ArticlePDF Available

Hepatic Notch Signaling Correlates With Insulin Resistance and Nonalcoholic Fatty Liver Disease

Authors:

Abstract and Figures

Hepatic Notch signaling is inappropriately activated in obese/insulin-resistant mouse models. Genetic or pharmacologic inhibition of hepatic Notch signaling in obese mice simultaneously improves glucose tolerance and reduces hepatic triglyceride content. As such, we predicted that Notch signaling in human liver would be positively associated with both insulin resistance and hepatic steatosis. Here, we systematically survey Notch signaling in liver biopsy specimens, and show active Notch signaling in both lean and obese adults, with expression of multiple Notch receptors and ligands. In morbidly obese patients undergoing bariatric surgery, we show that Notch activation positively correlates with Glucose-6-phosphatase (G6PC) and Phosphoenolpyruvate carboxykinase (PCK1) expression, key regulators of hepatic glucose output. We used immunofluorescence to identify active Notch signaling in hepatocytes, and show highest activity in hyperglycemia, which we confirmed is a direct effect of hyperglycemia and insulin resistance. In a validation cohort of leaner individuals undergoing percutaneous liver biopsy for suspected nonalcoholic fatty liver disease, Notch activity showed independent positive association with both insulin resistance and hepatic steatosis. Notably, Notch activity showed stronger correlation with nonalcoholic fatty liver disease activity score and ALT levels than with steatosis alone, suggesting that Notch activity is associated with nonalcoholic steatohepatitis. In summary, this study establishes that Notch signaling is activated in, and may represent a therapeutic target for patients with obesity-related liver disease.
Content may be subject to copyright.
Hepatic Notch Signaling Correlates With Insulin
Resistance and Nonalcoholic Fatty Liver Disease
Luca Valenti,
1
Rosa M. Mendoza,
2
Raffaela Rametta,
1
Marco Maggioni,
3
Chris Kitajewski,
4
Carrie J. Shawber,
4
and Utpal B. Pajvani
2
Hepatic Notch signaling is inappropriately activated in obese/
insulin-resistant mouse models. Genetic or pharmacologic in-
hibition of hepatic Notch signaling in obese mice simultaneously
improves glucose tolerance and reduces hepatic triglyceride
content. As such, we predicted that Notch signaling in human
liver would be positively associated with insulin resistance and
hepatic steatosis. Here, we systematically survey Notch signaling
in liver biopsy specimens, and show active Notch signaling in
lean and obese adults, with expression of multiple Notch receptors
and ligands. In morbidly obese patients undergoing bariatric
surgery, we show that Notch activation positively correlates with
glucose-6-phosphatase (G6PC) and phosphoenolpyruvate carboxy-
kinase (PCK1) expression, key regulators of hepatic glucose out-
put. We used immunouorescence to identify active Notch
signaling in hepatocytes and show highest activity in hyperglyce-
mia, which we conrmed is a direct effect of hyperglycemia and
insulin resistance. In a validation cohort of leaner individuals
undergoing percutaneous liver biopsy for suspected nonalcoholic
fatty liver disease (NAFLD), Notch activity showed independent
positive association with insulin resistance and hepatic steatosis.
Notably, Notch activity showed stronger correlation with the
NAFLD activity score and alanine aminotransferase levels than
with steatosis alone, suggesting that Notch activity is associated
with nonalcoholic steatohepatitis. In summary, this study estab-
lishes that Notch signaling is activated in and may represent
a therapeutic target for patients with obesity-related liver disease.
Diabetes 62:40524062, 2013
O
besity manifests as multiple pathologic states in
the liver. Insulin resistance in adipocytes
results in unrestrained lipolysis, with conse-
quent excess free fatty acid ux to the liver (1).
In a parallel pathogenic process, ex cess adiposity leads to
insulin resistance, which begets the fasting hyperglycemia
of type 2 diabetes (T2D) (2). Compensatory hyper-
insulinemia drives de novo lipogenesis (3), and coupled
with an impaired ability to catabolize and export fatty
acids (4), results in excess hepatocyte triglyceride accu-
mulation, or nonalcoholic fatty liver disease (NAFLD), in
the presence of a predisposing genetic background (5).
Steatosis may be associated with hepatocellular damage
and necroinammatory changes, dening nonalcoholic
steatohepatitis (NASH), which predisposes to cirrhosis
and hepatocellular cancer (1). Interestingly, NASH further
exacerbates hepatic insulin resistance through activation
of forkhead box class O (FoxO) 1, the key transcriptional
activator of glucose-6-phosphatase (G6PC) and phospho-
enolpyruvate carboxykinase (PCK1), rate-limiting enzymes
of gluconeogenesis and glycogenolysis, which combined
regulate hepatic glucose output. This vicious cycle results in
coincident NAFLD and T2D, which show independent asso-
ciations with cardiovascular disease (6). No approved phar-
macologic therapy is approved for NALFD, and although
multiple T2D therapies are available, few show durability and
long-term efcacy (7). As such, novel therapeutic directions
are necessary to reduce overall obesity-related morbidity .
We previously showed that inhibition of hepatic Notch
signaling protects from both obesity-induced glucose in-
tolerance, by suppressing hepatic glucose output (8), and
fatty liver, by reducing de novo lipogenesis (9). Notch
signaling is highly conserved from lower organisms to
primates and is critical for cell fate decision making, in-
cluding regulation of cell specication and lineage restric-
tion, depending on the cellular context (10). In mammals,
cell surface Notch ligands of the Jagged (-1 and -2) and
Delta-like (-1, -3, -4) families bind one of four Notch re-
ceptors (Notch14) on a neighboring cell, resulting in a se-
ries of cleavage events that culminate in the transcription of
canonical Notch targets, the hairy enhancer of split (HES)
and Hes-related (HEY) family of genes (11). Homozygous
null alleles in this pathway result in embryonic lethality in
mice (1214) and loss-of-function mutations in severe de-
velopmental defects in affected individuals (15,16), proving
the critical role of Notch signaling to regulate cell fate
decisions in normal development.
Less is known about Notch signaling in matur e tissue.
Increased Notch expression and fun ction has been shown
in cancer and tumor angiogenesis (1719), but there are
few reports of expression analysis of Notch pathway pro-
teins in developed, non-neoplastic tissue. In the liver,
Notch proteins are constitutively expressed in multiple cell
types (20,21), with increased expression in hepatocytes
after partial hepatectomy (22). In rat models, normal liver
regeneration was prevented by genetic inhibition of he-
patocyte Notch signaling (23). As such, we pred icted that
the Notch-signaling apparatus is present in mature liver
and may be induced by obesity-induced hepatocyte dam-
age. Our initial characterization of Notch signaling in mu-
rine liver demonstrated that Notch target gene expression
is, in fact, increased in mouse models of obesity and in-
sulin resistance (8). Interestingly, genetic (8) or pharma-
cologic (11,24,25) blockade of hepatocyte Notch signaling
resulted in parallel inhibition of hepatic glucose pro-
duction (8) as well as triglyceride accumulation (9), low-
ering the overall atherosclerotic burden in obese mice (26).
From the
1
Department of Pathophysiology and Transplantation, Università
degli Studi di Milano, Internal Medicine, Fondazione Istituto di Ricovero e
Cura a Carattere Scientico Ca Granda, Milano, Italy; the
2
Department of
Medicine, Columbia University, New York, New York; the
3
Department of
Pathology, Università degli Studi di Milano, Internal Medicine, Fondazione
Istituto di Ricovero e Cura a Carattere Scientico Ca Granda, Milano, Italy;
and the
4
Department of Obstetrics and Gynecology, Columbia University,
New York, New York.
Corresponding author: Utpal B. Pajvani, up2104@columbia.edu.
Received 13 May 2013 and accepted 22 August 2013.
DOI: 10.2337/db13-0769
Ó 2013 by the American Diabetes Association. Readers may use this article as
long as the work is properly cited, the use is educational and not for prot,
and the work is not altered. See http://creativecommons.org/licenses/by
-nc-nd/3.0/ for details.
4052 DIABETES, VOL. 62, DECEMBER 2013 diabetes.diabetesjournals.org
ORIGINAL ARTICLE
Conversely, constitutive activation of hepatocyte Notch
signaling caused glucose intolerance and fatty liver (8,9).
These studies suggested that the Notch pathway is active
through adulthood in rodent liver, is inappropriately
stimulated by obesity, and may be manipulated to reduce
the obesity-related metabolic disease burden.
On the basis of these rodent studies, we hypothesized
that Notch signaling is similarly functional and may cor-
relate with disease severity in patients with hepatic insulin
resistance an d NAFLD. In this study, we show that Notch
proteins and ligands are expressed in lean and obese
subjects and that increased activation of this pathway, as
assessed by expression of Notch target genes of the HES/
HEY family, positively correlates with gluconeogenic gene
expression and hyperglycemia in a cohort of morbidly
obese patients undergoing bariatric surgery. In a validation
cohort across a range of BMIs, we conrmed the positive
association between HES/HEY family genes and insulin
resistance as well as demonstrated an independent pos i-
tive association with hepatic fat content. Finally, we show
that hepatic Notch signal activation correl ates better with
measures of liver inammation than with simple steatosis
(SS), suggestin g that it may represent a marker of the
transition from SS to NASH. This work establishes that
Notch signaling correlates and potentially represents
a novel therapeutic target for both arms of obesity-related
liver disease, T2D and NAFLD.
RESEARCH DESIGN AND METHODS
Subjects. The study conforms to the ethical guidelines of the 1975 Declaration
of Helsinki and was approved by the institutional review board and ethical
committee of the Fondazione Istituto di Ricovero e Cura a Carattere Scientico
(IRCCS) Ca Granda. Each subject gave written informed consent. Demographic
and anthropo met ric feat ures, ar teria l blood pr essu re, med ical hi story, an d
medication s were r ecorde d for a ll patients and are summarized in Table 1.
A needle liver biopsy was performed in all patients. The specimen was
formalin-preserved for histological and immunouorescence analysis. Part of
the sample was included in RNAlater (Ambion, Carlsbad, CA), immediately
frozen in liquid nitrogen, and stored at 280°C for RNA analysis.
Bariatric surgery clinic. We recruited 44 of 48 conse cutive patients who
underwent ba riatric surgery at the IRCCS Ca Granda Ospedale Policlinico d i
Milano, between 2006 and 2008. Indications for bariatric surgery included
BMI .40 kg/m
2
or BMI .35 kg/m
2
in the prese nce of metabolic complica -
tions (T2D, uncontro lled hypertensi on, severe dyslipidemia, obstructive
sleep apnea). We e xclude d subjects with alcohol consumption .30 g/day for
men and 20 g/day for wome n (n = 1) and chronic viral h epatitis (n =3).
Fasting glucose, HDL and total chole sterol, triglycerides, alanine amino-
transferase (ALT), and as partate aminotransferase (AST) were assessed the
day of surgery, and ne edle liver biopsy (16 -gaug e) was p erformed during
the bar iatric surgery. Insulin-resistance status was c lassi e d according to
fasting glucose levels and oral glucose tolerance test results. These patients
are part of a previously reported c ohort in which we characterized insulin-
dependent signaling and the re gulation of lipid metabolism according to liver
histology (27).
Hepatology clinic patients. We recruited 38 unrelated patients followed up
at the Metabolic Liver Diseases outpatient service, Fondazione IRCCS Ca
Granda, who underwent percutaneous liver biopsy because of suspected
NAFLD due to persistently abnormal liver enzymes/serum ferritin test results
or a history of steatosis associated with severe metabolic abnormalities, be-
tween January 2011 and January 2012. Other causes of liver disease were
excluded, including increased alcohol intake (.30 g/day for men or 20 g/day
for women), viral and autoimmune hepatitis, hereditary hemochromatosis,
and a1-antitrypsin deciency. Fasting glucose and insulin levels, HDL and total
cholesterol, triglycerides, and ALT and AST levels were assessed the day of
the biopsy. Patients were classied as insulin-sensitive based on homeostasis
model assessmentinsulin resis tanc e (HOMA-I R) ,2.5 ([fasting insulin
(mU/mL) 3 fasting glucose (mmol/L)]/22.5) (28) without a history of impaired
glucose tolerance or impaired fasting glucose, insulin resistance (HOMA-IR
.2.5, or impaired fasting glucose or impaired glucose tolerance, but no di-
agnosis of diabetes) or T2D.
Histological analysis. A single expert pathologist, unaware of gene expres -
sion and immunouorescence data, evaluated all biopsy specimens according
to Kleiner et al. (29), based on the determination of the NAFLD activity score
(NAS) as the result of the sum of steatosis severity (03), intralobular necro-
inammation (03), and hepatocellular ballooning (02). Steatosis percentage
was determined in at least 10 hepatic lobules per patient. Subjects were clas-
sied in three groups according to liver histology: histologically normal liver, SS,
and NASH.
Immunouorescence. Parafnized sections were deparafnized, rehydrated,
and stained as previously described (30). To determine Notch signal activation,
tissues were stained with Hey1 (#5714, 1:100 dilution) or HeyL (#10094, 1:150
dilution) antibodies from Millipore and detected with donkey anti-rabbit Alexa
Fluor 488 (1:1,000 dilution) or donkey anti-mouse Alexa Fluor 594 (1:1,000)
from Invitrogen. Slides were mounted with Vectashield with DAPI (Vector
TABLE 1
Demographic and clinical features of subjects included in the study subdivided according to the case series (bariatric surgery and
hepatology clinic) and liver histology
Bariatric surgery Hepatology clinic
Normal SS NASH P value Normal SS NASH P value
n (%) 5 (12) 14 (33) 23 (55) 7 (18) 11 (29) 20 (53)
Sex (n) 0.035 0.23
Female 5 13 14 0 1 0
Male 0 1 9 7 10 20
Age (years) 44 6 4406 9426 10 0.73 45 6 12 54 6 9516 12 0.33
BMI (kg/m
2
) 40.8 6 10 40.5 6 10 43.5 6 8 0.54 25.1 6 2.4 27.5 6 3.4 28.7 6 4.2 0.11
Abdominal circumference (cm) —— 94 6 6 102 6 6 112 6 21 0.05
Glucose (mg/dL) 83 6 8886 5 117 6 56 0.09 92 6 7 116 6 35 111 6 30 0.24
Insulin (IU/mL) —— 8.4 6 2.2 14.1 6 1.9 16.0 6 1.5 0.034
HOMA-IR —— 1.7 6 0.4 3.6 6 1.7 3.8 6 1.7 0.018
Glucose tolerance 0.001
IS [n (%)] —— 7 (100) 2 (18) 4 (20)
IR [n (%)] —— 0 5 (46) 10 (50)
T2D [n (%)] 0 0 9 (39) 0 4 (36) 6 (30)
Cholesterol (mg/dL) 215 6 31 211 6 36 214 6 41 0.96 191 6 47 201 6 37 188 6 40 0.66
Triglyceride (mg/dL) 99 6 54 155 6 95 157 6 54 0.30 94 6 27 98 6 48 115 6 54 0.51
HDL (mg/dL) 73 6 11 52 6 9466
12 ,0.0001 55 6 15 57 6 28 44 6 14 0.17
Steatosis (%) 2 6 1176 13 61 6 21 ,0.0001 3 6 2196 14 52 6 23 ,0.0001
ALT (IU/mL) 18 6 5266 9396 25 0.03 22 6 8406 32 66 6 46 0.02
Data represent average 6 SD unless indicated otherwise. IR, insulin-resistant; IS, insulin-sensitive.
L. VALENTI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 62, DECEMBER 2013 4053
Laboratories). Images were captured with a Nikon ECLIPSE E800 microscope
and a Nikon DXM 1200 digital camera and processed with Image ProPlus
software. Staining was quantitated in a blinded fashion from 1 (low expres-
sion) to 4 (high expression), independently by three investigators, and scores
were averaged.
Quantitative RT-PCR. We isolated RNA with Trizol (Invitrogen), synthesized
cDNA with Superscript III RT (Invitrogen), and performed quantitative PCR
with a DNA Engine Opticon 2 System (Bio-Rad) and GoTaq SYBR Green
(Promega). Absolute mRNA levels were determine for each gene using species-
and primer-specic standard curves, then normalized to 18S, and are presented
as relative transcript levels (fg/ng 18S). Primer sequences are available upon
request.
Luciferase assays. Hepa1c1c7 cells were transfected with the Rbp-Jk reporter
luciferase construct, as previously described (8), and then incubated in serum-
free medium with variable glucose content in the presence or absence of
10 nmol/L insulin or 10 nmol/L glucagon.
Hepatocyte studies. We isolated and cultured primary mouse hepatocytes as
described (8) and obtained human primary hepatocytes from Invitrogen. For
gene and protein expression studies, we treated hepatocytes with variable
concentrations of glucose, and/or 10 nmol/L insulin (Sigma-Aldrich) for 3 h,
with all experiments completed by 24 h after isolation.
Statistical analysis. Results are shown as mean 6 SEM. ANOVA was used for
comparison of means among groups. Gene expression levels were correlated
by the Pearson correlation. The association between HES1 expression (above
the median) and insulin resistance of T2D was evaluated by multivariate lo-
gistic regression analysis adjusted for age, steatosis, and BMI. Independent
predictors of HES1 expression were evaluated at multivariate regression
analysis (generalized linear model), including NAS and insulin levels, the
variables most signicantly associated at univariate analysis. Differences were
considered signicant at P , 0.05 (two-tailed).
RESULTS
Human liver has a functional, evolutionarily conserved
Notch-signaling apparatus. To clarify the role of
Notch signaling in the postdevelopment human liver,
we characterized liver Notchandligandexpressionin
insulin-sensitive subjects with a histologically normal
liver. We found that in these subjects, all four Notch
receptors were expressed, with relative higher lev els of
NOTCH1 and NOTCH2 (Fig. 1A, left panel). In addition, all
ve Notch ligands were detectable by quantitative PCR,
with JAG1 and JAG2 showing greater expression than
Delta-like ligands. Of note, this expression pattern is
broadly similar to the distribution observed in murine liver
(Fig. 1A, right panel) as well as in mouse hepatocytes (8).
Generally, expression of NOTCH1 and other Notch genes
covaried (Fig. 1B)aswellaswithHES/HEY family target
genes (Fig. 1C), but showed no correlation with house-
keeping genes (ACTB, GAPDH)andanonsignicant trend
toward a negative correlation with Notch ligand expres-
sion (data not shown). These data show that Notch sig-
naling components are present in mature liver and that
relative expression is evolutionarily conserved.
Hepatic Notch signaling correlates with G6PC/PCK1
expression and is increased in pat ients with T2D.
Because Notch proteins and ligands are expressed in hu-
man liver, we hypothesized that Notch signaling may be
FIG. 1. Notch pathway in human liver. A: Notch receptor and ligand expression is similar in human (left) and mouse liver (right). Hepatic NOTCH1
expression positively correlates with the expression of other Notch receptors (P < 0.001 by ANOVA for all comparisons) (B) and canonical Notch
target HES1 (P < 0.001 by ANOVA for all comparisons) (C). Data show means 6 SEM. AU, arbitrary units.
NOTCH ACTIVATION IN DIABETES AND FATTY LIVER
4054 DIABETES, VOL. 62, DECEMBER 2013 diabetes.diabetesjournals.org
increased in obesity and insuli n resistance as in mouse
models. We performed a pilot study in 42 morbidly obese
patients undergoing liver biopsy at time of gastric banding.
Full demographic and clinical information is summarized
in Table 1, but most of the subjects were female (76%),
with SS or NASH (88%), and nondiabetic (79%). Expres-
sion of canonical Notch target genes of the HES/HEY
family positively correlated with hepatic expression of
G6PC and PC K1 (Table 2 a nd Fig. 2A). Of note, we
saw no correlation in severel y obese patients between
Notch signaling and age, sex, or BMI (data not shown),
which suggests that the correlations between HES/HEY
and G6PC/ PCK1 were in dependen t of overall adiposity,
but correlations persisted or even strengthened when
FIG. 1. Continued.
L. VALENTI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 62, DECEMBER 2013 4055
diabetic patients were excluded from the analysis
(Table 2).
Liver consists primarily of hepatocytes, but also non-
hepatocyte residents, including endothelial, phagocytic
Kupffer, and stellate cells (31). The relative contribution of
hepatocytes to overall hepatic Notch signaling was unclear
from gene expression studies from the whole liver, so we
performed immunouorescence for Notch targets on
a representative subset of patients across a range of gly-
cemic control. We observed predominantly hepatocyte
staining of Notch targets HEY1 and HEYL (Fig. 2B). HEY1
hepatocyte staining and HEY1 gene expression was very
well correlated (Fig. 2C), as was HEYL staining and HE YL
expression (not shown), suggesting he patic gene expres-
sion is a good surrogate for hepatocyte protein levels.
Further, HEY1 gene expression strongly correlated with
HES1, HEY1, and other Notch targets (F ig. 2D and data
not shown), allowing HEY1 staining as a surrog ate for
global hepatocyte Notch activation. Having validated
the technique, we next examined sections from age-
and BMI-matched normoglycemic and diabetic patients
and noted a marked increase in HEY1 and HEYL
staining in hyperglycemic patients (Fig. 2E and F and
data not shown ). These data establish that Notch sig-
naling is present in adult hepatocytes and increased in
T2D.
FIG. 2. Notch activity in liver increases with hyperglycemia. A: Correlation of Notch targets of the HES/HEY family with the expression of genes
controlling hepatic glucose production, G6PC and PCK1. B: Representative liver sections, stained with antibodies to HEY1 (green, left) or HEYL
(red, middle) and counterstained with DAPI (blue), with merged image (right). Hepatic HEY1 expression correlates with Hey1 staining (P < 0.001
by ANOVA) (C) and HES1 expression (P < 0.001 by ANOVA) (D). Immunouorescence (E) and quantitation (F) of staining for HEY1 in formalin-
xed liver sections from non-T2D and T2D patients undergoing liver biopsy during gastric bypass surgery. The very bright spots are auto-
uorescent erythrocytes and are not included in the quantitation. Data show means 6 SEM. *P < 0.05 vs. non-T2D patients. AU, arbitrary units.
TABLE 2
Correlation of Notch gene targets with G6PC and PCK1 expression in 42 patients undergoing bariatric surgery
Overall series Patients without diabetes
G6PC PCK1 G6PC PCK1
Gene r Pearson r Pearson r Pearson r Pearson
HES1 +0.39 0.011 +0.47 0.0016 +0.44 0.006 +0.35 0.03
HES6 +0.24 NS +0.33 0.018 +0.35 0.03 +0.46 0.003
HES7 +0.27 NS +0.35 0.033 +0.51 0.007 +0.45 0.008
HEY1 +0.27 NS +0.42 0.002 +0.44 0.006 +0.41 0.01
NOTCH ACTIVATION IN DIABETES AND FATTY LIVER
4056 DIABETES, VOL. 62, DECEMBER 2013 diabetes.diabetesjournals.org
Hepatic Notch signaling is increased in insulin
resistance. The positive correlation of HES/HEY family
genes with G6PC/PCK1 in obese patients (Table 2), cou-
pled with increased HEY1 staining in hyperglycemia,
suggested that Notch s ignaling is i ncreased in insulin-
resistant li ver. To d etermine w hether hepat ic Notch
signaling was similarly induced in leaner patients and
m ight precede development of frank diabetes in insulin
FIG. 2. Continued.
TABLE 3
HES1 expression correlates independently with HOMA-IR as well as NAS scores
Correlation coefcient SE Unadjusted P value* Adjusted P value**
Age (per 10 years) 0.03 0.05 NS NA
BMI (kg/m
2
) 0.01 0.02 NS NA
Glucose (per 10 mg/dL increase) 0.00 0.02 NS NA
Insulin 0.02 0.01 0.002 0.03
ALT (per 10 IU/L increase) 0.07 0.02 0.003 NA
HOMA-IR 0.11 0.03 0.002 NA
% Steatosis (per 10% increase) 0.05 0.02 0.011 NA
NAS 0.11 0.03 ,0.0001 0.03
NA, not addressed. *At univariate analysis (generalized linear model). **At multivariate analysis (generalized linear model) including insulin
levels and NAS score, the strongest variables related to insulin resistance and histological damage, respectively, at univariate analysis.
L. VALENTI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 62, DECEMBER 2013 4057
resistance, we analyzed specimens from 38 consecutive
outpatients undergoing percutaneous liver biopsy. Full
demographics can be found in Table 1, but this group was
97% male, with an average BMI in the overweight range
(2530 kg/m
2
). In these patients, we again noted that HES/
HEY family genes correlated with G6PC and PCK1, as well
as with each other (data not shown), suggesting that Notch
and gluconeogenic gene expression coregulation is not
specic to obese patients. As in the bariatric surgery co-
hort, Notch signaling did not vary by age, BMI, or abdominal
circumference, but as predicted, we found a signicant
positive correlation with Notch targets (HES1) and plasma
FIG. 3. Notch activity in hepatocytes increases with hyperglycemia. Notch-luciferase reporter (Csl-luc) expression in Hepa1c1c7 hepatoma cells
(A), and Notch target gene expression in primary hepatocytes from mice (B) or human donors (C) is increased with transient exposure to hy-
perglycemic (25 mmol/L glucose) conditions. Insulin reduces Notch activation in Hepa1c1c7 hepatoma cells when cultured in normoglycemic (D),
but not hyperglycemic conditions (E), even as glucagon has a synergistic effect. F: Insulin fails to repress Hes1 expression in hepatocytes derived
from mice lacking hepatic Notch signaling. G: Hyperglycemic-induced Notch reporter expression in Hepa1c1c7 hepatoma cells is abrogated by
transduction with N1-decoy, which blocks ligand-dependent Notch signaling. Data show means 6 SEM. *P < 0.05, **P < 0.01, ***P < 0.001 vs.
5 mmol/L glucose or control (Cre- or GFP-transduced) cells. AU, arbitrary units.
NOTCH ACTIVATION IN DIABETES AND FATTY LIVER
4058 DIABETES, VOL. 62, DECEMBER 2013 diabetes.diabetesjournals.org
insulin levels (not shown) and HOMA-IR, independent of
confounding factors (Table 3), which was not mitigated by
exclusion of diabetic patients (data not shown). Multivari-
ate logistic regression analysis showed increased HES1
expression was positively associated with the presence of
insulin resistance of diabetes (odds ratio [OR] 2.11 [95% CI
1.538]), together with age (OR 1.17 [95% CI 1.051.40]) and
steatosis (OR 5.5 [95% CI 1.353]), independently of BMI.
These data establish that Notch activation is found in the
insulin-resistant liver, preceding frank hyperglycemia and
the development of diabetes.
Insulin and hyperglycemia directly and reciprocally
affect Notch signaling. To test the hypot hesis that
the apparent regulation of hepatic Notch signaling by
insulin resistance and hyperglycemia is direct and cell-
autonomous, we transfected hepatoma cells with a Notch-
reporter luciferase construct and found increased Notch
activation in hyperglycemic as opposed to basal conditions
(Fig. 3A). Similarly, endogenous Notch target expression
was higher in primary mouse or human hepatocytes tran-
siently exposed to hyperglycemia (Fig. 3B and C). Insulin
treatment had the opposite effect on Notch signaling, with
insulin-treated cells showing decreased reporter activation
(Fig. 3D). Interestingly, the inhibitory effect of insulin, but
not a synergistic effect of glucagon, was lost in cells
chronically cultured in hyperglycemic conditions (Fig. 3E),
Further, insulin was no longer able to repress Hes1 ex-
pression in primary hepatocytes derived from mice lacking
Rbp-Jk (8), the common transcriptional effector of Notch14
signaling (Fig. 3F). Similarly, pharmacologic application
of a novel Notch antagonist, an ectodomain decoy that
quenches ligand-dependent Notch signaling (24), abrogated
hyperglycemia-induced Notch-reporter activity (Fig. 3G). In
sum, these data suggest a cell-autonomous, dynamic regu-
lation of hepatic Notch signaling by metabolic stimuli.
Hepatic Notch signaling is positively correlated with
hepatic steatosis and inammation. Beyond the posi-
tive correlation between HES/HEY gene expression and
measures of insulin resistance and hyperglycemia, we
predicted that Notc h signaling would correlate with he-
patic lipid content and markers of necroinammation. In-
deed, when patients are subdivided by liver histology, we
nd that HES1 and other Notch target genes are strongly
upregulated across the spectrum from normal liver to SS
to NASH, and further still in insulin resistance (Fig. 4A)or
in patients with T2D (Fig. 4B). Notch target staining was
similarly increased in hepatocytes of NASH patients com-
pared with SS pa tients (Fig. 4C).
In fact, HES1 expression more closely correlated to
measures of hepatic inammation than to steatosis, with
stronger coefcients of correlation with NAS and ALT
levels than with the percentage of steatosis (Fig. 5AC and
Table 3). As such, patients with a NAS score of 02, cor-
relating with a low risk of NASH (29,32), had lower Notch,
ligand, and HES/HEY family gene expression than those
with scores of 3 or higher (Fig. 5DF). Furthermore, the NAS
score was associated with HES1 expression independently of
insulin resistance (Table 3). In sum, these data suggest that
hepatic Notch signaling is elevated in insulin resistance as
well as in NASH .
FIG. 4. Notch-dependent gene expression is progressively increased in insulin resistance and NAFLD severity. Quantitative PCR for HES1 and
other Notch target genes from liver biopsy samples from patients with pathologically conrmed SS, NASH, or normal liver, further subdivided as insulin-
sensitive (HOMA-IR <2.5) vs. insulin-resistant (HOMA-IR >2.5) (A), or T2D patients (B), are shown. C: Hepatocyte Notch target expression is in-
creased in patients with NASH. Scale bars are 50 mm long. Data show means 6 SEM. *P < 0.05 vs. T2D/SS; ***P < 0.001 by ANOVA. AU, arbitrary units.
L. VALENTI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 62, DECEMBER 2013 4059
DISCUSSION
Notch signaling has been extensively studied in the con-
text of differentiation or cancer (10), but its metabolic
functions are novel. Rodent studies have demonstrated
a postdevelopment role for Notch in the regulation of
obesity-induced insulin resistance/diabetes (8,26) and in
liver fat accumulation (9), but whether this would translate
to human disease was unclear. This work answers two
important questions: hepatic Notch signaling is 1) present
in human liver/hepatocytes, and 2) its activation is tied
FIG. 5. Notch activity correlates with hepatocyte necroinammation. Liver HES1 expression was plotted against NAS (A), serum ALT level (B),
and percentage of hepatic steatosis in patients undergoing percutaneous liver biopsy (C). Notch target (D), protein (E), and ligand gene expression
(F) are increased in liver from patients with higher NAS (NAS 3+) compared with patients at low risk for steatohepatitis (NAS 02). Data show
means 6 SEM. *P < 0.05, **P < 0.01, ***P < 0.001 vs. NAS 02. AU, arbitrary units.
NOTCH ACTIVATION IN DIABETES AND FATTY LIVER
4060 DIABETES, VOL. 62, DECEMBER 2013 diabetes.diabetesjournals.org
to the metabolic state of the organism, with an inde-
pendent positive correlation with measures of insulin
resistance and hepatic steatosis/inammation. Notch sig-
naling appears to be inappropriately reactivated in the
insulin-resistant liver, reprising its developmental role and
reassociating with its molecular partners from differentia-
tion that drives the metabolic effects of Notch signaling. For
instance, the Notch transcriptional effector Rbp-Jk binds to
and activates FoxO1 (33), a key transcriptional activator of
hormone-stimulated hepatic glucose production (2,34), in-
creasing functional hepatic insulin resistance (8). Similarly,
Notch signaling activates the nutrient-sensitive mTorc1
pathway in the liver, increasing de novo lipogenesis and
hepatic triglyceride (9). Mouse models with reduced hepatic
glucose production often have compensatory fatty liver due
to reoriented carbon ux; for instance, FoxO-knockout
mice (35,36) or mice treated with glucokinase activators
(37). Reduced Notch action allows the dissociation of
insulin-signaling pathways in the liver, redressing insulin
resistance without causing undue nutrient sensitivity, al-
lowing for this rare dual therapeutic benet independent of
effects on body weight or adiposity.
It is intriguing that Notch activation more strongly cor-
relates with markers of steatohepatitis, including the NAS
and ALT levels, than with steatosis itself. With better im-
aging techniques (38), clinicians can increasingly di-
agnose excess hepatic fat (5). NAFLD is extraordinarily
common, with a prevalence approaching 30% in many
populations (39), leaving a therapeutic dilemma because
only a subset of patients progress to NASH (40). As such,
increased Notch signaling may represent a biomarker or
even a causative factor for progression from SS to NASH. It
is noteworthy that Notch pathway activation has similarly
been found in human hepatocellular carcinoma (41) and
causes hepatic brosis and tumor formation in mice (4143),
which potentially explains the predisposition of patients
with NASH to develop cirrhosis and hepatocellular carci -
noma (44,45).
One of the major limitations of this type of observational
study is that we are able to detect Notch signaling, as well
as markers of insuli n sensitivity and hepatic fat, at one
moment in time. Longitudinal studies (46) are required to
determine whether altered Notch signaling predates or
predicts the development of worsening steatosis or pro-
gression to NASH and hepatocellular carcinoma. Similarly,
whether increased Notch signaling precedes developm ent
of insulin resistance or heralds the transition to frank di-
abetes is unknown. In addition, it would be of interest to
know whethe r interventions to reduce insulin resistance,
or steatosis and associated inammation (47,48), also re-
duce hepatic Notch signaling. Finally, whether higher
Notch signaling seen in NASH patients reects both he-
patocyte and nonhepatocyte contribution requires clari-
cation, because this may shed light on how Notch signals
are transduced. These future studies will inform the
question whether Notch may be a therapeutic target for
insulin-resistance/diabetes or NASH.
It is premature, given these lingering questions and po-
tential safety concerns, to propose a clinical trial with the
use of Notch inhibitors, which are already in advanced
clinical development for cancer (49,50), for treatmen t of
T2D or NASH. This temptation exists, however, in the
current era of pandemic obesity. Our data suggest the
possibility of alternative uses for these existing therapeu-
tics to combat the various faces of obesity-related meta-
bolic disease in the 21st century.
ACKNOWLEDGMENTS
The work was partly funded by National Institutes of
Health grants DK-093604 (U.B.P.) and 5R01-CA136673
(C.J.S.), a Columbia Diabetes and Endocrinology Research
Center Pilot and Feasibility Grant (U.B.P.), the Edward
Mallinckrodt, Jr., Foundation Grant (U.B.P.), and Ricerca
Corrente Fondazione IRCCS Ca Granda, Milano (L.V.).
No potential conicts of interest relevant to this article
were reported.
L.V. and C.J.S. analyzed data and wrote the manuscript.
R.M.M. and C.K. designed and performed experiments and
analyzed data. R.R. collected and managed biological
samples for RNA analysis. M.M. selected and collected
histological samples and reviewed histological samples.
U.B.P. designed and performed experiments, analyzed data,
and wrot e the manuscript. U.B.P. is the g uarantor of this
work and, as such, had full access to all of the data in the
study and takes responsibility for the integrity of the data
and the accuracy of the data analysis.
The authors thank Domenico Accili and members of the
Accili Laboratory at Columbia University, as well as mem-
bers of the Metabolic Liver Diseases Laboratory at the
University of Milan , for insightful discussion of the data,
and Robin Goland at Columbia University for assistance in
study design and implementation. The authors acknowl-
edge excellent technical support from Jessie Lee and Taylor
Lu at Columbia University, and Rosa Lombardi, Anna
Ludovica Fracanzani, Enrico Mozzi, and Silvia Fargion at
the University of Milan for colle ction of clinical data and
biological samples.
REFERENCES
1. Savage DB, Semple RK. Recent insights into fatty liver, metabolic dyslipidae-
mia and their links to insuli n resistance. Curr Opin Lipidol 2010;21: 329336
2. Lin HV, Accili D. Hormonal regulation of hepatic glucose production in
health and disease. Cell Metab 2011;14:919
3. Li S, Brown MS, Goldstein JL. Bifurcation of insulin signaling pathway in
rat liver: mTORC1 required for stimulation of lipogenesis, but not in-
hibition of gluconeogenesis. Proc Natl Acad Sci U S A 2010;107:34413446
4. Bugianesi E, Gastaldelli A, Vanni E, et al. Insulin resistance in non-diabetic
patients with non-alcoholic fatty liver disease: sites and mechanisms. Di-
abetologia 2005;48:634642
5. Dongiovanni P, Anstee QM, Valenti L. Genetic Predisposition in NAFLD
and NASH: impact on severity of liver disease and response to treatment.
Curr Pharm Des 2013;19:52195238
6. Villanova N, Moscatiello S, Ramilli S, et al. Endothelial dysfunction and
cardiovascular risk prole in nonalcoholic fatty liver disease. Hepatology
2005;42:473480
7. Kahn SE, Haffner SM, Heise MA, et al.; ADOPT Study Group . Glycemic
durability of rosiglitazone, metformin, or glyburide monotherapy. N Engl J
Med 2006;355:24272443
8. Pajvani UB, Shawber CJ, Samuel VT, et al. Inhibition of Notch signaling
ameliorates insulin resistance in a FoxO1-dependent manner. Nat Med
2011;17:961967
9. Pajvani UB, Qiang L, Kangsamaksin T, Kitajewski J, Ginsberg HN, Accili D.
Inhibition of Notch uncouples Akt activation from hepatic lipid accumu-
lation by decreasing mTorc1 stability. Nat Med 2013;19:10541060
10. Bolós V, Grego-Bessa J, de la Pompa JL. Notch signaling in development
and cancer. Endocr Rev 2007;28:339363
11. Dufraine J, Funahashi Y, Kitajewski J. Notch signaling regulates tumor
angiogenesis by diverse mechanisms. Oncogene 2008;27:51325137
12. Swiatek PJ, Lindsell CE, del Amo FF, Weinmaster G, Gridley T. Notch1 is
essential for postimplantation development in mice. Genes Dev 1994 ;8:
707719
13. Oka C, Nakano T, Wakeham A, et al. Disruption of the mouse RBP-Jk gene
results in early embryonic death. Development 1995;121:32913301
14. Shen J, Bronson RT, Chen DF, Xia W, Selkoe DJ, Tonegawa S. Skeletal and
CNS defects in Presenilin-1-decient mice. Cell 1997;89:629639
15. Shawber CJ, Kitajewski J. Notch function in the vasculature: insights from
zebrash, mouse and man. Bioessays 2004;26:225-234
L. VALENTI AND ASSOCIATES
diabetes.diabetesjournals.org DIABETES, VOL. 62, DECEMBER 2013 4061
16. Flynn DM, Nijjar S, Hubscher SG, et al. The role of Notch receptor ex-
pression in bile duct development and disease. J Pathol 2004;204:5564
17. Ellisen LW, Bird J, West DC, et al. TAN-1, the human homolog of the
Drosophila notch gene, is broken by chromosomal translocations in T
lymphoblastic neoplasms. Cell 1991;66:649661
18. Haruki N, Kawaguchi KS, Eichenberger S, et al. Dominant-negative Notch3
receptor inhibits mitogen-activated protein kinase pathway and the growth
of human lung cancers. Cancer Res 2005;65:35553561
19. Gallahan D, Callahan R. The mouse mammary tumor associated gene INT3
is a unique member of the NOTCH gene family (NOTCH4). Oncogene 1997;
14:18831890
20. Nijjar SS, Crosby HA, Wallace L, Hubscher SG, Strain AJ. Notch receptor
expression in adult human liver: a possible role in bile duct formation and
hepatic neovascularization. Hepatology 2001;34:11841192
21. Nijjar SS, Wallace L, Crosby HA, Hubscher SG, Strain AJ. Altered Notch
ligand expression in human liver disease: further evidence for a role of the
Notch signaling pathway in hepatic neovascularization and biliary ductular
defects. Am J Pathol 2002;160:16951703
22. Köhler C, Bell AW, Bowen WC, Monga SP, Fleig W, Michalopoulos GK.
Expression of Notch-1 and its ligand Jagged-1 in rat liver during liver re-
generation. Hepatology 2004;39:10561065
23. Wang L, Wang CM, Hou LH, et al. Disruption of the transcription factor
recombination signal-binding protein-Jk (RBP-J) leads to veno-occlusive dis-
ease and interfered liver regeneration in mice. Hepatology 2009;49:268277
24. Funahashi Y, Hernandez SL, Das I, et al. A Notch1 ectodomain construct
inhibits endothelial notch signaling, tumor growth, and angiogenesis.
Cancer Res 2008;68:47274735
25. Funahashi Y, Shawber CJ, Sharma A, Kanamaru E, Choi YK, Kitajewski J.
Notch modulates VEGF action in endothelial cells by inducing matrix
metalloprotease activity. Vasc Cell 2011;3:2
26. Fukuda D, Aikawa E, Swirski FK, et al. Notch ligand delta-like 4 blockade
attenuates atherosclerosis and metabolic disorders. Proc Natl Acad Sci
U S A 2012;;109:E1868E1877.
27. Rametta R, Mozzi E, Dongiovanni P, et al. Increased insulin receptor
substrate 2 expression is associated with steatohepatitis and altered lipid
metabolism in obese subjects. Int J Obes 2013;37:986992.
28. Muniyappa R, Lee S, Chen H, Quon MJ. Current approaches for assessing
insulin sensitivity and resistance in vivo: advantages, limitations, and ap-
propriate usage. Am J Physiol Endocrinol Metab 2008;294:E15E26
29. Kleiner DE, Brunt EM, Van Natta M, et al.; Nonalcoholic Steatohepatitis
Clinical Research Network. Design and validation of a histological scoring
system for nonalcoholic fatty liver di sease. H epatology 2005;41:1313
1321
30. Shawber CJ, Funahashi Y, Francisco E, et al. Notch alters VEGF re-
sponsiveness in human and murine endothelial cells by direct regulation of
VEGFR-3 expression. J Clin Invest 2007;117:33693382
31. Baratta JL, Ngo A, Lopez B, Kasabwalla N, Longmuir KJ, Robertson RT.
Cellular organization of normal mouse liver: a histological, quantitative
immunocytochemical, and ne structural analysis. Histochem Cell Biol
2009;131:713726
32. Sanyal AJ, Brunt EM, Kleiner DE, et al. Endpoints and clinical trial design
for nonalcoholic steatohepatitis. Hepatology 2011;54:344353
33. Kitamura T, Kitamura YI, Funahashi Y, et al. A Foxo/Notch pathway
controls myogenic differentiation and ber type specication. J Clin Invest
2007;117:24772485
34. Matsumoto M, Pocai A, Rossetti L, Depinho RA, Accili D. Impaired regu-
lation of hepatic glucose production in mice lacking the forkhead tran-
scription factor Foxo1 in liver. Cell Metab 2007;6:208216
35. Tao R, Wei D, G ao H, Liu Y, DePinho RA, Dong XC. Hep atic FoxOs
regulate lipid metabolism via modu lation of expression of the nicotin-
amide phosphoribosyltransferase gene. J Biol Chem 2011;286:14681
14690
36. Haeusler RA, Pratt-Hyatt M, Welch CL, Klaassen CD, Accili D. Impaired
generation of 12-hydroxylated bile acids links hepatic insulin signaling
with dyslipidemia. Cell Metab 2012;15:65
74
37. De Ceuninck F, Kargar C, Ilic C, et al. Small molecule glucokinase acti-
vators disturb lipid homeostasis and induce fatty liver in rodents: a warn-
ing for therapeutic applications in humans. Br J Pharmacol 2013;168:
339353
38. Schwenzer NF, Springer F, Schraml C, Stefan N, Machann J, Schick F.
Non-invasive assessment and quantication of liver steatosis by ultra-
sound, computed tomography and magnetic resonance. J Hepatol 2009;51:
433445
39. Bhala N, Jouness RI, Bugianesi E. Epidemiology and natural history of
patients with NAFLD. Curr Pharm Des 2013;19:51695176.
40. Loria P, Adinol LE, Bellentani S, et al.; NAFLD Expert Commit tee of the
Associazione Italiana per lo studio del Fegato. Practice guidelines for the
diagnosis and management of nonalcoholic fatty liver disease. A decalogue
from the Italian Association for the Study of the Liver (AISF) Expert
Committee. Dig Liver Dis 2010;42:272282
41. Villanueva A, Alsinet C, Yanger K, et al. Notch signaling is activated in
human hepatocellular carcinoma and induces tumor formation in mice.
Gastroenterology 2012;143:16601669.e1667
42. Tschaharganeh DF, Chen X, Latzko P, et al. Yes-associated protein up-
regulates Jagged-1 and activates the Notch pathway in human hepatocel-
lular carcinoma. Gastroenterology 2013;144:15301542.e12
43. Chen Y, Zheng S, Qi D, et al. Inhibit ion of Notch signaling by a g-secretase
inhibitor attenuates hepatic brosis in rats. PLoS ONE 2012;7:e46512
44. Anstee QM, Targher G, Day CP. Progression of NAFLD to diabetes melli-
tus, cardiovascular disease or cirrhosis. Nat Rev Gastroenterol Hepatol
2013;10:330344
45. White DL, Kanwal F, El-Serag HB. Association between nonalcoholic fatty
liver disease and risk for hepatocellular cancer, based on systematic re-
view. Clin Gastroenterol Hepatol 2012;10:13421359.e1342
46. Musso G, Cassader M, Bo S, De Michieli F, Gambino R. Sterol regulatory
element-binding factor 2 (SREBF-2) predicts 7-year NAFLD incidence
and severity of liver disease and lipoprotein and glucose dysmetabolism.
Diabetes 2013;62:11091120
47. Lavine JE, Schwimmer JB, Van Natta ML, et al.; Nonalcoholic Steatohep-
atitis Clinical Research Network. Effect of vitamin E or metformin for
treatment of nonalcoholic fatty liver disease in children and adolescents:
the TONIC randomized controlled trial. JAMA 2011;305:16591668
48. Sanyal AJ, Chalasani N, Kowdley KV, et al.; NASH CRN. Pioglitazone,
vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med 2010;
362:16751685
49. Noguera-Troise I, Daly C, Papad opoulos NJ, et al. Blockade of Dll4 inhibits
tumour growth by promoting non-productive angiogenesis. Nature 2006;
444:10321037
50. Wu Y, Cain-Hom C, Choy L, et al. Therapeutic antibody targeting of in-
dividual Notch receptors. Nature 2010;464:10521057
NOTCH ACTIVATION IN DIABETES AND FATTY LIVER
4062 DIABETES, VOL. 62, DECEMBER 2013 diabetes.diabetesjournals.org
... Accumulating evidence points to a major involvement of the Notch pathway in hepatic insulin resistance, liver steatosis, and atherosclerosis [13][14][15]. Only a handful of studies, however, have focused on obesity and AT-related inflammation, and these from the adipocyte point of view. ...
Article
Full-text available
The importance of macrophages in adipose tissue (AT) homeostasis and inflammation is well established. However, the potential cues that regulate their function remain incompletely understood. To bridge this important gap, we sought to characterize novel pathways involved using a mouse model of diet‐induced obesity. By performing transcriptomics analysis of AT macrophages (ATMs), we found that late‐stage ATMs from high‐fat diet mice presented with perturbed Notch signaling accompanied by robust proinflammatory and metabolic changes. To explore the hypothesis that the deregulated Notch pathway contributes to the development of AT inflammation and diet‐induced obesity, we employed a genetic approach to abrogate myeloid Notch1 and Notch2 receptors. Our results revealed that the combined loss of Notch1 and Notch2 worsened obesity‐related metabolic dysregulation. Body and AT weight gain was higher, blood glucose levels increased and metabolic parameters were substantially worsened in deficient mice fed high‐fat diet. Moreover, serum insulin and leptin were elevated as were triglycerides. Molecular analysis of ATMs showed that deletion of Notch receptors escalated inflammation through the induction of an M1‐like pro‐inflammatory phenotype. Our findings thus support a protective role of myeloid Notch signaling in adipose tissue inflammation and metabolic dysregulation.
... We observed that the expression levels of key lipolytic genes (atgl, lpl, and ucp2) were decreased, indicating a slowed rate of lipolytic metabolism in the liver. Moreover, it has been shown that activation of notch signaling in hepatocytes causes decreased glucose metabolism and adipogenesis, leading to lipid accumulation [73]. This was also verified by the upregulation of notch1a expression. ...
Article
Full-text available
The appropriate level of dietary lipids is essential for the nutrient requirements, rapid growth, and health maintenance of aquatic animals, while excessive dietary lipid intake will lead to lipid deposition and affect fish health. However, the symptoms of excessive lipid deposition in the liver of freshwater drums (Aplodinotus grunniens) remain unclear. In this study, a 4-month rearing experiment feeding with high-fat diets and a 6-week starvation stress experiment were conducted to evaluate the physiological alteration and underlying mechanism associated with lipid deposition in the liver of A. grunniens. From the results, high-fat-diet-induced lipid deposition was associated with increased condition factor (CF), viscerosomatic index (VSI), and hepatosomatic index (HSI). Meanwhile, lipid deposition led to physiological and metabolic disorders, inhibited antioxidant capacity, and exacerbated the burden of lipid metabolism. Lipid deposition promoted fatty acid synthesis but suppressed catabolism. Specifically, the transcriptome and metabolome showed significant enrichment of lipid metabolism and antioxidant pathways. In addition, the interaction analysis suggested that peroxisome proliferator-activated receptor (PPAR)-mediated 13-S-hydroxyoctadecenoic acid (13 (s)-HODE) could serve as the key target in regulating lipid metabolism and oxidative stress during lipid deposition in A. grunniens. Inversely, with a lipid intake restriction experiment, PPARs were confirmed to regulate lipid expenditure and physiological homeostasis in A. grunniens. These results uncover the molecular basis of and provide specific molecular targets for fatty liver control and prevention, which are of great importance for the sustainable development of A. grunniens.
... Evidence reveals that the Notch signaling pathway is valuable in the development of MAFLD. For example, the Notch signal increases the synthesis of hepatocyte fatty acids and lipid deposition by activating the mechanistic target of rapamycin complex 1 pathway [76]. The Notch signal can regulate cell proliferation and inhibit apoptosis [77]. ...
Article
Full-text available
Metabolic-associated fatty liver disease (MAFLD) is currently the most common chronic liver disease worldwide. However, its pathophysiological mechanism is complicated, and currently, it has no FDA-approved pharmacological therapies. In recent years, mesenchymal stem cell (MSC) therapy has attracted increasing attention in the treatment of hepatic diseases. MSCs are multipotent stromal cells that originated from mesoderm mesenchyme, which have self-renewal and multipotent differentiation capability. Recent experiments and studies have found that MSCs have the latent capacity to be used for MAFLD treatment. MSCs have the potential to differentiate into hepatocytes, which could be induced into hepatocyte-like cells (HLCs) with liver-specific morphology and function under appropriate conditions to promote liver tissue regeneration. They can also reduce liver tissue injury and reverse the development of MAFLD by regulating immune response, antifibrotic activities, and lipid metabolism. Moreover, several advantages are attributed to MSC-derived exosomes (MSC-exosomes), such as targeted delivery, reliable reparability, and poor immunogenicity. After entering the target cells, MSC-exosomes help regulate cell function and signal transduction; thus, it is expected to become an emerging treatment for MAFLD. In this review, we comprehensively discussed the roles of MSCs in MAFLD, main signaling pathways of MSCs that affect MAFLD, and mechanisms of MSC-exosomes on MAFLD.
Preprint
Fat accumulation, de novo lipogenesis, and glycolysis are key drivers of hepatocyte reprogramming and the consequent metabolic dysfunction-associated steatotic liver disease (MASLD). Here we report that obesity leads to dysregulated expression of hepatic protein-tyrosine phosphatases (PTPs). PTPRK was found to be increased in steatotic hepatocytes in both humans and mice, and positively correlated with PPARγ-induced lipogenic signalling. High-fat-fed PTPRK knockout mice displayed reduced weight gain and hepatic fat accumulation. Phosphoproteomic analysis in primary hepatocytes and hepatic metabolomics identified fructose-1,6-bisphosphatase 1 and glycolysis as PTPRK targets in metabolic reprogramming. Silencing PTPRK in hepatoma cell lines resulted in reduced colony-forming ability and PTPRK knockout mice developed smaller tumours after diethylnitrosamine-induced hepatocarcinogenesis. Our study defines a novel role for PTPRK in regulating hepatic glycolysis, lipid metabolism, and tumour development. PTPRK inhibition may provide therapeutic possibilities in obesity-associated liver diseases. Highlights Hepatic receptor-type PTPs are increased in MASLD PTPRK is expressed in hepatocytes and upregulated in obesity PTPRK deficiency reduces body fat mass and liver steatosis in diet-induced obesity PTPRK regulates hepatic glycolysis and lipogenesis, promoting tumorigenesis
Article
Background & aims: Fatty acid translocase CD36 (CD36/FAT) is a widely expressed membrane protein with multiple immuno-metabolic functions. Genetic CD36 deficiency is associated with increased risk of metabolic dysfunction-associated fatty liver disease (MAFLD) in patients. Liver fibrosis severity mainly affects the prognosis in patients with MAFLD, but the role of hepatocyte CD36 in liver fibrosis of MAFLD remains unclear. Methods: A high-fat high-cholesterol diet and a high-fat diet with high-fructose drinking water were used to induce nonalcoholic steatohepatitis (NASH) in hepatocyte-specific CD36 knockout (CD36LKO) and CD36flox/flox (LWT) mice. Human hepG2 cell line was used to investigate the role of CD36 in regulating Notch pathway in vitro. Results: Compared to LWT mice, CD36LKO mice were susceptible to NASH diet-induced liver injury and fibrosis. The analysis of RNA-sequencing data revealed that Notch pathway was activated in CD36LKO mice. LY3039478, an inhibitor of γ-secretase, inhibited Notch1 protein S3 cleavage and Notch1 intracellular domain (N1ICD) production, alleviating liver injury and fibrosis in CD36LKO mice livers. Likewise, both LY3039478 and knockdown of Notch1 inhibited the CD36KO-induced increase of N1ICD production, causing the decrease of fibrogenic markers in CD36KO HepG2 cells. Mechanistically, CD36 formed a complex with Notch1 and γ-secretase in lipid rafts, and hence CD36 anchored Notch1 in lipid rafts domains and blocked Notch1/γ-secretase interaction, inhibiting γ-secretase-mediated cleavage of Notch1 and the production of N1ICD. Conclusions: Hepatocyte CD36 plays a key role in protecting mice from diet-induced liver injury and fibrosis, which may provide a potential therapeutic strategy for preventing liver fibrogenesis in MAFLD.
Article
Full-text available
The liver is the central organ for digestion and detoxification and has unique metabolic and regenerative capacities. The hepatobiliary system originates from the foregut endoderm, in which cells undergo multiple events of cell proliferation, migration, and differentiation to form the liver parenchyma and ductal system under the hierarchical regulation of transcription factors. Studies on liver development and diseases have revealed that SRY-related high-mobility group box 9 (SOX9) plays an important role in liver embryogenesis and the progression of hepatobiliary diseases. SOX9 is not only a master regulator of cell fate determination and tissue morphogenesis, but also regulates various biological features of cancer, including cancer stemness, invasion, and drug resistance, making SOX9 a potential biomarker for tumor prognosis and progression. This review systematically summarizes the latest findings of SOX9 in hepatobiliary development, homeostasis, and disease. We also highlight the value of SOX9 as a novel biomarker and potential target for the clinical treatment of major liver diseases.
Article
Di-(2-ethylhexyl) phthalate (DEHP) and mono-2-ethylhexyl phthalate (MEHP) can induce hepatic lipid metabolism disorders, while the molecular mechanism still remain unknown. We aim to explore the underlying mechanism of Notch signaling pathway on hepatic lipid accumulation induced by DEHP/MEHP. A total of 40 male wistar rats were exposed to DEHP (0, 5, 50, and 500 mg/kg/d) for 8 weeks, BRL-3A hepatocytes were exposed to MEHP (0, 10, 50, 100, and 200 μM) for 24 h. About 50 μM DAPT and 100 μg/mL Aspirin were used to inhibit Notch pathway and prevent inflammation, respectively. Real-Time PCR was performed to detect the mRNA expression, western blot and immunofluorescence were used to detect the protein expression. Lipids and inflammatory factors levels were determined by commercial kits. The results showed that DEHP/MEHP promoted the expression of Notch pathway molecules and lipids accumulation in rat livers/BRL-3A cells. The up-regulated Notch receptors were correlated with the TG levels in the rat liver. MEHP increased the levels of IL-8 and IL-1β. The lipids levels were reduced after anti-inflammation. The inhibition of Notch pathway reversed the elevation of inflammation and lipid accumulation caused by MEHP. In conclusion, this study demonstrated that DEHP/MEHP led to lipid accumulation in hepatocytes by up-regulating Notch pathway and the inflammation might play a key role in the process.
Article
Epidemiological evidence showed that patients suffering from obesity and T2DM are significantly at higher risk for chronic low-grade inflammation, oxidative stress, nonalcoholic fatty liver (NAFLD) and intestinal flora imbalance. Increasing evidence of pathological characteristics illustrates that some common signaling pathways participate in the occurrence, progression, treatment, and prevention of obesity and T2DM. These signaling pathways contain the pivotal players in glucose and lipid metabolism, e.g., AMPK, PI3K/AKT, FGF21, Hedgehog, Notch, and WNT; the inflammation response, for instance, Nrf2, MAPK, NF- kB, and JAK/STAT. Bioactive compounds from plants have emerged as key food components related to healthy status and disease prevention. They can act as signaling molecules to initiate or mediate signaling transduction that regulates cell function and homeostasis to repair and re-functionalize the damaged tissues and organs. Therefore, it is crucial to continuously investigate bioactive compounds as sources of new pharmaceuticals for obesity and T2DM. This review provides comprehensive information of the commonly shared signaling pathways between obesity and T2DM, and we also summarize the therapeutic bioactive compounds that may serve as anti-obesity and/or anti-diabetes therapeutics by regulating these associated pathways, which contribute to improving glucose and lipid metabolism, attenuating inflammation.
Article
Full-text available
NAFLD is a spectrum of progressive liver disease that encompasses simple steatosis, NASH, fibrosis and, ultimately, cirrhosis. NAFLD is recognized as the hepatic component of the metabolic syndrome, as these conditions have insulin resistance as a common pathophysiological mechanism. Therefore, NAFLD is strongly associated with type 2 diabetes mellitus and abdominal obesity. As lifestyles have become increasingly sedentary and dietary patterns have changed, the worldwide prevalence of NAFLD has increased dramatically and is projected to be the principal aetiology for liver transplantation within the next decade. Importantly, a growing body of clinical and epidemiological evidence suggests that NAFLD is associated not only with liver-related morbidity and mortality, but also with an increased risk of developing both cardiovascular disease and type 2 diabetes mellitus. This article reviews the evidence that suggests NAFLD is a multisystem disease and the factors that might determine interindividual variation in the development and progression of its major hepatic and extrahepatic manifestations (principally type 2 diabetes mellitus and cardiovascular disease).
Article
Full-text available
Liver fat deposition related to systemic insulin resistance defines non-alcoholic fatty liver disease (NAFLD) which, when associated with oxidative hepatocellular damage, inflammation, and activation of fibrogenesis, i.e. non-alcoholic steatohepatitis (NASH), can progress towards cirrhosis and hepatocellular carcinoma. Due to the epidemic of obesity, NAFLD is now the most frequent liver disease and the leading cause of altered liver enzymes in Western countries. Epidemiological, familial, and twin studies provide evidence for an element of heritability of NAFLD. Genetic modifiers of disease severity and progression have been identified through genome-wide association studies. These include the Patatin-like phosholipase domain-containing 3 (PNPLA3) gene variant I148M as a major determinant of inter-individual and ethnicity-related differences in hepatic fat content independent of insulin resistance and serum lipid concentration. Association studies confirm that the I148M polymorphism is also a strong modifier of NASH and progressive hepatic injury. Furthermore, a few large multicentre case-control studies have demonstrated a role for genetic variants implicated in insulin signalling, oxidative stress, and fibrogenesis in the progression of NAFLD towards fibrosing NASH, and confirm that hepatocellular fat accumulation and insulin resistance are key operative mechanisms closely involved in the progression of liver damage. It is now important to explore the molecular mechanisms underlying these associations between gene variants and progressive liver disease, and to evaluate their impact on the response to available therapies. It is hoped that this knowledge will offer further insights into pathogenesis, suggest novel therapeutic targets, and could help guide physicians towards individualised therapy that improves clinical outcome.
Article
The Notch family of cell surface receptors and its ligands are highly conserved proteins that regulate cell fate determination, including those involved in mammalian vascular development. We report that Notch induces VEGFR-3 expression in vitro in human endothelial cells and in vivo in mice. In vitro, Notch in complex with the DNA-binding protein CBF-1/suppressor of hairless/Lag1 (CSL) bound the VEGFR-3 promoter and transactivated VEGFR-3 specifically in endothelial cells. Through induction of VEGFR-3, Notch increased endothelial cell responsiveness to VEGF-C, promoting endothelial cell survival and morphological changes. In vivo, VEGFR-3 was upregulated in endothelial cells with active Notch signaling. Mice heterozygous for null alleles of both Notch1 and VEGFR-3 had significantly reduced viability and displayed midgestational vascular patterning defects analogous to Notch1 nullizygous embryos. We found that Notch1 and Notch4 were expressed in normal and tumor lymphatic endothelial cells and that Notch1 was activated in lymphatic endothelium of invasive mammary micropapillary carcinomas. These results demonstrate that Notch1 and VEGFR-3 interact genetically, that Notch directly induces VEGFR-3 in blood endothelial cells to regulate vascular development, and that Notch may function in tumor lymphangiogenesis.
Article
Background & Aims Nonalcoholic fatty liver disease (NAFLD) has been implicated as a cause of hepatocellular carcinoma (HCC). We performed a systematic review of epidemiology studies to confirm the association between these disorders. Methods We searched PubMed for original reports published from January 1992 to December 2011 that evaluated the association between NAFLD, nonalcoholic steatohepatitis (NASH), cryptogenic cirrhosis presumed to be NASH-related, and the risk of HCC. Studies were categorized as offering potential direct evidence (eg, cohort studies) or indirect evidence (eg, case-control, cross-sectional, or case-series studies) for an association. We analyzed data from a total of 17 cohort studies (3 population based, 9 clinic based [6 limited to patients with cirrhosis], and 5 natural history), 18 case-control and cross-sectional studies, and 26 case series. Results NAFLD or NASH cohorts with few or no cases of cirrhosis cases had a minimal risk for HCC (cumulative HCC mortality of 0%–3% for study periods up to 20 y). Cohorts with NASH and cirrhosis had a consistently higher risk (cumulative incidence ranging from 2.4% over 7 y to 12.8% over 3 y). However, the risk for HCC was substantially lower in these cohorts than for cohorts with hepatitis C–related cirrhosis. Factors that increased risk among cohorts with NASH and cirrhosis could not be determined, because most studies were not sufficiently powered for multivariate analysis. Conclusions This systematic review shows that despite several limitations, there is epidemiologic evidence to support an association between NAFLD or NASH and an increased risk of HCC; risk seems to be limited to individuals with cirrhosis.
Article
The Jagged and Delta family of transmembrane proteins are ligands for Notch receptors, which control the proliferation and/or differentiation of many cell lineages. Expression and localization of these ligands in the adult human liver has not been fully elucidated, nor whether dysregulation of these proteins contributes to liver disease processes. We have examined expression of the five known Notch ligands in human liver. Expression of Jagged-1 and Delta-4 mRNA was seen in normal and diseased liver tissue, whereas Jagged-2, Delta-1, and Delta-3 mRNA was undetectable. In primary liver cell isolates, Jagged-1 expression was found in all cell types, whereas Delta-4 was present in biliary epithelial and liver endothelial cells, but absent in hepatocytes. Interestingly, Jagged-1 mRNA expression was significantly up-regulated in diseased liver tissue. By immunohistochemistry, Jagged-1 expression was present on most structures in normal tissue. However in disease, strikingly strong Jagged-1 immunoreactivity was observed on many small neovessels and bile ductules. The expression of downstream modulators and effectors of Notch signaling was also detectable in purified cell isolates. This, together with aberrant Jagged-1 expression suggests that the Notch signaling pathway may play a role in the neovascularization and biliary defects observed in the liver during the development of cirrhosis.
Article
Increased hepatic lipid content is an early correlate of insulin resistance and can be caused by nutrient-induced activation of mammalian target of rapamycin (mTor). This activation of mTor increases basal Akt activity, leading to a self-perpetuating lipogenic cycle. We have previously shown that the developmental Notch pathway has metabolic functions in adult mouse liver. Acute or chronic inhibition of Notch dampens hepatic glucose production and increases Akt activity and may therefore be predicted to increase hepatic lipid content. Here we now show that constitutive liver-specific ablation of Notch signaling, or its acute inhibition with a decoy Notch1 receptor, prevents hepatosteatosis by blocking mTor complex 1 (mTorc1) activity. Conversely, Notch gain of function causes fatty liver through constitutive activation of mTorc1, an effect that is reversible by treatment with rapamycin. We demonstrate that Notch signaling increases mTorc1 complex stability, augmenting mTorc1 function and sterol regulatory element binding transcription factor 1c (Srebp1c)-mediated lipogenesis. These data identify Notch as a therapeutically actionable branch point of metabolic signaling at which Akt activation in the liver can be uncoupled from hepatosteatosis.
Article
Background & aims: Cancer cells often lose contact inhibition to undergo anchorage-independent proliferation and become resistant to apoptosis by inactivating the Hippo signaling pathway, resulting in activation of the transcriptional co-activator yes-associated protein (YAP). However, the oncogenic mechanisms of YAP activity are unclear. Methods: By using cross-species analysis of expression data, the Notch ligand Jagged-1 (Jag-1) was identified as a downstream target of YAP in hepatocytes and hepatocellular carcinoma (HCC) cells. We analyzed the functions of YAP in HCC cells via overexpression and RNA silencing experiments. We used transgenic mice that overexpressed a constitutively activated form of YAP (YAP(S127A)), and measured protein levels in HCC, colorectal and pancreatic tumor samples from patients. Results: Human HCC cell lines and mouse hepatocytes that overexpress YAP(S127A) up-regulated Jag-1, leading to activation of the Notch pathway and increased proliferation. Induction of Jag-1, activation of Notch, and cell proliferation required binding of YAP to its transcriptional partner TEA domain family member 4 (TEAD4); TEAD4 binding required the Mst1/2 but not β-catenin signaling. Levels of YAP correlated with Jag-1 expression and Notch signaling in human tumor samples and correlated with shorter survival times of patients with HCC or colorectal cancer. Conclusions: The transcriptional regulator YAP up-regulates Jag-1 to activate Notch signaling in HCC cells and mouse hepatocytes. YAP-dependent activity of Jag-1 and Notch correlate in human HCC and colorectal tumor samples with patient survival times, suggesting the use of YAP and Notch inhibitors as therapeutics for gastrointestinal cancer. Transcript profiling: microarray information was deposited at the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=jxepvsumwosqkve&acc=GSE35004).
Article
Non-alcoholic fatty liver disease (NAFLD) currently represents the most common liver disease in Western countries, being found in 25-30% of the general population. NAFLD embraces a wide range of metabolic hepatic damage characterised by steatosis and, in some cases, associated non-alcoholic steatohepatitis (NASH). The long-term hepatic prognosis of NAFLD patients depends on the histological stage at diagnosis: simple steatosis has a favourable outcome, whereas patients with NASH can develop cirrhosis and other liver-related complications, including hepatocellular carcinoma. Progression of fibrosis is thought to develop in up to one third of NASH patients, including the development of cirrhosis, but regression is also possible in pre-cirrhotic stages. Independent predictors of fibrosis are older age, diabetes, obesity, hypertension, and the degree of insulin resistance. Patients with NAFLD, particularly those with NASH, have a higher prevalence and incidence of clinically manifested cardiovascular disease, independently of classical cardiometabolic risk factors. Hepatocellular carcinoma (HCC) is usually diagnosed at a late stage, but it may also occur in non-cirrhotic NASH, as obesity and diabetes both independently increase the risk of developing HCC. Liver-related mortality is increased up to ten-fold in patients with NASH.