectopic lipid ac- to determine the role of ectodysplasin on lipid
cumulation in liver and muscle, enhances insulin metabolism and confirm the relevance of this protein
signaling, and improves glycemic control (, , ), in NAFLD and type  diabetes.
and anti-ANGPTL antibody therapy in obese and
diabetic mice recapitulates this favorable metabolic Fetuin A
phenotype (). Further studies are clearly required Fetuin A (also known as a--HS-glycoprotein) is a
to fully understand the discrepancy in these disparate liver-secreted glycoprotein encoded by the ASHG
findings and to ascertain the potential of ANGPTL gene. Fetuin A is positively associated with circulating
therapeutic applications in dyslipidemia and glycemic triglycerides, the severity of NAFLD (, ), and
control. insulin resistance (, ) in rodents and humans. In
this regard, lipid oversupply induces ER stress, which
Dipeptidyl peptidase-4 activates extracellular signal–regulated kinase (ERK)/
Dipeptidyl peptidase- (DPP) is a ubiquitous serine and JNK to drive fetuin A production (). The he-
protease secreted by the liver that rapidly inactivates the patic expression and secretion of fetuin A is also
circulating incretin hormones glucagon-like peptide regulated by F-box and WD repeat domain-containing
(GLP)- and gastric inhibitory peptide (GIP) ().  (FBXW), a ubiquitin protein ligase that degrades
Incretins are important regulators of whole-body glu- fetuin A. FBXW is suppressed in obese mice and in
cose homeostasis, as GLP- and GIP promote insulin humans with obesity in parallel with increased fetuin
secretion and suppress glucagon secretion, resulting in A levels (). Fetuin A induces insulin resistance
peripheral glucose uptake and reduced hepatic glucose by several mechanisms (, –) (Fig. ). Fetuin
output (). Individuals with NAFLD and insulin re- A inhibits insulin receptor tyrosine kinase activity,
sistance have elevated plasma DPP activity (), which resulting in lower autophosphorylation and impaired
is consistent with lower GLP and GIP levels in the insulin signaling (). Fetuin A also acts as an en-
blood of these individuals (). Consistent with human dogenous ligand for Toll-like receptor (TLR), which

1372 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

Figure 2. The influence of hepatokines on insulin resistance in skeletal muscle or adipose tissue. Hepatokines target pathways
involved in regulating insulin action. Fetuin A (in the presence of palmitate) and RBP4 can activate TLR4 and result in JNK phosphorylation,

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
greater serine phosphorylation of IRS1, and suppression of insulin signaling and GLUT4 trafficking to the sarcolemma. Fetuin A and SeP can
directly inhibit the insulin receptor, resulting in lower insulin signaling and GLUT4 trafficking. Fetuin A can promote ER stress through
unknown mechanisms, resulting in greater JNK activation and impaired insulin signaling. Follistatin, HFREP1, LECT2, PEDF, and ectodysplasin
are also implicated in impairing insulin signaling secondary to activation of JNK; however, the upstream signaling is unresolved.

enables saturated free fatty acids to activate TLR humans (). This indicates that fetuin B’s primary
signaling to induce insulin resistance (). This fetuin function is the suppression of glucose effectiveness
A/free fatty acid interaction predicts the development (), which refers to the ability of glucose to promote
of insulin resistance in humans (). In the pancreas, its own disposal, independently of insulin (Fig. ). This
fetuin A signals to b-cells and impairs glucose sensing process is at least as important as insulin for glucose
(), which results in impaired insulin secretion (, clearance, accounting for ~% of an oral glucose
) in response to inflammatory processes, including tolerance test in normal individuals and ~% in
TLR, JNK, and nuclear factor kB (NF-kB) activation insulin-resistant individuals with obesity (). Ad-
and the accumulation of lipotoxic lipids (, ). ditionally, there is evidence that fetuin B impairs first-
Mice with global fetuin A deletion show improved phase glucose-stimulated insulin secretion (), signifying
insulin sensitivity and are resistant to diet-induced a role in b-cell function. The importance of fetuin B in
obesity (, ), highlighting the likely therapeutic NAFLD-induced insulin resistance is confirmed by
potential of fetuin A antagonists. studies showing that short hairpin RNA suppression
of liver fetuin B protein and reduced fetuin B secretion
Fetuin B improve glycemic control in obese, insulin-resistant
Fetuin B is encoded by the FETUB gene and shares mice ().
% homology with fetuin A. Fetuin B is increased in
patients with NAFLD (, , , ), type  di- Fibroblast growth factor 21
abetes (, ), and gestational diabetes (, ), Fibroblast growth factor (FGF) regulates systemic
and it correlates positively with insulin resistance lipid metabolism in response to diet, exercise, and cold
(). Although administration of fetuin B induces exposure (, ), and its role in metabolism has
insulin resistance in myotubes and hepatocytes in been extensively reviewed (). FGF expression is
vitro, administration of fetuin B in lean mice caused increased by activation of PPARa (), and consistent
whole-body glucose intolerance but not insulin re- with this finding, humans with NAFLD have increased
sistance (), observations that were recapitulated in circulating FGF levels (, ). This may be a

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1373


REVIEW

compensatory response designed to limit the impact of NAFLD and systemic insulin resistance. HFREP is
lipotoxic stress, as FGF reduces adipose tissue li- elevated in human NAFLD () and NASH (), and

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
polysis (, ), increases fatty acid oxidation, lowers circulating concentrations correlate positively with
hepatic lipids such as diacylglycerol, enhances insulin plasma glucose levels and insulin resistance (). In
sensitivity, and improves glycemic control (, , mice, high-fat feeding promotes liver HFREP ex-
). FGF also reduces hepatic VLDL secretion and pression and NAFLD (), suggesting that elevated
accelerates VLDL disposal in both white and brown circulating lipids induce hepatic HFREP expression.
adipose tissue via coordinated upregulation of CD In this context, the fatty acid palmitate (C:) dose-
and lipoprotein lipase (). The importance of FGF dependently increases HFREP expression in hepa-
is highlighted in mouse studies where the deletion of tocytes (). Lipid-dependent HFREP expression is
Fgf, its receptor FGF receptor-c, or the coreceptor mediated by ER stress and the resultant activation of
b-klotho, results in greater adiposity, hepatic steatosis, the P and CCAAT/enhancer-binding protein b in
and liver insulin resistance, increased hepatic glucose hepatocytes, and pharmacological blockade of this
production, and hyperglycemia (). Finally, FGF pathway blunts HFREP expression and partially re-
promotes pancreatic b-cell function and insulin se- stores insulin action (). This provides a plausible
cretion (). It is for these reasons that FGF has mechanism for NAFLD-induced HFREP production,
emerged as a therapeutic agent for the treatment of given that ER stress is elevated in humans with
type  diabetes and the metabolic syndrome (), conditions characterized by dyslipidemia such as
although FGF analogs have failed to lower blood NAFLD (), diabetes (), and obesity ().
glucose in humans. Moreover, HFREP promotes lipogenesis through
ERK/ activation (), indicating the presence of a
Follistatin feed-forward mechanism that drives NAFLD. HFREP
Follistatin is a member of the TGFb family and was has also been shown to cause insulin resistance in
originally recognized for its inhibitory effect on FSH immortalized CC myotubes and skeletal muscle ex
production in the pituitary () and, later, suppres- vivo (), which is mediated by decreased 9-AMP–
sion of myostatin to support skeletal muscle growth, activated protein kinase (AMPK) phosphorylation
which demonstrated actions outside of the re- and enhanced JNK activation in a process dependent
productive system (). Follistatin is increased in on epidermal growth factor receptor and FOXO
serum of individuals with NAFLD () and type  phosphorylation (). Administration of recombinant
diabetes (), potentially through forkhead box O HFREP or genetic overexpression of HFREP causes
(FOXO)–mediated transcriptional activation (). liver and skeletal muscle insulin resistance (), whereas
In contrast, weight loss following bariatric surgery deletion of liver HFREP protects against diet-induced
leads to a reduction in serum follistatin, which is insulin resistance in mice () (Fig. ). Reducing cir-
accompanied by improvements in insulin sensitivity culating HFREP could therefore be a viable approach
and glycemic control (, ). It remains to be for insulin resistance.
determined whether the decrease in serum follistatin
is a direct mediator of the improvements in glycemic High-mobility group box 1 protein
control. However, it is known that follistatin promotes Sterile inflammation caused by free fatty acids,
proinflammatory cytokine expression, such as IL-b, chemokines, and cytokines stimulates the release of
that is implicated in fibrosis progression (, ) and endogenous molecules termed damage-associated
the development of insulin resistance in adipose tissue molecular patterns, and these molecules can activate
() and skeletal muscle (). Similarly, follistatin TLR signaling in a variety of cell types that promote
overexpression in isolated hepatocytes or livers of mice inflammatory responses. High-mobility group box 
impairs signaling in white adipose tissue, insulin- protein (HMGB) was originally discovered as a
mediated suppression of hepatic glucose production, protein that binds to nucleosomes to stabilize DNA
and whole-body glucose tolerance, whereas follistatin structure and modulate transcription (), but more
knockdown improves insulin sensitivity () (Fig. ). recently it has been shown to be secreted from he-
Taken together, these studies provide evidence that patocytes (). HMGB expression and secretion are
follistatin is increased with NAFLD and can pro- increased in NAFLD, and this process is mediated by
mote inflammation, insulin resistance, and glucose increased free fatty acid availability (). The secreted
intolerance. HMGB activates TLR signaling and the resultant
NF-kB activation drives inflammation in neighboring
Hepatocyte-derived fibrinogen-related protein 1 hepatocytes. Given that TLR activation dampens
Hepatocyte-derived fibrinogen-related protein  insulin signaling (), it is possible that HMGB–
(HFREP), also referred to as hepassocin or fibrinogen- TLR signaling may contribute to insulin resistance
like protein , is secreted by hepatocytes and is known in NAFLD, although this requires formal testing.
to promote cell growth and proliferation (–). Blocking HMGB reduces lipotoxic effects in hepa-
More recently, HFREP has been implicated in tocytes () and protects against NAFLD progression

1374 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

in rats (, ), which supports a prominent autocrine/ via its interaction with adipose triglyceride lipase
paracrine role. Whether the pathogenic effects of (ATGL), the rate-limiting enzyme for triglyceride

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
HMGB extend beyond the liver in humans is hydrolysis, and this coincides with increased ceramide
questionable, as HMGB levels are not associated with and diacylglycerol accumulation in liver and muscle
histological severity in NAFLD (). (, , , ). Neutralizing PEDF with monoclonal
antibodies reverses these effects (). Whereas circu-
Inhibin bE lating PEDF correlates with hepatic steatosis and in-
The hepatokine inhibin bE is a member of the TGFb sulin resistance, liver-specific overexpression of Pedf
family and is positively associated with body mass reverses hepatic lipid accumulation (), which likely
index and insulin resistance in rodents (, ) and reflects higher intrahepatic lipolysis, and it aligns with
humans (). Knockdown of hepatic inhibin bE lower NASH development (, ). Taken together,
using small interfering RNA attenuates fat mass gain this suggests that the increased production of PEDF
in parallel with greater whole-body fat oxidation in with NAFLD is directed toward secretion and not
obese db/db mice (); however, no overt metabolic retained within the liver, where it promotes insulin
phenotype was reported in Inhbe knockout mice (). resistance and dyslipidemia.
Additional work is needed to determine whether in-
hibin bE indeed increases fat oxidation and whether Retinol binding protein 4
blocking inhibin bE’s actions has therapeutic utility Retinol binding protein  (RBP) is a liver and ad-
for NAFLD and perhaps obesity. ipose tissue secreted protein that transports vitamin
A in the form of retinol (). Serum RBP levels are
Leukocyte cell–derived chemotaxin 2 increased with NAFLD (, ), insulin resistance
Leukocyte cell–derived chemotaxin  (LECT) is a (), and in type  diabetes (), and reduced with
-kDa hepatokine originally described as chemotactic diet-induced weight loss (), bariatric surgery
for neutrophils (). LECT has since been impli- (), and exercise (). Although these findings in
cated in NAFLD (, ) and insulin sensitivity (, humans suggest a potential role for RBP in reducing
), and circulating LECT expression is positively insulin action, direct examination of RBP effects
correlated with body weight and insulin resistance in in mice are equivocal. RBP overexpression causes
humans (). Studies in mice show that whole-body inflammation and insulin resistance in mouse adi- “…ketone bodies signal to
deletion of Lect enhances insulin-stimulated AKT pose tissue due to activation of JNK and TLR peripheral tissues and the
phosphorylation in skeletal muscle, whereas admin- signaling (), and genetic deletion of Rbp enhances central nervous system to
istration of recombinant LECT activates JNK () insulin sensitivity () (Fig. ). However, in a recent regulate metabolism.”
and inhibits insulin signaling via increased serine study where RBP was overexpressed specifically in
phosphorylation of IRS () (Fig. ). Interestingly, the liver of mice, glycemic control was not affected
pharmacological inhibition of DPP improves glucose despite significant increases in circulating RBP levels
metabolism in mice in parallel with a reduction in (). In light of these conflicting data, additional
hepatic LECT protein content, and this occurs via research is warranted to determine RBP’s role in the
activation of AMPK and suppression of JNK activity pathogenesis of NAFLD and type  diabetes.
(). Thus, DPP may regulate LECT expression,
indicating likely cross-talk between hepatokines in Selenoprotein P
NAFLD. Circulating selenoprotein P (SeP) levels are positively
correlated with type  diabetes (), insulin resistance,
Pigment epithelium-derived factor and blood glucose levels in humans (–), which
Pigment epithelium–derived factor (PEDF) is a agrees with studies in mice reporting increased liver
-kDa noninhibitory serine protease originally found expression and secretion of SeP in response to high-fat
to be secreted by retinal epithelial cells, but it is also feeding (), NAFLD (), and type  diabetes ().
highly expressed in, and secreted by, liver and adipose SeP expression and secretion are increased in response
tissue (). Liver PEDF expression () and circu- to ER stress and JNK activation, subsequent to TLR
lating PEDF levels (–) are increased in humans stimulation (). Moreover, activation of AMPK via
with obesity, insulin resistance, and NAFLD, and it is the anti-inflammatory drug salsalate or the insulin-
reduced with weight loss (). PEDF is implicated in sensitizing drug metformin protects against ER stress
the development of insulin resistance and glucose and SeP production (). Taken together, this links
intolerance in mice (). PEDF increases JNK and proinflammatory and/or dyslipidemic states to in-
ERK/ activity in skeletal muscle and liver, as well creased SeP expression.
as NF-kB activity in adipocytes, which corresponds Consistent with a role in metabolic disease, acute
with reduced insulin signal transduction (, ) administration of recombinant SeP causes insulin
(Fig. ). Increasing plasma PEDF by administration of resistance in mice (). In vitro, SeP administration
recombinant PEDF or overexpression of Pedf in ad- impairs insulin receptor phosphorylation in HepG
ipose tissue of mice increases adipose tissue lipolysis hepatocyte-like cells, suggesting direct inhibition of

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1375


REVIEW

autophosphorylation at this proximal step of insulin by the improvements in systemic metabolism rather
signaling () (Fig. ). Additionally, SeP impairs than direct actions targeting proinflammatory, profi-

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
glucose-stimulated insulin secretion in pancreatic brotic, or insulin signaling pathways. Future work
b-cells, effects that are reversed by monoclonal anti- including the identification of the putative TSK re-
body neutralization of circulating SeP (). Genetic ceptor is required to determine the role of TSK in lipid
deletion and RNA interference–mediated knockdown metabolism and insulin signaling in peripheral tissues.
of SeP improves systemic insulin sensitivity and glu-
cose tolerance (), whereas SeP-neutralizing anti- Hepatocyte exosomes and protein secretion
bodies improve insulin secretion and glycemic control Exosomes are emerging as an important mode of
in diabetic mice (), collectively highlighting the intercellular and intertissue communication. Proteo-
potential therapeutic utility of this hepatokine. mic analysis of exosomes derived from rat primary
hepatocytes identified ~ proteins, some of
Sex hormone binding globulin which are also denoted as classically secreted hep-
Sex hormone binding globulin (SHBG) is best known atokines (e.g., DPP). Bioinformatic analysis predicts
as a transporter of sex steroids (, ). Liver () that these hepatocyte-derived exosomal proteins are
and serum () SHBG levels are lower in individuals implicated in intracellular transport, lipid metabolism,
with hepatic steatosis when compared with individuals carbohydrate metabolism, metabolite availability, and
with no adverse liver pathology, and plasma SHBG protein turnover (). Although the influence of
negatively predicts insulin resistance and hyperinsulinemia NAFLD or NASH on liver-secreted exosomes is
(–). This is accompanied by lower hepatic SHBG poorly understood, in vitro studies showed that pal-
content and lower expression of the transcription factor mitate exposure increases exosome/extracellular vesicle
hepatocyte nuclear factor a, a key transcriptional reg- secretion (–), and that these exosomes contained
ulator of SHBG (). Additionally, low circulating SHBG a greater proportion of proteins involved in regulating
is independently associated with obesity, and weight loss fibrosis (i.e., a-SMA, TGFb, Cola) (). These
following bariatric surgery increases circulating SHBG exosomes were found to communicate with “healthy”
levels (). Collectively, these data describe a clear as- hepatocytes to promote the progression of fibrosis
sociation between obesity, NAFLD, insulin resistance, and (–), suggesting a potential autocrine effect of
lower SHBG levels in liver and blood. exosomes in liver disease. However, this is very much an
The lower expression of SHBG in NAFLD may emerging field, and the importance of liver-derived
occur secondary to inflammation, as an increase in exosomes in regulating cell metabolism and influenc-
TNFa in response to JNK and NF-kB activation re- ing whole-body glucose homeostasis is currently un-
duced SHBG production in HepG cells (). Given known. Future work should examine how liver disease
that insulin promotes SHBG production in vitro (), affects the protein composition in liver-secreted exo-
and people with type  diabetes have a stronger re- somes, and whether these exosomes are important
lationship between insulin levels and SHBG than do mediators of insulin resistance.
those with type  diabetes (), it is possible that
hepatic insulin resistance could precede a reduction in Summary
SHBG and exacerbate lipid accumulation. This notion Hepatokines clearly exert pleiotropic effects on lipid
is at least partially supported by the finding that and glucose metabolism and insulin action, and their
resveratrol, a polyphenol that improves insulin sen- secretion is impacted by NAFLD (Table ). Although
sitivity, also increases SHBG levels in mice (). The there has been one comprehensive examination of the
mechanisms underpinning the effects of SHBG on hepatocyte protein secretome (), which demon-
glycemic control are unknown. With respect to lipid strates marked changes in response to simple steatosis,
metabolism, overexpression of SHBG suppresses li- rapid developments in mass spectrometry now
pogenesis (, ) that could reduce hepatic steatosis. permit a deeper examination of cell/tissue secretomes,
enabling more detailed understanding of the breadth
Tsukushi of protein secretion during the progression of NAFL to
Tsukushi (TSK) is a highly conserved proteoglycan in NASH. Performing such experiments in human liver
mammals and a newly discovered hepatokine that is tissues will be important in driving our understanding
increased in rodent obesity () and NASH (). of hepatokines in physiology and NAFLD-related
Studies in Tsk-null mice highlight an important role comorbidities and, by extension, the identification
for TSK in regulating energy balance under obesogenic of therapeutic targets to treat cardiometabolic diseases.
conditions. TSK is induced in response to increases in Such studies will also be important for identifying
energy expenditure, which blunts sympathetic outflow novel biomarkers that could be used in conjunction
and innervation of adipose tissue, thereby reducing with readily available clinical parameters to identify
thermogenesis and energy expenditure (). Ablation the presence and staging of NAFLD/NASH with
of TSK also prevents diet-induced NASH and whole- higher sensitivity and specificity than current risk-
body insulin resistance (), which may be mediated stratification algorithms used in routine clinical care.

1376 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

Metabolite secretion, NAFLD, and oxidative stress (), a well-documented mediator of


insulin resistance insulin resistance (). Second, b-hydroxybutyrate

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
The liver plays critical roles in regulating systemic directly modulates lysine residues on histones via a
glucose and lipid metabolism, processes that have been process known as lysine b-hydroxybutyrylation,
extensively reviewed in the context of NAFLD (). leading to activation of starvation-regulated meta-
Glucose and VLDL aside, liver-derived major me- bolic pathways, including amino acid catabolism,
tabolite classes, including lipoproteins, ketones, acyl- PPAR signaling, and oxidative phosphorylation ().
carnitines, and bile acids, appear to transduce specific Although correlative, HDAC inhibition is reported with
metabolic signals; however, much of the literature on the consumption of ketogenic diets, with subsequent
this topic remains correlative and circumstantial with activation of PPARa and the expected sequalae of
regard to their endocrine functions. This mode of increased expression of lipid metabolism genes (),
intertissue communication is discussed below with this greater hepatic fatty acid oxidation and plasma tri-
caveat in mind. glyceride clearance, and FGF production (, ).
Thus, b-hydroxybutyrate links changes in metabolite-
Ketones directed histone modifications to changes in cellular
Ketone bodies such as acetone, acetoacetate, and metabolism.
b-hydroxybutyrate are produced in the liver using Although b-hydroxybutyrate is commonly used in
b-oxidation–derived acetyl-CoA as substrate, and they metabolic studies to represent “ketones,” acetoacetate
are then secreted from the liver for transport to other constitutes % to % of the total hepatic ketone
peripheral tissues. Upon their uptake by peripheral body pool (, ) and is thereby a significant source
tissues, ketone bodies are converted back to acetyl- of acetyl-CoA and a potential signaling molecule.
CoA, which provides substrate for energy production Acetoacetate can inhibit glucose uptake in skeletal
through the tricarboxylic acid (TCA) cycle and ox- muscle and heart (, ), although others show no
idative phosphorylation. The excess acetyl-CoA also effect (). Recent work has shown that hepatocyte-
increases the capacity for protein acetylation, par- secreted acetoacetate, but not b-hydroxybutyrate,
ticularly of mitochondrial proteins, which regulates ameliorates diet-induced hepatic fibrosis (), but
the activity of transcription factors and transcrip- these studies did not assess metabolism, and a better
“Increased circulating
tional coactivators such as FOXO, PPARg coac- understanding of acetoacetate roles in metabolism is
ceramide and/or ceramide
tivator a (PGCa), and PPARg, all of which play needed. Similarly, another ketone, acetone, can be accumulation in rodents
critical roles in regulating the expression of metabolic taken up by tissues but its effects on glycemic control induces inflammation and
genes, particularly in response to changes in nutrient and insulin sensitivity are unknown. Further detailed insulin resistance.”
availability (, ). In this regard, ketogenesis is information on ketones and metabolism is available
important when carbohydrate is limiting, including elsewhere ().
starvation and with the consumption of extremely The role of ketones in regulating insulin action and
low-carbohydrate diets, which are often referred to as glycemic control is equivocal (–). These con-
“ketogenic” diets (, ). Ketone bodies con- flicting findings result from discrepancies in study
tribute up to % of total energy expenditure under designs, the composition of ketogenic diets, the like-
these conditions (, ). lihood of “positive” and “negative” responders to ke-
Moving beyond their direct role as an energy togenic dietary intervention, the duration of the
substrate, ketone bodies signal to peripheral tissues ketogenic diet, and the myriad of “off-target” changes
and the central nervous system to regulate metab- that accompany ketogenic diet consumption.
olism. Ketones can cross the blood–brain barrier and Ketone bodies in NAFLD. Hepatic ketogen-
are sensed in the hypothalamus to stimulate food esis and plasma b-hydroxybutyrate are reportedly
intake by increasing the expression of the orexigenic increased (, ), unchanged (–), or decreased
neuropeptides Npy and Agrp () and through the (, ) in rodents and humans with NAFLD. There
potentiation of hypothalamic leptin and insulin sig- is similar uncertainty in NASH, with reports of higher
naling () (Fig. ). Such signaling is associated with (, ) or lower levels () of circulating ketone
reduced adiposity and improved systemic insulin bodies compared with individuals with steatosis or no
sensitivity (). In the periphery, b-hydroxybutyrate liver pathology. Future studies are required to clarify
regulates lipid metabolism in adipocytes via activa- this ambiguity and to also identify the potential links
tion of hydroxy-carboxylic acid receptor  (HCA, between specific ketones and insulin action.
GPRA), which sequentially decreases adenylyl
cyclase activity, protein kinase A activity, and lipolysis Lipoproteins
(, ). b-Hydroxybutyrate is also implicated in the The liver secretes lipids primarily within VLDLs, but
longer-term regulation of metabolism by modifying also within extracellular vesicles. Although most lipids
histones through two distinct epigenetic processes. within VLDLs are triglycerides (~%), VLDLs also
First, b-hydroxybutyrate inhibits class I histone deace- contain cholesterol and cholesterol esters (~%)
tylase (HDAC) (), which is associated with reduced and phospholipids (~%), with most being :-

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1377


REVIEW

Figure 3. Hepatic metabolite secretion. Different pathways for metabolite secretion are color-coded. Fatty acids (FA) are taken up
by hepatocytes and can be converted to acylcarnitine for oxidation within the mitochondria. These acylcarnitines can be secreted

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
from the liver and induce insulin resistance in peripheral tissues (green). Ketone bodies are produced during mitochondrial fatty
acid oxidation and can be secreted from hepatocytes, affecting oxidative metabolism, and they may affect insulin sensitivity in
peripheral tissues (orange). The liver plays a significant role in lipoprotein metabolism by taking up chylomicron remnants from the
circulation (blue) and by secreting VLDLs that transport triglycerides (and other lipids) to peripheral tissues (yellow). Cholesterol
synthesized within the liver or taken up as part lipoproteins can be converted to bile acids, which are secreted and affect intestinal
lipid absorption and bile flow, as well as peripheral metabolism through activation of various receptors (blue). Lastly, the liver
secretes exosomes that carry thousands of metabolites, including ceramide and LPC, that were shown to lead to stellate cell
activation and fibrosis in a paracrine manner (red). Changes in those pathways with NAFLD are shown with red arrows (↑ increased,
↓ decreased with NAFLD, ? unknown or controversial). HSC, hepatic stellate cell; TAG, triacylglycerol.

containing phosphatidylethanolamine (). Addi- availability, as shown experimentally by coinfusing


tionally, % to % of liver sphingolipids, mainly Intralipid (triglyceride emulsion) and heparin
ceramide and free sphingosine, can be released as (lipoprotein lipase activator) (, ) (Fig. ).
components of VLDLs (). Increased circulating VLDLs, and thereby tri-
Fatty acids are cleaved from triglyceride con- glycerides, is associated with hepatic steatosis,
tained within VLDLs by lipoprotein lipase that is obesity, and insulin resistance (, ). Once
localized to the surface of endothelial cells in depleted from triglyceride, lipoprotein particles are
capillaries and are transported into cells residing in enriched in cholesterol and cholesterol esters to
close proximity or carried in the blood bound form intermediate-density lipoproteins or low-
to albumin. Increasing VLDL-triacylglycerol se- density lipoproteins (LDLs). Insulin resistance in
cretion and delivery to tissues can cause periph- humans is characterized by high levels of plasma
eral insulin resistance by increasing fatty acid cholesterol esters (), and high circulating cholesterol

1378 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

levels are associated with reduced insulin secretion, and/or ceramide administration in rodents induces
effects that can be normalized with cholesterol depletion inflammation and insulin resistance, particularly in

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
(). Cholesterol accumulation within the plasma skeletal muscle (, –), whereas a reduction
membrane of skeletal muscle is associated with insulin in liver and plasma ceramide after weight loss is
resistance secondary to reduced GLUT insertion associated with reduced inflammation and im-
within the membrane (, ). These studies suggest proved insulin sensitivity (, , ). Addi-
that circulating cholesterol (liver derived or from other tionally, these patients showed reductions in
sources) has a negative impact on glycaemic control. We plasma cholesterol, triglycerides, LDLs, and free
are not aware of any studies assessing the direct impact fatty acids (), increased ketone bodies and
of liver-derived VLDL/LDL-associated cholesterol on acylcarnitines, and reduced levels of branched-
glucose metabolism. However, as the liver is a major site chain amino acids and (lyso)glycerophospholipids
of endogenous cholesterol synthesis and secretion (), (), suggesting that reductions in plasma ceramide
most peripheral effects are most likely related to liver- is one of many changes that mediate systemic meta-
derived cholesterol. bolic improvements with weight loss. Similarly,
Intracellular ceramide accumulation causes insulin induction of hepatic ceramide degradation through
resistance (), and circulating ceramides appear to increased expression of acid ceramidase within the
induce similar effects. Ceramides contained within liver and subsequent reductions in circulating
LDLs are transferred to the plasma membrane of ceramide are associated with improvements in
skeletal muscle, which leads to a reduction in insulin- hepatic steatosis and systemic insulin sensitivity
stimulated GLUT translocation (). LDL ceramides (, ).
also induce inflammation in macrophages (), per-
haps via a TLR-dependent mechanism, which can in Acylcarnitines
turn induce insulin resistance in peripheral tissues (). Acylcarnitines are generated through coupling of
Additionally, treatment of macrophages with VLDLs acyl-CoA to carnitine for import into the mito-
increases macrophage ceramide content and promotes chondrial matrix. Once inside the mitochondria,
an M-like macrophage polarization, leading to adipose carnitine and acyl-CoA are regenerated and acyl-
tissue inflammation and insulin resistance in diet- CoA is oxidized through b-oxidation. Acylcarni-
induced obese mice (). tines are an important energy source within the
Lipoproteins in NAFLD and insulin re- mitochondria, and they are also secreted into the
sistance. Hepatic steatosis is associated with in- circulation (), either directly into the blood
creased secretion of VLDLs and increased plasma through the acylcarnitine transporter SLCA
triglycerides (, ), and patients with NAFLD () or within extracellular vesicles (, ). The
have a reduced capacity for insulin-mediated suppres- liver (, ), but not skeletal muscle (, ),
sion of VLDL secretion (), which may contribute to is the major source of circulating acylcarnitine and
the insulin resistance and glucose intolerance that is this is most pronounced with fasting (). Acyl-
commonly associated with NAFLD (, ). Fur- carnitines can provide up to % of the circulating
thermore, increased VLDL secretion is associated with carbon product from fatty acids () and are taken
inflammation, particularly increased TNFa production up by skeletal muscle, heart, and brown adipose
(), and increases in TNFa can further drive hepatic tissue (, ). Acylcarnitine accumulation in
VLDL secretion () and insulin resistance (). muscle has been linked to skeletal muscle insulin
VLDL and LDL particles carry ceramide (, resistance (), which is thought to result as a
), and hepatic ceramide secretion is increased in product of a mismatch between fatty acid oxidation
the presence of lipid oversupply (, ). Fur- and TCA flux, resulting in mitochondrial stress and
thermore, ceramide transported in LDLs is in- reactive oxygen species production () (Fig. ).
creased in the plasma of individuals with obesity Some studies demonstrate increased plasma acyl-
with type  diabetes and correlates with insulin carnitines in humans with insulin resistance (,
resistance (). The liver is the major contributor ), and treating CC myotubes with acylcar-
to circulating ceramide levels (), and liver nitine modestly impairs insulin signaling ().
ceramide synthesis is highly dependent on fatty acid Whether circulating acylcarnitines impair systemic
availability (), linking dysregulated adipose insulin action and glucose homeostasis is ques-
tissue lipolysis to liver-derived ceramide in the tionable because increasing plasma acylcarnitines
circulation and NAFLD. Increased ceramide se- via g-butyrobetaine supplementation in mice ()
cretion in conditions of hepatic steatosis/hepatic or carnitine supplementation in rats () does not
lipid oversupply may reflect an attempt by the liver affect glucose homeostasis. In addition to their
to protect itself from the deleterious consequences potential role in (dys)regulating insulin action,
of intracellular ceramide accumulation (), al- acylcarnitines are taken up by pancreatic b-cells,
though there is presently no known ceramide leading to insulin depletion as a result of a com-
sensing mechanism. Increased circulating ceramide bination of diminished insulin refill and enhanced

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1379


REVIEW

insulin granule release (). Acylcarnitines acti- this area are contradicting. Most studies report
vate proinflammatory signaling in macrophages increases in liver and circulating bile acids with

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
(), and liver-derived acylcarnitines are a likely progressive NAFLD (–), and circulating bile
fuel source for brown fat thermogenesis during cold acids appear to be related to the metabolic phe-
exposure, suggesting a role for acylcarnitines in notype associated with NAFLD/NASH, especially
regulating energy homeostasis () (Fig. ). insulin resistance (). Further work is clearly
Acylcarnitines in NAFLD and insulin re- needed to bridge the gap between the un-
sistance. Plasma acylcarnitines have been reported derstanding of bile acid secretion and their met-
to be decreased in individuals with hepatic stea- abolic functions and influence on metabolic
tosis () and increased in patients with NASH diseases when considering the development of
(–), while others report acylcarnitine species- targeted therapies in NAFLD.
specific changes with NAFLD (). The increase in
NASH patients could be attributed to the down- Metabolite secretion in exosomes
regulation of carnitine palmitoyltransferase  (CPT) Most hepatic lipids are secreted within lipoprotein
and a subsequent decrease in mitochondrial fatty acid particles, and far less is known about the secretion of
entry and oxidation (). These reported differences hepatic lipids or other metabolites contained within
may relate to changes in mitochondrial b-oxidation extracellular vesicles, including exosomes. In humans,
with NAFLD progression (), as b-oxidation is ~% of total lipids found in serum are contained
reduced in livers with steatosis/NAFL (, ) and within lipoproteins (), suggesting that only a minor
increased in NASH (, ). The mechanisms component of circulating lipids (~%) are transported
underpinning NAFLD stage-specific differences in in extracellular vesicles (Fig. ). This relative low
lipid metabolism remain unresolved, and it would be abundance of lipids does not exclude a meaningful
interesting to assess how and why b-oxidation (and contribution of exosomal lipids as signaling molecules
subsequently acylcarnitine secretion) is altered with because exosomes can be targeted to specific cell/tissue
the progression of NAFL to NASH. At this point, types (). Liquid chromatography–tandem mass
it is difficult to reconcile any clear association be- spectrometry and other biochemical approaches have
tween NAFLD, acylcarnitines, and impaired glycemic identified many lipid species in exosomes [e.g., 
control. lipid species from  lipid classes (),  lipid
species from  lipid classes ()], reflecting a
Bile acids complex lipid composition. The most abundant lipid
Oxidation of cholesterol within the liver results in the classes found within exosomes are cholesterol, phos-
generation of bile acids, including cholic acid, che- pholipids, sphingolipids, and the mitochondrial lipid,
nodeoxycholic acid, and deoxycholic acid. In addi- cardiolipin (–). Although the lipid composition
tion to well-documented roles in intestinal lipid of exosomes generally resembles the lipid composi-
absorption and cholesterol metabolism (), bile tion of the cell of origin, more so than the exosomal
acids act as signaling molecules by activating the protein content (, ), certain lipids have been
nuclear receptors farnesoid X receptor, pregnane shown to be enriched within all exosomes, as discussed
X receptor, and vitamin D receptor, as well as below.
the G-protein–coupled receptor TGR, which are Little is known about the composition of liver-
expressed within and external to the enterohepatic derived exosomes, especially in NAFLD. Hepatocytes
system (). For the most part, bile acids induce secrete exosomes that carry ceramides and preferen-
favorable metabolic outcomes such as reduced he- tially fuse in a paracrine manner with hepatocytes
patic gluconeogenesis and improved glucose ho- (), and one study examined the composition of
meostasis through farnesoid X receptor (, ) immortalized Huh hepatocellular carcinoma cell
and pregnane X receptor activation (), as well as exosomes and showed that they are enriched in long-
increased energy expenditure in brown adipose tis- chain saturated free fatty acids (C: and C:),
sue, and improved glucose metabolism and insulin distearoyl phosphatidic acid [PA(:/:)], various
sensitivity through TGR () (Fig. ). Notably, phospholipids [particularly the phosphatidyl serine
these metabolic effects are unlikely to be mediated via PS(:/:)], lysophospholipids [e.g., lysophos-
direct actions in muscle and adipose tissue because of phatidylcholines (LPCs), lysophosphatidylserines,
the very low expression of these nuclear receptors in lysophosphatidylglycerol, lysophosphatidylinositol,
these tissues. Readers should refer to de Aguiar lysophosphatidylethanolamine), as well as a variety of
Vallim et al. () for an expansive review of bile acid sphingomyelin and cardiolipin species (). Notably,
effects on metabolism. the composition of human liver-derived exosomes and
Bile acids in NAFLD and insulin resistance. the role of the vast majority of exosomal lipids in
Detailed studies on the interaction between bile intercellular communication are unknown.
acids in the enterohepatic system and NAFLD are One of the more abundant lipids within hepatocyte-
limited, and the small number of investigations in derived exosomes is LPC (). The high LPC

1380 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

Figure 4. Secretion of miRNA into the circulation. Secretion of miRNA encapsulated by (1) exosomes, (2) contained within HDL, or (3)
bound to argonaute 2 (Ago2), which is channeled through vesicle-associated membrane protein 3 (VAMP3) and synaptosomal-associated

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
protein 23 (SNAP23). DAG, diacylglycerol; LPC, lysophosphatidylcholine; TAG, triacylglycerol.

content within exosomes is not surprising, as LPC is abundance of amino acids and TCA intermediates
the most abundantly secreted phospholipid from the in exosomes, including acetate, citrate, pyruvate,
liver (). In addition to being found within exo- a-ketoglutarate, fumarate, and malate, and these can
somes, LPC is transported in the circulation bound to used as substrates by surrounding cells (–).
albumin and is the second most prevalent phospho- Exosomes in NAFLD and insulin resistance.
lipid in plasma (). Circulating LPCs activate Hepatic exosome secretion is increased with
GRP to enhance glucose-stimulated insulin se- NAFLD or lipid overload in vitro (, , ,
cretion () and to increase glucose uptake by in- ). The increase in exosome release with lipid
creasing GLUT translocation (, ), resulting overload is mediated by ER stress (), particularly
in lower blood glucose levels in mice with type  through IRE/XBP signaling (). These lipid
diabetes (). In humans, plasma LPC (and “overloaded” hepatocytes are enriched in C:
lysophosphatidylcholine/alkyl-phosphatidylcholine) ceramide, and they are packaged into exosomes in
levels are reduced in individuals with insulin resistance, either an IRE/XBP-dependent manner through
independent of obesity (), indicating that LPC se- ER stress-induced upregulation of the sphingolipid
cretion may be increased to protect against the insulin biosynthesis enzyme serine C-palmitoyltransferase
resistance associated with NAFLD. Irrespective, it re- (SPT) (), or through the activity of StAR-
mains uncertain as to how LPCs contained within related lipid transfer domain  (STARD), a
exosomes or other extracellular vesicles can be transferred ceramide transport protein (). Increased exo-
in meaningful quantities to impact cell functions in vivo. somal C: ceramide is also reported in mice and
The composition of nonlipid metabolites con- humans with NASH (); however, direct secre-
tained within liver-derived exosomes is not described. tion from the liver was not assessed in this study,
Metabolomic examination in other cells report high and the exosome source was not verified. Ceramides

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1381


REVIEW

stimulate exosome secretion (–), and hepatocyte- has been hindered by the inability to deliver lipids in
derived exosomes preferentially accumulate in stellate cells aqueous environments (e.g., blood, culture medium)

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
and hepatocytes, where they can promote stellate cell in a reproducible, physiologically relevant manner, and
activation and exacerbate liver fibrosis (, , ). This new technologies are urgently required to move this
raises the intriguing possibility that exosomal ceramides field forward.
promote NALFD progression to NASH through paracrine
interactions (, ). miRNA secretion
LPCs are present in hepatocyte-derived exosomes
(), but changes with NAFLD are currently un- miRNAs and intracellular metabolism
known. Patients with NAFLD show reductions in a miRNAs are noncoding RNAs of ~ nucleotides
variety of serum LPC and phosphatidylcholine that regulate gene expression by binding to the 9
species (–), which is related to increased gene untranslated region of mRNAs to repress translation
expression of proteins involved in LPC degradation or guide mRNAs for degradation in lysosomes ().
(). Interestingly, greater abundance of plasma miRNAs typically target mRNAs transcribed from
LPC-: can distinguish insulin-sensitive from insulin- gene clusters rather than single genes, which facili-
resistant NAFL patients, providing potential diagnostic tates critical roles in fundamental biological pro-
value and further supporting an insulin sensitizing role cesses, including cell proliferation, differentiation,
for LPCs (). and apoptosis. Intracellular accumulation of specific
miRNAs can regulate diverse metabolic functions
Summary in a variety of tissues, including skeletal muscle in-
Many studies in mice and humans have demonstrated sulin action, insulin secretion from pancreatic b-cells,
that several liver-derived lipids and metabolites can and adipocyte lipolysis (, ), thereby impli-
serve as signaling molecules to regulate insulin action cating miRNAs in the pathophysiology of metabolic
and other metabolic processes. Recent advances in diseases such as type  diabetes. One specific example
metabolomic technologies have progressed the field by is miR-a and miR-b, which are increased in
confirming correlational relationships in large human NAFLD and target IRS and mRNAs encoding en-
cohorts and for the identification of novel metabolites zymes essential for lipid transport and b-oxidation,
with relationships to metabolic diseases. However, the such as CPTa and AMPK a subunit, to simulta-
causality of most of these metabolites for the regu- neously inhibit insulin signaling and lipid-supported
lation of cell functions remains to be elucidated. This ATP synthesis in the liver ().

Regulation of miRNA secretion


It is now clear that miRNAs are secreted by cells and
can be delivered to recipient cells where they func-
tion as endogenous miRNAs. Although miRNAs
are rapidly degraded by ribonucleases in the plasma
(), miRNAs encapsulated by extracellular vesicles
() are highly stable in the circulation ().
Moreover, miRNAs can also travel through the blood
in association with proteins such as argonaute
(Ago), a key element of the RNA-induced silenc-
ing complex that suppresses gene expression, and
within high-density lipoproteins (HDLs) (, ).
The Ago–miRNA complex is secreted by the in-
teraction of vesicle-associated membrane protein 
(VAMP) and synaptosomal-associated protein 
(SNAP) to create a pore allowing export of the
complex (Fig. ).
Although a detailed discussion of the factors reg-
ulating miRNA secretion is beyond the scope of this
review, there is evidence that neutral sphingomyelinase
 (nSMase), an enzyme that catalyzes the synthesis of
ceramides from sphingomyelin, and ceramide accu-
mulation are important for exosome secretion and
miRNA sorting within vesicles (, ). Moreover,
nSMase can inhibit the export of miRNA by HDLs
Figure 5. Potential roles of miRNA in regulating metabolism in peripheral tissues. miRNAs can be (), suggesting an important role for the nSMase–
transported in the circulation and delivered to other tissues to influence metabolic functions. ceramide axis in regulating miRNA secretion. This has

1382 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

important implications for miRNA “crosstalk” in and insulin action, both in the liver and in distant
NAFLD/NASH, which is characterized by increased tissues. The discovery of new hepatokines and an

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
nSMase activity and ceramide accumulation (). understanding of their biological functions have
provided new targets for intervention strategies to stop
Liver-derived miRNAs and metabolic regulation the rise of metabolic disease, with FGF analogs
To our knowledge, no study has reported changes being a prominent example.
in miRNA secretion from the liver/hepatocytes However, we are only beginning to appreciate
with NAFLD. However, it is clear that many the sheer magnitude of factors secreted by the liver
miRNAs are increased in the liver with NAFLD, and how these are altered in NAFLD. Rapid de-
that many of these miRNAs are also increased in velopments in mass spectrometry have allowed for a
the circulation, and that these same miRNAs can deeper understanding of cell/tissue secretomes, and
regulate insulin secretion and insulin sensitivity in the depth of information is certain to grow. In this
various cell types (–) (Fig. ). For example, review, we have summarized the metabolic effects of
miR- (), miR- (), and miR- () individual hepatokines, and although this knowledge
impair insulin secretion in pancreatic b-cells, and is beneficial in advancing the understanding of
miR- (, ), miR-a (), miR- (), metabolism, both in health and NAFLD, these dis-
miR-b (), miR- (), and miR- () coveries are tempered by the reality that a diverse
impair insulin sensitivity in the liver, skeletal array of changes link NAFLD to insulin resistance. In
muscle, or adipose tissue. Although these data this context, transomic approaches using tissues
suggest a potentially important role for liver- obtained from well-characterized, clinically relevant
derived miRNAs in regulating glycemic control human cohorts will need to be incorporated using
in NAFLD, definitive evidence for causality is systems biology approaches to interrogate the com-
missing, as these miRNAs are also expressed by, plex control underpinning changes in liver-secreted
and secreted from, other tissues. Approaches products and their relationship with metabolic
aimed at tracking the destination cells of adipose- diseases ().
produced miRNAs have been developed (), A central problem with NAFLD is that it rarely
and the use of this technology would help to clarify manifests specific symptoms and diagnosis is
the capacity for liver-derived miRNAs to regulate frequently incidental. Currently, the only reliable
gene expression and metabolism in distant tissues. means of diagnosing and staging NAFLD is by
miRNAs constitute a minor fraction of all noncoding liver biopsy, which is unsuitable for routine use on
RNAs and other regulatory nucleic acids such as individuals at risk for NAFLD. Hence, noninvasive
P-element–induced wimpy testis (PIWI)-interacting tests are increasingly being used in clinical
RNAs and long noncoding RNA have the capacity to practice to assess NAFLD, leading to concerted
regulate gene expression and to regulate glycaemic efforts to identify new serum biomarkers. Aside
control and lipid homeostasis (–), and they from informing on basic biology, future studies
have been detected in exosomes. Given their rela- that interrogate the hepatocyte or liver secretomes
tively recent discovery, it is not surprising that the using “omics” approaches are well positioned to
relevance of these noncoding RNAs for the physi- identify new biomarkers for the development of
ological and pathophysiological regulation of meta- readily available serum tests able to differentiate
bolism remains to be elucidated. the clinically significant forms of NAFLD and to
monitor disease progression noninvasively.
In closing, the field of “hepatokines” biology is
Conclusion rapidly evolving, and it is our hope that future
work in this domain will help unravel the com-
NAFLD is the most common chronic liver disorder in plexities associated with hepatokine regulation of
developed nations, and the notion that NAFLD is insulin resistance in NAFLD, to delineate pre-
closely linked to the development of insulin resistance viously unappreciated communication between
and type  diabetes is well accepted. Work during the the liver and other organs in metabolic control,
last  years has identified a number of hepatokines and advance the clinical management for the
that play critical roles in regulating lipid metabolism treatment of NAFLD-related comorbidities.

References and Notes


1. Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt Association for the Study of Liver Diseases; liver disease: practice guideline by the American
EM, Cusi K, Charlton M, Sanyal AJ; Ameri- American College of Gastroenterology. The di- Gastroenterological Association, American Associ-
can Gastroenterological Association; American agnosis and management of non-alcoholic fatty ation for the Study of Liver Diseases, and American

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1383


REVIEW

College of Gastroenterology. Gastroenterology. Fay DF, Feigin VL, Flaxman A, Forouzanfar MH, 23. Bae JC, Cho YK, Lee WY, Seo HI, Rhee EJ, Park SE,
2012;142(7):1592–1609. Goto A, Green MA, Gupta R, Hafezi-Nejad N, Park CY, Oh KW, Sung KC, Kim BI. Impact of

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
2. Loomba R, Sanyal AJ. The global NAFLD epi- Hankey GJ, Harewood HC, Havmoeller R, Hay S, nonalcoholic fatty liver disease on insulin resistance
demic.Nat Rev Gastroenterol Hepatol. 2013;10(11): Hernandez L, Husseini A, Idrisov BT, Ikeda N, Islami in relation to HbA1c levels in nondiabetic subjects.
686–690. F, Jahangir E, Jassal SK, Jee SH, Jeffreys M, Jonas JB, Am J Gastroenterol. 2010;105(11):2389–2395.
3. Szczepaniak LS, Nurenberg P, Leonard D, Browning Kabagambe EK, Khalifa SE, Kengne AP, Khader YS, 24. Dongiovanni P, Stender S, Pietrelli A, Mancina RM,
JD, Reingold JS, Grundy S, Hobbs HH, Dobbins RL. Khang YH, Kim D, Kimokoti RW, Kinge JM, Kokubo Cespiati A, Petta S, Pelusi S, Pingitore P, Badiali S,
Magnetic resonance spectroscopy to measure Y, Kosen S, Kwan G, Lai T, Leinsalu M, Li Y, Liang X, Maggioni M, Mannisto V, Grimaudo S, Pipitone
hepatic triglyceride content: prevalence of hepatic Liu S, Logroscino G, Lotufo PA, Lu Y, Ma J, Mainoo RM, Pihlajamaki J, Craxi A, Taube M, Carlsson LM,
steatosis in the general population. Am J Physiol NK, Mensah GA, Merriman TR, Mokdad AH, Fargion S, Romeo S, Kozlitina J, Valenti L. Causal
Endocrinol Metab. 2005;288(2):E462–E468. Moschandreas J, Naghavi M, Naheed A, Nand D, relationship of hepatic fat with liver damage and
4. Meex RCR, Watt MJ. Hepatokines: linking non- Narayan KM, Nelson EL, Neuhouser ML, Nisar MI, insulin resistance in nonalcoholic fatty liver. J Intern
alcoholic fatty liver disease and insulin resistance. Ohkubo T, Oti SO, Pedroza A, Prabhakaran D, Roy Med. 2018;283(4):356–370.
Nat Rev Endocrinol. 2017;13(9):509–520. N, Sampson U, Seo H, Sepanlou SG, Shibuya K, Shiri 25. Manchanayake J, Chitturi S, Nolan C, Farrell GC.
5. Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi R, Shiue I, Singh GM, Singh JA, Skirbekk V, Postprandial hyperinsulinemia is universal in non-
K, Rinella M, Harrison SA, Brunt EM, Sanyal AJ. The Stapelberg NJ, Sturua L, Sykes BL, Tobias M, Tran BX, diabetic patients with nonalcoholic fatty liver dis-
diagnosis and management of nonalcoholic fatty Trasande L, Toyoshima H, van de Vijver S, Vasankari ease. J Gastroenterol Hepatol. 2011;26(3):510–516.
liver disease: practice guidance from the American TJ, Veerman JL, Velasquez-Melendez G, Vlassov VV, 26. Bril F, Barb D, Portillo-Sanchez P, Biernacki D,
Association for the Study of Liver Diseases. Hep- Vollset SE, Vos T, Wang C, Wang X, Weiderpass E, Lomonaco R, Suman A, Weber MH, Budd JT, Lupi ME,
atology. 2018;67(1):328–357. Werdecker A, Wright JL, Yang YC, Yatsuya H, Yoon Cusi K. Metabolic and histological implications of
6. Estes C, Razavi H, Loomba R, Younossi Z, Sanyal AJ. J, Yoon SJ, Zhao Y, Zhou M, Zhu S, Lopez AD, intrahepatic triglyceride content in nonalcoholic fatty
Modeling the epidemic of nonalcoholic fatty liver Murray CJ, Gakidou E. Global, regional, and national liver disease. Hepatology. 2017;65(4):1132–1144.
disease demonstrates an exponential increase in prevalence of overweight and obesity in children 27. Stefan N, Fritsche A, Schick F, Häring HU. Pheno-
burden of disease. Hepatology. 2018;67(1):123–133. and adults during 1980–2013: a systematic analysis types of prediabetes and stratification of car-
7. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, for the Global Burden of Disease Study 2013. Lancet. diometabolic risk. Lancet Diabetes Endocrinol. 2016;
Henry L, Wymer M. Global epidemiology of non- 2014;384(9945):766–781. 4(9):789–798.
alcoholic fatty liver disease—meta-analytic assess- 14. Altamirano-Barrera A, Barranco-Fragoso B, Méndez- 28. Stefan N, Schick F, Häring HU. Causes, character-
ment of prevalence, incidence, and outcomes. Sánchez N. Management strategies for liver fibrosis. istics, and consequences of metabolically unhealthy
Hepatology. 2016;64(1):73–84. Ann Hepatol. 2017;16(1):48–56. normal weight in humans. Cell Metab. 2017;26(2):
8. Bugianesi E, Gastaldelli A, Vanni E, Gambino R, 15. Singh S, Allen AM, Wang Z, Prokop LJ, Murad MH, 292–300.
Cassader M, Baldi S, Ponti V, Pagano G, Ferrannini E, Loomba R. Fibrosis progression in nonalcoholic 29. Kraegen EW, Clark PW, Jenkins AB, Daley EA,
Rizzetto M. Insulin resistance in non-diabetic pa- fatty liver vs nonalcoholic steatohepatitis: a sys- Chisholm DJ, Storlien LH. Development of muscle
tients with non-alcoholic fatty liver disease: sites tematic review and meta-analysis of paired-biopsy insulin resistance after liver insulin resistance in
and mechanisms. Diabetologia. 2005;48(4):634–642. studies. Clin Gastroenterol Hepatol.2015;13(4): high-fat-fed rats. Diabetes. 1991;40(11):1397–1403.
9. Korenblat KM, Fabbrini E, Mohammed BS, Klein S. 643–654.e1–9. 30. Turner N, Kowalski GM, Leslie SJ, Risis S, Yang C,
Liver, muscle, and adipose tissue insulin action is 16. Ooi GJ, Burton PR, Earnest A, Laurie C, Kemp WW, Lee-Young RS, Babb JR, Meikle PJ, Lancaster GI,
directly related to intrahepatic triglyceride content Nottle PD, McLean CA, Roberts SK, Brown WA. Henstridge DC, White PJ, Kraegen EW, Marette A,
in obese subjects. Gastroenterology. 2008;134(5): Visual liver score to stratify non-alcoholic steato- Cooney GJ, Febbraio MA, Bruce CR. Distinct pat-
1369–1375. hepatitis risk and determine selective intraoperative terns of tissue-specific lipid accumulation during
10. Seppälä-Lindroos A, Vehkavaara S, Häkkinen AM, liver biopsy in obesity. Obes Surg. 2018;28(2): the induction of insulin resistance in mice by high-
Goto T, Westerbacka J, Sovijärvi A, Halavaara J, Yki- 427–436. fat feeding. Diabetologia. 2013;56(7):1638–1648.
Järvinen H. Fat accumulation in the liver is asso- 17. Ooi GJ, Burton PR, Bayliss J, Raajendiran A, Earnest A, 31. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ,
ciated with defects in insulin suppression of glucose Laurie C, Kemp WW, McLean CA, Roberts SK, Watt Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H.
production and serum free fatty acids independent MJ, Brown WA. Effect of body mass index, meta- Chronic inflammation in fat plays a crucial role in
of obesity in normal men. J Clin Endocrinol Metab. bolic health and adipose tissue inflammation on the development of obesity-related insulin re-
2002;87(7):3023–3028. the severity of non-alcoholic fatty liver disease in sistance. J Clin Invest. 2003;112(12):1821–1830.
11. Marchesini G, Brizi M, Bianchi G, Tomassetti S, bariatric surgical patients: a prospective study. Obes 32. Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J,
Bugianesi E, Lenzi M, McCullough AJ, Natale S, Surg. 2019;29(1):99–108. Boldt MD, Parks EJ. Sources of fatty acids stored in
Forlani G, Melchionda N. Nonalcoholic fatty liver 18. Schwenger KJ, Fischer SE, Jackson TD, Okrainec A, liver and secreted via lipoproteins in patients with
disease: a feature of the metabolic syndrome. Di- Allard JP. Non-alcoholic fatty liver disease in mor- nonalcoholic fatty liver disease. J Clin Invest. 2005;
abetes. 2001;50(8):1844–1850. bidly obese individuals undergoing bariatric surgery: 115(5):1343–1351.
12. Abarca-Gómez L, Abdeen ZA, Hamid ZA, Abu- prevalence and effect of the pre-bariatric very low 33. Shimomura I, Bashmakov Y, Horton JD. Increased
Rmeileh NM, Acosta-Cazares B, Acuin C, Adams RJ, calorie diet. Obes Surg. 2018;28(4):1109–1116. levels of nuclear SREBP-1c associated with fatty
Aekplakorn W, Afsana K, Aguilar-Salinas CA, 19. Musso G, Gambino R, Cassader M, Pagano G. Meta- livers in two mouse models of diabetes mellitus.
Agyemang C, Ahmadvand A, Ahrens W, Ajlouni K, analysis: natural history of non-alcoholic fatty liver J Biol Chem. 1999;274(42):30028–30032.
Akhtaeva N, Al-Hazzaa HM, Al-Othman AR, Al- disease (NAFLD) and diagnostic accuracy of non- 34. Vatner DF, Majumdar SK, Kumashiro N, Petersen
Raddadi R, Al Buhairan F, Al Dhukair S, Ali MM, Ali invasive tests for liver disease severity. Ann Med. MC, Rahimi Y, Gattu AK, Bears M, Camporez JP,
O; NCD Risk Factor Collaboration (NCD-RisC). 2011;43(8):617–649. Cline GW, Jurczak MJ, Samuel VT, Shulman GI.
Worldwide trends in body-mass index, un- 20. Portillo-Sanchez P, Bril F, Maximos M, Lomonaco R, Insulin-independent regulation of hepatic tri-
derweight, overweight, and obesity from 1975 to Biernacki D, Orsak B, Subbarayan S, Webb A, Hecht glyceride synthesis by fatty acids. Proc Natl Acad Sci
2016: a pooled analysis of 2416 population-based J, Cusi K. High prevalence of nonalcoholic fatty liver USA. 2015;112(4):1143–1148.
measurement studies in 128.9 million children, disease in patients with type 2 diabetes mellitus and 35. Sunny NE, Parks EJ, Browning JD, Burgess SC. Ex-
adolescents, and adults. Lancet. 2017;390(10113): normal plasma aminotransferase levels. J Clin cessive hepatic mitochondrial TCA cycle and glu-
2627–2642. Endocrinol Metab. 2015;100(6):2231–2238. coneogenesis in humans with nonalcoholic fatty
13. Ng M, Fleming T, Robinson M, Thomson B, Graetz 21. Ortiz-Lopez C, Lomonaco R, Orsak B, Finch J, Chang liver disease. Cell Metab. 2011;14(6):804–810.
N, Margono C, Mullany EC, Biryukov S, Abbafati C, Z, Kochunov VG, Hardies J, Cusi K. Prevalence of 36. Satapati S, Kucejova B, Duarte JA, Fletcher JA,
Abera SF, Abraham JP, Abu-Rmeileh NM, Achoki T, prediabetes and diabetes and metabolic profile of Reynolds L, Sunny NE, He T, Nair LA, Livingston KA,
AlBuhairan FS, Alemu ZA, Alfonso R, Ali MK, Ali R, patients with nonalcoholic fatty liver disease Fu X, Merritt ME, Sherry AD, Malloy CR, Shelton JM,
Guzman NA, Ammar W, Anwari P, Banerjee A, (NAFLD). Diabetes Care. 2012;35(4):873–878. Lambert J, Parks EJ, Corbin I, Magnuson MA,
Barquera S, Basu S, Bennett DA, Bhutta Z, Blore J, 22. Masarone M, Rosato V, Aglitti A, Bucci T, Caruso R, Browning JD, Burgess SC. Mitochondrial meta-
Cabral N, Nonato IC, Chang JC, Chowdhury R, Salvatore T, Sasso FC, Tripodi MF, Persico M. Liver bolism mediates oxidative stress and inflammation
Courville KJ, Criqui MH, Cundiff DK, Dabhadkar KC, biopsy in type 2 diabetes mellitus: steatohepatitis in fatty liver [published correction appears in J Clin
Dandona L, Davis A, Dayama A, Dharmaratne SD, represents the sole feature of liver damage. PLoS Invest. 2016;126(4):1605]. J Clin Invest. 2015;125(12):
Ding EL, Durrani AM, Esteghamati A, Farzadfar F, One. 2017;12(6):e0178473. 4447–4462.

1384 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

37. Sanyal AJ, Campbell-Sargent C, Mirshahi F, Rizzo WB, metabolic regulation. Physiology (Bethesda). expression of fetuin-A to develop insulin resistance.
Contos MJ, Sterling RK, Luketic VA, Shiffman ML, 2019;34(2):134–149. Endocrinology. 2012;153(7):2974–2984.

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
Clore JN. Nonalcoholic steatohepatitis: association of 50. Ungerleider NA, Bonomi LM, Brown ML, Schneyer 63. Auberger P, Falquerho L, Contreres JO, Pages G, Le
insulin resistance and mitochondrial abnormalities. AL. Increased activin bioavailability enhances he- Cam G, Rossi B, Le Cam A. Characterization of a
Gastroenterology. 2001;120(5):1183–1192. patic insulin sensitivity while inducing hepatic natural inhibitor of the insulin receptor tyrosine
38. Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, steatosis in male mice. Endocrinology. 2013;154(6): kinase: cDNA cloning, purification, and anti-
Pennacchio LA, Boerwinkle E, Cohen JC, Hobbs HH. 2025–2033. mitogenic activity. Cell. 1989;58(4):631–640.
Genetic variation in PNPLA3 confers susceptibility 51. Hashimoto O, Funaba M, Sekiyama K, Doi S, Shindo 64. Pal D, Dasgupta S, Kundu R, Maitra S, Das
to nonalcoholic fatty liver disease. Nat Genet. 2008; D, Satoh R, Itoi H, Oiwa H, Morita M, Suzuki C, G, Mukhopadhyay S, Ray S, Majumdar SS,
40(12):1461–1465. Sugiyama M, Yamakawa N, Takada H, Matsumura S, Bhattacharya S. Fetuin-A acts as an endogenous
39. Stefan N, Häring HU. The metabolically benign and Inoue K, Oyadomari S, Sugino H, Kurisaki A. Activin ligand of TLR4 to promote lipid-induced insulin
malignant fatty liver. Diabetes. 2011;60(8):2011– E controls energy homeostasis in both brown and resistance. Nat Med. 2012;18(8):1279–1285.
2017. white adipose tissues as a hepatokine. Cell Reports. 65. Meex RC, Hoy AJ, Morris A, Brown RD, Lo JC, Burke
40. Kantartzis K, Peter A, Machicao F, Machann J, 2018;25(5):1193–1203. M, Goode RJ, Kingwell BA, Kraakman MJ, Febbraio
Wagner S, Königsrainer I, Königsrainer A, Schick F, 52. Kumar KG, Trevaskis JL, Lam DD, Sutton GM, Koza MA, Greve JW, Rensen SS, Molloy MP, Lancaster GI,
Fritsche A, Häring HU, Stefan N. Dissociation be- RA, Chouljenko VN, Kousoulas KG, Rogers PM, Bruce CR, Watt MJ. Fetuin B is a secreted hepa-
tween fatty liver and insulin resistance in humans Kesterson RA, Thearle M, Ferrante AW Jr, Mynatt tocyte factor linking steatosis to impaired glucose
carrying a variant of the patatin-like phospholipase RL, Burris TP, Dong JZ, Halem HA, Culler MD, Heisler metabolism. Cell Metab. 2015;22(6):1078–1089.
3 gene. Diabetes. 2009;58(11):2616–2623. LK, Stephens JM, Butler AA. Identification of 66. Badman MK, Pissios P, Kennedy AR, Koukos G, Flier
41. Kantartzis K, Machicao F, Machann J, Schick F, adropin as a secreted factor linking dietary mac- JS, Maratos-Flier E. Hepatic fibroblast growth factor
Fritsche A, Häring HU, Stefan N. The DGAT2 gene ronutrient intake with energy homeostasis and lipid 21 is regulated by PPARa and is a key mediator of
is a candidate for the dissociation between fatty metabolism. Cell Metab. 2008;8(6):468–481. hepatic lipid metabolism in ketotic states. Cell
liver and insulin resistance in humans. Clin Sci 53. Gao S, McMillan RP, Zhu Q, Lopaschuk GD, Hulver Metab. 2007;5(6):426–437.
(Lond). 2009;116(6):531–537. MW, Butler AA. Therapeutic effects of adropin on 67. Arner P, Pettersson A, Mitchell PJ, Dunbar JD,
42. Kozlitina J, Smagris E, Stender S, Nordestgaard BG, glucose tolerance and substrate utilization in diet- Kharitonenkov A, Rydén M. FGF21 attenuates li-
Zhou HH, Tybjærg-Hansen A, Vogt TF, Hobbs HH, induced obese mice with insulin resistance. Mol polysis in human adipocytes—a possible link to
Cohen JC. Exome-wide association study identifies a Metab. 2015;4(4):310–324. improved insulin sensitivity. FEBS Lett. 2008;582(12):
TM6SF2 variant that confers susceptibility to 54. Mandard S, Zandbergen F, van Straten E, Wahli W, 1725–1730.
nonalcoholic fatty liver disease. Nat Genet. 2014; Kuipers F, Müller M, Kersten S. The fasting-induced 68. Xu J, Lloyd DJ, Hale C, Stanislaus S, Chen M, Sivits G,
46(4):352–356. adipose factor/angiopoietin-like protein 4 is phys- Vonderfecht S, Hecht R, Li YS, Lindberg RA, Chen JL,
43. Stefan N, Haring HU, Cusi K. Non-alcoholic fatty ically associated with lipoproteins and governs Jung DY, Zhang Z, Ko HJ, Kim JK, Véniant MM.
liver disease: causes, diagnosis, cardiometabolic plasma lipid levels and adiposity. J Biol Chem. 2006; Fibroblast growth factor 21 reverses hepatic stea-
281(2):934–944. tosis, increases energy expenditure, and improves
consequences, and treatment strategies. Lancet
55. Xu A, Lam MC, Chan KW, Wang Y, Zhang J, Hoo insulin sensitivity in diet-induced obese mice. Di-
Diabetes Endocrinol. 2019;7(4):313–324.
RLC, Xu JY, Chen B, Chow WS, Tso AW, Lam KS. abetes. 2009;58(1):250–259.
44. Kim MS, Pinto SM, Getnet D, Nirujogi RS, Manda SS,
Angiopoietin-like protein 4 decreases blood glucose 69. Wente W, Efanov AM, Brenner M, Kharitonenkov
Chaerkady R, Madugundu AK, Kelkar DS, Isserlin R,
and improves glucose tolerance but induces hy- A, Köster A, Sandusky GE, Sewing S, Treinies I, Zitzer
Jain S, Thomas JK, Muthusamy B, Leal-Rojas P, Kumar
perlipidemia and hepatic steatosis in mice. Proc Natl H, Gromada J. Fibroblast growth factor-21 improves
P, Sahasrabuddhe NA, Balakrishnan L, Advani J,
Acad Sci USA. 2005;102(17):6086–6091. pancreatic b-cell function and survival by activa-
George B, Renuse S, Selvan LD, Patil AH, Nanjappa V,
56. Wang Y, Liu LM, Wei L, Ye WW, Meng XY, Chen F, tion of extracellular signal-regulated kinase 1/2 and
Radhakrishnan A, Prasad S, Subbannayya T, Raju R,
Xiao Q, Chen JY, Zhou Y. Angiopoietin-like protein Akt signaling pathways. Diabetes. 2006;55(9):2470–
Kumar M, Sreenivasamurthy SK, Marimuthu A, Sathe
4 improves glucose tolerance and insulin resistance 2478.
GJ, Chavan S, Datta KK, Subbannayya Y, Sahu A,
but induces liver steatosis in high-fat-diet mice. Mol 70. Miyamae T, Marinov AD, Sowders D, Wilson DC,
Yelamanchi SD, Jayaram S, Rajagopalan P, Sharma J,
Med Rep. 2016;14(4):3293–3300. Devlin J, Boudreau R, Robbins P, Hirsch R. Follistatin-
Murthy KR, Syed N, Goel R, Khan AA, Ahmad S, Dey 57. Aryal B, Singh AK, Zhang X, Varela L, Rotllan N, like protein-1 is a novel proinflammatory molecule.
G, Mudgal K, Chatterjee A, Huang TC, Zhong J, Wu X, Goedeke L, Chaube B, Camporez JP, Vatner DF, J Immunol. 2006;177(7):4758–4762.
Shaw PG, Freed D, Zahari MS, Mukherjee KK, Shankar Horvath TL, Shulman GI, Suárez Y, Fernández- 71. Wu HT, Lu FH, Ou HY, Su YC, Hung HC, Wu JS,
S, Mahadevan A, Lam H, Mitchell CJ, Shankar SK, Hernando C. Absence of ANGPTL4 in adipose Yang YC, Wu CL, Chang CJ. The role of hepassocin
Satishchandra P, Schroeder JT, Sirdeshmukh R, Maitra tissue improves glucose tolerance and attenuates in the development of non-alcoholic fatty liver
A, Leach SD, Drake CG, Halushka MK, Prasad TS, atherogenesis. JCI Insight. 2018;3(6):97918. disease [published correction appears in J Hepatol.
Hruban RH, Kerr CL, Bader GD, Iacobuzio-Donahue 58. Singh AK, Aryal B, Chaube B, Rotllan N, Varela L, 2017;66(2):468]. J Hepatol. 2013;59(5):1065–1072.
CA, Gowda H, Pandey A. A draft map of the human Horvath TL, Suárez Y, Fernández-Hernando C. 72. Wu HT, Ou HY, Hung HC, Su YC, Lu FH, Wu JS,
proteome. Nature. 2014;509(7502):575–581. Brown adipose tissue derived ANGPTL4 controls Yang YC, Wu CL, Chang CJ. A novel hepatokine,
45. Azimifar SB, Nagaraj N, Cox J, Mann M. Cell-type- glucose and lipid metabolism and regulates ther- HFREP1, plays a crucial role in the development of
resolved quantitative proteomics of murine liver. mogenesis. Mol Metab. 2018;11:59–69. insulin resistance and type 2 diabetes. Diabetologia.
Cell Metab. 2014;20(6):1076–1087. 59. Baumeier C, Schlüter L, Saussenthaler S, Laeger T, 2016;59(8):1732–1742.
46. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Rödiger M, Alaze SA, Fritsche L, Häring HU, Stefan 73. Zeng W, Shan W, Gao L, Gao D, Hu Y, Wang G,
Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, N, Fritsche A, Schwenk RW, Schürmann A. Elevated Zhang N, Li Z, Tian X, Xu W, Peng J, Ma X, Yao J.
Sjöstedt E, Asplund A, Olsson I, Edlund K, Lundberg hepatic DPP4 activity promotes insulin resistance Inhibition of HMGB1 release via salvianolic acid
E, Navani S, Szigyarto CA, Odeberg J, Djureinovic D, and non-alcoholic fatty liver disease. Mol Metab. B-mediated SIRT1 up-regulation protects rats
Takanen JO, Hober S, Alm T, Edqvist PH, Berling H, 2017;6(10):1254–1263. against non-alcoholic fatty liver disease. Sci Rep.
Tegel H, Mulder J, Rockberg J, Nilsson P, Schwenk 60. Drucker DJ, Nauck MA. The incretin system: 2015;5(1):16013.
JM, Hamsten M, von Feilitzen K, Forsberg M, glucagon-like peptide-1 receptor agonists and 74. Zhang W, Wang LW, Wang LK, Li X, Zhang H, Luo LP,
Persson L, Johansson F, Zwahlen M, von Heijne G, dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Song JC, Gong ZJ. Betaine protects against high-fat-
Nielsen J, Pontén F. Tissue-based map of the human Lancet. 2006;368(9548):1696–1705. diet-induced liver injury by inhibition of high-mobility
proteome. Science. 2015;347(6220):1260419. 61. Awazawa M, Gabel P, Tsaousidou E, Nolte H, Krüger group box 1 and Toll-like receptor 4 expression in rats.
47. Michael DJ, Cai H, Xiong W, Ouyang J, Chow RH. M, Schmitz J, Ackermann PJ, Brandt C, Altmüller J, Dig Dis Sci. 2013;58(11):3198–3206.
Mechanisms of peptide hormone secretion. Trends Motameny S, Wunderlich FT, Kornfeld JW, Blüher 75. Li L, Chen L, Hu L, Liu Y, Sun HY, Tang J, Hou YJ,
Endocrinol Metab. 2006;17(10):408–415. M, Brüning JC. A microRNA screen reveals that Chang YX, Tu QQ, Feng GS, Shen F, Wu MC, Wang
48. Hessvik NP, Llorente A. Current knowledge on elevated hepatic ectodysplasin A expression con- HY. Nuclear factor high-mobility group box1 me-
exosome biogenesis and release. Cell Mol Life Sci. tributes to obesity-induced insulin resistance in diating the activation of Toll-like receptor 4 sig-
2018;75(2):193–208. skeletal muscle. Nat Med. 2017;23(12):1466–1473. naling in hepatocytes in the early stage of
49. Montgomery MK, De Nardo W, Watt MJ. Im- 62. Ou HY, Wu HT, Hung HC, Yang YC, Wu JS, Chang nonalcoholic fatty liver disease in mice. Hepatology.
pact of lipotoxicity on tissue “cross talk” and CJ. Endoplasmic reticulum stress induces the 2011;54(5):1620–1630.

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1385


REVIEW

76. Lan F, Misu H, Chikamoto K, Takayama H, Kikuchi Kanson T, Ladoux A, Elabd C, Scheideler M, Trajanoski Schneider M, Van Sligtenhorst I, Suwanichkul A,
A, Mohri K, Takata N, Hayashi H, Matsuzawa- Z, Takashima Y, Amri EZ, Lacasa D, Sengenes C, Vogel P, Wilganowski N, Wingert J, Zambrowicz BP,

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
Nagata N, Takeshita Y, Noda H, Matsumoto Y, Ailhaud G, Clément K, Bouloumie A, Kirkland JL, Dani Landes G, Powell DR. Lipid-lowering effects of anti-
Ota T, Nagano T, Nakagen M, Miyamoto K, C. Activin a plays a critical role in proliferation and angiopoietin-like 4 antibody recapitulate the lipid
Takatsuki K, Seo T, Iwayama K, Tokuyama K, differentiation of human adipose progenitors. Di- phenotype found in angiopoietin-like 4 knockout
Matsugo S, Tang H, Saito Y, Yamagoe S, Kaneko S, abetes. 2010;59(10):2513–2521. mice. Proc Natl Acad Sci USA. 2007;104(28):
Takamura T. LECT2 functions as a hepatokine that 89. Yndestad A, Haukeland JW, Dahl TB, Bjøro K, 11766–11771.
links obesity to skeletal muscle insulin resistance. Gladhaug IP, Berge C, Damås JK, Haaland T, Løberg 102. Kersten S, Mandard S, Tan NS, Escher P, Metzger D,
Diabetes. 2014;63(5):1649–1664. EM, Linnestad P, Birkeland K, Konopski Z, Halvorsen Chambon P, Gonzalez FJ, Desvergne B, Wahli W.
77. Hirosumi J, Tuncman G, Chang L, Görgün CZ, Uysal B, Berge RK, Aukrust P. A complex role of activin A Characterization of the fasting-induced adipose
KT, Maeda K, Karin M, Hotamisligil GS. A central in non-alcoholic fatty liver disease. Am J Gastro- factor FIAF, a novel peroxisome proliferator-
role for JNK in obesity and insulin resistance. Nature. enterol. 2009;104(9):2196–2205. activated receptor target gene. J Biol Chem. 2000;
2002;420(6913):333–336. 90. Stevens JR, Kearney ML, St-Onge MP, Stanhope KL, 275(37):28488–28493.
78. Borg ML, Andrews ZB, Duh EJ, Zechner R, Meikle PJ, Havel PJ, Kanaley JA, Thyfault JP, Weiss EP, Butler AA. 103. Mizutani N, Ozaki N, Seino Y, Fukami A, Sakamoto
Watt MJ. Pigment epithelium-derived factor reg- Inverse association between carbohydrate con- E, Fukuyama T, Sugimura Y, Nagasaki H, Arima H,
ulates lipid metabolism via adipose triglyceride li- sumption and plasma adropin concentrations in Oiso Y. Reduction of insulin signaling upregulates
pase. Diabetes. 2011;60(5):1458–1466. humans. Obesity (Silver Spring). 2016;24(8):1731–1740. angiopoietin-like protein 4 through elevated free
79. Chung C, Doll JA, Gattu AK, Shugrue C, Cornwell M, 91. Butler AA, Tam CS, Stanhope KL, Wolfe BM, Ali MR, fatty acids in diabetic mice. Exp Clin Endocrinol
Fitchev P, Crawford SE. Anti-angiogenic pigment O’Keeffe M, St-Onge MP, Ravussin E, Havel PJ. Low Diabetes. 2012;120(3):139–144.
epithelium-derived factor regulates hepatocyte circulating adropin concentrations with obesity 104. Bernsmeier C, Meyer-Gerspach AC, Blaser LS, Jeker L,
triglyceride content through adipose triglyceride and aging correlate with risk factors for metabolic Steinert RE, Heim MH, Beglinger C. Glucose-
lipase (ATGL). J Hepatol. 2008;48(3):471–478. disease and increase after gastric bypass surgery in induced glucagon-like peptide 1 secretion is de-
80. Crowe S, Wu LE, Economou C, Turpin SM, Matzaris humans. J Clin Endocrinol Metab. 2012;97(10): ficient in patients with non-alcoholic fatty liver
M, Hoehn KL, Hevener AL, James DE, Duh EJ, Watt 3783–3791. disease. PLoS One. 2014;9(1):e87488.
MJ. Pigment epithelium-derived factor contributes 92. Chen S, Zeng K, Liu QC, Guo Z, Zhang S, Chen XR, 105. Lamers D, Famulla S, Wronkowitz N, Hartwig S, Lehr
to insulin resistance in obesity. Cell Metab. 2009; Lin JH, Wen JP, Zhao CF, Lin XH, Gao F. Adropin S, Ouwens DM, Eckardt K, Kaufman JM, Ryden M,
10(1):40–47. deficiency worsens HFD-induced metabolic defects. Müller S, Hanisch FG, Ruige J, Arner P, Sell H, Eckel J.
81. Fedders R, Muenzner M, Weber P, Sommerfeld M, Cell Death Dis. 2017;8(8):e3008. Dipeptidyl peptidase 4 is a novel adipokine po-
Knauer M, Kedziora S, Kast N, Heidenreich S, Raila J, 93. Ganesh Kumar K, Zhang J, Gao S, Rossi J, tentially linking obesity to the metabolic syndrome.
Weger S, Henze A, Schupp M. Liver-secreted RBP4 McGuinness OP, Halem HH, Culler MD, Mynatt RL, Diabetes. 2011;60(7):1917–1925.
does not impair glucose homeostasis in mice. J Biol Butler AA. Adropin deficiency is associated with 106. Murgia A, Caboni P, Cadoni E, Serra M, Marongiu F,
Chem. 2018;293(39):15269–15276. increased adiposity and insulin resistance. Obesity Laconi E. A GC-MS untargeted metabolomics
82. Norseen J, Hosooka T, Hammarstedt A, Yore MM, (Silver Spring). 2012;20(7):1394–1402. analysis in the plasma and liver of rats lacking
Kant S, Aryal P, Kiernan UA, Phillips DA, Maruyama 94. Gao S, McMillan RP, Jacas J, Zhu Q, Li X, Kumar GK, dipeptidyl-peptidase type IV enzyme activity.
H, Kraus BJ, Usheva A, Davis RJ, Smith U, Kahn BB. Casals N, Hegardt FG, Robbins PD, Lopaschuk GD, J Physiol Biochem. 2017;73(4):575–582.
Retinol-binding protein 4 inhibits insulin signaling Hulver MW, Butler AA. Regulation of substrate 107. Marques AP, Cunha-Santos J, Leal H, Sousa-Ferreira
in adipocytes by inducing proinflammatory cyto- oxidation preferences in muscle by the peptide L, Pereira de Almeida L, Cavadas C, Rosmaninho-
kines in macrophages through a c-Jun N-terminal hormone adropin. Diabetes. 2014;63(10):3242– Salgado J. Dipeptidyl peptidase IV (DPP-IV) in-
kinase- and Toll-like receptor 4-dependent and 3252. hibition prevents fibrosis in adipose tissue of obese
retinol-independent mechanism. Mol Cell Biol. 2012; 95. Ingerslev B, Hansen JS, Hoffmann C, Clemmesen JO, mice. Biochim Biophys Acta, Gen Subj. 2018;1862(3):
32(10):2010–2019. Secher NH, Scheler M, Hrabĕ de Angelis M, Häring HU, 403–413.
83. Misu H, Takamura T, Takayama H, Hayashi H, Pedersen BK, Weigert C, Plomgaard P. Angiopoietin- 108. Zheng W, Zhou J, Song S, Kong W, Xia W, Chen L,
Matsuzawa-Nagata N, Kurita S, Ishikura K, Ando H, like protein 4 is an exercise-induced hepatokine in Zeng T. Dipeptidyl-peptidase 4 inhibitor sitagliptin
Takeshita Y, Ota T, Sakurai M, Yamashita T, humans, regulated by glucagon and cAMP. Mol Metab. ameliorates hepatic insulin resistance by modu-
Mizukoshi E, Yamashita T, Honda M, Miyamoto K, 2017;6(10):1286–1295. lating inflammation and autophagy in ob/ob mice.
Kubota T, Kubota N, Kadowaki T, Kim HJ, Lee IK, 96. McQueen AE, Kanamaluru D, Yan K, Gray NE, Wu L, Int J Endocrinol. 2018;2018:8309723.
Minokoshi Y, Saito Y, Takahashi K, Yamada Y, Li ML, Chang A, Hasan A, Stifler D, Koliwad SK, 109. Mori K, Emoto M, Yokoyama H, Araki T, Teramura
Takakura N, Kaneko S. A liver-derived secretory Wang JC. The C-terminal fibrinogen-like domain of M, Koyama H, Shoji T, Inaba M, Nishizawa Y. As-
protein, selenoprotein P, causes insulin resistance. angiopoietin-like 4 stimulates adipose tissue lipol- sociation of serum fetuin-A with insulin resistance
Cell Metab. 2010;12(5):483–495. ysis and promotes energy expenditure. J Biol Chem. in type 2 diabetic and nondiabetic subjects. Di-
84. Mita Y, Nakayama K, Inari S, Nishito Y, Yoshioka Y, 2017;292(39):16122–16134. abetes Care. 2006;29(2):468.
Sakai N, Sotani K, Nagamura T, Kuzuhara Y, Inagaki 97. Robal T, Larsson M, Martin M, Olivecrona G, 110. Peter A, Kovarova M, Staiger H, Machann J, Schick F,
K, Iwasaki M, Misu H, Ikegawa M, Takamura T, Lookene A. Fatty acids bind tightly to the N- Königsrainer A, Königsrainer I, Schleicher E, Fritsche
Noguchi N, Saito Y. Selenoprotein P-neutralizing terminal domain of angiopoietin-like protein 4 and A, Häring HU, Stefan N. The hepatokines fetuin-A
antibodies improve insulin secretion and glucose modulate its interaction with lipoprotein lipase. and fetuin-B are upregulated in the state of hepatic
sensitivity in type 2 diabetes mouse models. Nat J Biol Chem. 2012;287(35):29739–29752. steatosis and may differently impact on glucose
Commun. 2017;8(1):1658. 98. Hansen JS, Pedersen BK, Xu G, Lehmann R, Weigert homeostasis in humans. Am J Physiol Endocrinol
85. Sáez-López C, Salcedo-Allende MT, Hernandez C, C, Plomgaard P. Exercise-induced secretion of Metab. 2018;314(3):E266–E273.
Simó-Servat O, Simó R, Selva DM. Sex hormone– FGF21 and follistatin are blocked by pancreatic 111. Jung CH, Kim BY, Kim CH, Kang SK, Jung SH, Mok
binding globulin expression correlates with acetyl- clamp and impaired in type 2 diabetes. J Clin JO. Associations of serum fetuin-A levels with in-
coenzyme A carboxylase and triglyceride content Endocrinol Metab. 2016;101(7):2816–2825. sulin resistance and vascular complications in pa-
in human liver. J Clin Endocrinol Metab. 2019;104(5): 99. Okamoto H, Cavino K, Na E, Krumm E, Kim S, Stevis tients with type 2 diabetes. Diab Vasc Dis Res. 2013;
1500–1507. PE, Harp J, Murphy AJ, Yancopoulos GD, Gromada J. 10(5):459–467.
86. Xiong X, Wang Q, Wang S, Zhang J, Liu T, Guo L, Yu Angptl4 does not control hyperglucagonemia or 112. Shim YS, Kang MJ, Oh YJ, Baek JW, Yang S, Hwang IT.
Y, Lin JD. Mapping the molecular signatures of diet- a-cell hyperplasia following glucagon receptor in- Fetuin-A as an alternative marker for insulin resistance
induced NASH and its regulation by the hepatokine hibition. Proc Natl Acad Sci USA. 2017;114(10): and cardiovascular risk in prepubertal children.
Tsukushi. Mol Metab. 2019;20:128–137. 2747–2752. J Atheroscler Thromb. 2017;24(10):1031–1038.
87. Fang J, Wang SQ, Smiley E, Bonadio J. Genes coding 100. Romeo S, Pennacchio LA, Fu Y, Boerwinkle E, 113. Zhao J, Xiong X, Li Y, Liu X, Wang T, Zhang H, Jiao Y,
for mouse activin bC and bE are closely linked and Tybjaerg-Hansen A, Hobbs HH, Cohen JC. Jiang J, Zhang H, Tang Q, Gao X, Li X, Lu Y, Liu B, Hu
exhibit a liver-specific expression pattern in adult Population-based resequencing of ANGPTL4 un- C, Li X. Hepatic F-box protein FBXW7 maintains
tissues. Biochem Biophys Res Commun. 1997;231(3): covers variations that reduce triglycerides and in- glucose homeostasis through degradation of fetuin-
655–661. crease HDL. Nat Genet. 2007;39(4):513–516. A. Diabetes. 2018;67(5):818–830.
88. Zaragosi LE, Wdziekonski B, Villageois P, Keophiphath 101. Desai U, Lee EC, Chung K, Gao C, Gay J, Key B, 114. Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H,
M, Maumus M, Tchkonia T, Bourlier V, Mohsen- Hansen G, Machajewski D, Platt KA, Sands AT, Flier JS. TLR4 links innate immunity and fatty

1386 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

acid-induced insulin resistance. J Clin Invest. 2006; 129. Staiger H, Keuper M, Berti L, Hrabe de Angelis M, 144. Wu H, Ballantyne CM. Skeletal muscle in-
116(11):3015–3025. Häring HU. Fibroblast growth factor 21—metabolic flammation and insulin resistance in obesity. J Clin

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
115. Mathews ST, Singh GP, Ranalletta M, Cintron VJ, role in mice and men. Endocr Rev. 2017;38(5): Invest. 2017;127(1):43–54.
Qiang X, Goustin AS, Jen KL, Charron MJ, Jahnen- 468–488. 145. Hara H, Yoshimura H, Uchida S, Toyoda Y, Aoki M,
Dechent W, Grunberger G. Improved insulin sen- 130. Dushay J, Chui PC, Gopalakrishnan GS, Varela-Rey Sakai Y, Morimoto S, Shiokawa K. Molecular cloning
sitivity and resistance to weight gain in mice null for M, Crawley M, Fisher FM, Badman MK, Martinez- and functional expression analysis of a cDNA for
the Ahsg gene. Diabetes. 2002;51(8):2450–2458. Chantar ML, Maratos-Flier E. Increased fibroblast human hepassocin, a liver-specific protein with
116. Pierre N, Deldicque L, Barbé C, Naslain D, Cani PD, growth factor 21 in obesity and nonalcoholic fatty hepatocyte mitogenic activity. Biochim Biophys
Francaus M. Toll-like receptor 4 knockout mice are liver disease. Gastroenterology. 2010;139(2):456–463. Acta. 2001;1520(1):45–53.
protected against endoplasmic reticulum stress 131. Li H, Fang Q, Gao F, Fan J, Zhou J, Wang X, Zhang H, 146. Yu HT, Yu M, Li CY, Zhan YQ, Xu WX, Li YH, Li W,
induced by a high-fat diet. Plos One. 2013;8(5): Pan X, Bao Y, Xiang K, Xu A, Jia W. Fibroblast growth Wang ZD, Ge CH, Yang XM. Specific expression
e65061. factor 21 levels are increased in nonalcoholic fatty and regulation of hepassocin in the liver and
117. Stefan N, Häring HU. Circulating fetuin-A and free liver disease patients and are correlated with he- down-regulation of the correlation of HNF1a with
fatty acids interact to predict insulin resistance in patic triglyceride. J Hepatol. 2010;53(5):934–940. decreased levels of hepassocin in human hepa-
humans. Nat Med. 2013;19(4):394–395. 132. Li X, Ge H, Weiszmann J, Hecht R, Li YS, Véniant tocellular carcinoma. J Biol Chem. 2009;284(20):
118. Gerst F, Wagner R, Kaiser G, Panse M, Heni M, MM, Xu J, Wu X, Lindberg R, Li Y. Inhibition of 13335–13347.
Machann J, Bongers MN, Sartorius T, Sipos B, Fend F, lipolysis may contribute to the acute regulation of 147. Gao M, Zhan YQ, Yu M, Ge CH, Li CY, Zhang JH,
Thiel C, Nadalin S, Königsrainer A, Stefan N, Fritsche plasma FFA and glucose by FGF21 in ob/ob mice. Wang XH, Ge ZQ, Yang XM. Hepassocin activates
A, Häring HU, Ullrich S, Siegel-Axel D. Metabolic FEBS Lett. 2009;583(19):3230–3234. the EGFR/ERK cascade and induces proliferation of
crosstalk between fatty pancreas and fatty liver: 133. Coskun T, Bina HA, Schneider MA, Dunbar JD, Hu L02 cells through the Src-dependent pathway. Cell
effects on local inflammation and insulin secretion. CC, Chen Y, Moller DE, Kharitonenkov A. Fibroblast Signal. 2014;26(10):2161–2166.
Diabetologia. 2017;60(11):2240–2251. growth factor 21 corrects obesity in mice. Endo- 148. Sreekumar R, Rosado B, Rasmussen D, Charlton M.
119. Shen X, Yang L, Yan S, Zheng H, Liang L, Cai X, Liao crinology. 2008;149(12):6018–6027. Hepatic gene expression in histologically pro-
M. Fetuin A promotes lipotoxicity in b cells 134. Berglund ED, Li CY, Bina HA, Lynes SE, Michael MD, gressive nonalcoholic steatohepatitis. Hepatology.
through the TLR4 signaling pathway and the role of Shanafelt AB, Kharitonenkov A, Wasserman DH. 2003;38(1):244–251.
pioglitazone in anti-lipotoxicity. Mol Cell Endocrinol. Fibroblast growth fact or 21 controls glycemia via 149. Jung TW, Chung YH, Kim HC, Abd El-Aty AM, Jeong
2015;412:1–11. regulation of hepatic glucose flux and insulin JH. Hyperlipidemia-induced hepassocin in the liver
120. Mukhuty A, Fouzder C, Mukherjee S, Malick C, sensitivity. Endocrinology. 2009;150(9):4084–4093. contributes to insulin resistance in skeletal muscle.
Mukhopadhyay S, Bhattacharya S, Kundu R. Pal- 135. Schlein C, Talukdar S, Heine M, Fischer AW, Krott Mol Cell Endocrinol. 2018;470:26–33.
mitate induced fetuin-A secretion from pancreatic LM, Nilsson SK, Brenner MB, Heeren J, Scheja L. 150. Puri P, Mirshahi F, Cheung O, Natarajan R, Maher
FGF21 lowers plasma triglycerides by accelerating JW, Kellum JM, Sanyal AJ. Activation and dysre-
b-cells adversely affects its function and elicits in-
lipoprotein catabolism in white and brown adipose gulation of the unfolded protein response in
flammation. Biochem Biophys Res Commun. 2017;
tissues. Cell Metab. 2016;23(3):441–453. nonalcoholic fatty liver disease. Gastroenterology.
491(4):1118–1124.
136. Camporez JP, Asrih M, Zhang D, Kahn M, Samuel 2008;134(2):568–576.
121. Mathews ST, Rakhade S, Zhou X, Parker GC, Coscina
VT, Jurczak MJ, Jornayvaz FR. Hepatic insulin re- 151. Back SH, Kang SW, Han J, Chung HT. Endoplasmic
DV, Grunberger G. Fetuin-null mice are protected
sistance and increased hepatic glucose production reticulum stress in the b-cell pathogenesis of type 2
against obesity and insulin resistance associated
in mice lacking Fgf21. J Endocrinol. 2015;226(3): diabetes. Exp Diabetes Res. 2012;2012:618396.
with aging. Biochem Biophys Res Commun. 2006;
207–217. 152. Tsutsumi A, Motoshima H, Kondo T, Kawasaki S,
350(2):437–443.
137. Sonoda J, Chen MZ, Baruch A. FGF21-receptor Matsumura T, Hanatani S, Igata M, Ishii N, Kinoshita
122. Zhu J, Wan X, Wang Y, Zhu K, Li C, Yu C, Li Y. Serum
agonists: an emerging therapeutic class for H, Kawashima J, Taketa K, Furukawa N, Tsuruzoe K,
fetuin B level increased in subjects of nonalcoholic
obesity-related diseases. Horm Mol Biol Clin Investig. Nishikawa T, Araki E. Caloric restriction decreases ER
fatty liver disease: a case-control study. Endocrine.
2017;30(2):/j/hmbci.2017.30.issue-2/hmbci-2017- stress in liver and adipose tissue in ob/ob mice.
2017;56(1):208–211.
0002/hmbci-2017-0002.xml. Biochem Biophys Res Commun. 2011;404(1):339–
123. Li Z, Lin M, Liu C, Wang D, Shi X, Chen Z, Liu Y, Yang
138. Ueno N, Ling N, Ying SY, Esch F, Shimasaki S, 344.
S, Li X. Fetuin-B links nonalcoholic fatty liver disease
Guillemin R. Isolation and partial characterization of 153. Andersson U, Tracey KJ. HMGB1 is a therapeutic
to type 2 diabetes via inducing insulin resistance: follistatin: a single-chain Mr 35,000 monomeric target for sterile inflammation and infection. Annu
association and path analyses. Cytokine. 2018;108: protein that inhibits the release of follicle- Rev Immunol. 2011;29(1):139–162.
145–150. stimulating hormone. Proc Natl Acad Sci USA. 154. Yates KP, Deppe R, Comerford M, Masuoka H,
124. Qu H, Qiu Y, Wang Y, Liao Y, Zheng Y, Zheng H. 1987;84(23):8282–8286. Cummings OW, Tonascia J, Chalasani N, Vuppalanchi
Plasma fetuin-B concentrations are associated with 139. Lee SJ, McPherron AC. Regulation of myostatin R; NASH CRN. Serum high mobility group box 1
insulin resistance and first-phase glucose-stimulated activity and muscle growth. Proc Natl Acad Sci USA. protein levels are not associated with either histo-
insulin secretion in individuals with different de- 2001;98(16):9306–9311. logical severity or treatment response in children and
grees of glucose tolerance. Diabetes Metab. 2018; 140. Tao R, Wang C, Stöhr O, Qiu W, Hu Y, Miao J, Dong adults with nonalcoholic fatty liver disease. PLoS One.
44(6):488–492. XC, Leng S, Stefater M, Stylopoulos N, Lin L, Copps 2017;12(11):e0185813.
125. Kralisch S, Hoffmann A, Lössner U, Kratzsch J, Blüher KD, White MF. Inactivating hepatic follistatin al- 155. Sugiyama M, Kikuchi A, Misu H, Igawa H, Ashihara
M, Stumvoll M, Fasshauer M, Ebert T. Regulation of leviates hyperglycemia [published correction ap- M, Kushima Y, Honda K, Suzuki Y, Kawabe Y,
the novel adipokines/hepatokines fetuin A and pears in Nat Med. 2018;24(10):1628]. Nat Med. 2018; Kaneko S, Takamura T. Inhibin bE (INHBE) is a
fetuin B in gestational diabetes mellitus. Metab- 24(7):1058–1069. possible insulin resistance-associated hepatokine
olism. 2017;68:88–94. 141. Perakakis N, Kokkinos A, Peradze N, Tentolouris N, identified by comprehensive gene expression
126. Šimják P, Cinkajzlová A, Anderlová K, Kloučková J, Ghaly W, Tsilingiris D, Alexandrou A, Mantzoros CS. analysis in human liver biopsy samples. PLoS One.
Kratochvı́lová H, Lacinová Z, Kaválková P, Krejčı́ H, Follistatins in glucose regulation in healthy and 2018;13(3):e0194798.
Mráz M, Pařı́zek A, Kršek M, Haluzı́k M. Changes in obese individuals. Diabetes Obes Metab. 2019;21(3): 156. Hashimoto O, Sekiyama K, Matsuo T, Hasegawa Y.
plasma concentrations and mRNA expression of 683–690. Implication of activin E in glucose metabolism:
hepatokines fetuin A, fetuin B and FGF21 in 142. Ramnath D, Irvine KM, Lukowski SW, Horsfall LU, transcriptional regulation of the inhibin/activin
physiological pregnancy and gestational diabetes Loh Z, Clouston AD, Patel PJ, Fagan KJ, Iyer A, Lampe betaE subunit gene in the liver. Life Sci. 2009;
mellitus. Physiol Res. 2018;67(Suppl 3):S531–S542. G, Stow JL, Schroder K, Fairlie DP, Powell JE, Powell 85(13–14):534–540.
127. Best JD, Kahn SE, Ader M, Watanabe RM, Ni TC, EE, Sweet MJ. Hepatic expression profiling identifies 157. Lau AL, Kumar TR, Nishimori K, Bonadio J, Matzuk
Bergman RN. Role of glucose effectiveness in the steatosis-independent and steatosis-driven ad- MM. Activin bC and bE genes are not essential for
determination of glucose tolerance. Diabetes Care. vanced fibrosis genes. JCI Insight. 2018;3(14):120274. mouse liver growth, differentiation, and re-
1996;19(9):1018–1030. 143. Jager J, Grémeaux T, Cormont M, Le Marchand- generation. Mol Cell Biol. 2000;20(16):6127–6137.
128. Nishimura T, Nakatake Y, Konishi M, Itoh N. Brustel Y, Tanti JF. Interleukin-1b-induced insulin 158. Yamagoe S, Yamakawa Y, Matsuo Y, Minowada J,
Identification of a novel FGF, FGF-21, preferentially resistance in adipocytes through down-regulation Mizuno S, Suzuki K. Purification and primary amino
expressed in the liver. Biochim Biophys Acta. 2000; of insulin receptor substrate-1 expression. Endocri- acid sequence of a novel neutrophil chemotactic
1492(1):203–206. nology. 2007;148(1):241–251. factor LECT2. Immunol Lett. 1996;52(1):9–13.

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1387


REVIEW

159. Yoo HJ, Hwang SY, Choi JH, Lee HJ, Chung HS, Seo 171. Ho TC, Chen SL, Shih SC, Wu JY, Han WH, Cheng sex hormone-binding globulin gene promoter. J Biol
JA, Kim SG, Kim NH, Baik SH, Choi DS, Choi KM. HC, Yang SL, Tsao YP. Pigment epithelium-derived Chem. 1998;273(51):34105–34114.

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
Association of leukocyte cell-derived chemotaxin 2 factor is an intrinsic antifibrosis factor targeting 186. Selva DM, Hammond GL. Peroxisome-proliferator
(LECT2) with NAFLD, metabolic syndrome, and hepatic stellate cells. Am J Pathol. 2010;177(4): receptor g represses hepatic sex hormone-binding
atherosclerosis. PLoS One. 2017;12(4):e0174717. 1798–1811. globulin expression. Endocrinology. 2009;150(5):
160. Chikamoto K, Misu H, Takayama H, Kikuchi A, Ishii 172. Kanai M, Raz A, Goodman DS. Retinol-binding 2183–2189.
KA, Lan F, Takata N, Tajima-Shirasaki N, Takeshita Y, protein: the transport protein for vitamin A in 187. Luo J, Chen Q, Shen T, Wang X, Fang W, Wu X, Yuan
Tsugane H, Kaneko S, Matsugo S, Takamura T. human plasma. J Clin Invest. 1968;47(9):2025–2044. Z, Chen G, Ling W, Chen Y. Association of sex
Rapid response of the steatosis-sensing hepatokine 173. Cai H, Lu S, Chen Y, Das Mbbs Mrcog S, Niu Z, Zhuo hormone-binding globulin with nonalcoholic fatty
LECT2 during diet-induced weight cycling in mice. G, Lai L, Zhang Z. Serum retinol binding protein 4 liver disease in Chinese adults. Nutr Metab (Lond).
Biochem Biophys Res Commun. 2016;478(3):1310– and galectin-3 binding protein as novel markers for 2018;15(1):79.
1316. postmenopausal nonalcoholic fatty liver disease. 188. Hua X, Li M, Pan F, Xiao Y, Cui W, Hu Y. Non-
161. Hwang HJ, Jung TW, Kim BH, Hong HC, Seo JA, Kim Clin Biochem. 2018;56:95–101. alcoholic fatty liver disease is an influencing factor
SG, Kim NH, Choi KM, Choi DS, Baik SH, Yoo HJ. A 174. Perseghin G, Lattuada G, De Cobelli F, Esposito A, for the association of SHBG with metabolic syn-
dipeptidyl peptidase-IV inhibitor improves he- Belloni E, Canu T, Ragogna F, Scifo P, Del Maschio A, drome in diabetes patients. Sci Rep. 2017;7(1):14532.
patic steatosis and insulin resistance by AMPK- Luzi L. Serum retinol-binding protein-4, leptin, and 189. Daka B, Rosen T, Jansson PA, Råstam L, Larsson CA,
dependent and JNK-dependent inhibition of adiponectin concentrations are related to ectopic Lindblad U. Inverse association between serum
LECT2 expression. Biochem Pharmacol. 2015;98(1): fat accumulation. J Clin Endocrinol Metab. 2007; insulin and sex hormone-binding globulin in a
157–166. 92(12):4883–4888. population survey in Sweden. Endocr Connect. 2012;
162. Tombran-Tink J, Mazuruk K, Rodriguez IR, Chung D, 175. Wu H, Jia W, Bao Y, Lu J, Zhu J, Wang R, Chen Y, 2(1):18–22.
Linker T, Englander E, Chader GJ. Organization, Xiang K. Serum retinol binding protein 4 and 190. Kavanagh K, Espeland MA, Sutton-Tyrrell K, Barinas-
evolutionary conservation, expression and unusual nonalcoholic fatty liver disease in patients with type Mitchell E, El Khoudary SR, Wildman RP. Liver fat
Alu density of the human gene for pigment 2 diabetes mellitus. Diabetes Res Clin Pract. 2008; and SHBG affect insulin resistance in midlife
epithelium-derived factor, a unique neurotrophic 79(2):185–190. women: the Study of Women’s Health Across the
serpin. Mol Vis. 1996;2:11. 176. Graham TE, Yang Q, Blüher M, Hammarstedt A, Nation (SWAN). Obesity (Silver Spring). 2013;21(5):
163. Yilmaz Y, Eren F, Ayyildiz T, Colak Y, Kurt R, Senates Ciaraldi TP, Henry RR, Wason CJ, Oberbach A, 1031–1038.
E, Tuncer I, Dolar E, Imeryuz N. Serum pigment Jansson PA, Smith U, Kahn BB. Retinol-binding 191. Osuna JA, Gómez-Pérez R, Arata-Bellabarba G,
epithelium-derived factor levels are increased in protein 4 and insulin resistance in lean, obese, Villaroel V. Relationship between BMI, total tes-
patients with biopsy-proven nonalcoholic fatty liver and diabetic subjects. N Engl J Med. 2006;354(24): tosterone, sex hormone-binding-globulin, leptin,
disease and independently associated with liver 2552–2563. insulin and insulin resistance in obese men. Arch
177. Lee JW, Lee HR, Shim JY, Im JA, Lee DC. Abdominal Androl. 2006;52(5):355–361.
steatosis. Clin Chim Acta. 2011;412(23–24):2296–
visceral fat reduction is associated with favorable 192. Niskanen L, Laaksonen DE, Punnonen K, Mustajoki
2299.
changes of serum retinol binding protein-4 in P, Kaukua J, Rissanen A. Changes in sex hormone-
164. Yamagishi S, Adachi H, Abe A, Yashiro T, Enomoto
nondiabetic subjects. Endocr J. 2008;55(5):811–818. binding globulin and testosterone during weight
M, Furuki K, Hino A, Jinnouchi Y, Takenaka K,
178. Haider DG, Schindler K, Prager G, Bohdjalian A, loss and weight maintenance in abdominally obese
Matsui T, Nakamura K, Imaizumi T. Elevated serum
Luger A, Wolzt M, Ludvik B. Serum retinol-binding men with the metabolic syndrome. Diabetes Obes
levels of pigment epithelium-derived factor in the
protein 4 is reduced after weight loss in morbidly Metab. 2004;6(3):208–215.
metabolic syndrome. J Clin Endocrinol Metab. 2006;
obese subjects. J Clin Endocrinol Metab. 2007;92(3): 193. Simó R, Barbosa-Desongles A, Sáez-Lopez C, Lecube
91(6):2447–2450.
1168–1171. A, Hernandez C, Selva DM. Molecular Mechanism
165. Yang M, Xu D, Liu Y, Guo X, Li W, Guo C, Zhang H,
179. Yang Q, Graham TE, Mody N, Preitner F, Peroni OD, of TNFa-induced down-regulation of SHBG ex-
Gao Y, Mao Y, Zhao J. Combined serum biomarkers
Zabolotny JM, Kotani K, Quadro L, Kahn BB. Serum pression. Mol Endocrinol. 2012;26(3):438–446.
in non-invasive diagnosis of non-alcoholic steato-
retinol binding protein 4 contributes to insulin 194. Plymate SR, Matej LA, Jones RE, Friedl KE. Inhibition
hepatitis. PLoS One. 2015;10(6):e0131664.
resistance in obesity and type 2 diabetes. Nature. of sex hormone-binding globulin production in the
166. Sabater M, Moreno-Navarrete JM, Ortega FJ, Pardo
2005;436(7049):356–362. human hepatoma (Hep G2) cell line by insulin and
G, Salvador J, Ricart W, Frühbeck G, Fernández-Real 180. Oo SM, Misu H, Saito Y, Tanaka M, Kato S, Kita Y, prolactin. J Clin Endocrinol Metab. 1988;67(3):
JM. Circulating pigment epithelium-derived factor Takayama H, Takeshita Y, Kanamori T, Nagano T, 460–464.
levels are associated with insulin resistance and Nakagen M, Urabe T, Matsuyama N, Kaneko S, 195. Saez-Lopez C, Brianso-Llort L, Torres-Torronteras J,
decrease after weight loss. J Clin Endocrinol Metab. Takamura T. Serum selenoprotein P, but not se- Simó R, Hammond GL, Selva DM. Resveratrol in-
2010;95(10):4720–4728. lenium, predicts future hyperglycemia in a general creases hepatic SHBG expression through human
167. Dai Z, Zhou T, Li C, Qi W, Mao Y, Lu J, Yao Y, Li L, Japanese population. Sci Rep. 2018;8(1):16727. constitutive androstane receptor: a new contribu-
Zhang T, Hong H, Li S, Cai W, Yang Z, Ma J, Yang X, 181. Chen M, Liu B, Wilkinson D, Hutchison AT, tion to the French paradox [published correction
Gao G. Intracellular pigment epithelium-derived Thompson CH, Wittert GA, Heilbronn LK. Sele- appears in Sci Rep. 2018;8(1):5046]. Sci Rep. 2017;
factor contributes to triglyceride degradation. Int noprotein P is elevated in individuals with obesity, 7(1):12284.
J Biochem Cell Biol. 2013;45(9):2076–2086. but is not independently associated with insulin 196. Saez-Lopez C, Barbosa-Desongles A, Hernandez C,
168. Lakeland TV, Borg ML, Matzaris M, Abdelkader A, resistance. Obes Res Clin Pract. 2017;11(2):227–232. Dyer RA, Innis SM, Simó R, Selva DM. Sex hormone-
Evans RG, Watt MJ. Augmented expression and 182. Yang SJ, Hwang SY, Choi HY, Yoo HJ, Seo JA, Kim SG, binding globulin reduction in metabolic disorders
secretion of adipose-derived pigment epithelium- Kim NH, Baik SH, Choi DS, Choi KM. Serum sele- may play a role in NAFLD development. Endocri-
derived factor does not alter local angiogenesis or noprotein P levels in patients with type 2 diabetes nology. 2017;158(3):545–559.
contribute to the development of systemic met- and prediabetes: implications for insulin resistance, 197. Wang Q, Sharma VP, Shen H, Xiao Y, Zhu Q, Xiong
abolic derangements. Am J Physiol Endocrinol inflammation, and atherosclerosis. J Clin Endocrinol X, Guo L, Jiang L, Ohta K, Li S, Shi H, Rui L, Lin JD. The
Metab. 2014;306(12):E1367–E1377. Metab. 2011;96(8):E1325–E1329. hepatokine Tsukushi gates energy expenditure via
169. Yoshida T, Akiba J, Matsui T, Nakamura K, 183. Choi HY, Hwang SY, Lee CH, Hong HC, Yang SJ, Yoo brown fat sympathetic innervation. Nat Metab.
Hisamoto T, Abe M, Ikezono Y, Wada F, Iwamoto H, HJ, Seo JA, Kim SG, Kim NH, Baik SH, Choi DS, Choi 2019;1:251–260.
Nakamura T, Koga H, Yamagishi SI, Torimura T. KM. Increased selenoprotein P levels in subjects 198. Conde-Vancells J, Rodriguez-Suarez E, Embade N, Gil
Pigment epithelium-derived factor (PEDF) prevents with visceral obesity and nonalcoholic fatty liver D, Matthiesen R, Valle M, Elortza F, Lu SC, Mato JM,
hepatic fat storage, inflammation, and fibrosis in disease. Diabetes Metab J. 2013;37(1):63–71. Falcon-Perez JM. Characterization and compre-
dietary steatohepatitis of mice. Dig Dis Sci. 2017; 184. Jung TW, Lee SY, Hong HC, Choi HY, Yoo HJ, Baik hensive proteome profiling of exosomes secreted
62(6):1527–1536. SH, Choi KM. AMPK activator-mediated inhibition by hepatocytes. J Proteome Res. 2008;7(12):5157–
170. Tsai TH, Shih SC, Ho TC, Ma HI, Liu MY, Chen SL, Tsao of endoplasmic reticulum stress ameliorates 5166.
YP. Pigment epithelium-derived factor 34-mer pep- carrageenan-induced insulin resistance through the 199. Hirsova P, Ibrahim SH, Krishnan A, Verma VK, Bronk
tide prevents liver fibrosis and hepatic stellate cell suppression of selenoprotein P in HepG2 hepato- SF, Werneburg NW, Charlton MR, Shah VH, Malhi
activation through down-regulation of the PDGF cytes. Mol Cell Endocrinol. 2014;382(1):66–73. H, Gores GJ. Lipid-induced signaling causes release
receptor [published correction appears in PLoS One. 185. Jänne M, Hammond GL. Hepatocyte nuclear factor- of inflammatory extracellular vesicles from hepa-
2014;9(9):e108835]. PLoS One. 2014;9(4):e95443. 4 controls transcription from a TATA-less human tocytes. Gastroenterology. 2016;150(4):956–967.

1388 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

200. Kakazu E, Mauer AS, Yin M, Malhi H. Hepatocytes b-hydroxybutyrylation. Mol Cell. 2016;62(2):194– Liver fat and lipid oxidation in humans. Liver Int.
release ceramide-enriched pro-inflammatory ex- 206. 2009;29(9):1439–1446.

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
tracellular vesicles in an IRE1a-dependent manner. 216. Tognini P, Murakami M, Liu Y, Eckel-Mahan KL, 231. Dahlhoff C, Worsch S, Sailer M, Hummel BA,
J Lipid Res. 2016;57(2):233–245. Newman JC, Verdin E, Baldi P, Sassone-Corsi P. Fiamoncini J, Uebel K, Obeid R, Scherling C, Geisel J,
201. Lee YS, Kim SY, Ko E, Lee JH, Yi HS, Yoo YJ, Je J, Suh SJ, Distinct circadian signatures in liver and gut clocks Bader BL, Daniel H. Methyl-donor supplementation
Jung YK, Kim JH, Seo YS, Yim HJ, Jeong WI, Yeon JE, revealed by ketogenic diet. Cell Metab. 2017;26(3): in obese mice prevents the progression of NAFLD,
Um SH, Byun KS. Exosomes derived from palmitic 523–538.e5. activates AMPK and decreases acyl-carnitine levels.
acid-treated hepatocytes induce fibrotic activation 217. Evans RM, Barish GD, Wang YX. PPARs and the Mol Metab. 2014;3(5):565–580.
of hepatic stellate cells. Sci Rep. 2017;7(1):3710. complex journey to obesity. Nat Med. 2004;10(4): 232. Wu JW, Wang SP, Alvarez F, Casavant S, Gauthier N,
202. Jones JG. Hepatic glucose and lipid metabolism. 355–361. Abed L, Soni KG, Yang G, Mitchell GA. Deficiency of
Diabetologia. 2016;59(6):1098–1103. 218. McGarry JD, Foster DW. The regulation of keto- liver adipose triglyceride lipase in mice causes
203. Gomes AR, Yong JS, Kiew KC, Aydin E, Khongkow genesis from octanoic acid. The role of the tri- progressive hepatic steatosis. Hepatology. 2011;
M, Laohasinnarong S, Lam EW. Sirtuin1 (SIRT1) in carboxylic acid cycle and fatty acid synthesis. J Biol 54(1):122–132.
the acetylation of downstream target proteins. Chem. 1971;246(4):1149–1159. 233. Newman JC, Covarrubias AJ, Zhao M, Yu X, Gut P,
Methods Mol Biol. 2016;1436:169–188. 219. Krebs HA, Hems R. Fatty acid metabolism in the Ng CP, Huang Y, Haldar S, Verdin E. Ketogenic diet
204. Menzies KJ, Zhang H, Katsyuba E, Auwerx J. Protein perfused rat liver. Biochem J. 1970;119(3):525–533. reduces midlife mortality and improves memory in
acetylation in metabolism—metabolites and co- 220. Maizels EZ, Ruderman NB, Goodman MN, Lau D. aging mice. Cell Metab. 2017;26(3):547–557.e8.
factors. Nat Rev Endocrinol. 2016;12(1):43–60. Effect of acetoacetate on glucose metabolism in the 234. Croci I, Byrne NM, Choquette S, Hills AP, Chachay VS,
205. Reichard GA Jr, Owen OE, Haff AC, Paul P, Bortz soleus and extensor digitorum longus muscles of Clouston AD, O’Moore-Sullivan TM, Macdonald GA,
WM. Ketone-body production and oxidation in the rat. Biochem J. 1977;162(3):557–568. Prins JB, Hickman IJ. Whole-body substrate meta-
fasting obese humans. J Clin Invest. 1974;53(2): 221. Russell RR III, Cline GW, Guthrie PH, Goodwin GW, bolism is associated with disease severity in patients
508–515. Shulman GI, Taegtmeyer H. Regulation of exoge- with non-alcoholic fatty liver disease. Gut. 2013;
206. Youm YH, Nguyen KY, Grant RW, Goldberg EL, nous and endogenous glucose metabolism by in- 62(11):1625–1633.
Bodogai M, Kim D, D’Agostino D, Planavsky N, sulin and acetoacetate in the isolated working rat 235. Boudaba N, Marion A, Huet C, Pierre R, Viollet B,
Lupfer C, Kanneganti TD, Kang S, Horvath TL, heart. A three tracer study of glycolysis, glycogen Foretz M. AMPK re-activation suppresses hepatic
Fahmy TM, Crawford PA, Biragyn A, Alnemri E, Dixit metabolism, and glucose oxidation. J Clin Invest. steatosis but its downregulation does not promote
VD. The ketone metabolite b-hydroxybutyrate 1997;100(11):2892–2899. fatty liver development. EBioMedicine. 2018;28:
blocks NLRP3 inflammasome-mediated inflamma- 222. Berger M, Hagg SA, Goodman MN, Ruderman NB. 194–209.
tory disease. Nat Med. 2015;21(3):263–269. Glucose metabolism in perfused skeletal muscle. 236. Chalasani N, Gorski JC, Asghar MS, Asghar A,
207. Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Effects of starvation, diabetes, fatty acids, acetoa- Foresman B, Hall SD, Crabb DW. Hepatic cyto-
Smith A, Murray AJ, Stubbs B, West J, McLure SW, cetate, insulin and exercise on glucose uptake and chrome P450 2E1 activity in nondiabetic patients
King MT, Dodd MS, Holloway C, Neubauer S, disposition. Biochem J. 1976;158(2):191–202. with nonalcoholic steatohepatitis. Hepatology. 2003;
Drawer S, Veech RL, Griffin JL, Clarke K. Nutritional 223. Puchalska P, Martin SE, Huang X, Lengfeld JE, 37(3):544–550.
ketosis alters fuel preference and thereby endurance Daniel B, Graham MJ, Han X, Nagy L, Patti GJ, 237. Dasarathy S, Yang Y, McCullough AJ, Marczewski S,
performance in athletes. Cell Metab. 2016;24(2): Crawford PA. Hepatocyte-macrophage acetoace- Bennett C, Kalhan SC. Elevated hepatic fatty acid
256–268. tate shuttle protects against tissue fibrosis. Cell oxidation, high plasma fibroblast growth factor 21,
208. Balasse EO, Fery F, Neef MA. Changes induced by Metab. 2019;29(2):383–398.e7. and fasting bile acids in nonalcoholic steatohepatitis.
exercise in rates of turnover and oxidation of ke- 224. Puchalska P, Crawford PA. Multi-dimensional roles Eur J Gastroenterol Hepatol. 2011;23(5):382–388.
tone bodies in fasting man. J Appl Physiol. 1978; of ketone bodies in fuel metabolism, signaling, and 238. Christie WW. Plasma lipoproteins. Available at:
44(1):5–11. therapeutics. Cell Metab. 2017;25(2):262–284. http://www.lipidhome.co.uk/lipids/simple/lipoprot/
209. Carneiro L, Geller S, Hébert A, Repond C, Fioramonti 225. Kinzig KP, Honors MA, Hargrave SL. Insulin sensi- index.htm. Accessed 13 January 2019.
X, Leloup C, Pellerin L. Hypothalamic sensing of tivity and glucose tolerance are altered by main- 239. Merrill AH Jr, Lingrell S, Wang E, Nikolova-
ketone bodies after prolonged cerebral exposure tenance on a ketogenic diet. Endocrinology. 2010; Karakashian M, Vales TR, Vance DE. Sphingolipid
leads to metabolic control dysregulation. Sci Rep. 151(7):3105–3114. biosynthesis de novo by rat hepatocytes in culture.
2016;6(1):34909. 226. Ellenbroek JH, van Dijck L, Töns HA, Rabelink TJ, Ceramide and sphingomyelin are associated with,
210. Park S, Kim DS, Daily JW. Central infusion of ketone Carlotti F, Ballieux BE, de Koning EJ. Long-term but not required for, very low density lipoprotein
bodies modulates body weight and hepatic insulin ketogenic diet causes glucose intolerance and re- secretion. J Biol Chem. 1995;270(23):13834–13841.
sensitivity by modifying hypothalamic leptin and duced b- and a-cell mass but no weight loss in 240. Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y,
insulin signaling pathways in type 2 diabetic rats. mice. Am J Physiol Endocrinol Metab. 2014;306(5): Bergeron R, Kim JK, Cushman SW, Cooney GJ,
Brain Res. 2011;1401:95–103. E552–E558. Atcheson B, White MF, Kraegen EW, Shulman GI.
211. Ahmed K. Biological roles and therapeutic potential 227. Garbow JR, Doherty JM, Schugar RC, Travers S, Mechanism by which fatty acids inhibit insulin
of hydroxy-carboxylic Acid receptors. Front Endo- Weber ML, Wentz AE, Ezenwajiaku N, Cotter DG, activation of insulin receptor substrate-1 (IRS-1)-
crinol (Lausanne). 2011;2:51. Brunt EM, Crawford PA. Hepatic steatosis, in- associated phosphatidylinositol 3-kinase activity in
212. Beylot M, Chassard D, Chambrier C, Guiraud M, flammation, and ER stress in mice maintained long muscle. J Biol Chem. 2002;277(52):50230–50236.
Odeon M, Beaufrère B, Bouletreau P. Metabolic term on a very low-carbohydrate ketogenic diet. 241. Roden M, Price TB, Perseghin G, Petersen KF,
effects of a D-b-hydroxybutyrate infusion in septic Am J Physiol Gastrointest Liver Physiol. 2011;300(6): Rothman DL, Cline GW, Shulman GI. Mechanism of
patients: inhibition of lipolysis and glucose pro- G956–G967. free fatty acid-induced insulin resistance in humans.
duction but not leucine oxidation. Crit Care Med. 228. Goday A, Bellido D, Sajoux I, Crujeiras AB, Burguera J Clin Invest. 1996;97(12):2859–2865.
1994;22(7):1091–1098. B, Garcı́a-Luna PP, Oleaga A, Moreno B, Casanueva 242. Choi SH, Ginsberg HN. Increased Very Low Density
213. Shimazu T, Hirschey MD, Newman J, He W, FF. Short-term safety, tolerability and efficacy of a Lipoprotein Secretion, Hepatic Steatosis, and Insulin
Shirakawa K, Le Moan N, Grueter CA, Lim H, very low-calorie-ketogenic diet interventional Resistance. Trends Endocrinol Metab. 2011;22(9):
Saunders LR, Stevens RD, Newgard CB, Farese RV Jr, weight loss program versus hypocaloric diet in 353–363.
de Cabo R, Ulrich S, Akassoglou K, Verdin E. Sup- patients with type 2 diabetes mellitus. Nutr Di- 243. Kurosaki Y, Tsukushi T, Munekata S, Kanoh Y,
pression of oxidative stress by b-hydroxybutyrate, abetes. 2016;6(9):e230. Moriya T, Nishinari M, Aoyama N, Ogawa Z. Is
an endogenous histone deacetylase inhibitor. Sci- 229. Männistö VT, Simonen M, Hyysalo J, Soininen P, there a relation between triglyceride concentrations
ence. 2013;339(6116):211–214. Kangas AJ, Kaminska D, Matte AK, Venesmaa S, in very low density lipoprotein and the index of
214. Ye J. Mechanisms of insulin resistance in obesity. Käkelä P, Kärjä V, Arola J, Gylling H, Cederberg H, insulin resistance in nondiabetic subjects? J Clin Lab
Front Med. 2013;7(1):14–24. Kuusisto J, Laakso M, Yki-Järvinen H, Ala-Korpela M, Anal. 2014;28(4):269–274.
215. Xie Z, Zhang D, Chung D, Tang Z, Huang H, Dai L, Pihlajamäki J. Ketone body production is differen- 244. Tonks KT, Coster AC, Christopher MJ, Chaudhuri R,
Qi S, Li J, Colak G, Chen Y, Xia C, Peng C, Ruan H, tially altered in steatosis and non-alcoholic stea- Xu A, Gagnon-Bartsch J, Chisholm DJ, James DE,
Kirkey M, Wang D, Jensen LM, Kwon OK, Lee S, tohepatitis in obese humans. Liver Int. 2015;35(7): Meikle PJ, Greenfield JR, Samocha-Bonet D. Skeletal
Pletcher SD, Tan M, Lombard DB, White KP, Zhao 1853–1861. muscle and plasma lipidomic signatures of insulin
H, Li J, Roeder RG, Yang X, Zhao Y. Metabolic 230. Kotronen A, Seppälä-Lindroos A, Vehkavaara S, resistance and overweight/obesity in humans.
regulation of gene expression by histone lysine Bergholm R, Frayn KN, Fielding BA, Yki-Järvinen H. Obesity (Silver Spring). 2016;24(4):908–916.

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1389


REVIEW

245. Hao M, Head WS, Gunawardana SC, Hasty AH, role of serine-palmitoyl transferase. Arch Biochem effects of dietary change and fasting on serum
Piston DW. Direct effect of cholesterol on insulin Biophys. 2003;419(2):120–128. triglycerides, unesterified carnitine, esterified (acyl)

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
secretion: a novel mechanism for pancreatic b-cell 261. Watt MJ, Barnett AC, Bruce CR, Schenk S, Horowitz carnitine, and b-hydroxybutyrate. Am J Clin Nutr.
dysfunction. Diabetes. 2007;56(9):2328–2338. JF, Hoy AJ. Regulation of plasma ceramide levels 1982;36(1):115–121.
246. Ambery AG, Tackett L, Penque BA, Brozinick JT, with fatty acid oversupply: evidence that the liver 275. Xu G, Hansen JS, Zhao XJ, Chen S, Hoene M, Wang
Elmendorf JS. Exercise training prevents skeletal detects and secretes de novo synthesised ceramide. XL, Clemmesen JO, Secher NH, Häring HU, Pedersen
muscle plasma membrane cholesterol accumulation, Diabetologia. 2012;55(10):2741–2746. BK, Lehmann R, Weigert C, Plomgaard P. Liver and
cortical actin filament loss, and insulin resistance in 262. Promrat K, Longato L, Wands JR, de la Monte SM. muscle contribute differently to the plasma acyl-
C57BL/6J mice fed a western-style high-fat diet. Weight loss amelioration of non-alcoholic steato- carnitine pool during fasting and exercise in
Physiol Rep. 2017;5(16):e13363. hepatitis linked to shifts in hepatic ceramide ex- humans. J Clin Endocrinol Metab. 2016;101(12):
247. Barrientos G, Sánchez-Aguilera P, Jaimovich E, pression and serum ceramide levels. Hepatol Res. 5044–5052.
Hidalgo C, Llanos P. Membrane cholesterol in 2011;41(8):754–762. 276. Soeters MR, Sauerwein HP, Duran M, Wanders RJ,
skeletal muscle: a novel player in excitation- 263. Bikman BT, Guan Y, Shui G, Siddique MM, Holland Ackermans MT, Fliers E, Houten SM, Serlie MJ.
contraction coupling and insulin resistance. WL, Kim JY, Fabriàs G, Wenk MR, Summers SA. Muscle acylcarnitines during short-term fasting in
J Diabetes Res. 2017;2017:3941898. Fenretinide prevents lipid-induced insulin re- lean healthy men. Clin Sci (Lond). 2009;116(7):
248. Arnold DR, Kwiterovich PO. Cholesterol: absorp- sistance by blocking ceramide biosynthesis. J Biol 585–592.
tion, function, and metabolism. In: Caballero B, ed. Chem. 2012;287(21):17426–17437. 277. Eaton S, Bartlett K, Pourfarzam M. Mammalian
Encyclopedia of Food Sciences and Nutrition. 2nd ed. 264. Bruce CR, Risis S, Babb JR, Yang C, Kowalski GM, mitochondrial beta-oxidation. Biochem J. 1996;
Oxford, United Kingdom: Academic Press; 2003: Selathurai A, Lee-Young RS, Weir JM, Yoshioka K, 320(Pt 2):345–357.
1226–1237. Takuwa Y, Meikle PJ, Pitson SM, Febbraio MA. 278. Bartlett K, Bhuiyan AK, Aynsley-Green A, Butler PC,
249. Chaurasia B, Summers SA. Ceramides—lipotoxic Overexpression of sphingosine kinase 1 prevents Alberti KG. Human forearm arteriovenous differ-
inducers of metabolic disorders [published cor- ceramide accumulation and ameliorates muscle ences of carnitine, short-chain acylcarnitine and
rection appears in Trends Endocrinol Metab. 2018; insulin resistance in high-fat diet-fed mice. Diabetes. long-chain acylcarnitine. Clin Sci (Lond). 1989;77(4):
29(1):66–67]. Trends Endocrinol Metab. 2015;26(10): 2012;61(12):3148–3155. 413–416.
538–550. 265. Chavez JA, Siddique MM, Wang ST, Ching J, 279. Simcox J, Geoghegan G, Maschek JA, Bensard CL,
250. Boon J, Hoy AJ, Stark R, Brown RD, Meex RC, Shayman JA, Summers SA. Ceramides and gluco- Pasquali M, Miao R, Lee S, Jiang L, Huck I, Kershaw
Henstridge DC, Schenk S, Meikle PJ, Horowitz JF, sylceramides are independent antagonists of insulin EE, Donato AJ, Apte U, Longo N, Rutter J, Schreiber
Kingwell BA, Bruce CR, Watt MJ. Ceramides con- signaling. J Biol Chem. 2014;289(2):723–734. R, Zechner R, Cox J, Villanueva CJ. Global analysis of
tained in LDL are elevated in type 2 diabetes and 266. Huang H, Kasumov T, Gatmaitan P, Heneghan HM, plasma lipids identifies liver-derived acylcarnitines
promote inflammation and skeletal muscle insulin Kashyap SR, Schauer PR, Brethauer SA, Kirwan JP. as a fuel source for brown fat thermogenesis. Cell
resistance. Diabetes. 2013;62(2):401–410. Gastric bypass surgery reduces plasma ceramide Metab. 2017;26(3):509–522.e6.
251. Osborn O, Olefsky JM. The cellular and signaling subspecies and improves insulin sensitivity in se- 280. Aguer C, McCoin CS, Knotts TA, Thrush AB, Ono-
networks linking the immune system and meta- verely obese patients. Obesity (Silver Spring). 2011; Moore K, McPherson R, Dent R, Hwang DH, Adams
bolism in disease. Nat Med. 2012;18(3):363–374. 19(11):2235–2240. SH, Harper ME. Acylcarnitines: potential implica-
252. Shin KC, Hwang I, Choe SS, Park J, Ji Y, Kim JI, Lee GY, 267. Tulipani S, Griffin J, Palau-Rodriguez M, Mora- tions for skeletal muscle insulin resistance. FASEB J.
Choi SH, Ching J, Kovalik JP, Kim JB. Macrophage Cubillos X, Bernal-Lopez RM, Tinahones FJ, 2015;29(1):336–345.
VLDLR mediates obesity-induced insulin resistance Corkey BE, Andres-Lacueva C. Metabolomics- 281. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale
with adipose tissue inflammation. Nat Commun. guided insights on bariatric surgery versus behav- M, Ilkayeva O, Bain J, Stevens R, Dyck JR, Newgard
2017;8(1):1087. ioral interventions for weight loss. Obesity (Silver CB, Lopaschuk GD, Muoio DM. Mitochondrial
253. Fabbrini E, Magkos F. Hepatic steatosis as a marker Spring). 2016;24(12):2451–2466. overload and incomplete fatty acid oxidation
of metabolic dysfunction. Nutrients. 2015;7(6): 268. Xia JY, Holland WL, Kusminski CM, Sun K, Sharma contribute to skeletal muscle insulin resistance. Cell
4995–5019. AX, Pearson MJ, Sifuentes AJ, McDonald JG, Gordillo Metab. 2008;7(1):45–56.
254. Poulsen MK, Nellemann B, Stødkilde-Jørgensen H, R, Scherer PE. Targeted induction of ceramide 282. Hoppel CL, Genuth SM. Carnitine metabolism in
Pedersen SB, Grønbæk H, Nielsen S. Impaired insulin degradation leads to improved systemic metab- normal-weight and obese human subjects during
suppression of VLDL-triglyceride kinetics in non- olism and reduced hepatic steatosis. Cell Metab. fasting. Am J Physiol. 1980;238(5):E409–E415.
alcoholic fatty liver disease. J Clin Endocrinol Metab. 2015;22(2):266–278. 283. Mihalik SJ, Goodpaster BH, Kelley DE, Chace DH,
2016;101(4):1637–1646. 269. Chavez JA, Holland WL, Bär J, Sandhoff K, Summers Vockley J, Toledo FGS, DeLany JP. Increased levels of
255. Gaggini M, Morelli M, Buzzigoli E, DeFronzo RA, SA. Acid ceramidase overexpression prevents the plasma acylcarnitines in obesity and type 2 diabetes
Bugianesi E, Gastaldelli A. Non-alcoholic fatty liver inhibitory effects of saturated fatty acids on insulin and identification of a marker of glucolipotoxicity.
disease (NAFLD) and its connection with insulin signaling. J Biol Chem. 2005;280(20):20148–20153. Obesity (Silver Spring). 2010;18(9):1695–1700.
resistance, dyslipidemia, atherosclerosis and coro- 270. Eaton S, Bartlett K, Pourfarzam M, Markley MA, 284. Schooneman MG, Houtkooper RH, Hollak CEM,
nary heart disease. Nutrients. 2013;5(5):1544–1560. New KJ, Quant PA. Production and export of Wanders RJA, Vaz FM, Soeters MR, Houten SM. The
256. Nielsen S, Karpe F. Determinants of VLDL- acylcarnitine esters by neonatal rat hepatocytes. In: impact of altered carnitine availability on acylcar-
triglycerides production. Curr Opin Lipidol. 2012; Quant PA, Eaton S, eds. Current Views of Fatty Acid nitine metabolism, energy expenditure and glucose
23(4):321–326. Oxidation and Ketogenesis: From Organelles to Point tolerance in diet-induced obese mice. Biochim
257. Song Y, Zhao M, Cheng X, Shen J, Khound R, Zhang Mutations. Boston, MA: Springer, 2002:155–159. Biophys Acta. 2016;1862(8):1375–1382.
K, Su Q. CREBH mediates metabolic inflammation 271. Kim HI, Raffler J, Lu W, Lee JJ, Abbey D, Saleheen D, 285. Cahova M, Chrastina P, Hansikova H, Drahota Z,
to hepatic VLDL overproduction and hyper- Rabinowitz JD, Bennett MJ, Hand NJ, Brown C, Rader Trnovska J, Skop V, Spacilova J, Malinska H,
lipoproteinemia. J Mol Med (Berl). 2017;95(8): DJ. Fine mapping and functional analysis reveal a Oliyarnyk O, Papackova Z, Palenickova E, Kazdova L.
839–849. role of SLC22A1 in acylcarnitine transport. Am J Carnitine supplementation alleviates lipid metab-
258. Qin B, Anderson RA, Adeli K. Tumor necrosis factor- Hum Genet. 2017;101(4):489–502. olism derangements and protects against oxidative
a directly stimulates the overproduction of hepatic 272. Brzozowski JS, Jankowski H, Bond DR, McCague SB, stress in non-obese hereditary hypertriglyceridemic
apolipoprotein B100-containing VLDL via impair- Munro BR, Predebon MJ, Scarlett CJ, Skelding KA, rats. Appl Physiol Nutr Metab. 2015;40(3):280–291.
ment of hepatic insulin signaling. Am J Physiol Weidenhofer J. Lipidomic profiling of extracellular 286. Aichler M, Borgmann D, Krumsiek J, Buck A,
Gastrointest Liver Physiol. 2008;294(5):G1120– vesicles derived from prostate and prostate cancer MacDonald PE, Fox JE, Lyon J, Light PE, Keipert S,
G1129. cell lines. Lipids Health Dis. 2018;17(1):211. Jastroch M, Feuchtinger A, Mueller NS, Sun N,
259. Akash MS, Rehman K, Liaqat A. Tumor necrosis 273. Haraszti RA, Didiot MC, Sapp E, Leszyk J, Shaffer SA, Palmer A, Alexandrov T, Hrabe de Angelis M,
factor-alpha: role in development of insulin resis- Rockwell HE, Gao F, Narain NR, DiFiglia M, Kiebish Neschen S, Tschöp MH, Walch A. N-acyl taurines
tance and pathogenesis of type 2 diabetes mellitus. MA, Aronin N, Khvorova A. High-resolution pro- and acylcarnitines cause an imbalance in insulin
J Cell Biochem. 2018;119(1):105–110. teomic and lipidomic analysis of exosomes and synthesis and secretion provoking b cell dysfunc-
260. Lightle S, Tosheva R, Lee A, Queen-Baker J, microvesicles from different cell sources. J Extracell tion in type 2 diabetes. Cell Metab. 2017;25(6):
Boyanovsky B, Shedlofsky S, Nikolova-Karakashian Vesicles. 2016;5(1):32570. 1334–1347.e4.
M. Elevation of ceramide in serum lipoproteins 274. Bell FP, DeLucia A, Bryant LR, Patt CS, Greenberg HS. 287. Rutkowsky JM, Knotts TA, Ono-Moore KD, McCoin
during acute phase response in humans and mice: Carnitine metabolism in Macaca arctoides: the CS, Huang S, Schneider D, Singh S, Adams SH, Hwang

1390 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

DH. Acylcarnitines activate proinflammatory signal- repress small heterodimer partner (SHP) activation 318. Soga T, Ohishi T, Matsui T, Saito T, Matsumoto M,
ing pathways. Am J Physiol Endocrinol Metab. 2014; and adiponectin counteracts bile acid-induced liver Takasaki J, Matsumoto S, Kamohara M, Hiyama H,

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
306(12):E1378–E1387. injury in superobese patients with nonalcoholic Yoshida S, Momose K, Ueda Y, Matsushime H,
288. Gonzalez-Granda A, Damms-Machado A, Basrai M, steatohepatitis. Hepatology. 2013;57(4):1394–1406. Kobori M, Furuichi K. Lysophosphatidylcholine
Bischoff SC. Changes in plasma acylcarnitine and 304. Jahnel J, Zöhrer E, Alisi A, Ferrari F, Ceccarelli S, De enhances glucose-dependent insulin secretion via
lysophosphatidylcholine levels following a high- Vito R, Scharnagl H, Stojakovic T, Fauler G, Trauner an orphan G-protein-coupled receptor. Biochem
fructose diet: a targeted metabolomics study in M, Nobili V. Serum bile acid levels in children with Biophys Res Commun. 2005;326(4):744–751.
healthy women. Nutrients. 2018;10(9):1254. nonalcoholic fatty liver disease. J Pediatr Gastro- 319. Takahashi H, Goto T, Yamazaki Y, Kamakari K,
289. Kalhan SC, Guo L, Edmison J, Dasarathy S, enterol Nutr. 2015;61(1):85–90. Hirata M, Suzuki H, Shibata D, Nakata R, Inoue H,
McCullough AJ, Hanson RW, Milburn M. Plasma 305. Caussy C, Hsu C, Singh S, Bassirian S, Kolar J, Faulkner Takahashi N, Kawada T. Metabolomics reveal 1-
metabolomic profile in nonalcoholic fatty liver C, Sinha N, Bettencourt R, Gara N, Valasek MA, palmitoyl lysophosphatidylcholine production by
disease. Metabolism. 2011;60(3):404–413. Schnabl B, Richards L, Brenner DA, Hofmann AF, peroxisome proliferator-activated receptor a. J Lipid
290. Peng KY, Watt MJ, Rensen S, Greve JW, Huynh K, Loomba R. Serum bile acid patterns are associated Res. 2015;56(2):254–265.
Jayawardana KS, Meikle PJ, Meex RCR. Mitochon- with the presence of NAFLD in twins, and dose- 320. Yea K, Kim J, Yoon JH, Kwon T, Kim JH, Lee BD, Lee
drial dysfunction-related lipid changes occur in dependent changes with increase in fibrosis stage in HJ, Lee SJ, Kim JI, Lee TG, Baek MC, Park HS, Park KS,
nonalcoholic fatty liver disease progression. J Lipid patients with biopsy-proven NAFLD. Aliment Ohba M, Suh PG, Ryu SH. Lysophosphatidylcholine
Res. 2018;59(10):1977–1986. Pharmacol Ther. 2019;49(2):183–193. activates adipocyte glucose uptake and lowers
291. Fujiwara N, Nakagawa H, Enooku K, Kudo Y, Hayata 306. Lu LP, Wan YP, Xun PC, Zhou KJ, Chen C, Cheng SY, blood glucose levels in murine models of diabetes.
Y, Nakatsuka T, Tanaka Y, Tateishi R, Hikiba Y, Zhang MZ, Wu CH, Lin WW, Jiang Y, Feng HX, Wang J Biol Chem. 2009;284(49):33833–33840.
Misumi K, Tanaka M, Hayashi A, Shibahara J, JL, He K, Cai W. Serum bile acid level and fatty acid 321. Zhao H, Yang L, Baddour J, Achreja A, Bernard V,
Fukayama M, Arita J, Hasegawa K, Hirschfield H, composition in Chinese children with non-alcoholic Moss T, Marini JC, Tudawe T, Seviour EG, San Lucas
Hoshida Y, Hirata Y, Otsuka M, Tateishi K, Koike K. fatty liver disease. J Dig Dis. 2017;18(8):461–471. FA, Alvarez H, Gupta S, Maiti SN, Cooper L, Peehl D,
CPT2 downregulation adapts HCC to lipid-rich 307. Puri P, Daita K, Joyce A, Mirshahi F, Santhekadur PK, Ram PT, Maitra A, Nagrath D. Tumor microenvi-
environment and promotes carcinogenesis via Cazanave S, Luketic VA, Siddiqui MS, Boyett S, Min ronment derived exosomes pleiotropically modu-
acylcarnitine accumulation in obesity. Gut. 2018; HK, Kumar DP, Kohli R, Zhou H, Hylemon PB, late cancer cell metabolism. eLife. 2016;5:e10250.
67(8):1493–1504. Contos MJ, Idowu M, Sanyal AJ. The presence and 322. Achreja A, Zhao H, Yang L, Yun TH, Marini J,
292. Morris EM, Meers GM, Koch LG, Britton SL, Fletcher severity of nonalcoholic steatohepatitis is associ- Nagrath D. Exo-MFA—a 13C metabolic flux analysis
JA, Fu X, Shankar K, Burgess SC, Ibdah JA, Rector RS, ated with specific changes in circulating bile acids. framework to dissect tumor microenvironment-
Thyfault JP. Aerobic capacity and hepatic mito- Hepatology. 2018;67(2):534–548. secreted exosome contributions towards cancer
chondrial lipid oxidation alters susceptibility for cell metabolism. Metab Eng. 2017;43(Pt B):156–172.
308. Legry V, Francque S, Haas JT, Verrijken A, Caron S,
chronic high-fat diet-induced hepatic steatosis. Am 323. Mayr M, Grainger D, Mayr U, Leroyer AS, Leseche G,
Chávez-Talavera O, Vallez E, Vonghia L, Dirinck E,
J Physiol Endocrinol Metab. 2016;311(4):E749–E760. Sidibe A, Herbin O, Yin X, Gomes A, Madhu B,
Verhaegen A, Kouach M, Lestavel S, Lefebvre P, Van
293. Geisler CE, Renquist BJ. Hepatic lipid accumulation: Griffiths JR, Xu Q, Tedgui A, Boulanger CM. Pro-
Gaal L, Tailleux A, Paumelle R, Staels B. Bile acid
cause and consequence of dysregulated glucor- teomics, metabolomics, and immunomics on mi-
alterations are associated with insulin resistance,
egulatory hormones. J Endocrinol. 2017;234(1):R1–R21. croparticles derived from human atherosclerotic
but not with NASH, in obese subjects. J Clin
294. Fucho R, Casals N, Serra D, Herrero L. Ceramides and plaques. Circ Cardiovasc Genet. 2009;2(4):379–388.
Endocrinol Metab. 2017;102(10):3783–3794.
mitochondrial fatty acid oxidation in obesity. FASEB 324. Luo X, An M, Cuneo KC, Lubman DM, Li L. High-
309. Maseki M, Nishigaki I, Hagihara M, Tomoda Y, Yagi
J. 2017;31(4):1263–1272. performance chemical isotope labeling liquid
K. Lipid peroxide levels and lipids content of serum
295. Patterson RE, Kalavalapalli S, Williams CM, Nautiyal chromatography mass spectrometry for exosome
lipoprotein fractions of pregnant subjects with or
M, Mathew JT, Martinez J, Reinhard MK, McDougall metabolomics. Anal Chem. 2018;90(14):8314–8319.
without pre-eclampsia. Clin Chim Acta. 1981;115(2):
DJ, Rocca JR, Yost RA, Cusi K, Garrett TJ, Sunny NE. 325. Povero D, Eguchi A, Li H, Johnson CD, Papouchado
Lipotoxicity in steatohepatitis occurs despite an 155–161. BG, Wree A, Messer K, Feldstein AE. Circulating
increase in tricarboxylic acid cycle activity. Am J 310. Antimisiaris SG, Mourtas S, Marazioti A. Exosomes extracellular vesicles with specific proteome and
Physiol Endocrinol Metab. 2016;310(7):E484–E494. and exosome-inspired vesicles for targeted drug liver microRNAs are potential biomarkers for liver
296. de Aguiar Vallim TQ, Tarling EJ, Edwards PA. delivery. Pharmaceutics. 2018;10(4):218. injury in experimental fatty liver disease. PLoS One.
Pleiotropic roles of bile acids in metabolism. Cell 311. Skotland T, Sandvig K, Llorente A. Lipids in exo- 2014;9(12):e113651.
Metab. 2013;17(5):657–669. somes: current knowledge and the way forward. 326. Eguchi A, Feldstein AE. Extracellular vesicles in non-
297. Zhou H, Hylemon PB. Bile acids are nutrient sig- Prog Lipid Res. 2017;66:30–41. alcoholic and alcoholic fatty liver diseases. Liver Res.
naling hormones. Steroids. 2014;86:62–68. 312. Dang VD, Jella KK, Ragheb RRT, Denslow ND, Alli 2018;2(1):30–34.
298. Ma K, Saha PK, Chan L, Moore DD. Farnesoid X AA. Lipidomic and proteomic analysis of exosomes 327. Fukushima M, Dasgupta D, Mauer AS, Kakazu E,
receptor is essential for normal glucose homeo- from mouse cortical collecting duct cells. FASEB J. Nakao K, Malhi H. StAR-related lipid transfer do-
stasis. J Clin Invest. 2006;116(4):1102–1109. 2017;31(12):5399–5408. main 11 (STARD11)-mediated ceramide transport
299. Zhang Y, Lee FY, Barrera G, Lee H, Vales C, Gonzalez 313. Llorente A, Skotland T, Sylvänne T, Kauhanen D, mediates extracellular vesicle biogenesis. J Biol Chem.
FJ, Willson TM, Edwards PA. Activation of the Róg T, Orłowski A, Vattulainen I, Ekroos K, Sandvig 2018;293(39):15277–15289.
nuclear receptor FXR improves hyperglycemia and K. Molecular lipidomics of exosomes released by 328. Cheng Q, Li X, Wang Y, Dong M, Zhan FH, Liu J. The
hyperlipidemia in diabetic mice. Proc Natl Acad Sci PC-3 prostate cancer cells. Biochim Biophys Acta. ceramide pathway is involved in the survival, ap-
USA. 2006;103(4):1006–1011. 2013;1831(7):1302–1309. optosis and exosome functions of human multiple
300. Hakkola J, Rysä J, Hukkanen J. Regulation of hepatic 314. Janas AM, Sapoń K, Janas T, Stowell MHB, Janas T. myeloma cells in vitro. Acta Pharmacol Sin. 2018;
energy metabolism by the nuclear receptor PXR. Exosomes and other extracellular vesicles in neural 39(4):561–568.
Biochim Biophys Acta. 2016;1859(9):1072–1082. cells and neurodegenerative diseases. Biochim Bio- 329. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel
301. Jiao N, Baker SS, Chapa-Rodriguez A, Liu W, Nugent phys Acta. 2016;1858(6):1139–1151. D, Wieland F, Schwille P, Brügger B, Simons M.
CA, Tsompana M, Mastrandrea L, Buck MJ, Baker 315. Nojima H, Freeman CM, Schuster RM, Japtok L, Ceramide triggers budding of exosome vesicles into
RD, Genco RJ, Zhu R, Zhu L. Suppressed hepatic bile Kleuser B, Edwards MJ, Gulbins E, Lentsch AB. multivesicular endosomes. Science. 2008;319(5867):
acid signalling despite elevated production of pri- Hepatocyte exosomes mediate liver repair and 1244–1247.
mary and secondary bile acids in NAFLD. Gut. 2018; regeneration via sphingosine-1-phosphate. J Hep- 330. Wang G, Dinkins M, He Q, Zhu G, Poirier C,
67(10):1881–1891. atol. 2016;64(1):60–68. Campbell A, Mayer-Proschel M, Bieberich E. As-
302. Ferslew BC, Xie G, Johnston CK, Su M, Stewart PW, 316. Sekas G, Patton GM, Lincoln EC, Robins SJ. Origin of trocytes secrete exosomes enriched with proapo-
Jia W, Brouwer KL, Barritt AS IV. Altered bile acid plasma lysophosphatidylcholine: evidence for direct ptotic ceramide and prostate apoptosis response 4
metabolome in patients with nonalcoholic stea- hepatic secretion in the rat. J Lab Clin Med. 1985; (PAR-4): potential mechanism of apoptosis in-
tohepatitis. Dig Dis Sci. 2015;60(11):3318–3328. 105(2):190–194. duction in Alzheimer disease (AD). J Biol Chem.
303. Bechmann LP, Kocabayoglu P, Sowa JP, Sydor S, Best 317. Brindley DN. Hepatic secretion of lysophosphati- 2012;287(25):21384–21395.
J, Schlattjan M, Beilfuss A, Schmitt J, Hannivoort RA, dylcholine: a novel transport system for poly- 331. Chen L, Chen R, Kemper S, Brigstock DR. Pathways
Kilicarslan A, Rust C, Berr F, Tschopp O, Gerken G, unsaturated fatty acids and choline. J Nutr Biochem. of production and delivery of hepatocyte exosomes.
Friedman SL, Geier A, Canbay A. Free fatty acids 1993;4(8):442–449. J Cell Commun Signal. 2018;12(1):343–357.

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1391


REVIEW

332. Povero D, Panera N, Eguchi A, Johnson CD, 345. Tsui NB, Ng EK, Lo YM. Stability of endogenous and 362. Jampoka K, Muangpaisarn P, Khongnomnan K,
Papouchado BG, de Araujo Horcel L, Pinatel EM, added RNA in blood specimens, serum, and plasma. Treeprasertsuk S, Tangkijvanich P, Payungporn S.

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
Alisi A, Nobili V, Feldstein AE. Lipid-induced Clin Chem. 2002;48(10):1647–1653. Serum miR-29a and miR-122 as potential bio-
hepatocyte-derived extracellular vesicles regulate 346. Sung S, Kim J, Jung Y. Liver-derived exosomes and markers for non-alcoholic fatty liver disease
hepatic stellate cells via microRNA targeting per- their implications in liver pathobiology. Int J Mol Sci. (NAFLD). MicroRNA. 2018;7(3):215–222.
oxisome proliferator-activated receptor-g. Cell Mol 2018;19(12):E3715. 363. Nie J, Li CP, Li JH, Chen X, Zhong X. Analysis of non-
Gastroenterol Hepatol. 2015;1(6):646–663.e4. 347. Vickers KC, Palmisano BT, Shoucri BM, Shamburek alcoholic fatty liver disease microRNA expression
333. Seo W, Eun HS, Kim SY, Yi HS, Lee YS, Park SH, Jang RD, Remaley AT. MicroRNAs are transported in spectra in rat liver tissues. Mol Med Rep. 2018;18(3):
MJ, Jo E, Kim SC, Han YM, Park KG, Jeong WI. plasma and delivered to recipient cells by high- 2669–2680.
Exosome-mediated activation of Toll-like receptor 3 density lipoproteins. Nat Cell Biol. 2011;13(4):423–433. 364. Cermelli S, Ruggieri A, Marrero JA, Ioannou GN,
in stellate cells stimulates interleukin-17 production 348. Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard Beretta L. Circulating microRNAs in patients with
by gd T cells in liver fibrosis. Hepatology. 2016;64(2): CC, Gibson DF, Mitchell PS, Bennett CF, Pogosova- chronic hepatitis C and non-alcoholic fatty liver
616–631. Agadjanyan EL, Stirewalt DL, Tait JF, Tewari M. disease. PLoS One. 2011;6(8):e23937.
334. Apostolopoulou M, Gordillo R, Koliaki C, Gancheva Argonaute2 complexes carry a population of cir- 365. Xu Q, Li Y, Shang YF, Wang HL, Yao MX. miRNA-
S, Jelenik T, De Filippo E, Herder C, Markgraf D, culating microRNAs independent of vesicles in 103: molecular link between insulin resistance and
Jankowiak F, Esposito I, Schlensak M, Scherer PE, human plasma. Proc Natl Acad Sci USA. 2011; nonalcoholic fatty liver disease. World J Gastro-
Roden M. Specific hepatic sphingolipids relate to 108(12):5003–5008. enterol. 2015;21(2):511–516.
insulin resistance, oxidative stress, and in- 349. Kosaka N, Iguchi H, Yoshioka Y, Takeshita F, 366. Soronen J, Yki-Järvinen H, Zhou Y, Sädevirta S, Sarin
flammation in nonalcoholic steatohepatitis. Di- Matsuki Y, Ochiya T. Secretory mechanisms and AP, Leivonen M, Sevastianova K, Perttilä J, Laurila PP,
abetes Care. 2018;41(6):1235–1243. intercellular transfer of microRNAs in living cells. Sigruener A, Schmitz G, Olkkonen VM. Novel hepatic
335. Pagadala M, Kasumov T, McCullough AJ, Zein NN, J Biol Chem. 2010;285(23):17442–17452. microRNAs upregulated in human nonalcoholic fatty
Kirwan JP. Role of ceramides in nonalcoholic fatty 350. Frost RJ, Olson EN. Control of glucose homeostasis and liver disease. Physiol Rep. 2016;4(1):e12661.
liver disease. Trends Endocrinol Metab. 2012;23(8): insulin sensitivity by the Let-7 family of microRNAs. Proc 367. Kornfeld JW, Baitzel C, Könner AC, Nicholls HT,
365–371. Natl Acad Sci USA. 2011;108(52):21075–21080. Vogt MC, Herrmanns K, Scheja L, Haumaitre C,
336. Feldman A, Eder SK, Felder TK, Kedenko L, 351. Croce CM, Calin GA. miRNAs, cancer, and stem cell Wolf AM, Knippschild U, Seibler J, Cereghini S,
Paulweber B, Stadlmayr A, Huber-Schönauer U, division. Cell. 2005;122(1):6–7. Heeren J, Stoffel M, Brüning JC. Obesity-induced
Niederseer D, Stickel F, Auer S, Haschke-Becher E, 352. He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, overexpression of miR-802 impairs glucose metab-
Patsch W, Datz C, Aigner E. Clinical and metabolic Xue W, Zender L, Magnus J, Ridzon D, Jackson AL, olism through silencing of Hnf1b. Nature. 2013;
characterization of lean caucasian subjects with Linsley PS, Chen C, Lowe SW, Cleary MA, Hannon GJ. 494(7435):111–115.
non-alcoholic fatty liver. Am J Gastroenterol. 2017; A microRNA component of the p53 tumour sup- 368. Karolina DS, Armugam A, Tavintharan S, Wong MT,
112(1):102–110. pressor network. Nature. 2007;447(7148):1130–1134. Lim SC, Sum CF, Jeyaseelan K. MicroRNA 144 im-
337. Orešič M, Hyötyläinen T, Kotronen A, Gopalacharyulu 353. Cheung O, Puri P, Eicken C, Contos MJ, Mirshahi F, pairs insulin signaling by inhibiting the expression of
P, Nygren H, Arola J, Castillo S, Mattila I, Hakkarainen Maher JW, Kellum JM, Min H, Luketic VA, Sanyal AJ. insulin receptor substrate 1 in type 2 diabetes
A, Borra RJ, Honka MJ, Verrijken A, Francque S, Iozzo Nonalcoholic steatohepatitis is associated with al- mellitus [published correction appears in PLoS One.
P, Leivonen M, Jaser N, Juuti A, Sørensen TIA, tered hepatic microRNA expression. Hepatology. 2011;6(9): doi: 10.1371/annotation/698b7123-174f-
Nuutila P, Van Gaal L, Yki-Järvinen H. Prediction of 2008;48(6):1810–1820. 4a09-95c9-fd6f5017d622]. PLoS One. 2011;6(8):
non-alcoholic fatty-liver disease and liver fat con- 354. Xie H, Lim B, Lodish HF. MicroRNAs induced during e22839.
tent by serum molecular lipids. Diabetologia. 2013; adipogenesis that accelerate fat cell development 369. Poy MN, Eliasson L, Krutzfeldt J, Kuwajima S, Ma X,
56(10):2266–2274. are downregulated in obesity. Diabetes. 2009;58(5): Macdonald PE, Pfeffer S, Tuschl T, Rajewsky N,
338. Tanaka N, Matsubara T, Krausz KW, Patterson AD, 1050–1057. Rorsman P, Stoffel M. A pancreatic islet-specific
Gonzalez FJ. Disruption of phospholipid and bile 355. Lovis P, Roggli E, Laybutt DR, Gattesco S, Yang JY, microRNA regulates insulin secretion. Nature. 2004;
acid homeostasis in mice with nonalcoholic stea- Widmann C, Abderrahmani A, Regazzi R. Alter- 432(7014):226–230.
tohepatitis. Hepatology. 2012;56(1):118–129. ations in microRNA expression contribute to fatty 370. Plaisance V, Abderrahmani A, Perret-Menoud V,
339. Zhou Y, Orešič M, Leivonen M, Gopalacharyulu P, acid-induced pancreatic b-cell dysfunction. Di- Jacquemin P, Lemaigre F, Regazzi R. MicroRNA-9
Hyysalo J, Arola J, Verrijken A, Francque S, Van Gaal L, abetes. 2008;57(10):2728–2736. controls the expression of Granuphilin/Slp4 and the
Hyötyläinen T, Yki-Järvinen H. Noninvasive detection 356. Rottiers V, Näär AM. MicroRNAs in metabolism and secretory response of insulin-producing cells. J Biol
of nonalcoholic steatohepatitis using clinical markers metabolic disorders [published correction appears in Chem. 2006;281(37):26932–26942.
and circulating levels of lipids and metabolites. Clin Nat Rev Mol Cell Biol. 2012;13(5): doi:10.1038/ 371. Jordan SD, Krüger M, Willmes DM, Redemann N,
Gastroenterol Hepatol. 2016;14(10):1463–1472.e6. nrm3328]. Nat Rev Mol Cell Biol. 2012;13(4):239–250. Wunderlich FT, Brönneke HS, Merkwirth C, Kashkar
340. Lehmann R, Franken H, Dammeier S, Rosenbaum L, 357. Ding J, Li M, Wan X, Jin X, Chen S, Yu C, Li Y. Effect of H, Olkkonen VM, Böttger T, Braun T, Seibler J,
Kantartzis K, Peter A, Zell A, Adam P, Li J, Xu G, miR-34a in regulating steatosis by targeting PPARa Brüning JC. Obesity-induced overexpression of
Königsrainer A, Machann J, Schick F, Hrabé de expression in nonalcoholic fatty liver disease. Sci miRNA-143 inhibits insulin-stimulated AKT acti-
Angelis M, Schwab M, Staiger H, Schleicher E, Rep. 2015;5(1):13729. vation and impairs glucose metabolism. Nat Cell
Gastaldelli A, Fritsche A, Häring HU, Stefan N. 358. Liu XL, Pan Q, Zhang RN, Shen F, Yan SY, Sun C, Xu Biol. 2011;13(4):434–446.
Circulating lysophosphatidylcholines are markers ZJ, Chen YW, Fan JG. Disease-specific miR-34a as 372. He A, Zhu L, Gupta N, Chang Y, Fang F. Overexpression of
of a metabolically benign nonalcoholic fatty liver. diagnostic marker of non-alcoholic steatohepatitis micro ribonucleic acid 29, highly up-regulated in diabetic
Diabetes Care. 2013;36(8):2331–2338. in a Chinese population. World J Gastroenterol. 2016; rats, leads to insulin resistance in 3T3-L1 adipocytes. Mol
341. Guo H, Ingolia NT, Weissman JS, Bartel DP. Mam- 22(44):9844–9852. Endocrinol. 2007;21(11):2785–2794.
malian microRNAs predominantly act to decrease 359. Ao R, Wang Y, Tong J, Wang BF. Altered 373. Herrera BM, Lockstone HE, Taylor JM, Ria M, Barrett
target mRNA levels. Nature. 2010;466(7308):835– microRNA-9 expression level is directly correlated A, Collins S, Kaisaki P, Argoud K, Fernandez C,
840. with pathogenesis of nonalcoholic fatty liver Travers ME, Grew JP, Randall JC, Gloyn AL, Gauguier
342. Raajendiran A, Tsiloulis T, Watt MJ. Adipose tissue disease by targeting Onecut2 and SIRT1. Med Sci D, McCarthy MI, Lindgren CM. Global microRNA
development and the molecular regulation of lipid Monit. 2016;22:3804–3819. expression profiles in insulin target tissues in a
metabolism. Essays Biochem. 2016;60(5):437–450. 360. Mehta R, Otgonsuren M, Younoszai Z, Allawi H, spontaneous rat model of type 2 diabetes. Dia-
343. Hashimoto N, Tanaka T. Role of miRNAs in the Raybuck B, Younossi Z. Circulating miRNA in pa- betologia. 2010;53(6):1099–1109.
pathogenesis and susceptibility of diabetes mellitus. tients with non-alcoholic fatty liver disease and 374. Lee J, Padhye A, Sharma A, Song G, Miao J, Mo YY,
J Hum Genet. 2017;62(2):141–150. coronary artery disease. BMJ Open Gastroenterol. Wang L, Kemper JK. A pathway involving farnesoid
344. Dávalos A, Goedeke L, Smibert P, Ramı́rez CM, 2016;3(1):e000096. X receptor and small heterodimer partner positively
Warrier NP, Andreo U, Cirera-Salinas D, Rayner K, 361. López-Riera M, Conde I, Quintas G, Pedrola L, regulates hepatic sirtuin 1 levels via microRNA-34a
Suresh U, Pastor-Pareja JC, Esplugues E, Fisher EA, Zaragoza Á, Perez-Rojas J, Salcedo M, Benlloch S, inhibition. J Biol Chem. 2010;285(17):12604–12611.
Penalva LO, Moore KJ, Suárez Y, Lai EC, Fernández- Castell JV, Jover R. Non-invasive prediction of 375. Trajkovski M, Hausser J, Soutschek J, Bhat B, Akin A,
Hernando C. miR-33a/b contribute to the regula- NAFLD severity: a comprehensive, independent Zavolan M, Heim MH, Stoffel M. MicroRNAs 103
tion of fatty acid metabolism and insulin signaling. validation of previously postulated serum micro- and 107 regulate insulin sensitivity. Nature. 2011;
Proc Natl Acad Sci USA. 2011;108(22):9232–9237. RNA biomarkers. Sci Rep. 2018;8(1):10606. 474(7353):649–653.

1392 Watt et al Liver Hepatokines and Metabolic Disease Endocrine Reviews, October 2019, 40(5):1367–1393
REVIEW

376. Zhang Y, Yang L, Gao YF, Fan ZM, Cai XY, Liu MY, 381. Parker BL, Calkin AC, Seldin MM, Keating MF, Melbourne, Medical Building, Gratton Street, Melbourne,
Guo XR, Gao CL, Xia ZK. MicroRNA-106b induces Tarling EJ, Yang P, Moody SC, Liu Y, Zerenturk EJ, Victoria 3010, Australia. E: matt.watt@unimelb.edu.au.

Downloaded from https://academic.oup.com/edrv/article/40/5/1367/5479344 by BOEHRINGER INGELHEIM PHARMA GmbH CO. KG, Scientific Media Centre user on 22 June 2023
mitochondrial dysfunction and insulin resistance in Needham EJ, Miller ML, Clifford BL, Morand P, Disclosure Summary: The authors have nothing to
C2C12 myotubes by targeting mitofusin-2. Mol Cell Watt MJ, Meex RCR, Peng KY, Lee R, Jayawardana disclose.
Endocrinol. 2013;381(1–2):230–240. K, Pan C, Mellett NA, Weir JM, Lazarus R, Lusis AJ,
377. Thomou T, Mori MA, Dreyfuss JM, Konishi M, Meikle PJ, James DE, de Aguiar Vallim TQ, Drew Abbreviations
Sakaguchi M, Wolfrum C, Rao TN, Winnay JN, BG. An integrative systems genetic analysis of AKT, protein kinase B; AMPK, 59-AMP–activated protein
Garcia-Martin R, Grinspoon SK, Gorden P, Kahn CR. mammalian lipid metabolism. Nature. 2019; kinase; ANGPTL4, angiopoietin-like protein 4; CoA, co-
Adipose-derived circulating miRNAs regulate gene 567(7747):187–193. enzyme A; CPT, carnitine palmytoyl transferase; DPP4,
expression in other tissues [published correction dipeptidyl peptidase-4; ER, endoplasmic reticulum; FGF,
appears in Nature. 2017;545(7653):252]. Nature. fibroblast growth factor; FOXO1, forkhead box O1; GIP,
2017;542(7642):450–455. Acknowledgments gastric inhibitory peptide; GLP, glucagon-like peptide;
378. Henaoui IS, Jacovetti C, Guerra Mollet I, Guay C, Financial Support: M.J.W. is supported by National Health GLUT4, glucose transporter type 4; HDL, high-density li-
Sobel J, Eliasson L, Regazzi R. PIWI-interacting RNAs and Medical Research Council of Australia Senior Research poprotein; HFREP1, hepatocyte-derived fibrinogen-related
as novel regulators of pancreatic beta cell function. Fellowship APP1077703. P.M.M. is supported by a Natural protein 1; HMGB1, high-mobility group box 1 protein; JNK,
Diabetologia. 2017;60(10):1977–1986. Sciences and Engineering Research Council of Canada c-Jun N-terminal kinase; LECT2, leukocyte cell–derived
379. Li P, Ruan X, Yang L, Kiesewetter K, Zhao Y, Luo H, Chen Postdoctoral Fellowship. W.D.N. is supported by a Melbourne chemotaxin 2; LDL, low-density lipoprotein; LECT2, leu-
Y, Gucek M, Zhu J, Cao H. A liver-enriched long non- Research Scholarship (University of Melbourne). M.K.M. is kocyte cell–derived chemotaxin 2; LPC, lysophosphati-
coding RNA, lncLSTR, regulates systemic lipid meta- supported by National Health and Medical Research Council dylcholine; NAFL, nonalcoholic fatty liver; NAFLD,
bolism in mice. Cell Metab. 2015;21(3):455–467. of Australia Career Development Fellowship APP1143224. nonalcoholic fatty liver disease; NASH, nonalcoholic
380. Zhu X, Li H, Wu Y, Zhou J, Yang G, Wang W. lncRNA The authors acknowledge the support of National Health steatohepatitis; NF-kB, nuclear factor kB; nSMase2, neutral
MEG3 promotes hepatic insulin resistance by serving and Medical Research Council of Australia Grants sphingomyelinase 2; PEDF, pigment epithelium–derived
as a competing endogenous RNA of miR-214 to reg- APP1162511 (to M.J.W.) and APP1156508 (to M.J.W.). factor; PPAR, peroxisome proliferator-activated receptor;
ulate ATF4 expression. Int J Mol Med. 2019;43(1): Correspondence and Reprint Requests: Matthew RBP4, retinol binding protein 4; SeP, selenoprotein P; TCA,
345–357. J. Watt, PhD, Department of Physiology, University of tricarboxylic acid; TLR, Toll-like receptor; TSK, Tsukushi.

doi: 10.1210/er.2019-00034 https://academic.oup.com/edrv 1393

You might also like