You are on page 1of 16

REVIEWS

Regulation of hepatic glucose


metabolism in health and disease
Max C.Petersen1,2, Daniel F.Vatner1 and Gerald I.Shulman13
Abstract | The liver is crucial for the maintenance of normal glucose homeostasis it produces
glucose during fasting and stores glucose postprandially. However, these hepatic processes are
dysregulated in type1 and type2 diabetes mellitus, and this imbalance contributes to
hyperglycaemia in the fasted and postprandial states. Net hepatic glucose production is the
summation of glucose fluxes from gluconeogenesis, glycogenolysis, glycogen synthesis,
glycolysis and other pathways. In this Review, we discuss the invivo regulation of these hepatic
glucose fluxes. In particular, we highlight the importance of indirect (extrahepatic) control of
hepatic gluconeogenesis and direct (hepatic) control of hepatic glycogen metabolism. We also
propose a mechanism for the progression of subclinical hepatic insulin resistance to overt fasting
hyperglycaemia in type2 diabetes mellitus. Insights into the control of hepatic gluconeogenesis
by metformin and insulin and into the role of lipid-induced hepatic insulin resistance in modifying
gluconeogenic and net hepatic glycogen synthetic flux are also discussed. Finally, we consider
the therapeutic potential of strategies that target hepatosteatosis, hyperglucagonaemia and
adipose lipolysis.

Hepatic glucose production (HGP) accounts for ~90% thus facilitating the consumption of residual exogenous
of endogenous glucose production1, and it is crucial for glucose by extrahepatic tissues, such as skeletal muscle
systemic glucose homeostasis2. Net HGP is the sum- and adiposetissue.
mation of fluxes from gluconeogenesis, glycogenolysis, Key regulators of hepatic glucose metabolism act
glycogen synthesis, glycolysis and other pathways. In the through diverse mechanisms. For example, HGP is reg-
fasted state, the liver provides glucose to maintain eugly- ulated by the provision of substrates, such as glucose or
caemia and fuel obligate glucose-consuming cell types, glycerol; allosteric control by metabolites, such as acetyl-
such as neurons, red blood cells and renal medullary CoA, glucose and glucose6phosphate; the balance of
cells3. Postprandially, the liver contributes to normal glu- hormones, including insulin, glucagon, catecholamines
cose tolerance4. The liver contributes to the disposal of and corticosteroids; and cellular redox state, which can
enteral glucose loads by increasing the rate of glycogen be modified by treatment with metformin. This list is
synthesis and suppressing hepatic glucose output; these not comprehensive, which highlights the complexity
result in a net switch from hepatic glucose output to of the physiological regulation of HGP. In addition, the
hepatic glucose uptake2. The suppression of hepatic glu- processes that contribute to net HGP, including glycogen
cose output involves the suppression of hepatic glyco synthesis, glycogenolysis and gluconeogenesis, are regu-
1
Department of Internal genolysis and gluconeogenesis. As both glycogenolysis lated by independent mechanisms1012. As a result of this
Medicine, Yale School
of Medicine.
and gluconeogenesis contribute to HGP in humans complexity, hepatic glucose uptake is maximally stim-
2
Department of Cellular & that have fasted for less than 24h (REF.5), knowledge ulated by conditions that mimic the postprandial state,
Molecular Physiology, Yale of the mechanisms that mediate the postprandial sup- such as portal venous hyperglycaemia and hyperinsuli-
School of Medicine. pression of both processes is relevant to understand- naemia8,13. Of note, the gold-standard test of peripheral
3
Howard Hughes Medical
ing the hyperglycaemia observed in diabetes mellitus. and hepatic insulin sensitivity the hyperinsulinaemic
Institute, Yale School of
Medicine, New Haven, Net hepatic glucose uptake, as measured by splanchnic euglycaemic clamp technique14 fails to recreate the
Connecticut 06520, USA. arteriovenous balance and tracer methods, is estimated postprandial hepatic sinusoidal milieu. For example,
Correspondence to G.I.S. to be approximately one-third of a moderate enteral glu- maintaining euglycaemia rather than achieving portal
gerald.shulman@yale.edu cose load in humans and dogs2,4,69. However, the liver venous hyperglycaemia does not promote net hepatic
doi:10.1038/nrendo.2017.80 also contributes to the systemic disposal of an enteral glycogen synthesis2. Furthermore, the infusion of insu-
Published online 21 Jul 2017 glucose load through the suppression of glucose output, lin through a peripheral catheter decreases the normal

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 1



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Key points insulin resistance than visceral adiposity or BMI in indi-


viduals without diabetes mellitus and with obesity 21,
Hepatic glucose metabolism encompasses several catabolic and anabolic fluxes that and interventions that decrease IHTG content are asso-
have distinct modes of hepatocyte-autonomous (direct) and hepatocyte- ciated with the reversal of hepatic insulin resistance in
non-autonomous (indirect) regulatory mechanisms humans with NAFLD and rodent models of this dis-
Acute regulation of hepatic glucose metabolism is achieved through changes in ease20,2226. Hepatic levels of diacylglycerol the penul-
protein phosphorylation, substrate availability, allostery and redox state timate intermediate in the triglyceride synthesis pathway
Chronic regulation of hepatic glucose metabolism occurs through transcriptional have been proposed to mediate lipid-induced hepatic
mechanisms and the development of insulin resistance insulin resistance27. In the four human studies that have
Acute suppression of hepatic gluconeogenesis by insulin is largely an indirect effect measured hepatic diacylglycerol content and hepatic
that is mediated mostly through the suppression of adipose lipolysis, which reduces insulin sensitivity, diacylglycerol content was strongly
delivery of nonesterified fatty acids and glycerol to the liver
correlated with hepatic insulin resistance in all four,
The major direct effect of insulin on hepatic glucose metabolism is the acute whereas other potential mediators of hepatic insulin
regulation of hepatic glycogen metabolism; however, hyperglycaemia and
resistance (such as ceramides) showed an inconsistent
hyperinsulinaemia are required to maximally stimulate net hepatic glycogenesis
relationship21,2830. An increase in the level of hepatic dia-
Lipid-induced hepatic insulin resistance, hyperglucagonaemia and excessive
cylglycerol activates protein kinase C (PKC), which
adipose lipolysis represent three pathophysiological processes that might be
impairs the tyrosine kinase activity of the insulin recep-
amenable to pharmacological intervention in humans who have impaired hepatic
glucose metabolism tor (INSR)31,32 through inhibitory phosphorylation of
INSR at Thr1160; mice homozygous for a Thr1150Ala
(the homologous residue to human Thr1160) muta-
insulin concentration gradient between the portal vein tion in Insr were protected from lipid-induced hepatic
and systemic vein (normally approximately threefold insulin resistance33.
higher relative to the concentration in the portal vein)15. Hepatic insulin resistance is also associated with other
Consequently, for hyperinsulinaemiceuglycaemic abnormalities that might contribute to dysregulated glu-
clamps that only increase peripheral insulin concentra- cose metabolism. For example, the progression of NAFLD
tion by approximately threefold, the insulin level that is to liver fibrosis and non-alcoholic steatohepatitis (NASH)
detected by the liver will not change. These limitations is common, and is associated with increased morbidity
must be considered in studies of hepatic glucose metab- and mortality 34. The increased deposition of extracellular
olism using the hyperinsulinaemiceuglycaemic clamp. matrix (ECM) due to high-fat diet (HFD)-induced
This Review aims to detail both the established mech- hepatic fibrosis results in interactions between the ECM
anistic knowledge and newer insights regarding the and mediators of insulin signalling, such as AKT, through
regulation of HGP in health and in diabetes mellitus. the scaffolding pseudokinase integrin-linked protein
We begin by discussing lipid-induced hepatic insulin kinase (ILK)35. The liver-specific deletion of Ilk protected
resistance, which is one of the primary pathophysiolog- mice from HFD-induced hepatic steatosis, and therefore
ical processes that are involved in the dysregulation of from HFD-induced hepatic insulin resistance, hinting at
hepatic glucose metabolism in type2 diabetes mellitus a bidirectional relationship between fibrosis and hepatic
(T2DM). We then consider elements of the regulation of lipid deposition in this model35.
hepatic gluconeogenesis and hepatic glycogen metabo- Improvements in our understanding of the regulation
lism, and use this knowledge to propose a unified frame- of hepatic glucose metabolism and of hepatic insulin
work for understanding hepatic insulin action. Finally, resistance might inform potential therapeutic strategies
we highlight some of the therapeutic implications of for normalizing hepatic glucose production in T2DM.
these advances. The physiological and pathophysiological regulation of
hepatic gluconeogenesis and glycogen metabolism are
Hepatosteatosis and insulin resistance examinedbelow.
Although both insulin and glucose are required for
net hepatic glucose uptake, insulin alone suppresses Control of hepatic gluconeogenesis
HGP in a dose-dependent manner 16,17. In individuals Gluconeogenesis contributes approximately half of the
with T2DM, the rate of HGP is increased under basal total HGP in humans following an overnight fast and
physiological conditions, and insulin-dependent sup- is primarily responsible for the increase in fasting HGP
pression of HGP is impaired at both physiological and in individuals with T2DM5,19,3639. Major gluconeogenic
Hyperinsulinaemic modest supraphysiological plasma levels of insulin3,18,19. precursors, including lactate, alanine and glycerol, are
euglycaemic clamp Although the insulin-dependent suppression of HGP subject to diverse regulatory mechanisms. Below, we
technique has several physiological components (see below), consider the recent progress in our understanding of
A technique in which insulin is
impaired suppression of HGP is generally considered gluconeogenic regulation (FIG.1).
infused at a constant rate to
achieve hyperinsulinaemia and to represent hepatic insulin resistance. Accordingly,
glucose is infused at a variable the suppression of HGP is widely used in mechanistic Indirect control of hepatic gluconeogenesis by lipolysis.
rate to maintain euglycaemia; studies as a measure of hepatic insulin action20. Impaired Indirect control (that is, non-hepatocyte-autonomous
once steady-state euglycaemia suppression of HGP is reproducibly associated with control) of HGP was first hypothesized to occur more
has been achieved, the glucose
infusion rate is proportional to
increased intrahepatic triglyceride (IHTG) content, than half a century ago by Levine and Fritz40. It is now
the whole-body insulin which is a hallmark of non-alcoholic fatty liver disease clear that multiple indirect mechanisms contribute
sensitivity of the individual. (NAFLD). IHTG content is a better predictor of hepatic to the physiological regulation of HGP. For example,

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

White adipose tissue

From Cori cycling

WAT lipolysis Glucose


Lactate
GLUT2 GCGR
NEFA Glycerol
INSR

Lactate

NEFA Glycerol DHAP [cAMP]

Metformin PFK2/
[NADH]:[NAD+] LDH PKA AKT
FBPase-2

mGPD Gluconeogenesis IP3R


Glycolysis

[NADH]:[NAD+] Ca2+
L-PK
Pyruvate
-oxidation
CRTC2 FOXO1
CREB CBP

Acetyl-CoA Oxaloacetate
+
PGC1a
Nature Reviews | Endocrinology
Gluconeogenic
PC capacity
Acetyl-CoA PCK1

Pyruvate
G6PC

Mitochondrion

Substrate Allostery Transcriptional Redox

Figure 1 | Control of hepatic gluconeogenesis. Hepatic gluconeogenesis is regulated byNature Reviews of


the availability | Endocrinology
substrates
(light blue boxes), allostery from metabolites (green boxes), transcriptional mechanisms (purple boxes) and cellular redox
state (dark blue boxes). Lipolysis in white adipose tissue (WAT) produces nonesterified fatty acids (NEFA) and glycerol, both
of which can stimulate gluconeogenesis. The oxidation of NEFA yields mitochondrial acetyl-CoA, which promotes
gluconeogenesis by allosterically activating pyruvate carboxylase (PC), which, in turn, catalyses the conversion of
pyruvate to the gluconeogenic substrate oxaloacetate. Glycerol can be phosphorylated and converted into the
gluconeogenic precursor dihydroxyacetone phosphate (DHAP). This process is inhibited by metformin, a non-competitive
inhibitor of mitochondrial glycerol3phosphate dehydrogenase (mGPD). Inhibition of mGPD impairs the production of
DHAP, which results in a decrease in gluconeogenesis from glycerol. Furthermore, the increased cytosolic redox state
([NADH+]:[NAD+]) that results from the inhibition of mGPD inhibits the redox-dependent enzyme lactate dehydrogenase
(LDH), thus limiting the production of pyruvate, and thus gluconeogenesis, from lactate. The transcriptional regulation of
gluconeogenesis by glucagon and insulin is relatively slow compared with the effects of these hormones on hepatic
glycogen metabolism, acting primarily through transcriptional activation and repression, respectively, of the genes that
encode the gluconeogenic cytosolic enzymes phosphoenolpyruvate carboxykinase (PCK1) and glucose6phosphatase
(G6PC). The binding of insulin to the insulin receptor (INSR) leads to the activation of AKT, which phosphorylates and
excludes the transcription factor Forkhead box O1 (FOXO1) from the nucleus. In the absence of insulin, FOXO1 promotes
gluconeogenic gene transcription with its coactivator peroxisome proliferator-activated receptor- co-activator 1
(PGC1; encoded by PGC1a). Glucagon binding to the glucagon receptor (GCGR) increases intracellular concentrations
of cAMP and activates protein kinase A (PKA), which phosphorylates the inositol1,4,5trisphosphate receptor (IP3R),
increasing cytosolic Ca2+ levels and activating CREB-regulated transcription co-activator 2 (CRTC2). However,
GCGR-dependent activation of PKA also acts acutely by inducing inhibitory phosphorylation of glycolytic regulatory
enzymes, including 6phosphofructo2kinase/fructose2,6bisphosphatase (PFK2/FBPase2) and liver-type pyruvate
kinase (LPK), to decrease glucose oxidation and thereby favour net glucose production. Small up and down arrows
represent an increase or decrease, respectively, in protein level or activity. Dotted arrows represent glycolysis. CBP,
CREB-binding protein; CREB, cAMP-responsive element-binding protein 1; GLUT2, glucose transporter 2.

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 3



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

insulin strongly regulates the secretion of glucagon of lipolysis52,53. These concepts were then extended to
from pancreatic cells, and the loss of this paracrine the normal physiological suppression of hepatic gluco
mode of regulation contributes to the development of neogenesis by insulin. Infusion of insulin in fasted rats
hyperglucagonaemia and increased HGP in diabetes rapidly and markedly suppressed plasma concentra-
mellitus41,42. HGP is also indirectly regulated by the tions of NEFA and glycerol, which was accompanied
products of lipolysis glycerol and nonesterified fatty by decreases in hepatic acetyl-CoA concentration and
acids (NEFA). Progress in this field accelerated when HGP51. Functionally blocking the insulin-dependent
investigators began to compare the suppression of HGP suppression of lipolysis using acetate and glycerol infu-
during portal venous versus peripheral venous insu- sions that were calibrated to match basal hepatic levels
lin infusions. In an early canine study, these modes of of acetyl-CoA and rates of whole-body glycerol turnover
infusion achieved similar peripheral concentrations completely prevented insulin-mediated suppression of
of insulin but markedly different portal concentra- HGP51. Importantly, these studies were carried out in
tions of insulin; however, both methods of infusion fasted rats that had minimal hepatic glycogen stores;
resulted in similar magnitudes of HGP suppression43. In therefore, insulin-dependent suppression of HGP solely
healthy humans, portal and peripheral venous infusions reflected the suppression of gluconeogenesis.
achieved similar portal insulin concentrations and dif- Further evidence for this indirect mechanism of
ferent peripheral insulin concentrations, and revealed gluconeogenic suppression was provided by studies in
that individuals who had the highest peripheral insu- rodents with genetic defects in hepatocellular insulin
lin concentrations also displayed the greatest degree of signalling. Mice in which Akt1, Akt2, and Forkhead box
HGP suppression44. Although a specific increase in portal O1 (Foxo1) genes that encode three crucial effec-
venous insulin levels also caused the suppression of HGP tors of hepatocellular insulin action were knocked
in dogs45 and humans44, a role for the indirect control of out behaved similarly to wild-type mice, suppressing
HGP by insulin was apparent46. Attention quickly turned HGP normally in hyperinsulinaemiceuglycaemic
to the role of lipolytic products (NEFA and glycerol) in clamp studies but showing increased HGP when ace-
the regulation of hepatic gluconeogenesis. In humans, tate and glycerol infusions were superimposed on the
low-dose infusion of the lipid emulsion Intralipid clamp51,58. Moreover, rats in which Insr was ablated in
attenuated fasting-induced decreases in gluconeogen- liver and white adipose tissue (WAT) by antisense oligo-
esis, which indicates that NEFA and/or glycerol have a nucleotides regained the ability to suppress HGP in an
physiological effect on gluconeogenesis47. Furthermore, insulin-dependent manner when adipose lipolysis was
low-dose infusion of an Intralipid and heparin mixture pharmacologically blocked51. This finding is consistent
to prevent insulin-mediated decreases in plasma levels with other studies in mice that reported that the ablation
of NEFA abolished the enhanced suppression of HGP of hepatic Insr does not prevent the insulin-dependent
observed with peripheral infusion compared with equi- suppression of HGP59, and that liver-specific rescue of
molar tolbutamide-stimulated portal insulin secretion in Insr expression does not restore acute insulin-dependent
humans48. Insulin-dependent suppression of HGP was suppression of HGP60. These results demonstrate that
also impaired by this fatty acid clamp in dogs49,50, which hepatic insulin signalling is not essential for the suppres-
implicates insulin-mediated suppression of lipolysis as a sion of HGP by insulin in an overnight fasted rodent,
mediator of insulin-dependent suppression ofHGP. which is depleted of hepatic glycogen and is mostly
Additional studies have advanced the hypothesis dependent on hepatic gluconeogenesis. Overall, sub-
that the control of hepatic gluconeogenesis by lipolysis stantial data point to a crucial role for the inhibition of
involves the conversion of glycerol to glucose, and, quan- adipose lipolysis, and resultant decreases in the turno-
titatively, the acetyl-CoA-mediated allosteric activation ver of NEFA and glycerol, in the acute insulin-mediated
of pyruvate carboxylase5153. Conceptually, the effect of suppression of hepatic gluconeogenesis.
lipolysis to increase hepatic gluconeogenesis from glyc- Whether the impaired lipolytic control of hepatic
erol is mechanistically straightforward; it involves a sub- gluconeogenesis contributes to the increase in HGP
strate push mechanism, in which increased substrate that is associated with T2DM is unclear. Humans
availability drives an increase in the formation of prod- who have poorly controlled T2DM (fasting plasma
uct 54. However, the mechanism by which an increased glucose >250mg dl1) have higher plasma concentra-
turnover and oxidation of fatty acids might drive gluco tions of NEFA than healthy controls throughout the day,
neogenesis from pyruvate cannot be explained by a and increased plasma concentrations of NEFA are an
substrate push mechanism, as acetyl-CoA and acetate, independent predictor of incident T2DM6167. Insulin-
the products of oxidation of fatty acids, are not gluco dependent suppression of glycerol turnover, which is a
neogenic substrates; they do not contribute net carbon readout of lipolytic flux, is impaired in individuals who
to gluconeogenesis. Instead, fatty acids can activate have insulin resistance, with or without diabetes mel-
hepatic gluconeogenesis by increasing mitochondrial litus6871. The rates of glycerol turnover and of gluco
levels of acetyl CoA and, consequently, by allosterically neogenesis from glycerol are also increased in overnight
activating pyruvate carboxylase, as was described in the fasted humans with T2DM72,73. Although these data are
1960s52,5557. Indeed, hepatic concentrations of acetyl- largely correlative, they suggest that chronic increases in
CoA and pyruvate carboxylase activity were increased the rate of lipolysis might promote an increased rate of
in rat models of insulinopenic diabetic ketoacidosis hepatic gluconeogenesis in T2DM. Of note, the plasma
and suppressed following pharmacological inhibition turnover of hydroxybutyrate might be a non-invasive

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

surrogate biomarker for hepatic concentrations of However, some reports suggest that metformin-dependent
acetyl-CoA74. This method might be useful for human inhibition of mitochondrial complex I is observed only
studies that examine the lipolytic control of hepatic at suprapharmacological concentrations (millimolar) of
gluconeogenesis. metformin, in contrast to the clinically relevant plasma
concentration range of 50100M, which is observed in
Suppression of hepatic gluconeogenesis by metformin. patients who take 2g of metformin daily 83,87. Moreover,
The biguanide compound metformin is used as a first- many studies disagree on whether pharmacologically
line therapy for T2DM75, and acts primarily through the relevant concentrations of biguanides can increase intra-
suppression of hepatic gluconeogenesis76,77. However, cellular levels of AMP83,84,89. In one study 89, long-term
the molecular mechanism that underlies this effect treatment of mouse hepatocytes with physiologically
remains a subject of active investigation. Perhaps the relevant biguanide concentrations increased intracellular
best-studied potential mechanism of metformin action is levels of AMP, and decreased intracellular levels of cAMP
the stimulatory phosphorylation of AMP-activated pro- (consistent with the allosteric inhibition of adenylyl
tein kinase (AMPK) at Thr172 (REF.78). This mechanism cyclase by AMP); this decrease in the concentration of
was supported by studies in mice with a liver-specific cAMP was hypothesized to reduce HGP by antagonizing
deletion of the gene that encodes serine/threonine liver the action of glucagon. However, other studies in mouse
kinase B1 (LKB1; also known as STK11), which phos- hepatocytes84 and rats83 have not observed a link between
phorylates AMPK at Thr172; these mice were refractory metformin-dependent suppression of HGP and changes
to metformin therapy and displayed substantial tran- in intracellular concentrations of cAMP. Furthermore,
scriptional dysregulation as a result of chronic inactiva- metformin did not inhibit glucagon-stimulated HGP
tion of AMPK79. The link between metformin, AMPK in a randomized placebo-controlled double-blind trial in
and gluconeogenesis has been proposed to involve humans with prediabetes90.
both the AMPK-mediated disassembly of the cAMP- Metformin is less potent than other related guanide
responsive element-binding protein 1 (CREB)CREB- and biguanide compounds, such as galegine and phen-
binding protein (CBP)CREB-regulated transcription formin91. In particular, phenformin was withdrawn
co-activator 2 (CRTC2) transcriptional complex, which from clinical use owing to adverse effects, including
positively regulates the expression of the gluconeogenic lactic acidosis91. However, the observation that acute
genes phosphoenolpyruvate carboxykinase 1 (Pck1) and galegine treatment decreased hepatic gluconeogenesis
glucose6phosphatase (G6pc)80, and improvements in and increased plasma concentrations of lactate in rats
lipid-induced hepatic insulin resistance81. However, met- within 30min of administration provided an initial
formin continues to inhibit glucose production in Ampk- clue to a third proposed mechanism for the action of
knockout primary mouse hepatocytes and improves metformin83. In rats, metformin increased the cytoplas-
glucose tolerance in Ampk-knockout mice to a similar mic redox state and decreased the mitochondrial redox
extent to wild-type control mice82. Furthermore, met- state, which suggests that metformin inhibits one of the
formin suppressed glucose production in mouse hepato metabolic shuttles that are involved in equilibrating
cytes that overexpressed the gluconeogenic enzymes cytoplasmic and mitochondrial redox states83. Indeed,
cytosolic PCK1 and G6PC, which challenges the hypo metformin was shown to non-competitively inhibit
thesis that transcriptional mechanisms are essential for mitochondrial glycerol3phosphate dehydrogenase
the action of metformin82. In addition, both galegine (a (mGPD) in isolated hepatocytes with a clinically rele-
related guanide compound) and metformin suppressed vant Ki of ~50M83. An important distinguishing feature
hepatic gluconeogenesis within 20min of intravenous of this mechanism, in contrast to all previous proposed
infusion in rats, a time frame that is inconsistent with mechanisms, is that inhibition of mGPD was predicted
that of transcriptional mechanisms83. In hepatocytes, the to impair gluconeogenesis only from redox-dependent
activation of AMPK in response to guanide and bigua- substrates: lactate, as lactate dehydrogenase requires
nide compounds is well established78,80,82,8486 and occurs the reduction of NAD+, and glycerol, as mGPD pro-
rapidly following the intravenous administration of duces the gluconeogenic precursor dihydroxyacetone
galegine83. However, interestingly, pharmacological acti- phosphate (DHAP)83. This prediction was confirmed
vation of AMPK with A-769662 (which activates AMPK in cultured hepatocytes that were treated with met-
to a similar extent to galegine) did not suppress HGP in formin or small interfering RNA (siRNA) targeting
awake hepatic glycogen-depleted rats83. Thus, several lines the mGPD transcript 83. Consistent with this model,
of evidence have cast doubt on the necessity of AMPK knockdown of hepatic mGPD using antisense oligo-
activation for the metformin-dependent suppression of nucleotides phenocopied metformin treatment in rats,
hepatic gluconeogenesis, warranting the investigation and Gpd2knockout mice displayed reduced HGP dur-
of alternative mechanisms ofaction. ing fasting that was unaltered by treatment with met-
An alternative but related hypothesis posits that met- formin83, which is consistent with a previous report of
formin inhibits the activity of mitochondrial complex I decreased fasting glycaemia in Gpd2knockout mice92.
and thus alters the adenine nucleotide energy charge As the importance of the glycerol3phosphate shuttle in
(that is, the cellular [AMP]:[ATP] and [ADP]:[ATP] gluconeogenesis in the human liver is unclear 93,94, stud-
ratios)87. An increase in the [AMP]:[ATP] ratio would ies in humans are required to determine whether met-
activate AMPK and inhibit the activity of fructose1,6 formin regulates gluconeogenesis by modulating cellular
bisphosphatase, which is a key gluconeogenic enzyme88. redox state. A unique testable prediction of the redox

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 5



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

hypothesis is that treatment with metformin should gluconeogenesis110,111. Overall, the place of the FOXO
decrease gluconeogenesis from lactate and glycerol, but PGC1 axis in the hierarchy of gluconeogenic control
not from pyruvate, alanine orDHAP. mechanisms remains ambiguous. For example, it is
unclear whether dysregulation of FOXO transcription
Hormonal control of hepatic gluconeogenesis. Glucagon factors contributes to the increased rate of gluconeo-
and catecholamines positively regulate hepatic gluconeo- genesis observed in T2DM. Rodents that are fed a HFD
genesis through cAMP-dependent activation of protein have hepatic insulin resistance but unaltered levels of the
kinase A (PKA)95. Glucagon action is essential for hyper gluconeogenic enzymes G6PC and PCK1 (REFS24,51).
glycaemia in rodent models of T2DM; db/db diabetic mice Similarly, the expression levels of G6PC and PCK1 were
that lack the glucagon receptor (Gcgr) did not develop not altered in humans with T2DM112. Available evidence,
hyperinsulinaemia or hyperglycaemia96. Glucagon and although limited, does not currently support a central
catecholamines stimulate net hepatic gluconeogenic flux role for perturbed gluconeogenic gene transcription in
in the acute setting by promoting the phosphorylation the increased gluconeogenesis involved inT2DM.
of PKA, inhibition of the liver-type isozyme of pyruvate Glucagon and catecholamines also participate in
kinase (LPK; with glucagon only)97 and phosphoryl- the slow transcriptional control of gluconeogenesis by
ation of the bifunctional enzyme 6phosphofructo2 stimulating the cAMPPKA-dependent activation of
kinase/fructose2,6bisphosphatase 2 (PFK2/FBPase2)98 the CREBCBPCRTC2 complex 80,113. The key medi-
at Ser36. Phosphorylation of PFK2/FBPase2 favours its ator of hormonal control in the CREBCBPCRTC2
phosphatase activity, enabling it to decrease the production module seems to be CRTC2. Similarly to FOXO1,
of fructose2,6bisphosphate, an allosteric inhibitor of CRTC2 is dephosphorylated and localized to the
the gluconeogenic enzyme fructose2,6bisphosphatase 1 nucleus during fasting 113, and is phosphorylated and
(FBPase1)98,99; phosphorylated PFK2/FBPase2 also facil- excluded from the nucleus in response to insulin114. The
itates the nuclear translocation and consequent functional dephosphorylation of CRTC2 during fasting involves
inactivation of glucokinase (GCK)100. These mechanisms the PKA-dependent inhibition of the serine/threonine
might also underlie the stimulatory effect of asprosin kinase SIK2, which normally phosphorylates CRTC2,
(an adipose-derived peptide hormone that activates and the PKA-dependent phosphorylation of inositol
hepatocellular cAMPPKA signalling) on HGP101. The 1,4,5trisphosphate receptors (IP3R) with a resultant
observation that asprosin stimulated HGP in mice that increase in intracellular Ca2+ levels, which activates the
were fasted for 18h (REF.101) suggests that asprosin CRTC2specific phosphatase calcineurin115. Crosstalk
stimulates gluconeogenesis; however, cAMPPKA- between the FOXO1 and CRTC2 pathways includes the
dependent stimulation of hepatic glycogenolysis (see induction of PGC1 expression by CREB113. Consistent
below) would also be expected to contribute to HGP with this crosstalk, the CREBCBPCRTC2 module
in non-fastedmice. exerts stronger control over the expression of G6pc dur-
By contrast, the acute and direct negative control of ing the first 6h of fasting in mice, whereas the FOXO1
gluconeogenesis by insulin is not prominent invivo11,102. PGC1 module has a larger role in stimulating G6pc
High concentrations of insulin can counteract cAMP- transcription over longer durations (~18h) of fasting
mediated effects, such as the phosphorylation of LPK in mice116. As predicted, Crtc2knockout mice had
and PFK2/FBPase2, within 30min (REFS97,102,103); fasting hypoglycaemia and mice with adeno-associated
however, the insulin-dependent regulation of gluco- virus (AAV)-mediated overexpression of constitutively
neogenesis primarily occurs through slow transcrip- active CRTC2 were hyperglycaemic117,118.
tional mechanisms. The best-characterized pathway for
insulin-dependent transcriptional control of gluconeo- Gluconeogenic capacity of the liver. Oscillations in the
genic gene expression involves members of the FOXO FOXO- and CREB-regulated expression of gluconeo-
family of transcription factors (FOXO1, FOXO3a and genic enzymes have been proposed to dictate the gluco
FOXO4)11. FOXO proteins, in concert with the transcrip- neogenic capacity of the liver. These transcriptional
tional coactivator peroxisome proliferator-activated oscillations are probably superimposed on normal
receptor- co-activator 1 (PGC1; also known as circadian oscillations119; the circadian clock modulates
PPARGC1A), positively regulate the expression of PCK1 the activity of CREB (and therefore glucagon and gluco
and G6PC104107. FOXO proteins are phosphorylated by corticoid function)120,121. Liver-specific knockout of the
AKT following stimulation with insulin, which induces circadian clock component aryl hydrocarbon receptor
their nuclear exclusion and consequent inactivation106. translocator-like protein 1 (Bmal1, also known as Arntl)
Gainoffunction or lossoffunction perturbations of in mice was associated with fasting hypoglycaemia; how-
the FOXOPGC1 axis have marked effects on G6PC ever, this effect was primarily attributable to a marked
and PCK1 protein levels and glycaemia in rodent stud- decrease in the expression of glucose transporter 2
ies105,108,109. However, normalization of hyperglycaemia (Glut2; also known as Slc2a2) rather than changes in
and re-sensitization of HGP to insulin stimulation were gluconeogenic gene transcription122. Interesting associ-
observed following the deletion of Foxo1 in mice with ations between the circadian control of gluconeogenesis
liver-specific knockout of Insr, which illustrates both and lipogenesis have been uncovered. Mice with hepatic
the profound consequences of continuous activation knockout of the gene that encodes histone deacetylase 3
of FOXO and the dispensability of the FOXOPGC1 (Hdac3), which regulates the circadian rhythm of lipo-
axis for acute insulin-mediated suppression of hepatic genesis, rerouted gluconeogenic substrates to lipid

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

storage, resulting in modest decreases in fasting glycae- synthesis in the liver 138. Net hepatic glycogen deposition
mia and improved glucose tolerance123. However, inter- depends on the coordinated suppression of glycogeno-
estingly, these alterations in gluconeogenic flux were lytic flux and the stimulation of glycogen synthetic flux.
not associated with changes in the expression of G6pc Both glycogenolysis and glycogen synthesis are subject
or Pck1 (REF.123). Overall, no direct evidence indicates to complex regulatory mechanisms; however, generally,
that modest oscillations in the transcription of gluco- a useful simplification is to consider glucose as the pri-
neogenic genes drive changes in gluconeogenic flux mary suppressor of hepatic glycogenolysis and insulin
the pyruvate tolerance test, which is used in many studies as the principal activator of hepatic glycogen synthesis
of circadian control of gluconeogenesis, is highly respon- invivo. This paradigm was illustrated in a study that used
sive to changes in gluconeogenic capacity but might not 13
C magnetic resonance spectroscopy (MRS) measure-
reflect physiological rates of gluconeogenesis. Indeed, ments of hepatic glycogen in healthy individuals who
indirect evidence suggests that gluconeogenic capacity had fasted overnight and who were subjected to hyper-
might not be a major regulator of gluconeogenic flux in glycaemia (10mM glucose), hyperinsulinaemia (400pM
mice. In one study, a >90% decrease in the expression insulin), both hyperglycaemia and hyperinsulinaemia,
of Pck1 in mice resulted in only a moderate 40% reduc- or neither 10. Endogenous secretion of insulin and gluca-
tion in gluconeogenesis124. Similarly, in G6pc/ mice, gon was suppressed using somatostatin. Control indi-
adenoviral-mediated restoration of G6pc expression viduals who were not subjected to hyperglycaemia or
to only 20% of the activity in wild-type mice alleviated hyperinsulinaemia exhibited net hepatic glycogenolysis,
fasting hypoglycaemia125. These remarkable pheno- as expected. Hyperinsulinaemia was required for the
types suggest that modest oscillations in the expression stimulation of glycogen synthesis but did not suppress
of Pck1 and G6pc, mediated by insulin, the circadian glycogenolysis. By contrast, hyperglycaemia was required
clock, glucagon and other mechanisms, exert limited for the suppression of glycogenolysis; consequently, the
control of gluconeogenic flux in the normal liver; how- hyperinsulinaemichyperglycaemic group achieved
ever, compensations, such as the activation of alternative maximal hepatic glycogen synthesis10. Of note, the degree
metabolic pathways in these mouse models, potentially of hepatic hyperinsulinaemia that was achieved in this
complicate this simple interpretation. study was modest; higher concentrations of exogenous
insulin might suppress glycogenolysis, as observed in
Neural control of HGP. A role for the central nerv- cultured human hepatocytes139.
ous system (CNS) in the regulation of hepatic glucose The conclusion that both hyperglycaemia and
metabolism has been appreciated since the 1850s126, and hyperinsulinaemia are necessary for maximal hepatic
this subject has experienced a resurgence in research in glycogen synthesis also has implications for the use of
the past few years127,128. Leptin, an adipose-derived hor- hyperinsulinaemiceuglycaemic clamps. The action
mone that controls satiety, acts through several mecha- of hepatic insulin during the hyperinsulinaemic
nisms to modulate energy balance and hepatic glucose euglycaemic clamp is often measured as suppression
metabolism129. Insulin is transcytosed across the blood of HGP; however, suppression of HGP has a large
brain barrier 130 and can bind to INSR on neurons and extrahepatic component, owing largely to the lipolytic
glial cells131. Neuron-specific deletion of Insr predisposes control of gluconeogenesis. Insulin-dependent stim-
mice to diet-induced obesity and concomitant hepatic ulation of hepatic glycogen synthesis would be a use-
insulin resistance, probably through increasing appe- ful readout of direct hepatic insulin action, but under
tite132. However, CNS-dependent regulation of hepatic hyperinsulinaemiceuglycaemic conditions both glyco-
insulin action might occur through mechanisms that gen synthesis and glycogenolysis are active, which results
are independent of energy balance. For example, insu- in glycogen cycling and attenuated net hepatic glycogen
lin action in hypothalamic nuclei can potently suppress synthesis10. Thus, a hyperinsulinaemichyperglycaemic
HGP127,133. Hepatic vagotomy might also alter hepatic clamp is necessary to achieve maximal net hepatic gly-
insulin action, although the mechanisms that underlie cogen synthesis and, accordingly, this technique has
this are currently unclear 134. The intracerebroventricu- been used to measure absolute rates of hepatic glycogen
lar infusion of insulin suppressed HGP in rodents135; synthesis through both direct and indirect glycogen syn-
however, a physiological increase in levels of insulin in thesis pathways in rodents33 and dogs140. The prevention
the brain achieved through intra-arterial infusion did of lipid-induced hepatic insulin resistance in InsrT1150A
not alter HGP in dogs136,137. Although the physiolog- mice (which are insensitive to INSR inhibition by PKC)
ical mechanisms that link insulin action in the brain was associated with improvements in insulin-stimulated
and liver are unclear, they involve direct sympathetic and hepatic glycogen synthesis compared with HFD-fed
Pyruvate tolerance test parasympathetic efferents, as well as the activation of the wild-type controls during hyperinsulinaemichyperg-
A test in which a large bolus of
hypothalamicpituitaryadrenal axis127,128. lycaemic clamp studies33. Although they are infrequently
the gluconeogenic substrate
pyruvate is administered and used, these invivo measurements of net hepatic glyco-
plasma levels of glucose are Control of hepatic glycogen metabolism gen synthesis probably provide the best direct readout of
measured at defined time Hepatic glycogen can be synthesized from glucose directly hepatic insulinaction.
intervals; plasma glucose (glucose to glucose6phosphate to UDP-glucose to gly- Insulin, glucagon and glucose regulate hepatic
excursion is assumed to be
proportional to the rate of
cogen) or indirectly (glucose to glucose6phosphate to glycogenolysis and glycogen synthesis through mecha-
pyruvate-stimulated hepatic pyruvate to glucose6phosphate to UDP-glucose to gly- nisms that alter the activity of GCK, glycogen synthase
gluconeogenesis. cogen); these pathways contribute similarly to glycogen and glycogen phosphorylase (FIG.2).

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 7



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

GCK activity and glycogenesis. The transport of glu- glucose metabolism in hepatocytes141,142. GCK strongly
cose into hepatocytes occurs by facilitated diffusion regulates hepatic glycogen deposition. Transgenic mice
through GLUT2, and, consequently, intrahepatic glu- that have an extra copy of the Gck displayed more
cose concentrations parallel those in the plasma. GCK than threefold greater hepatic glycogen deposition than
which catalyses the phosphorylation of glucose to wild-type mice during hyperglycaemic clamp studies143.
glucose6phosphate is therefore the gatekeeper for Similarly, the glucose intolerance of mice that have

a b
Fasting Fed
Glycogenolysis > Glycogen synthesis Glycogen synthesis > Glycogenolysis
Glucagon tone > Insulin tone Insulin tone > Glucagon tone

Glucose
GCGR
GLUT2
INSR

GKRP GKRP
GCK GCK

GCK GCK AKT


Nucleus GCK
PDE3B
Direct Glucose-6-
pathway phosphate

[cAMP] [cAMP]
Indirect
pathway
UDP-glucose Allostery
Phosphorylase PKA Pyruvate
kinase
Allostery Glycogen pSer
Glycogen pSer15 synthase
phosphorylase
PP1 Glycogen pSer15
GL synthase
PP1
pSer GL
PP1 G
L
Glycogen
Glycogen
phosphorylase

Nature Reviews | Endocrinology


Figure 2 | Control of hepatic glycogen metabolism. a|Under fasted involves the conversion of glucose to glucose6phosphate and its subsequent
conditions, glycogenolysis is activated and glycogen synthesis is suppressed. incorporation into glycogen, with all six carbons of the glucosyl unit intact. The
Activation of the glucagon receptor (GCGR) induces increased intracellular indirect pathway of glycogen synthesis involves the conversion of glucose to
concentrations of cyclic AMP (cAMP; indicated by an up arrow), which leads pyruvate, and pyruvate to glucose6phosphate, before incorporation into
to the activation of protein kinase A (PKA). Activated PKA inhibits the glycogen. Glucose6phosphate both allosterically activates glycogen
transcription of the glucokinase (GCK) gene; inhibits the dissociation of GCK synthase and is a substrate for glycogen synthesis. Insulin activates AKT, which,
from glucokinase regulatory protein (GKRP), and thus induces the nuclear in turn, induces the transcription of GCK and the cytoplasmic translocation
sequestration of GCK; phosphorylates, and thus inactivates, glycogen of GCK and activates phosphodiesterase 3B (PDE3B), which decreases
synthase; and phosphorylates and activates phosphorylase kinase, which intracellular levels of cAMP (indicated by a down arrow) and leads to the
activates glycogen phosphorylase by phosphorylating Ser15. Phosphorylated, inhibition of the PKA-dependent processes described in part a. Active PP1 with
active glycogen phosphorylase also binds to and inhibits the GL subunit of its GL targeting subunit dephosphorylates and inactivates glycogen
protein phosphatase 1 (PP1), which prevents the PP1dependent phosphorylase, and dephosphorylates and activates glycogen synthase. The
dephosphorylation and inactivation of glycogen synthase. The coordinated coordinated inhibition of glycogen phosphorylase and activation of glycogen
activation of glycogen phosphorylase and inhibition of glycogen synthase synthase result in net hepatic glycogen synthesis. In parts a, b the insulin
result in net glycogenolysis. b|Under fed conditions, hormonal and allosteric receptor (INSR) and glucagon receptor (GCGR) are shown in faded colours for
mechanisms coordinate the stimulation of glycogen synthesis through direct context, and grey inhibitory arrows depict the processes in which they are not
and indirect pathways. Glucose promotes the dissociation of GCK from GKRP, dominant. The dashed arrows indicate dephosphorylation. Small up and down
which leads to the cytoplasmic translocation of GCK; glucose also allosterically arrows indicate an increase or decrease, respectively, in protein level or activity.
inhibits glycogen phosphorylase. The direct pathway of glycogen synthesis GLUT2, glucose transporter 2; pSer15, phosphorylated Ser15.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

liver-specific deletions of the genes that encode AKT glycogen synthesis, which demonstrates the necessity of
(Akt1//Akt2/) is normalized by the overexpression of AKT for insulin-dependent stimulation of glycogen syn-
Gck 144. Conversely, patients with maturity onset diabe- thesis; however, Gsk3Ser21Ala/Gsk3Ser9Ala mice displayed
tes mellitus of the young type2 (MODY2) which is normal hepatic glycogen metabolism, suggesting that
caused by lossoffunction mutations in GCK have inhibitory phosphorylation of GSK3 by AKT is unnec-
reduced hepatic glycogen synthesis in the postprandial essary for glycogen synthesis156. The mechanism for the
state, which is attributable to both the loss of hepatic GCK insulin-dependent activation of GYS2 might involve
activity and a reduction in pancreatic insulin secretion145. the activation of phosphodiesterase 3B (PDE3B) by AKT,
GCK activity is largely regulated by its subcellular locali- which antagonizes the cAMP-mediated phosphorylation
zation. Glucokinase regulatory protein (GKRP) binds to of GYS2 (REF.157) In addition, this process might involve
GCK in the nucleus when cytoplasmic concentrations the dephosphorylation of GYS2 by protein phosphatase 1
of glucose are low 146 (FIG.2a). Glucose induces the disso- (PP1), which involves targeting subunits of PP1,
ciation of the GCKGKRP complex, thus enabling the especially GL (REF.158) (FIG.2b).
translocation of GCK to the cytosol, where it is active147
Glycogen phosphorylase and glycogenolysis. The
(FIG.2b). Insulin facilitates this glucose-induced dissoci-
ation of the GCKGKRP complex and upregulates the stimulation of hepatic glycogenolysis during fast-
expression of Gck in rat hepatocytes through unclear ing or adrenergic stimulation occurs largely through
AKT-dependent mechanisms147. Conversely, glucagon well-characterized pathways downstream of cAMP
inhibits the dissociation of the GCKGKRP complex and PKA signalling; both GYS2 and glycogen phospho-
represses Gck expression in rodents147. GCK exerts a high rylase are phosphorylated by PKA, which results in
degree of metabolic control over glycogen synthesis, and the coordinated inhibition of GYS2 and activation of
only small increases in cytosolic free GCK are required glycogen phosphorylase. This elegant reciprocal control
to stimulate glycogen synthesis148. of glycogen synthesis and glycogenolysis is mediated by
the GL targeting subunit of PP1, which regulates both
Glycogen synthase and glycogenesis. Although GCK GYS2 and glycogen phosphorylase through dephos-
has a high metabolic control coefficient for glycogen phorylation. The phosphorylated active form of gly-
synthesis in the hepatocyte, liver glycogen synthase cogen phosphorylase binds to and inhibits PP1GL,
(GYS2) also shares control147. Indeed, the concomi- preventing PP1dependent dephosphorylation and
tant overexpression of both Gck and Gys2 in mouse activation ofGYS2 (REF.159); this process prevents futile
models enhanced hepatocellular glycogen synthesis glycogen cycling (FIG.2a). Glucose also regulates glyco-
more than the overexpression of either gene alone141. gen phosphorylase activity by allosterically binding to
This shared control of glycogen synthesis is probably glycogen phosphorylase, which stabilizes a conforma-
partly attributable to the GCK-dependent production of tion that enables dephosphorylation and inactivation of
glucose6phosphate, which is the key allosteric activa- the enzyme147,160. In this way, glycogen phosphorylase
tor of GYS2 (REFS141,149). Mice than have an Arg582Ala senses plasma levels of glucose to ensure that glycog-
mutation in GYS2, which renders the enzyme insen- enolysis is halted when glucose is abundant in plasma
sitive to allosteric activation by glucose6phosphate, (FIG.2b). Interestingly, a prediction of this model is that
displayed severely impaired hepatic glycogen synthesis hyperglycaemia should lead to the dephosphorylation
and unstable GYS2, which highlights the essential role of glycogen phosphorylase, the release of PP1GL and
of allostery in mediating the activity of GYS2 (REF.150). the consequent dephosphorylation and activation of
GYS2 is also regulated by phosphorylation at seven GYS2; however, in humans, when the release of insulin
sites; phosphorylated GYS2 has a decreased maximum and glucagon was prevented by somatostatin infusion,
reaction rate (Vmax)151 (FIG.2a). This mode of regulation hyperglycaemia could block hepatic glycogenolysis but
of GYS2 is complex, as it can be phosphorylated by it could not stimulate hepatic glycogen synthesis10. This
PKA, PKC isoforms, AMPK, glycogen synthase kinase 3 finding suggests that the role of insulin in stimulating
(GSK3) and possibly other kinases151. However, site- hepatic glycogen synthesis extends beyond its ability
directed mutagenesis studies indicate that the most to promote the PDE-mediated attenuation of glucagon
important phosphorylation site for GYS2 activity is signalling. Therefore, further research is required to elu-
Ser7, which is a substrate of AMPK and PKA152,153; mice cidate the complete mechanistic basis for the regulation
with adenoviral overexpression of GYS2 with a Ser7Ala of hepatic glycogen synthesis by insulin and glucagon.
mutation displayed increased levels of liver glycogen
under both fasting and fed conditions154. These mice Understanding hepatic insulin action
had markedly improved glucose tolerance and lower A clinically and therapeutically relevant perspective on
plasma levels of glucose in the fed state but not in the hepatic glucose metabolism requires an understanding
fasted state, which highlights the crucial role of hepatic of how hepatic glucose fluxes fit together in states of
glycogen synthesis in postprandial glucose disposal154. health and disease. On the basis of data described in
Insulin, which is essential for hepatic glycogen synthase this Review, we attempt to construct a unified frame-
flux invivo10, was long thought to act through the AKT- work to describe the control of hepatic glucose metab-
dependent inhibition of GSK3, which would favour olism by insulin, both in healthy physiological states
the dephosphorylation of GYS2 (REF.155). In fasting and in the state of metabolic dysfunction that causes
refeeding studies, Akt2/ mice had impaired hepatic T2DM (FIG.3).

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 9



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

The most important aspect of this framework is to response to insulin as a direct effect that is mediated by
distinguish the direct (cell intrinsic) effects of insulin on the activation of the INSR in hepatocytes. This is a sim-
hepatocytes from the indirect (cell non-autonomous) plification: for example, insulin also alters the phospho-
effects: that is, to regard the acute suppression of hepatic rylation status of several gluconeogenic and glycolytic
gluconeogenesis in response to insulin as primarily an enzymes (as described above), and suppresses lipolysis in
indirect effect that is mediated by the insulin-dependent hepatocytes. However, as discussed above, physiological
inhibition of adipocyte lipolysis, and to regard the acute data indicate that these mechanisms contribute relatively
stimulation of net hepatic glycogen metabolism in little to the insulin-mediated suppression of gluconeo-
genesis compared with the insulin-mediated inhibition
of adipose lipolysis. Insulin-mediated suppression of
adipose lipolysis suppresses hepatic gluconeogenesis
a Normal hepatic insulin action
through two main mechanisms: first, it decreases the
delivery of NEFA to the liver, which results in an acute
reduction in hepatic mitochondrial acetyl-CoA levels
and a consequent decrease in pyruvate carboxylase activ-
ity; second, it decreases the turnover of glycerol, which
decreases hepatic gluconeogenesis from this substrate.
Concomitantly, the direct action of insulin in the liver,

INSR Glycogen Figure 3 | Framework for understanding the


FOXO1 GCK insulin-dependent regulation of hepatic glucose
synthase
metabolism. a|Under normal physiological conditions,
Slow the direct actions of hepatocellular insulin (dark blue
Glycogen boxes) primarily facilitate net hepatic glycogen synthesis
Lipolysis Gluconeogenesis
synthesis through the activation of glucokinase (GCK) and glycogen
synthase. Insulin receptor (INSR)-dependent inactivation of
PC activity Forkhead box O1 (FOXO1) decreases the transcription
White adipose tissue of gluconeogenic genes, but this is a relatively slow
mechanism that does not mediate the acute suppression
Acetyl-CoA
of hepatic gluconeogenesis by insulin. Suppression of
lipolysis in white adipose tissue (WAT), which is an indirect
Glycerol HGP mechanism of hepatic insulin action (green boxes), acutely
suppresses hepatic gluconeogenesis by decreasing the
Hepatocyte
delivery of nonesterified fatty acids (NEFA) and glycerol to
the liver, which results in reduced acetyl-CoA-dependent
activation of pyruvate carboxylase (PC), and decreased
b Hepatic insulin action in type 2 diabetes mellitus gluconeogenesis from glycerol. The direct stimulation
of net hepatic glycogen synthesis and the indirect
suppression of hepatic gluconeogenesis collectively
suppress hepatic glucose production (HGP). b|In type2
PKC diabetes mellitus (T2DM), the direct (dark blue boxes) and
indirect (green boxes) effects of insulin are impaired
P through different mechanisms. Lipid-induced hepatic
DAG Thr1160 insulin resistance increases hepatic levels of diacylglycerol
DAG
(DAG), which results in the activation of protein kinase C
Hepatic lipid (PKC) and thereby impairs direct hepatic insulin signalling
accumulation through PKC-dependent phosphorylation of INSR at
Thr1160. This inhibits the INSR-dependent stimulation of
FOXO1 GCK
Glycogen hepatic glycogen synthesis in response to insulin. Impaired
synthase INSR signalling is indicated by grey arrows. In WAT,
macrophage activation and consequent inflammatory
Lipolysis Gluconeogenesis signalling, as well as intrinsic adipocyte dysfunction,
Glycogen increase lipolysis and promote adipocyte insulin resistance,
synthesis resulting in the continued delivery of NEFA and glycerol to
PC activity
the liver despite high plasma concentrations of insulin.
Continued delivery of NEFA and glycerol to the liver
Acetyl-CoA promotes hepatic lipid accumulation and PKC-mediated
hepatic insulin resistance, and promotes gluconeogenesis
Activated
macrophage
by increasing hepatic acetyl-CoA and gluconeogenesis
Glycerol HGP
from glycerol. Impaired net hepatic glycogen synthesis and
unrestrained hepatic gluconeogenesis together lead to
increases in HGP. Small up and down arrows indicate an
increase or decrease, respectively, in protein level or
Indirect insulin action Direct insulin action activity. DAG, diacylglycerol.
Nature Reviews | Endocrinology

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

which occurs through insulin receptor signalling, stim- for example, it is focused on the acute effects of insulin
ulates hepatic glycogen synthesis, mostly through the and does not account for the many relevant transcrip-
activation of both GCK and glycogen synthase. As noted, tional mechanisms that are discussed above, which prob-
maximal net hepatic glycogen synthesis also requires ably participate in the chronic control of hepatic glucose
hyperglycaemia to inhibit glycogen phosphorylase in a metabolism. However, we believe that this framework
glucose-dependentmanner. might prove to be a useful heuristic tool for scientists and
This framework can be adapted to understand the clinicians who wish to understand and further probe the
physiological basis of impaired hepatic glucose metabo- complexities of hepatic glucose metabolism in physiolog-
lism in T2DM. Lipid-induced hepatic insulin resistance, ical states of health and disease, especially those who seek
which involves diacylglycerol-dependent activation to interpret hyperinsulinaemic clamp data in rodents.
of PKC and the resulting inhibitory phosphorylation of The framework also has therapeutic implications, which
INSR at Thr1160, would be primarily expected to impair we considerbelow.
insulin-stimulated hepatic glycogen synthesis33. However,
lipid-induced hepatocellular insulin resistance would not Targeting hepatic glucose production
be expected to substantially alter the acute suppression of The dysregulated hepatic glucose metabolism in indi-
hepatic gluconeogenesis by insulin because that process viduals with T2DM is an attractive therapeutic target.
is mostly controlled indirectly. Rather, impaired insulin- Three major pathophysiological mechanisms that can
dependent suppression of hepatic gluconeogenesis would be targeted include the excessive action of glucagon,
be expected to result from adipocyte insulin resistance; lipid-induced hepatic insulin resistance and excess adi-
that is, an inability of the adipocyte to suppress lipolysis pose lipolysis. Hyperglucagonaemia is a hallmark of
following insulin stimulation. Although the mechanisms both T1DM and T2DM, and antagonizing the action
of adipose insulin resistance are incompletely under- of glucagon has been remarkably effective at decreasing
stood, and are beyond the scope of this Review, they prob- hyperglycaemia in diverse rodent models of diabetes
ably involve macrophage activation and inflammatory mellitus164. Similarly, the case for targeting lipid-induced
signalling, and intrinsic adipocyte defects161,162. hepatic insulin resistance is well supported. Weight
Inflammatory signalling might also drive adipose loss following lifestyle modification improves, or even
lipolysis independent of insulin signalling and might resolves, T2DM in humans, in part, by reversing lipid-
therefore have implications for understanding the pro- induced hepatic insulin resistance165. A study that exam-
gression of metabolic disease from subclinical insulin ined the mechanism that underlies these improvements
resistance to overt T2DM. The development of NAFLD is demonstrated that modest weight loss (~8kg) led to the
an early step in the progression of metabolic disease; most resolution of NAFLD and the normalization of fasting
individuals with NAFLD do not have T2DM, although plasma concentrations of glucose. The improvement
the majority of individuals with both obesity and T2DM in plasma glucose was attributed to decreases in rates of
have NAFLD163. Lipid-induced hepatic insulin resistance, fasting HGP and gluconeogenesis, and increased insulin-
but not necessarily inflammation-associated adipose mediated suppression of HGP22. The mechanism by
insulin resistance, accompanies NAFLD. Accordingly, which the resolution of NAFLD improves the dysregu-
lipid-induced hepatic insulin resistance might precede lated hepatic glucose metabolism that is associated with
impairments in the insulin-mediated suppression of T2DM might involve the reversal of INSR inhibition
gluconeogenesis. Individuals who have isolated hepatic through the inactivation of the diacylglycerolPKC
insulin resistance (that is, without adipose insulin axis33, and reductions in hepatic acetyl-CoA content that
resistance) would be expected to display fasting eug- result in decreased activity of pyruvate carboxylase45,52.
lycaemia, and might even show normal suppression of The American Diabetes Association (ADA) recommends
HGP in hyperinsulinaemiceuglycaemic clamp studies, a target of 5% weight loss through lifestyle interventions
owing to the fact that fasted glycogen-depleted indi- for individuals with obesity and diabetes mellitus who are
viduals are reliant on gluconeogenesis for the majority ready to lose weight166. However, readiness to lose weight
of HGP. We hypothesize that progression to the state of is often elusive; the multitude of physiological, societal
fasting hyperglycaemia that defines T2DM involves and psychosocial barriers to weight loss frequently ren-
inflammation-associated lipolysis and associated adi- der even a 5% long-term weight loss target by lifestyle
pose insulin resistance (in addition to cell dysfunction). intervention elusive. Therefore, searching for additional
Importantly, there are likely to be interrelations between therapies that target hepatic steatosis and lipid-induced
the direct and indirect modes of control of hepatic glu- hepatic insulin resistance is warranted (FIG.4).
cose metabolism. For example, the indirect regulation
of hepatic NEFA delivery by adipose lipolysis might drive Glucagon antagonism and HGP. The antagonism of cir-
hepatic lipid accumulation and therefore impair direct culating glucagon or of GCGR to reduce excess HGP is
hepatocellular insulinaction. a long-standing and active area of investigation42,167170.
This framework incorporates observations from Plasma levels of glucagon are aberrantly increased in
human, dog and rodent studies. It is heavily depend- individuals with T2DM171174. In addition, animal mod-
ent on insights from tracer studies of the integrated els of reduced or ablated glucagon action show improve-
physiology of insulin action invivo, without which ments in T2DM96,175,176 (FIG.4a,b). Although the complete
the indirect effects of insulin would not have been elu- prevention of T2DM observed in some of these rodent
cidated. Importantly, this framework is incomplete; models contrasts with the insulin dependence observed

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 11



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Untreated b Treated
DAG Mixed agonist of GCGR,
INSR PKC GIPR and GLP-1R

Intrahepatic lipid Mitochondrion


Thr1160 DAG
P

Intrahepatic lipid Intrahepatic lipid Liver-targeted


uncoupler
Hepatic Gluconeogenesis Hepatic Gluconeogenesis
insulin insulin
resistance sensitivity Energy
PC activity PC activity expenditure

Acetyl-CoA HGP Acetyl-CoA


HGP

Muscle insulin Intrahepatic Muscle insulin


resistance lipid VLDL export sensitivity
Glucagon
GCGR antagonist
Intramyocellular lipid Intramyocellular lipid
TZD
PPAR
NEFA
Glycerol Lipid storage NEFA
Glycerol
TAG
Lipolysis GPR109A
TAG
agonist
Lipolysis

White adipose tissue

Figure 4 | Therapeutic opportunities for dysregulated hepatic glucose Nature Reviews


(GCGR), gastric inhibitory polypeptide receptor | Endocrinology
(GIPR) and glucagon-like
metabolism. a|In patients with type2 diabetes mellitus (T2DM), peptide 1 receptor (GLP1R)) to promote fat oxidation. A second
lipid-induced hepatic insulin resistance might result from activation of the therapeutic strategy is to target excessive lipolysis (light green ovals) by
diacylglycerol (DAG)protein kinase C (PKC) axis and the consequent promoting the sequestration of lipids in white adipose tissue (WAT). The
inhibition of insulin receptor (INSR) signalling through inhibitory inhibition of lipolysis or the stimulation of lipogenesis in WAT is predicted
phosphorylation of INSR at Thr1160. This leads to impaired insulin to decrease hepatic lipid accumulation and reverse lipid-induced hepatic
stimulation of hepatic glycogen synthesis. In parallel, inappropriate insulin resistance by decreasing delivery of lipids to the liver. The
increases in adipose lipolysis can drive hepatic gluconeogenesis thiazolidinediones (TZD) are agonists of the lipogenic transcription factor
through increases in hepatic acetyl-CoA and pyruvate carboxylase (PC) peroxisome proliferator-activated receptor- (PPAR). The inhibition of
activity, and promote ectopic lipid accumulation in liver and muscle. These lipolysis with GPR109A agonists, or with WAT-specific adipose triglyceride
processes promote the increased hepatic glucose production (HGP) that lipase (ATGL) inhibitors, would also be predicted to reverse dysregulated
occurs in T2DM. b|These contributors to increased HGP can be glucose metabolism through these mechanisms. Glucagon antagonism
pharmacologically targeted. Mechanisms that target lipid-induced (red oval) represents a third potential therapeutic strategy to target the
hepatic insulin resistance (purple ovals) include the selective induction of hyperglucagonaemia and excessive hepatic glucose production
mitochondrial uncoupling in the liver (for example, with 2,4dinitrophenol associated with T2DM if it can be dissociated from on-target adverse
(DNP) analogues, such as DNP-methyl ether (DNPME) and other effects (for example, hepatic steatosis). Small up and down arrows
liver-targeted mitochondrial uncoupling agents) to increase hepatic fat indicate an increase or decrease, respectively, in protein level or activity.
oxidation, or mixed agonism of incretin, glucagon and/or thyroid The grey arrow depicts inhibition of lipolysis. ACC, acetyl-CoA
hormone receptors (for example, a mixed agonist of glucagon receptor carboxylase; TAG, triacylglycerol.

in humans with pancreoprivic diabetes mellitus, the to activate peroxisome proliferator-activated receptor-
marked antidiabetic phenotypes underscore the prom- (PPAR), which results in an increase in hepatic fatty
ise of such anti-glucagon therapeutics. Currently, data acid oxidation and a decrease in the export of VLDL181.
in humans is limited to three GCGR antagonists, BAY It is unclear whether this aspect of glucagon action in
279955 (REF.177), MK0893 (REF.178) and LY2409021 rodents translates to human physiology; however, hyper
(REFS 179,180) . BAY 279955 inhibited glucagon- triglyceridaemia has not been reported thus far in the
stimulated glucose production in humans177, but no human studies of LY2409021 (REFS179,180).
followup studies were published. MK0893 improved
hyperglycaemia in a human clinical trial, but was associ- Hepatosteatosis and T2DM. A rational therapeutic
Pancreoprivic diabetes ated with increases in plasma levels of LDL cholesterol178. approach for the treatment of NAFLD and its sequelae,
mellitus In phaseI and phase II trials, LY2409021 improved such as NASH, is to increase hepatic energy expendi-
Diabetes mellitus caused by glycaemic control in patients with T2DM, which was ture and thereby increase hepatic fat oxidation. Thyroid
medical or surgical loss of
pancreatic function, such as
complicated only by a modest and reversible increase in hormone receptor agonists have been tested preclini-
after a pancreatectomy or the levels of transaminases179,180. One potential caveat of cally for their ability achieve this and, although they can
pancreatitis. anti-glucagon therapies is that, in rodents, glucagon seems reverse NAFLD, none has been shown to improve the

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

suppression of HGP by insulin (in part due to increased were associated with an increase in extramyocellular lipid
adipose lipolysis)182,183. In addition, a T3glucagon con- content and an increase in the sensitivity of peripheral
jugate improved glycaemia and NAFLD, without caus- adipocytes to the inhibitory effects of insulin on lipoly-
ing the rapid development of cardiac hypertrophy and sis188. These results led to the hypothesis that thiazolidin-
osteopenia observed with T3 alone184; additional studies ediones enhance insulin sensitivity in patients with T2DM
to evaluate the safety and effects of this conjugate on by promoting increased insulin sensitivity in peripheral
HGP (which was not evaluated in this study 184) will help adipocytes, resulting in lower plasma concentrations of
to assess the promise of this class of compounds. fatty acids and the redistribution of intracellular lipids
Liver-targeted mitochondrial uncouplers, which from insulin-responsive organs into peripheral adipo-
can dissipate the inner mitochondrial membrane pro- cytes (FIG.4a,b). Consistent with these results, in patients
ton gradient in the liver, are another promising class of with NASH who had either impaired glucose tolerance
agents for the treatment of NAFLD, NASH and diabe- or overt T2DM, and who were fed a hypocaloric diet, a
tes mellitus. Two modified forms of the mitochondrial 6month treatment regimen with the thiazolidinedione
protonophore 2,4dinitrophenol (DNP), DNP-methyl pioglitazone improved hepatic insulin sensitivity and
ether (DNPME) and controlled-release mitochondrial hepatic steatosis compared with placebo189. In patients
protonophore (CRMP), reverse diabetes mellitus, hepatic with NASH, pioglitazone-mediated improvements in
inflammation and hepatic fibrosis in rodent models of hepatic steatosis and histological NASH were correlated
T2DM and NASH24,25. Importantly, the reversal of hyper- with improvements in adipose insulin responsiveness190.
glycaemia and hepatic insulin resistance by these drugs However, the clinical use of thiazolidinediones is limited
was attributed to decreases in diacylglycerol-dependent by concerns regarding adverse effects. For pioglitazone,
activation of PKC and decreases in hepatic acetyl-CoA the most widely prescribed thiazolidinedione compound
content 24,25 (FIG.4a,b). Both of these liver-targeted mito-
in the United States, adverse effects include weight gain
chondrial uncoupling agents promoted increased energy (in an already obese population), peripheral oedema,
expenditure exclusively in liver, and, importantly, the exacerbation of heart failure and increased frequency of
reversal of diabetes mellitus and NASH in these rodent bone fractures191. Thus, although pioglitazone effectively
models was not associated with detectable changes in improves adipose function, decreases excess lipolysis
body temperature, whole-body energy expenditure and improves insulin sensitivity, ample opportunities exist
or body weight, which minimizes the concerns of toxic- for development of better-tolerated therapies that target
ity that are inherent to all non-selective mitochondrial adipose tissue function inT2DM.
uncouplers24,25. These studies provide a proof of concept Direct inhibitors of lipolytic enzymes, such as adipose
for the development of novel liver-targeted mitochondrial triglyceride lipase (ATGL) and hormone-sensitive lipase
protonophores to treat NAFLD, NASH andT2DM. (HSL), are not used in clinical practice. However, niacin
Incretin receptor agonists, and in particular glucagon-
(or nicotinic acid) a drug that has been used for dec-
like peptide 1 (GLP1) analogues, are used in the treat- ades to increase HDL levels and decrease triglyceride
ment of diabetes mellitus and obesity, and might be levels also inhibits adipose lipolysis. Niacin binds
useful for the treatment of NAFLD. In a phaseII clinical to, and activates, a small G protein-coupled receptor,
trial in patients with NASH, the histology of NASH was GPR109A, which decreases intracellular levels of cAMP,
resolved more frequently in patients who were treated leading to a decrease in the activity of HSL in adipose tis-
with the GLP1 analogue liraglutide than in patients who sue and reduced adipocyte lipolysis192. However, rather
were treated with placebo185. In preclinical studies, a novel
than improving endogenous glucose production, nia-
compound agonist of the GLP1, gastric inhibitory pep- cin can increase plasma glucose concentrations in some
tide (GIP) and glucagon receptors improved glycaemic patients193,194. This paradoxical effect could be explained
control, decreased food intake, increased energy expend- by a rebound increase in adipose lipolysis following dis-
iture and reversed hepatosteatosis in rodents186. This sociation of the niacinGPR109A complex 195. Changes
strategy of mixed agonism, also used in the aforemen- in niacin dosing strategy might prevent this rebound
tioned glucagonT3 conjugate, might help to minimize increase in plasma levels of NEFA and might improve
the undesired effects of any one hormone (for example, glycaemic control195. Thus, novel formulations of niacin
glucagon-mediated hyperglycaemia and T3-mediated with different pharmacokinetic profiles, novel GPR109A
tachycardia), and thus warrants further exploration184. agonists or other anti-lipolytics (for example, WAT-specific
ATGL inhibitors) might indirectly improve hepatic glucose
Targeting the adipocyte. Thiazolidinediones, the best- metabolism and therefore merit further investigation.
established pharmacological agents for the treatment for
NAFLD and NASH, are PPAR agonists that have mul- Conclusions
tiple downstream effects. Thiazolidinediones increase Well-coordinated hepatic glucose metabolism is essential
adipose function and adipose mass by increasing their to health, and dysregulation of hepatic glucose metabo-
capacity for lipid storage and decreasing inappropriate lism is central to the pathogenesis and complications of
lipolysis187. The treatment of individuals with T2DM with T2DM. The key components of HGP reviewed in this arti-
the thiazolidinedione rosiglitazone markedly improved cle include gluconeogenesis, which exhibits complex reg-
insulin-stimulated glucose metabolism, which was asso- ulation by substrate provision, redox state, gluconeogenic
ciated with decreased plasma concentrations of fatty acids gene transcription and other mechanisms (FIG.1), and
and lowered hepatic triglyceride content188. These changes glycogen metabolism, which is remarkably sensitive to

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 13



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

both hormonal tone and plasma concentrations of glu- a liver-targeted mitochondrial protonophore is used to
cose (FIG.2). We have highlighted the importance of indi- reverse lipid-induced hepatic insulin resistance, a gluca-
rect extrahepatic control of gluconeogenesis and of direct gon antagonist is used to neutralize the excessive HGP
hepatic control of glycogen metabolism, and have pro- that results from hyperglucagonaemia and a thiazolidin-
posed a paradigm in which dysregulation of both direct edione or antilipolytic agent is used to reduce lipolytic
and indirect mechanisms contribute to the increased stimulation of hepatic gluconeogenesis (FIG.4). It is also
HGP of T2DM (FIG. 3). The multifaceted regulation conceivable that reducing hepatic diacylglycerol and
of HGP through both direct and indirect mechanisms lim- acetyl-CoA content using liver-targeted mitochondrial
its the use of invitro hepatocyte models that are removed uncoupling agents alone might be sufficient to reverse
from the normal hormonal, anatomical and biochemi- both hepatic insulin resistance and increased rates of
cal context of the insitu liver. Although much remains hepatic gluconeogenesis in T2DM, as shown in preclinical
to be elucidated, current knowledge of the multimodal studies24,25. Although many preclinical and clinical studies
control of HGP invites rational therapeutic targeting of will be required to test the viability of these novel agents
multiple mechanisms simultaneously. Thus, it is conceiv- and combinatorial approaches, the global need for effec-
able to imagine a future in which metformin is used to tive medical management of dysregulated HGP in T2DM
control redox-dependent regulation of gluconeogenesis, provides an urgent impetus for suchwork.

1. Ekberg,K. etal. Contributions by kidney and liver to 18. Basu,A., Shah,P., Nielsen,M., Basu,R. & 35. Williams,A.S. etal. Integrin-linked kinase is
glucose production in the postabsorptive state and Rizza,R.A. Effects of type2 diabetes on the necessary for the development of diet-induced hepatic
after 60h of fasting. Diabetes 48, 292298 (1999). regulation of hepatic glucose metabolism. insulin resistance. Diabetes 66, 325334 (2017).
2. Moore,M.C., Coate,K.C., Winnick,J.J., An,Z. & J.Investig. Med. 52, 366374 (2004). 36. Boden,G. Gluconeogenesis and glycogenolysis in health
Cherrington,A.D. Regulation of hepatic glucose uptake 19. Magnusson,I., Rothman,D.L., Katz,L.D., and diabetes. J.Investig. Med. 52, 375378 (2004).
and storage invivo. Adv. Nutr. 3, 286294 (2012). Shulman,R.G. & Shulman,G.I. Increased rate of 37. Chen,X., Iqbal,N. & Boden,G. The effects of free fatty
3. Rizza,R.A. Pathogenesis of fasting and postprandial gluconeogenesis in typeII diabetes mellitus. A 13C acids on gluconeogenesis and glycogenolysis in normal
hyperglycemia in type2 diabetes: implications for nuclear magnetic resonance study. J.Clin. Invest. 90, subjects. J.Clin. Invest. 103, 365372 (1999).
therapy. Diabetes 59, 26972707 (2010). 13231327 (1992). 38. Katz,J. & Tayek,J.A. Gluconeogenesis and the Cori
4. Moore,M.C. etal. Sources of carbon for hepatic 20. Perry,R.J., Samuel,V.T., Petersen,K.F. & cycle in 12-, 20-, and 40hfasted humans.
glycogen synthesis in the conscious dog. J.Clin. Invest. Shulman,G.I. The role of hepatic lipids in hepatic Am. J.Physiol. 275, E537E542 (1998).
88, 578587 (1991). insulin resistance and type2 diabetes. Nature 510, 39. Landau,B.R. etal. Contributions of gluconeogenesis
5. Rothman,D.L., Magnusson,I., Katz,L.D., 8491 (2014). to glucose production in the fasted state. J.Clin.
Shulman,R.G. & Shulman,G.I. Quantitation of hepatic 21. Kumashiro,N. etal. Cellular mechanism of insulin Invest. 98, 378385 (1996).
glycogenolysis and gluconeogenesis in fasting humans resistance in nonalcoholic fatty liver disease. 40. Levine,R. & Fritz,I.B. The relation of insulin to liver
with 13C NMR. Science 254, 573576 (1991). Proc. Natl Acad. Sci. USA 108, 1638116385 (2011). metabolism. Diabetes 5, 209219 (1956).
6. Mari,A., Wahren,J., DeFronzo,R.A. & Ferrannini,E. 22. Petersen,K.F. etal. Reversal of nonalcoholic hepatic 41. Gaisano,H., MacDonald,P.E. & Vranic,M. Glucagon
Glucose absorption and production following oral steatosis, hepatic insulin resistance, and hyperglycemia secretion and signaling in the development of
glucose: comparison of compartmental and by moderate weight reduction in patients with type2 diabetes. Front. Physiol. 3, 349 (2012).
arteriovenous-difference methods. Metabolism 43, diabetes. Diabetes 54, 603608 (2005). 42. Pearson,M.J., Unger,R.H. & Holland,W.L. Clinical
14191425 (1994). 23. Petersen,K.F. etal. Leptin reverses insulin resistance trials, triumphs, and tribulations of glucagon receptor
7. Ishida,T. etal. Differential effects of oral, peripheral and hepatic steatosis in patients with severe antagonists. Diabetes Care 39, 10751077 (2016).
intravenous, and intraportal glucose on hepatic lipodystrophy. J.Clin. Invest. 109, 13451350 (2002). 43. Ader,M. & Bergman,R.N. Peripheral effects of insulin
glucose uptake and insulin and glucagon extraction in 24. Perry,R.J., Zhang,D., Zhang,X.M., Boyer,J.L. & dominate suppression of fasting hepatic glucose
conscious dogs. J.Clin. Invest. 72, 590601 (1983). Shulman,G.I. Controlled-release mitochondrial production. Am. J.Physiol. 258, E1020E1032 (1990).
8. Pagliassotti,M.J. & Cherrington,A.D. Regulation of protonophore reverses diabetes and steatohepatitis in 44. Lewis,G.F., Zinman,B., Groenewoud,Y., Vranic,M. &
net hepatic glucose uptake invivo. Annu. Rev. Physiol. rats. Science 347, 12531256 (2015). Giacca,A. Hepatic glucose production is regulated
54, 847860 (1992). 25. Perry,R.J. etal. Reversal of hypertriglyceridemia, both by direct hepatic and extrahepatic effects of
9. Ferrannini,E. etal. The disposal of an oral glucose fatty liver disease, and insulin resistance by a liver- insulin in humans. Diabetes 45, 454462 (1996).
load in healthy subjects. A quantitative study. targeted mitochondrial uncoupler. Cell Metab. 18, 45. Sindelar,D.K., Balcom,J.H., Chu,C.A., Neal,D.W.
Diabetes 34, 580588 (1985). 740748 (2013). & Cherrington,A.D. A comparison of the effects of
10. Petersen,K.F., Laurent,D., Rothman,D.L., 26. Kim,J.K., Gavrilova,O., Chen,Y., Reitman,M.L. & selective increases in peripheral or portal insulin on
Cline,G.W. & Shulman,G.I. Mechanism by which Shulman,G.I. Mechanism of insulin resistance in hepatic glucose production in the conscious dog.
glucose and insulin inhibit net hepatic glycogenolysis AZIP/F-1 fatless mice. J.Biol. Chem. 275, Diabetes 45, 15941604 (1996).
in humans. J.Clin. Invest. 101, 12031209 (1998). 84568460 (2000). 46. Prager,R., Wallace,P. & Olefsky,J.M. Direct and
11. Lin,H.V. & Accili,D. Hormonal regulation of hepatic 27. Samuel,V.T. & Shulman,G.I. Mechanisms for insulin indirect effects of insulin to inhibit hepatic glucose
glucose production in health and disease. Cell Metab. resistance: common threads and missing links. Cell output in obese subjects. Diabetes 36, 607611
14, 919 (2011). 148, 852871 (2012). (1987).
12. Cherrington,A.D., Edgerton,D. & Sindelar,D.K. The 28. Magkos,F. etal. Intrahepatic diacylglycerol content is 47. Staehr,P. etal. Effects of free fatty acids per se on
direct and indirect effects of insulin on hepatic glucose associated with hepatic insulin resistance in obese glucose production, gluconeogenesis, and
production invivo. Diabetologia 41, 987996 (1998). subjects. Gastroenterology 142, 14441446.e2 glycogenolysis. Diabetes 52, 260267 (2003).
13. Pagliassotti,M.J., Moore,M.C., Neal,D.W. & (2012). 48. Lewis,G.F., Vranic,M., Harley,P. & Giacca,A. Fatty
Cherrington,A.D. Insulin is required for the liver to 29. Luukkonen,P.K. etal. Hepatic ceramides dissociate acids mediate the acute extrahepatic effects of insulin
respond to intraportal glucose delivery in the steatosis and insulin resistance in patients with non- on hepatic glucose production in humans. Diabetes
conscious dog. Diabetes 41, 12471256 (1992). alcoholic fatty liver disease. J.Hepatol. 64, 46, 11111119 (1997).
14. McGuinness,O.P., Ayala,J.E., Laughlin,M.R. & 11671175 (2016). 49. Sindelar,D.K. etal. The role of fatty acids in
Wasserman,D.H. NIH experiment in centralized 30. ter Horst,K.W. etal. Hepatic diacylglycerol- mediating the effects of peripheral insulin on hepatic
mouse phenotyping: the Vanderbilt experience and associated protein kinase C translocation links glucose production in the conscious dog. Diabetes 46,
recommendations for evaluating glucose homeostasis hepatic steatosis to hepatic insulin resistance in 187196 (1997).
in the mouse. Am. J.Physiol. Endocrinol. Metab. 297, humans. Cell Rep. 19, 19972004 (2017). 50. Rebrin,K., Steil,G.M., Mittelman,S.D. &
E849E855 (2009). 31. Samuel,V.T. etal. Inhibition of protein kinase C Bergman,R.N. Causal linkage between insulin
15. Kowalski,G.M. & Bruce,C.R. The regulation of prevents hepatic insulin resistance in nonalcoholic fatty suppression of lipolysis and suppression of liver glucose
glucose metabolism: implications and considerations liver disease. J.Clin. Invest. 117, 739745 (2007). output in dogs. J.Clin. Invest. 98, 741749 (1996).
for the assessment of glucose homeostasis in rodents. 32. Samuel,V.T. etal. Mechanism of hepatic insulin 51. Perry,R.J. etal. Hepatic acetyl coa links adipose
Am. J.Physiol. Endocrinol. Metab. 307, E859E871 resistance in non-alcoholic fatty liver disease. J.Biol. tissue inflammation to hepatic insulin resistance and
(2014). Chem. 279, 3234532353 (2004). type2 diabetes. Cell 160, 745758 (2015).
16. Steele,R. etal. Inhibition by insulin of hepatic glucose 33. Petersen,M.C. etal. Insulin receptor Thr1160 52. Perry,R.J. etal. Leptin reverses diabetes by
production in the normal dog. Am. J.Physiol. 208, phosphorylation mediates lipid-induced hepatic suppression of the hypothalamicpituitaryadrenal
301306 (1965). insulin resistance. J.Clin. Invest. 126, 43614371 axis. Nat. Med. 20, 759763 (2014).
17. Rizza,R.A., Mandarino,L.J. & Gerich,J.E. (2016). 53. Perry,R.J., Peng,L. & Shulman,G.I. Mechanism for
Doseresponse characteristics for effects of insulin on 34. Michelotti,G.A., Machado,M.V. & Diehl,A.M. leptins acute insulin-independent effect to reverse
production and utilization of glucose in man. NAFLD, NASH and liver cancer. Nat. Rev. diabetic ketoacidosis. J.Clin. Invest. 127, 657669
Am. J.Physiol. 240, E630E639 (1981). Gastroenterol. Hepatol. 10, 656665 (2013). (2017).

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

54. Previs,S.F., Cline,G.W. & Shulman,G.I. A critical 77. Inzucchi,S.E. etal. Efficacy and metabolic effects of 101. Romere,C. etal. Asprosin, a fasting-induced glucogenic
evaluation of mass isotopomer distribution analysis metformin and troglitazone in typeII diabetes protein hormone. Cell 165, 566579 (2016).
of gluconeogenesis invivo. Am. J.Physiol. 277, mellitus. N.Engl. J.Med. 338, 867872 (1998). 102. Edgerton,D.S. etal. Effects of insulin on the
E154E160 (1999). 78. Zhou,G. etal. Role of AMP-activated protein kinase in metabolic control of hepatic gluconeogenesis invivo.
55. Krebs,H.A., Speake,R.N. & Hems,R. Acceleration of mechanism of metformin action. J.Clin. Invest. 108, Diabetes 58, 27662775 (2009).
renal gluconeogenesis by ketone bodies and fatty 11671174 (2001). 103. Ramnanan,C.J. etal. Molecular characterization of
acids. Biochem. J. 94, 712720 (1965). 79. Shaw,R.J. etal. The kinase LKB1 mediates glucose insulin-mediated suppression of hepatic glucose
56. Keech,D.B. & Utter,M.F. Pyruvate carboxylase. II. homeostasis in liver and therapeutic effects of production invivo. Diabetes 59, 13021311 (2010).
Properties. J.Biol. Chem. 238, 26092614 (1963). metformin. Science 310, 16421646 (2005). 104. Puigserver,P. etal. Insulin-regulated hepatic
57. Williamson,J.R., Kreisberg,R.A. & Felts,P.W. 80. He,L. etal. Metformin and insulin suppress hepatic gluconeogenesis through FOXO1PGC1 interaction.
Mechanism for the stimulation of gluconeogenesis by gluconeogenesis through phosphorylation of CREB Nature 423, 550555 (2003).
fatty acids in perfused rat liver. Proc. Natl Acad. Sci. binding protein. Cell 137, 635646 (2009). 105. Yoon,J.C. etal. Control of hepatic gluconeogenesis
USA 56, 247254 (1966). 81. Fullerton,M.D. etal. Single phosphorylation sites in through the transcriptional coactivator PGC1. Nature
58. Lu,M. etal. Insulin regulates liver metabolism invivo Acc1 and Acc2 regulate lipid homeostasis and the 413, 131138 (2001).
in the absence of hepatic Akt and Foxo1. Nat. Med. insulin-sensitizing effects of metformin. Nat. Med. 19, 106. Nakae,J., Kitamura,T., Silver,D.L. & Accili,D. The
18, 388395 (2012). 16491654 (2013). forkhead transcription factor Foxo1 (Fkhr) confers
59. Buettner,C. etal. Severe impairment in liver insulin 82. Foretz,M. etal. Metformin inhibits hepatic insulin sensitivity onto glucose6phosphatase
signaling fails to alter hepatic insulin action in conscious gluconeogenesis in mice independently of the LKB1/ expression. J.Clin. Invest. 108, 13591367 (2001).
mice. J.Clin. Invest. 115, 13061313 (2005). AMPK pathway via a decrease in hepatic energy 107. Schmoll,D. etal. Regulation of glucose6phosphatase
60. Okamoto,H., Obici,S., Accili,D. & Rossetti,L. state. J.Clin. Invest. 120, 23552369 (2010). gene expression by protein kinase B and the
Restoration of liver insulin signaling in Insr knockout 83. Madiraju,A.K. etal. Metformin suppresses forkhead transcription factor FKHR. Evidence for
mice fails to normalize hepatic insulin action. J.Clin. gluconeogenesis by inhibiting mitochondrial insulin response unit-dependent and -independent
Invest. 115, 13141322 (2005). glycerophosphate dehydrogenase. Nature 510, effects of insulin on promoter activity. J.Biol. Chem.
61. Chen,Y.D., Golay,A., Swislocki,A.L. & Reaven,G.M. 542546 (2014). 275, 3632436333 (2000).
Resistance to insulin suppression of plasma free fatty 84. Cao,J. etal. Low concentrations of metformin 108. Matsumoto,M. Dual role of transcription factor FoxO1
acid concentrations and insulin stimulation of glucose suppress glucose production in hepatocytes through in controlling hepatic insulin sensitivity and lipid
uptake in noninsulin-dependent diabetes mellitus. AMP-activated protein kinase (AMPK). J.Biol. Chem. metabolism. J.Clin. Invest. 116, 24642472 (2006).
J.Clin. Endocrinol. Metab. 64, 1721 (1987). 289, 2043520446 (2014). 109. Haeusler,R.A., Kaestner,K.H. & Accili,D. FoxOs
62. Fraze,E. etal. Ambient plasma free fatty acid 85. Hawley,S.A., Gadalla,A.E., Olsen,G.S. & function synergistically to promote glucose production.
concentrations in noninsulin-dependent diabetes Hardie,D.G. The antidiabetic drug metformin J.Biol. Chem. 285, 3524535248 (2010).
mellitus: evidence for insulin resistance. J.Clin. activates the AMP-activated protein kinase cascade 110. O.Sullivan,I. etal. FoxO1 integrates direct and
Endocrinol. Metab. 61, 807811 (1985). via an adenine nucleotide-independent mechanism. indirect effects of insulin on hepatic glucose
63. Reaven,G.M., Hollenbeck,C., Jeng,C.Y., Wu,M.S. & Diabetes 51, 24202425 (2002). production and glucose utilization. Nat. Commun. 6,
Chen,Y.D. Measurement of plasma glucose, free fatty 86. Howell,J.J. etal. Metformin inhibits hepatic 7079 (2015).
acid, lactate, and insulin for 24h in patients with mTORC1 signaling via dose-dependent mechanisms 111. Titchenell,P.M., Chu,Q., Monks,B.R. &
NIDDM. Diabetes 37, 10201024 (1988). involving AMPK and the TSC complex. Cell Metab. Birnbaum,M.J. Hepatic insulin signalling is
64. Golay,A., Swislocki,A.L., Chen,Y.D. & 25, 463471 (2017). dispensable for suppression of glucose output by
Reaven,G.M. Relationships between plasma-free 87. ElMir,M.Y. etal. Dimethylbiguanide inhibits cell insulin invivo. Nat. Commun. 6, 7078 (2015).
fatty acid concentration, endogenous glucose respiration via an indirect effect targeted on the 112. Samuel,V.T. etal. Fasting hyperglycemia is not
production, and fasting hyperglycemia in normal and respiratory chain complex I. J.Biol. Chem. 275, associated with increased expression of PEPCK or
non-insulin-dependent diabetic individuals. 223228 (2000). G6Pc in patients with type2 diabetes. Proc. Natl
Metabolism 36, 692696 (1987). 88. Hardie,D.G., Ross,F.A. & Hawley,S.A. AMPK: a nutrient Acad. Sci. USA 106, 1212112126 (2009).
65. Swislocki,A.L., Chen,Y.D., Golay,A., Chang,M.O. & and energy sensor that maintains energy homeostasis. 113. Koo,S.H. etal. The CREB coactivator TORC2 is a key
Reaven,G.M. Insulin suppression of plasma-free fatty Nat. Rev. Mol. Cell Biol. 13, 251262 (2012). regulator of fasting glucose metabolism. Nature 437,
acid concentration in normal individuals and patients 89. Miller,R.A. etal. Biguanides suppress hepatic 11091111 (2005).
with type2 (non-insulin-dependent) diabetes. glucagon signalling by decreasing production of cyclic 114. Dentin,R. etal. Insulin modulates gluconeogenesis by
Diabetologia 30, 622626 (1987). AMP. Nature 494, 256260 (2013). inhibition of the coactivator TORC2. Nature 449,
66. Charles,M.A. etal. The role of non-esterified fatty 90. Konopka,A.R. etal. Hyperglucagonemia mitigates the 366369 (2007).
acids in the deterioration of glucose tolerance in effect of metformin on glucose production in 115. Wang,Y. etal. Inositol1,4,5trisphosphate receptor
Caucasian subjects: results of the Paris Prospective prediabetes. Cell Rep. 15, 13941400 (2016). regulates hepatic gluconeogenesis in fasting and
Study. Diabetologia 40, 11011106 (1997). 91. Pernicova,I. & Korbonits,M. Metformin mode of diabetes. Nature 485, 128132 (2012).
67. Paolisso,G. etal. A high concentration of fasting action and clinical implications for diabetes and cancer. 116. Liu,Y. etal. A fasting inducible switch modulates
plasma non-esterified fatty acids is a risk factor for Nat. Rev. Endocrinol. 10, 143156 (2014). gluconeogenesis via activator/coactivator exchange.
the development of NIDDM. Diabetologia 38, 92. Brown,L.J. etal. Normal thyroid thermogenesis but Nature 456, 269273 (2008).
12131217 (1995). reduced viability and adiposity in mice lacking the 117. Wang,Y. etal. Targeted disruption of the CREB
68. Jocken,J.W.E. etal. Insulin-mediated suppression of mitochondrial glycerol phosphate dehydrogenase. coactivator Crtc2 increases insulin sensitivity.
lipolysis in adipose tissue and skeletal muscle of obese J.Biol. Chem. 277, 3289232898 (2002). Proc. Natl Acad. Sci. USA 107, 30873092 (2010).
type2 diabetic men and men with normal glucose 93. Saheki,T. etal. Citrin/mitochondrial 118. Hogan,M.F. etal. Hepatic insulin resistance following
tolerance. Diabetologia 56, 22552265 (2013). glycerol3phosphate dehydrogenase double knock-out chronic activation of the CREB coactivator CRTC2.
69. Heptulla,R.A. etal. In situ evidence that peripheral mice recapitulate features of human citrin deficiency. J.Biol. Chem. 290, 2599726006 (2015).
insulin resistance in adolescents with poorly controlled J.Biol. Chem. 282, 2504125052 (2007). 119. Bass,J. & Lazar,M.A. Circadian time signatures of
type1 diabetes is associated with impaired 94. Baur,J.A. & Birnbaum,M.J. Control of fitness and disease. Science 354, 994999 (2016).
suppression of lipolysis: a microdialysis study. Pediatr. gluconeogenesis by metformin: does redox trump 120. Zhang,E.E. etal. Cryptochrome mediates circadian
Res. 53, 830835 (2003). energy charge? Cell Metab. 20, 197199 (2014). regulation of cAMP signaling and hepatic
70. Gelding,S.V., Coldham,N., Niththyananthan,R., 95. Exton,J.H. & Park,C.R. Control of gluconeogenesis in liver. gluconeogenesis. Nat. Med. 16, 11521156 (2010).
Anyaoku,V. & Johnston,D.G. Insulin resistance with II. Effects of glucagon, catecholamines, and adenosine 121. Lamia,K.A. etal. Cryptochromes mediate rhythmic
respect to lipolysis in non-diabetic relatives of 3,5monophosphate on gluconeogenesis in the perfused rat repression of the glucocorticoid receptor. Nature 480,
European patients with type2 diabetes. Diabet. Med. liver. J.Biol. Chem. 243, 41894196 (1968). 552556 (2011).
12, 6673 (1995). 96. Lee,Y. etal. Hyperglycemia in rodent models of type2 122. Lamia,K.A., Storch,K.F. & Weitz,C.J. Physiological
71. Robinson,C. etal. Effect of insulin on glycerol diabetes requires insulin-resistant alpha cells. Proc. significance of a peripheral tissue circadian clock. Proc.
production in obese adolescents. Am. J.Physiol. 274, Natl Acad. Sci. USA 111, 1321713222 (2014). Natl Acad. Sci. USA 105, 1517215177 (2008).
E737E743 (1998). 97. Blair,J.B., Cimbala,M.A., Foster,J.L. & Morgan,R.A. 123. Sun,Z. etal. Hepatic Hdac3 promotes
72. Puhakainen,I., Koivisto,V.A. & Yki-Jrvinen,H. Hepatic pyruvate kinase. Regulation by glucagon, cyclic gluconeogenesis by repressing lipid synthesis and
Lipolysis and gluconeogenesis from glycerol are adenosine 35monophosphate, and insulin in the sequestration. Nat. Med. 18, 934942 (2012).
increased in patients with noninsulin-dependent perfused rat liver. J.Biol. Chem. 251, 37563762 124. Burgess,S.C. etal. Cytosolic phosphoenolpyruvate
diabetes mellitus. J.Clin. Endocrinol. Metab. 75, (1976). carboxykinase does not solely control the rate of
789794 (1992). 98. Rider,M.H. etal. 6Phosphofructo2kinase/ hepatic gluconeogenesis in the intact mouse liver. Cell
73. Nurjhan,N., Consoli,A. & Gerich,J. Increased fructose2,6bisphosphatase: headtohead with a Metab. 5, 313320 (2007).
lipolysis and its consequences on gluconeogenesis in bifunctional enzyme that controls glycolysis. Biochem. J. 125. Zingone,A. etal. Correction of glycogen storage
non-insulin-dependent diabetes mellitus. J.Clin. 381, 561579 (2004). disease type1a in a mouse model by gene therapy.
Invest. 89, 169175 (1992). 99. Wu,C. etal. Perturbation of glucose flux in the liver by J.Biol. Chem. 275, 828832 (2000).
74. Perry,R.J., Peng,L., Cline,G.W., Petersen,K.F. & decreasing F26P2 levels causes hepatic insulin 126. Bernard,C. Leons de physiologie exprimentale
Shulman,G.I.A. Non-invasive method to assess resistance and hyperglycemia. Am. J.Physiol. applique a la mdecine (J.-B. Baillire, 1855).
hepatic acetyl-CoA invivo. Cell Metab. 25, 749756 Endocrinol. Metab. 291, E536E543 (2006). 127. Schwartz,M.W. etal. Cooperation between brain and
(2017). 100. Cullen,K.S., AlOanzi,Z.H., OHarte,F.P.M., islet in glucose homeostasis and diabetes. Nature
75. American Diabetes Association. 7. Approaches to Agius,L. & Arden,C. Glucagon induces translocation 503, 5966 (2013).
glycemic treatment. Diabetes Care 39, S52S59 of glucokinase from the cytoplasm to the nucleus of 128. Myers,M.G. & Olson,D.P. Central nervous system
(2016). hepatocytes by transfer between 6phosphofructo control of metabolism. Nature 491, 357363 (2012).
76. Hundal,R.S. etal. Mechanism by which metformin 2kinase/fructose 2,6bisphosphatase2 and the 129. Perry,R.J., Petersen,K.F. & Shulman,G.I. Pleotropic
reduces glucose production in type2 diabetes. glucokinase regulatory protein. Biochim. Biophys. effects of leptin to reverse insulin resistance and diabetic
Diabetes 49, 20632069 (2000). Acta 1843, 11231134 (2014). ketoacidosis. Diabetologia 59, 933937 (2016).

NATURE REVIEWS | ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION | 15



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

130. Duffy,K.R. & Pardridge,W.M. Bloodbrain barrier 156. Wan,M. etal. A noncanonical, GSK3independent 181. Longuet,C. etal. The glucagon receptor is required
transcytosis of insulin in developing rabbits. Brain Res. pathway controls postprandial hepatic glycogen for the adaptive metabolic response to fasting. Cell
420, 3238 (1987). deposition. Cell Metab. 18, 99105 (2013). Metab. 8, 359371 (2008).
131. Plum,L., Schubert,M. & Brning,J.C. The role of 157. Kitamura,T. etal. Insulin-induced phosphorylation 182. Vatner,D.F. etal. Thyroid hormone receptor
insulin receptor signaling in the brain. Trends and activation of cyclic nucleotide phosphodiesterase agonists prevent hepatic steatosis in fat-fed rats but
Endocrinol. Metab. 16, 5965 (2005). 3B by the serinethreonine kinase Akt. Mol. Cell. Biol. impair insulin sensitivity via discrete pathways.
132. Brning,J.C. etal. Role of brain insulin receptor in 19, 62866296 (1999). Am. J.Physiol. Endocrinol. Metab. 305, E89E100
control of body weight and reproduction. Science 158. Jurczak,M.J., Danos,A.M., Rehrmann,V.R. & (2013).
289, 21222125 (2000). Brady,M.J. The role of protein translocation in the 183. Martagn,A.J., Lin,J.Z., Cimini,S.L., Webb,P. &
133. Obici,S., Zhang,B.B., Karkanias,G. & Rossetti,L. regulation of glycogen metabolism. J.Cell. Biochem. Phillips,K.J. The amelioration of hepatic steatosis by
Hypothalamic insulin signaling is required for inhibition of 104, 435443 (2008). thyroid hormone receptor agonists is insufficient to
glucose production. Nat. Med. 8, 13761382 (2002). 159. Alemany,S. & Cohen,P. Phosphorylase a is an restore insulin sensitivity in ob/ob mice. PLoS ONE 10,
134. Pocai,A. etal. Hypothalamic KATP channels control allosteric inhibitor of the glycogen and microsomal e0122987 (2015).
hepatic glucose production. Nature 434, 10261031 forms of rat hepatic protein phosphatase1. FEBS Lett. 184. Finan,B. etal. Chemical hybridization of glucagon
(2005). 198, 194202 (1986). and thyroid hormone optimizes therapeutic impact
135. Kleinridders,A., Ferris,H.A., Cai,W. & Kahn,C.R. 160. Carabaza,A., Ciudad,C.J., Baqu,S. & for metabolic disease. Cell 167, 843857.e14
Insulin action in brain regulates systemic metabolism Guinovart,J.J. Glucose has to be phosphorylated to (2016).
and brain function. Diabetes 63, 22322243 (2014). activate glycogen synthase, but not to inactivate 185. Armstrong,M.J. etal. Liraglutide safety and efficacy in
136. Edgerton,D.S. etal. Insulins direct effects on the glycogen phosphorylase in hepatocytes. FEBS Lett. patients with non-alcoholic steatohepatitis (LEAN): a
liver dominate the control of hepatic glucose 296, 211214 (1992). multicentre, double-blind, randomised, placebo-
production. J.Clin. Invest. 116, 521527 (2006). 161. Samuel,V.T. & Shulman,G.I. The pathogenesis of controlled phase2 study. Lancet 387, 679690
137. Ramnanan,C.J. etal. Brain insulin action augments insulin resistance: integrating signaling pathways and (2016).
hepatic glycogen synthesis without suppressing substrate flux. J.Clin. Invest. 126, 1222 (2016). 186. Finan,B. etal. A rationally designed monomeric
glucose production or gluconeogenesis in dogs. J.Clin. 162. Olefsky,J.M. & Glass,C.K. Macrophages, peptide triagonist corrects obesity and diabetes in
Invest. 121, 37133723 (2011). inflammation, and insulin resistance. Annu. Rev. rodents. Nat. Med. 21, 2736 (2015).
138. Shulman,G.I. & Landau,B.R. Pathways of glycogen Physiol. 72, 219246 (2010). 187. Soccio,R.E., Chen,E.R. & Lazar,M.A. Thiazolidinediones
repletion. Physiol. Rev. 72, 10191035 (1992). 163. Tolman,K.G., Fonseca,V., Dalpiaz,A. & Tan,M.H. and the promise of insulin sensitization in type2 diabetes.
139. Syed,N.A. & Khandelwal,R.L. Reciprocal regulation Spectrum of liver disease in type2 diabetes and Cell Metab. 20, 573591 (2014).
of glycogen phosphorylase and glycogen synthase by management of patients with diabetes and liver 188. Mayerson,A.B. etal. The effects of rosiglitazone on
insulin involving phosphatidylinositol3 kinase and disease. Diabetes Care 30, 734743 (2007). insulin sensitivity, lipolysis, and hepatic and skeletal
protein phosphatase1 in HepG2 cells. Mol. Cell. 164. Lee,Y.H., Wang,M.Y., Yu,X.X. & Unger,R.H. muscle triglyceride content in patients with type2
Biochem. 211, 123136 (2000). Glucagon is the key factor in the development of diabetes. Diabetes 51, 797802 (2002).
140. Moore,M.C. etal. Hepatic glucose disposition during diabetes. Diabetologia 59, 13721375 (2016). 189. Belfort,R. etal. A placebo-controlled trial of pioglitazone
concomitant portal glucose and amino acid infusions 165. Henry,R.R., Scheaffer,L. & Olefsky,J.M. Glycemic in subjects with nonalcoholic steatohepatitis. N.Engl.
in the dog. Am. J.Physiol. 274, E893E902 (1998). effects of intensive caloric restriction and isocaloric J.Med. 355, 22972307 (2006).
141. Gomis,R.R., Ferrer,J.C. & Guinovart,J.J. Shared control refeeding in noninsulin-dependent diabetes mellitus. 190. Gastaldelli,A. etal. Importance of changes in
of hepatic glycogen synthesis by glycogen synthase and J.Clin. Endocrinol. Metab. 61, 917925 (1985). adipose tissue insulin resistance to histological
glucokinase. Biochem. J. 351, 811816 (2000). 166. American Diabetes Association. 6. Obesity response during thiazolidinedione treatment of
142. ODoherty,R.M. etal. Differential metabolic effects of management for the treatment of type2 diabetes. patients with nonalcoholic steatohepatitis.
adenovirus-mediated glucokinase and hexokinase I Diabetes Care 39, S47S51 (2016). Hepatology 50, 10871093 (2009).
overexpression in rat primary hepatocytes. J.Biol. 167. Lefebvre,P.J. & Luyckx,A.S. Glucagon and diabetes: 191. Rizos,C.V., Kei,A. & Elisaf,M.S. The current role of
Chem. 271, 2052420530 (1996). a reappraisal. Diabetologia 16, 347354 (1979). thiazolidinediones in diabetes management.
143. Niswender,K.D., Shiota,M., Postic,C., 168. Lotfy,M., Kalasz,H., Szalai,G., Singh,J. & Adeghate,E. Arch. Toxicol. 90, 18611881 (2016).
Cherrington,A.D. & Magnuson,M.A. Effects of Recent progress in the use of glucagon and glucagon 192. Tunaru,S. etal. PUMAG and HM74 are receptors for
increased glucokinase gene copy number on glucose receptor antagonists in the treatment of diabetes nicotinic acid and mediate its anti-lipolytic effect.
homeostasis and hepatic glucose metabolism. J.Biol. mellitus. Open Med. Chem. J. 8, 2835 (2014). Nat. Med. 9, 352355 (2003).
Chem. 272, 2257022575 (1997). 169. Bagger,J.I., Knop,F.K., Holst,J.J. & Vilsbll,T. 193. Phan,B.A.P. etal. Effects of niacin on glucose levels,
144. Titchenell,P.M. etal. Direct hepatocyte insulin Glucagon antagonism as a potential therapeutic target in coronary stenosis progression, and clinical events in
signaling is required for lipogenesis but is dispensable type2 diabetes. Diabetes Obes. Metab. 13, 965971 subjects with normal baseline glucose levels (<100mg/
for the suppression of glucose production. Cell Metab. (2011). dl): a combined analysis of the Familial Atherosclerosis
23, 11541166 (2016). 170. Habegger,K.M. etal. The metabolic actions of Treatment Study (FATS), HDL-Atherosclerosis Treatment
145. Velho,G. etal. Impaired hepatic glycogen synthesis in glucagon revisited. Nat. Rev. Endocrinol. 6, 689697 Study (HATS), Armed Forces Regression Study
glucokinase-deficient (MODY2) subjects. J.Clin. (2010). (AFREGS), and Carotid Plaque Composition by MRI
Invest. 98, 17551761 (1996). 171. Reaven,G.M., Chen,Y.D., Golay,A., Swislocki,A.L. during lipid-lowering (CPC) study. Am. J.Cardiol. 111,
146. Raimondo,A., Rees,M.G. & Gloyn,A.L. Glucokinase & Jaspan,J.B. Documentation of hyperglucagonemia 352355 (2013).
regulatory protein: complexity at the crossroads of throughout the day in nonobese and obese patients 194. Guyton,J.R. Niacin in cardiovascular prevention:
triglyceride and glucose metabolism. Curr. Opin. with noninsulin-dependent diabetes mellitus. J.Clin. mechanisms, efficacy, and safety. Curr. Opin. Lipidol.
Lipidol. 26, 8895 (2015). Endocrinol. Metab. 64, 106110 (1987). 18, 415420 (2007).
147. Agius,L. Glucokinase and molecular aspects of liver 172. Baron,A.D., Schaeffer,L., Shragg,P. & Kolterman,O.G. 195. Kroon,T., Kjellstedt,A., Thaln,P., Gabrielsson,J. &
glycogen metabolism. Biochem. J. 414, 118 (2008). Role of hyperglucagonemia in maintenance of increased Oakes,N.D. Dosing profile profoundly influences
148. Agius,L., Peak,M., Newgard,C.B., Gomez-Foix,A.M. rates of hepatic glucose output in typeII diabetics. nicotinic acids ability to improve metabolic control in
& Guinovart,J.J. Evidence for a role of glucose- Diabetes 36, 274283 (1987). rats. J.Lipid Res. 56, 16791690 (2015).
induced translocation of glucokinase in the control of 173. Woerle,H.J. etal. Mechanisms for abnormal
hepatic glycogen synthesis. J.Biol. Chem. 271, postprandial glucose metabolism in type2 diabetes. Acknowledgements
3047930486 (1996). Am. J.Physiol. Endocrinol. Metab. 290, E67E77 The authors apologize to those many colleagues whose
149. Hrndahl,L., Schmoll,D., Herling,A.W. & Agius,L. (2006). important contributions could not be discussed owing to
The role of glucose 6phosphate in mediating the 174. Menge,B.A. etal. Loss of inverse relationship between word and reference limits. The authors thank A.K. Madiraju
effects of glucokinase overexpression on hepatic pulsatile insulin and glucagon secretion in patients with for helpful comments. M.C.P. acknowledges grant support
glucose metabolism. FEBS J. 273, 336346 (2006). type2 diabetes. Diabetes 60, 21602168 (2011). from the US National Institutes of Health (NIH; grants F30
150. von Wilamowitz-Moellendorff,A. etal. 175. Lee,Y. etal. Metabolic manifestations of insulin DK104596 and T32 GM007205). D.F.V. acknowledges
Glucose6phosphate-mediated activation of liver deficiency do not occur without glucagon action. Proc. grant support from the NIH (grant K23 DK102874). G.I.S.
glycogen synthase plays a key role in hepatic glycogen Natl Acad. Sci. USA 109, 1497214976 (2012). acknowledges grant support from the NIH (grants R01
synthesis. Diabetes 62, 40704082 (2013). 176. Neumann,U.H. etal. Glucagon receptor gene DK40936, R01 DK113984 and P30 DK045735).
151. Bollen,M., Keppens,S. & Stalmans,W. Specific deletion in insulin knockout mice modestly reduces
features of glycogen metabolism in the liver. Biochem. blood glucose and ketones but does not promote Author contributions
J. 336, 1931 (1998). survival. Mol. Metab. 5, 731736 (2016). All authors contributed to all aspects of the preparation of
152. Ros,S., Garca-Rocha,M., Domnguez,J., Ferrer,J.C. & 177. Petersen,K.F. & Sullivan,J.T. Effects of a novel the article. The order of authorship and contribution is
Guinovart,J.J. Control of liver glycogen synthase activity glucagon receptor antagonist (Bay 279955) on M.C.P., D.F.V. and G.I.S.
and intracellular distribution by phosphorylation. J.Biol. glucagon-stimulated glucose production in humans.
Chem. 284, 63706378 (2009). Diabetologia 44, 20182024 (2001). Competing interests statement
153. Bultot,L. etal. AMP-activated protein kinase 178. Guan,H.P. etal. Glucagon receptor antagonism M.C.P. and D.F.V. declare no competing interests. G.I.S.
phosphorylates and inactivates liver glycogen induces increased cholesterol absorption. J.Lipid Res. serves on scientific advisory boards for Merck, Novo Nordisk,
synthase. Biochem. J. 443, 193203 (2012). 56, 21832195 (2015). Celgene, Aegerion and AstraZeneca, receives investigator-
154. Ros,S. etal. Hepatic overexpression of a 179. Kelly,R.P. etal. Short-term administration of the initiated support from Gilead Sciences, Inc., and is an inven-
constitutively active form of liver glycogen synthase glucagon receptor antagonist LY2409021 lowers blood tor on Yale patents for liver-targeted mitochondrial
improves glucose homeostasis. J.Biol. Chem. 285, glucose in healthy people and in those with type2 uncoupling agents for the treatment of NAFLD, NASH, type
3717037177 (2010). diabetes. Diabetes Obes. Metab. 17, 414422 (2015). 2 diabetes and related metabolic disorders.
155. Cohen,P. The Croonian Lecture 1998. Identification of 180. Kazda,C.M. etal. Evaluation of efficacy and safety of
a protein kinase cascade of major importance in the glucagon receptor antagonist LY2409021 in Publishers note
insulin signal transduction. Phil. Trans. R.Soc. B 354, patients with type2 diabetes: 12- and 24week phase2 Springer Nature remains neutral with regard to jurisdictional
485495 (1999). studies. Diabetes Care 39, 12411249 (2016). claims in published maps and institutional affiliations.

16 | ADVANCE ONLINE PUBLICATION www.nature.com/nrendo



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like