ArticlePDF Available

Mice Lacking Thioredoxin-interacting Protein Provide Evidence Linking Cellular Redox State to Appropriate Response to Nutritional Signals

Authors:

Abstract and Figures

Thioredoxin-interacting protein (Txnip) is a ubiquitous protein that binds with high affinity to thioredoxin and inhibits its ability to reduce sulfhydryl groups via NADPH oxidation. HcB-19 mice contain a nonsense mutation in Txnip that eliminates its expression. Unlike normal animals, HcB-19 mice have approximately 3-fold increase in insulin levels when fasted. The C-peptide/insulin ratio is normal, suggesting that the hyperinsulinemia is due to increased insulin secretion. Fasted HcB-19 mice are hypoglycemic, hypertriglyceridemic, and have higher than normal levels of ketone bodies. Ablation of pancreatic beta-cells with streptozotocin completely blocks the fasting-induced hypoglycemia/hypertriglyceridemia, suggesting that these abnormalities are due to excess insulin secretion. This is supported by increased hepatic mRNA levels of the insulin-inducible, lipogenic transcription factor sterol-responsive element-binding protein-1c and two of its targets, acetyl-CoA carboxylase and fatty acid synthase. During a prolonged fast, the hyperinsulinemia up-regulates lipogenesis but fails to down-regulate hepatic phosphoenolpyruvate carboxykinase mRNA expression. Hepatic ratios of reduced:oxidized glutathione, established regulators of gluconeogenic/glycolytic/lipogenic enzymes, were elevated 30% in HcB-19 mice, suggesting a loss of Txnip-enhanced sulfhydryl reduction. The altered hepatic enzymatic profiles of HcB-19 mice divert phosphoenolpyruvate to glyceroneogenesis and lipogenesis rather than gluconeogenesis. Our findings implicate Txnip-modulated sulfhydryl redox as a central regulator of insulin secretion in beta-cells and regulation of many of the branch-points of gluconeogenesis/glycolysis/lipogenesis.
No caption available
… 
No caption available
… 
Content may be subject to copyright.
Mice Lacking Thioredoxin-interacting Protein Provide Evidence
Linking Cellular Redox State to Appropriate Response to
Nutritional Signals*
Received for publication, February 5, 2004, and in revised form, March 24, 2004
Published, JBC Papers in Press, March 26, 2004, DOI 10.1074/jbc. M401280200
To Yuen Hui‡, Sonal S. Sheth§, J. Matthew Diffley‡, Douglas W. Potter‡, Aldons J. Lusis§,
Alan D. Attie, and Roger A. Davis‡
From the Mammalian Cell and Molecular Biology Laboratory, Department of Biology, Molecular Biology Institute and
Heart Institute, San Diego State University, San Diego, California 92182, §Molecular Biology Institute and Department
of Medicine, Microbiology and Human Genetics, University of California, Los Angeles, California 90095, and
Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706
Thioredoxin-interacting protein (Txnip) is a ubiqui-
tous protein that binds with high affinity to thioredoxin
and inhibits its ability to reduce sulfhydryl groups via
NADPH oxidation. HcB-19 mice contain a nonsense mu-
tation in Txnip that eliminates its expression. Unlike
normal animals, HcB-19 mice have 3-fold increase in
insulin levels when fasted. The C-peptide/insulin ratio is
normal, suggesting that the hyperinsulinemia is due to
increased insulin secretion. Fasted HcB-19 mice are hy-
poglycemic, hypertriglyceridemic, and have higher than
normal levels of ketone bodies. Ablation of pancreatic
-cells with streptozotocin completely blocks the fast-
ing-induced hypoglycemia/hypertriglyceridemia, sug-
gesting that these abnormalities are due to excess insu-
lin secretion. This is supported by increased hepatic
mRNA levels of the insulin-inducible, lipogenic tran-
scription factor sterol-responsive element-binding pro-
tein-1c and two of its targets, acetyl-CoA carboxylase
and fatty acid synthase. During a prolonged fast, the
hyperinsulinemia up-regulates lipogenesis but fails to
down-regulate hepatic phosphoenolpyruvate carboxy-
kinase mRNA expression. Hepatic ratios of reduced:oxi-
dized glutathione, established regulators of gluconeo-
genic/glycolytic/lipogenic enzymes, were elevated 30%
in HcB-19 mice, suggesting a loss of Txnip-enhanced
sulfhydryl reduction. The altered hepatic enzymatic
profiles of HcB-19 mice divert phosphoenolpyruvate to
glyceroneogenesis and lipogenesis rather than glucone-
ogenesis. Our findings implicate Txnip-modulated sulf-
hydryl redox as a central regulator of insulin secretion
in
-cells and regulation of many of the branch-points of
gluconeogenesis/glycolysis/lipogenesis.
The liver plays a central role in controlling the flow of nutri-
ents to and from peripheral tissues. The flux of carbon units
through hepatic pathways of lipid and carbohydrate metabo-
lism are rapidly changed in response to nutrition. In the tran-
sition from the fed to the fasted state, hepatocytes switch from
being primarily glycolytic and lipogenic to being gluconeogenic
and ketogenic. This occurs in response to a drop in blood insulin
and to a lesser degree, an increase in glucagon levels.
Lipids are exported by the liver in the form of triglyceride-
rich very low density lipoprotein (VLDL)
1
particles. Depending
on the metabolic state, there are two major sources of free fatty
acids that are channeled into hepatic triglyceride production.
In the fed state, hepatocytes up-regulate the expression of
glycolytic and lipogenic enzymes, primarily by increasing the
expression and activation of a master regulator, the transcrip-
tion factor SREBP-1c (1). This process converts carbohydrate
into fatty acids, which are then esterified to form triglycerides,
the major core lipid secreted by the liver as VLDL. In the fasted
state, adipose tissue releases free fatty acids from triglyceride
stores because insulin levels drop below a threshold required to
repress lipolysis and also because of glucagon action. Although
the liver does not actively synthesize fatty acids under fasting
conditions, it retains the ability to esterify adipose tissue-de-
rived free fatty acids and to export VLDL particles. As much as
50% of fatty acids taken up by the liver during fasting are
eventually released as VLDL triglycerides (2). Indeed, in some
animal species (e.g. ponies), prolonged fasting leads to hyper-
triglyceridemia (3).
Fasting leads to enhanced gluconeogenesis by the liver. This
is largely due to an increase in the expression of key gluconeo-
genic enzymes, phosphoenolpyruvate carboxykinase (PEPCK)
(4), fructose-1,6-bisphosphatase (FBPase) (5), and glucose-6-
phosphatase (G6Pase) (6). Thus, there is usually an inverse
relationship between gluconeogenesis and lipogenesis in the
liver (2, 6, 7).
Hypertriglyceridemia is one of the most common lipid disor-
ders in the human population. It is usually associated with
obesity, insulin resistance, and diabetes (8, 9) but is also ob-
served in people without these metabolic disorders (10, 11).
There is still a dearth of knowledge about the key genes that
contribute to hypertriglyceridemia. However, in the majority of
cases, excess triglycerides are derived from overproduction
rather than defective catabolism of VLDL (8, 9).
The HcB-19 mouse is a variant of the C3H strain that was
* This project was supported by National Institutes of Health Grant
HL51648 (to R. A. D.) and American Heart Association Scientist Devel-
opment Award 0330109N (to T. Y. H.). The costs of publication of this
article were defrayed in part by the payment of page charges. This
article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
To whom correspondence should be addressed: Mammalian Cell and
Molecular Biology Laboratory, Life Sciences Bldg. LS307, 5500 Cam-
panile Dr., San Diego State University, San Diego, CA 92182-4614. Tel.:
619-594-7936. Fax: 619-594-7937; E-mail: rdavis@sunstroke.sdsu.
edu.
1
The abbreviations used are: VLDL, very low density lipoproteins;
SREBP, sterol-responsive element-binding protein; FAS, fatty acid syn-
thase; ACC, acetyl-CoA carboxylase; G6P, glucose 6-phosphate;
G6Pase, glucose-6-phosphatase; F6P, fructose-6-phosphate; FBPase,
fructose-1,6-bisphosphatase; PEPCK, phosphoenolpyruvate carboxyki-
nase; Txnip, thioredoxin-interacting protein; STZ, streptozotocin;
DHAP, dihydroxyacetone phosphate.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 279, No. 23, Issue of June 4, pp. 24387–24393, 2004
© 2004 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.
This paper is available on line at http://www.jbc.org 24387
by guest on May 12, 2018http://www.jbc.org/Downloaded from
first described as a model of human familial combined hyper-
lipidemia (12). The gene responsible for the HcB-19 phenotype
is thioredoxin-interacting protein (Txnip) (13). A nonsense mu-
tation renders the mice Txnip-null, which may enhance the
activity of thioredoxin. Thioredoxin plays a role in buffering the
redox state of proteins in which the standard redox potential
(E°) of their disulfide bonds is within the physiological range
(14 16). The HcB-19 mice, therefore, provide a new window
into processes that are regulated by a redox rheostat.
EXPERIMENTAL PROCEDURES
Animals and Streptozotocin Treatment—HcB-19 and C3H/DiSnA
mice derived from the colony at the University of California, Los Ange-
les (12, 13, 17, 18) were maintained under standardized conditions and
fed with rodent chow 5015 and water ad libitum. The light-cycle hours
were between 6 a.m. and 6 p.m. Streptozotocin (STZ) was injected
intraperitoneally (50 mg/kg in 0.1 Msodium citrate buffer, pH 4.50)
daily for 5 consecutive days. Control animals received the same volume
of citrate buffer. Plasma glucose and triglyceride were assessed 21 days
after the initial treatment.
Plasma Metabolites and Insulin Assays—Blood samples were col-
lected from mice under isofluorane anesthesia in heparinized capillary
tubes. Plasma was obtained by centrifugation at 12,000 gfor 5 min at
4°C. Glucose and triglycerides in plasma were assayed with commer-
cial kits from Sigma. Insulin and C-peptide were assayed using the
ultra sensitive rat/mouse insulin RIA kit (Linco) and C-peptide RIA kit
(Linco), respectively.
Glucose and Galactose Tolerance Tests—Glucose or galactose (3 g/kg
of body weight in 0.9% saline) was injected (intraperitoneal) into fasting
C3H and HcB-19 mice. Blood samples were collected by retro-orbital
bleeding at 0, 15, 30, and 60 min after injection and centrifuged at
12,000 gfor 5 min at 4 °C. Glucose levels in the plasma were assayed
with a commercial glucose diagnostic kit from Sigma.
Glucose 6-Phosphate (G6P) Phosphohydrolase Assays—Microsomes
from overnight-fasted mice were thawed on ice and diluted with ice-cold
0.25 Msucrose solution to 30 ml/g of wet liver weight. The microsomal
membrane was disrupted by the addition of 0.2% (w/v) sodium deoxy-
cholate. Enzyme assays were carried out in 0.1 Msodium cacodylate
buffer, pH 6.5, containing 20 mMG6P containing 100
g of microsome
protein per 0.75-ml reaction mixture. Incubations were at 30 °C for 10
min. The reactions were stopped by the addition of 0.5 ml of ice-cold
trichloroacetic acid. Denatured protein was sedimented by centrifuga-
tion. Aliquots of the 200-
l supernatant fraction were assayed for
inorganic phosphate by a colorimetric kit from Sigma.
Measurement of Gluconeogenic Intermediates in the Liver—Animals
FIG.1.Plasma triglyceride (TG), glucose, and ketone body levels in HcB-19 and control C3H mice. C3H (control) and HcB-19 were fed
a chow diet and water ad libitum in a room with a 12 h light cycle (lights on from 0600 to 1800). Blood was obtained from fed (open bars) or fasted
(filled bars) mice at 1000 h. Plasma levels of triglyceride (panel A), glucose (panel B), and ketone bodies (panel C) were measured as described under
Experimental Procedures.Results are presented as the mean S.D. from 12 mice in each group. The asterisk denotes statistical difference, p
0.01.
FIG.2.Comparison of G6Pase activ-
ities and abilities to utilize galactose
in HcB-19 and control C3H mice.
Panel A, liver microsomes were prepared
from overnight fasted C3H and HcB-19
mice. G6Pase activity was measured by
following the amount of phosphate re-
leased over time and normalized to the
amount of microsomal protein in each as-
say. Results are presented as mean
S.D. from four mice in each group. The
asterisk denotes statistical difference, p
0.05. Panel B, galactose (3 g/kg of body
weight in 0.9% saline) was injected (intra-
peritoneal) into fasting C3H (filled dia-
monds) and HcB-19 mice (open squares).
Blood samples were collected by retro-or-
bital bleeding at 0, 15, 30, and 60 min
after injection, and the amount of glucose
was determined. Results are presented as
the mean S.D. from five mice in each
group.
Txnip Loss Disrupts Appropriate Metabolic Response to Fasting24388
by guest on May 12, 2018http://www.jbc.org/Downloaded from
were starved for 18 h before sacrifice. The livers were immediately
removed and frozen in liquid nitrogen. The frozen tissue was ground in
a porcelain mortar pre-cooled with liquid nitrogen. About1goftissue
powder was mixed with 5 ml of 0.6 Mperchloric acid and centrifuged.
The deproteinized supernatant fraction was removed and adjusted to
pH 3.50 with 5 Mpotassium carbonate. Metabolites were estimated by
standard spectrophotometric enzymatic assays as previously described
(19, 20). G6P and fructose 6-phosphate (F6P) assays were carried out in
0.2 Mtriethanolamine buffer containing 0.2 mMNADP-Na
2
and5mM
MgCl
2
. Glucose-6-phospate dehydrogenase was then added, and the
production of NADPH was monitored by following the absorbance at
339 nm. The amount of G6P in the samples was calculated from the
amount of NADPH generated. After all G6P in the sample has been
consumed, phosphoglucose mutase was added to convert F6P to G6P.
The amount of F6P in the samples was estimated by the additional
production of NADPH. Dihydroxyacetone phosphate (DHAP), glyceral-
dehyde 3-phosphate, and fructose-1,6-bisphosphate were assayed were
sequentially in 0.2 Mtriethanolamine buffer containing 20 mMEDTA
and 17
MNADH. Glycerol-3-phosphate dehydrogenase was then
added, and the conversion of NADH to NAD was followed by the change
in absorbance at 339 nm. The amount of DHAP in the samples was
calculated from the amount of NADH consumed. After the reaction has
been completed, triose-phosphate isomerase was added to convert glyc-
eraldehyde 3-phosphate into DHAP. The amount of glyceraldehyde
3-phosphate in the samples was estimated by the additional consump-
tion of NADH. Finally, for the estimation of fructose-1,6-bisphosphate,
aldolase was added to convert fructose-1,6-bisphosphate to DHAP and
the change in NADH was measured.
Measurement of Glutathione Levels in the LiverGlutathione levels
in the liver were determined spectrophotometrically as described by
Anderson (21). Briefly, mice were sacrificed and perfused with ice-cold
PBS. Livers were excised and deproteinized with 10 volumes of 5%
5-sulfosalicylic acid. Total glutathione was determined using the 5,5-
dithiobis(2-nitrobenzoic acid)-GSSG reductase recycling assay. The pro-
duction of 5-thio-2-nitrobenzoate was followed at 412 nm and is propor-
tional to the amount of total glutathione in the sample. Oxidized
glutathione was determined by the same assay after blocking the thiol
group of GSH with 2-vinylpyridine.
Quantitative Real-time PCRTotal RNA was isolated from the livers
of mice by using the RNeasy mini kit (Qiagen). cDNA was synthesized
using the Superscript First Strand Synthesis kit (Invitrogen) from 2
g
of total RNA (DNase I-treated) and random hexamer. Real-time PCR
was performed in 96-well plates using the Bio-Rad iCycler machine.
Typically, the 25-
l reaction contained 12.5
l of 2xSYBR PCR Green
Master Mix (Bio-Rad), 1
l of cDNA, and 125 nMforward and reverse
primers. All reactions were done in triplicate and optimized by melt
curve analysis to ensure nonspecific products and primer dimers were
not formed. The primer sequences used for SREBP-1a, SREBP-1c,
SREBP-2, FAS, ACC, PEPCK, and apoB have been described (1, 22).
ApoB mRNA was used as the invariant control. The relative amount of
mRNA in each sample was determined by the comparative C
T
method.
Statistical AnalysisResults are given as the mean S.D. Statisti-
cal significance was determined by two-tailed ttest. Values of p0.05
were considered to be significant.
RESULTS
Loss of Txnip Is Associated with Fasting-induced Hypertri-
glyceridemia, Hypoglycemia, and KetosisAs expected, over-
night fasting caused triglycerides to decrease 3-fold in control
mice (Fig. 1A). In striking contrast, plasma triglycerides in-
creased 4-fold in HcB-19 mice in response to fasting (Fig. 1A).
The fasting-induced hypertriglyceridemia uniquely displayed
by HcB-19 mice is consistent with previous findings showing
that the livers of HcB-19 mice secrete more VLDL particles (12,
13). However, the finding that HcB-19 mice showed no hyper-
lipidemia in the non-fasted state (Fig. 1A) suggests their met-
abolic disorder is distinct from familial combined hyperlipi-
demia proposed by earlier reports (12, 13).
Hepatic VLDL production rapidly adapts to nutritional sta-
tus (23). Long term fasting is associated with decreased VLDL
assembly and secretion (24, 25). The anomalous response to
fasting by HcB-19 mice suggests that Txnip plays a role in
eliciting the appropriate metabolic response to fasting.
Normal mice are able to maintain normal plasma glucose
levels during fasting in part by stimulating hepatic gluconeo-
genesis. In contrast to the plasma glucose levels in excess of
100 mg/dl in control mice, HcB-19 mice became hypoglycemic
(66.7 mg/dl) after a 14-h fast (Fig. 1B). Consistent with a
previous report (12, 13), HcB-19 mice also showed a striking
6.6-fold increase in ketone bodies when fasted and a 2-fold
increase when fed (Fig. 1C).
Fasting-induced Hypertriglyceridemia and Hypoglycemia
Associated with Loss of Txnip Are Not Caused by Defective
Transport or Metabolism of Glucose 6-PhosphateThe fasting-
induced hypoglycemia and hypertriglyceridemia phenotype of
HcB-19 mice is similar to that of glycogen storage diseases
caused by a deficiency in hepatic G6Pase and/or the glucose
6-phosphate transporter in the endoplasmic reticulum (26, 27).
Our findings showing that hepatic microsomes from fasted
HcB-19 mice contain higher levels of G6Pase activity than
FIG.3.Glucose tolerance test. Glucose (3 g/kg of body weight in
0.9% saline) was injected (intraperitoneal) into fasting C3H (filled di-
amonds) and HcB-19 mice (open squares). Blood samples were collected
by retro-orbital bleeding at 0, 15, 30, and 60 min after injection, and the
amount of glucose was determined. Results are presented as the
mean S.D. from five mice in each group.
FIG.4.Steady-state levels of glycolytic/gluconeogenic intermediates in the livers of fasted HcB-19 and control C3H mice. Hepatic
contents of glycolytic/gluconeogenic intermediates in overnight-fasted control C3H (open bars) and HcB-19 (filled bars) mice were measured. Panel
A, steady-state levels of DHAP, glyceraldehyde 3-phosphate (GAP), and fructose-1,6-bis-phosphate (F-1,6-P2). Results are presented as the mean
S.D. from five mice in each group. Panel B, steady-state levels of G6P and F6P were assayed. Results are presented as the mean S.D. from five
mice in each group. The asterisk denotes statistical difference, p0.01.
Txnip Loss Disrupts Appropriate Metabolic Response to Fasting 24389
by guest on May 12, 2018http://www.jbc.org/Downloaded from
those from control mice (Fig. 2A) suggest that impaired G6Pase
activity is not responsible for the fasting-induced hypertriglyc-
eridemia, hypoglycemia, or ketosis associated with loss of
Txnip.
We also determined how loss of Txnip affects the ability of
liver in vivo to transport G6P into the lumen of the endoplasmic
reticulum and export it as glucose. Galactose is selectively
taken up by the liver for conversion to G6P, which can subse-
quently be used for the production of glucose for secretion into
the bloodstream. This process requires an intact function of the
G6Pase complex. Thus, the increase in plasma glucose after a
bolus of galactose provides an estimation of the relative flux of
substrate through G6Pase (28). As shown in Fig. 2B, injection
of a bolus of galactose (3 g/kg of body weight, intraperitoneal)
resulted in a similar increment in plasma glucose in both
groups of mice. These findings indicate that the fasting-in-
duced hypertriglyceridemia, hypoglycemia, and ketosis associ-
ated with loss of Txnip are not caused by defective transport or
metabolism of G6P.
Fasting-induced Hypertriglyceridemia and Hypoglycemia
Associated with Loss of Txnip Are Not Caused by Altered Rates
of Removal of Glucose from PlasmaTo examine the ability of
each group of mice to remove glucose from plasma, fasted mice
were challenged with a bolus of glucose (3 g/kg of body weight,
intraperitoneal) (Fig. 3). Before glucose administration,
HcB-19 mice exhibited lower plasma glucose levels that were
50 mg/dl less than those of control mice (Fig. 3). Both groups
exhibited similar net increments of plasma glucose as well as
similar rates of removal of glucose from plasma after the bolus
injection of glucose (Fig. 3). Thus, it appears that the fasting-
induced hypoglycemia associated with loss of Txnip is not
caused by accelerated removal of glucose from plasma and may
be the result of decreased hepatic gluconeogenesis and glucose
export.
Livers of Fasted HcB-19 Mice Display a Block Near the End
Point of Hepatic GluconeogenesisTo determine whether stim-
ulation of hepatic gluconeogenesis is impaired in fasting
HcB-19 mice, steady-state levels of glycolytic/gluconeogenic in-
termediates in the livers of fasted mice were measured (Fig. 4).
Although the hepatic levels of fructose-1,6-bisphosphate (F-1,6-
P
2
) and triose phosphate intermediates (dihydroxyacetone
phosphate and glyceraldehyde 3-phosphate) were similar in
both groups, levels of G6P and F6P were markedly decreased in
HcB-19 mice (70%, p0.01) (Fig. 4). Together with the
finding that HcB-19 mice can utilize G6P from galactose for
glucose production (Fig. 2B), these data suggest that hypogly-
cemia is due to a lack of supply of G6P and F6P in the terminal
steps of gluconeogenesis.
The reduction in G6P and F6P in the fasting HcB-19 livers
could be due to decreased FBPase activity. The mRNA and
protein levels of FBPase were similar in C3H and HcB-19 mice
FIG.5.Sulfhydryl redox status in the livers of fasting HcB-19
and control C3H mice. Levels of total GSH and GSSG were measured
from perfused livers of overnight-fasted C3H (open bars) and HcB-19
(filled bars) mice. Results are presented as mean S.D. from five mice
in each group. The asterisk denotes statistical difference, p0.05.
FIG.6.Plasma levels of insulin and C-peptide. Panel A, plasma insulin levels. C3H (control) and HcB-19 were fed a chow diet and water ad
libitum in a room with a 12-h light cycle (lights on from 0600 to 1800). Blood was obtained from fed (open bars) or fasted (filled bars) mice at 1000 h.
Plasma levels of insulin were measured by RIA. Panel B, plasma C-peptide levels in fasting mice. Blood was obtained from fasting C3H (open bars)
and HcB-19 (filled bars) mice at 1000 h. Plasma levels of C-peptide were measured by radioimmunoassay. Results are presented as the mean
S.D. from six mice in each group. The asterisk denotes statistical difference, p0.01.
FIG.7.Effects of streptozotocin treatment on the hypertriglyceridemia and hypoglycemia phenotypes exhibited by fasted HcB-19
mice. STZ was injected intraperitoneally (50 mg/kg in 0.1 Msodium citrate buffer, pH 4.50) daily for 5 days into C3H (open bars) and HcB-19 mice
(filled bars). Control animals in each strain of mice received the same volume of citrate buffer. After 21 days, blood samples were collected from
overnight-fasted mice in each group. Plasma levels of triglycerides (panel A) and glucose (panel B) were determined. Results are presented as the
mean S.D. from five mice in each group. The asterisk denotes statistical difference, p0.01.
Txnip Loss Disrupts Appropriate Metabolic Response to Fasting24390
by guest on May 12, 2018http://www.jbc.org/Downloaded from
(Northern and Western blotting, data not shown), suggesting
that the change in FBPase activity is due to post-translational
modification and/or allosteric control.
Loss of Txnip Is Associated with Increased Thiol Redox
(GSH/GSSG) Status, an Allosteric Regulator of Gluconeogene-
sis/GlycolysisFBPase enzyme activity is negatively regulated
by fructose 2,6-bisphosphate (5). This inhibition is exquisitely
dependent upon the thiol (cysteine) redox state of the enzyme,
the reduced form being more susceptible to the inhibition by
fructose2,6-bisphosphate (29). Thus, gluconeogenesis and gly-
colysis are reciprocally responsive to the ratio of reduced glu-
tathione:oxidized glutathione (GSH/GSSG) (30). A 50% in-
crease in the ratio of GSH/GSSG in renal tubules is associated
with an up to 20-fold increased rate of gluconeogenesis (30).
We, therefore, measured the GSH/GSSG ratio in the livers of
both groups of fasted mice. In HcB-19 mice, the hepatic GSH/
GSSG ratio was increased by 30% (p0.05) (Fig. 5). The 30%
increased GSH/GSSG ratio in the liver displayed by HcB-19
mice is sufficient to account for the apparent block in FBPase
observed in the livers of fasted HcB-19 mice (Fig. 4).
Loss of Txnip Is Associated with Increased Plasma Levels of
Insulin and C-peptideBecause insulin suppresses gluconeo-
genesis and negatively regulates FBPase activity, we measured
the plasma insulin levels in HcB-19 mice. As shown in Fig. 6A,
plasma insulin concentrations were significantly elevated in
both fed and fasting HcB-19 mice. In the fed state, plasma
insulin levels in HcB-19 mice were about 50% higher than that
of control. After an 18-h fast, the difference in plasma insulin
levels between the 2 groups of mice was 3-fold (Fig. 6A).
Plasma C-peptide levels in fasted HcB-19 mice also showed a
similar 3-fold increase (Fig. 6B). These findings suggest that
elevated plasma insulin levels in HcB-19 mice are linked to
enhanced pancreatic
-cell secretion rather than reduced he-
patic insulin clearance.
Livers from Fasted HcB-19 Mice Display a Selective Resist-
ance to HyperinsulinemiaSurprisingly, the hyperinsulinemia
displayed by fasted HcB-19 mice was not associated with a
detectable increase in the hepatic content of fructose 2,6-
bisphosphate or reduced activity of fructose-1,6-bisphos-
phatase (data not shown).
Normally, insulin suppresses the expression of PEPCK.
However, despite the 3-fold increase in insulin in fasted
HcB-19 mice relative to normal mice, PEPCK expression in
fasted HcB-19 mice was unchanged (i.e. not suppressed) com-
pared with fasted controls (Table I). Interestingly, fed mice
from both groups exhibited decreased expression of PEPCK
mRNA and increased expression of lipogenic SREBP-1c,
SREBP-2, ACC, and FAS compared with the levels displayed
when fasted (Table I).
A second physiological function of PEPCK is glyceroneogen-
esis (2). The hyperlipidemic phenotype of the fasted HcB-19
mice would be consistent with an abnormally increased diver-
sion of PEP toward glycerol production (via glycerol-3-phos-
phate dehydrogenase) and lipogenesis (31, 32). Consistent with
increased lipogenesis, SREBP-1c levels in the fasted HcB-19
mice were 3.3-fold higher than those of control mice (Table I).
As expected, the expression of SREBP-1c target genes, acetyl-
CoA carboxylase, and fatty acid synthase were also increased
2-fold in fasted HcB-19 mice (Table I).
Because the changes in lipogenesis correlate with hyperin-
sulinemia, we ablated the insulin-producing
-cells of the normal
and HcB-19 mice with STZ. In addition to the expected hyper-
glycemia associated with loss of insulin signaling, STZ treatment
completely blocked the fasting-induced hypertriglyceridemia in
the HcB-19 mice (Fig. 7). Thus, the fasting-induced hypertriglyc-
eridemia of the HcB-19 mice is dependent upon insulin.
DISCUSSION
Animals are able to reprogram their metabolic pathways to
adapt to changes in nutrient availability, hormonal status, and
energy demands. The mechanisms require sensing these stim-
uli and bringing about a change in the abundance or activation
state of metabolic enzymes and regulators of gene expression. A
recurring theme in metabolism is that cells sense their energy
chargeby responding to changes in ATP or AMP or to changes
in redox potentialas reflected in NAD(P)H/NAD(P) and
GSH/GSSG. The cysteine residues of many proteins exist in a
dynamic state between the reduced free sulfhydryl and the
oxidized disulfide bridge. There are many in vivo processes that
allow redox potential to equilibrate NAD(P)H/NAD
(P), GSH/
GSSG, and protein sulfhydryl reduction/oxidation. The ubiqui-
tous enzyme thioredoxin is mainly responsible for equilibrating
NAD(P)H/NAD
(P) redox potential with protein sulfhydryl re-
dox state via the oxidation of NADPH (3335). Thioredoxin
function can itself be modulated by its interaction with its
inhibitory protein partner, Txnip (36, 37). The availability of a
FIG.8.Regulation of glycolysis, gluconeogenesis, and triglyc-
eride biosynthesis by insulin, SREBP-1c, and cellular sulfhydryl
redox. Activities of key enzymes in glycolysis, gluconeogenesis, and
triglyceride (TG) synthesis are modulated by insulin (blue arrows),
SREBP-1c (red arrows), and thiol redox status (yellow arrows). In the
context of fasting HcB-19 mice with elevated plasma insulin, increased
hepatic SREBP-1c mRNA expression combined with a greater reduced
thiol redox status act in concert to activate enzymes in glycolysis and
triglyceride biosynthesis and to reciprocally suppress gluconeogenic
enzymes. Note that SREBP-1c is required to induce glucose-6-phos-
phate dehydrogenase in response to refeeding but is not required for
expression in fasted mice (see Ref. 57). TCA, tricarboxylic acid; HK,
hexokinase; GK, glucokinase; PFK, phosphofructokinase; Gly3PDH,
glycerol-3-phosphate (Gly-3-P) dehydrogenase; PDH, pyruvate dehy-
drogenase; ME, malic enzyme; ATPCL, ATP:citrate lyase; GPAT, glyc-
erol-3-phosphate acyltransferase; OAA, oxaloacetate; 1,3-BPG, 1,3-
bisphosphoglycerate; PC, pyruvate carboxylase; GAP, glyceraldehyde
3-phosphate.
Txnip Loss Disrupts Appropriate Metabolic Response to Fasting 24391
by guest on May 12, 2018http://www.jbc.org/Downloaded from
Txnip-null mutant mouse (13) provided an opportunity to dis-
cover the metabolic pathways that are affected by the loss of
Txnip. This study establishes a causal connection between loss
of Txnip resulting in enhanced sulfhydryl reduction and dys-
regulated carbohydrate and lipid metabolism. The work sug-
gests that Txnip plays a role in at least two tissue sites, the
liver and pancreatic
-cells.
To maintain homeostasis the liver must balance the flux of
carbon units into gluconeogenesis with metabolic needs by
sensing blood glucose and integrating this information with
energy stores and redox state. Glucose sensing occurs both
directly and through insulin. The discovery that SREBP-1c is
an insulin target explained how hepatocytes modify the expres-
sion of enzymes in glycolysis and lipogenesis appropriate to the
level of blood glucose (1, 38, 39).
In addition to blood glucose, cells respond to their own inter-
nal energy status. Fasting is associated with a more oxidized
sulfhydryl redox state (40). Our results establish a connection
between redox status, Txnip, and metabolic adaptation to fast-
ing. Instead of becoming more glucogenic, the fasting liver of a
Txnip-null mouse is lipogenic. The dearth of glucose leads to
hypoglycemia and ketosis.
The fasted HcB-19 mice are hyperinsulinemic. Yet, they fail
to suppress the expression of PEPCK. Together with the induc-
tion of SREBP-1c and its regulated genes, this presents a
picture of selective insulin resistance analogous to those previ-
ously reported by Shimomura and co-workers (41) in lipodys-
trophic mice. PEPCK catalyzes the formation of PEP, a precur-
sor for both glucose and glycerol production. In fasted HcB-19
mice, PEP is selectively channeled into glyceroneogenesis due
to the block in FBPase (Fig. 4) and the increased demand for
triglyceride synthesis caused by increased plasma levels of
fatty acids observed in HcB-19 mice (12, 13) returning to the
liver and increased hepatic synthesis due to increased expres-
sion of ACC and FAS (Table I).
Sulfhydryl redox state, as reflected by the ratio of cellular
GSH/GSSG, reciprocally regulates enzymes involved in glu-
coneogenesis and glycolysis (for review, see Ref. 42) such as
hepatic FBPase (gluconeogenesis) (43) and phosphofructoki-
nase (glycolysis) (44). In addition, sulfhydryl redox state also
modulates the activity of enzymes involved in fatty acid and
glycerolipid biosynthesis such as ATP-citrate lyase (45, 46),
FAS (47), glycerol-3-phosphate dehydrogenase (48), and glyc-
erol-3-phosphate acyltransferase (49) (summarized in Fig. 8).
The findings showing that GSH in the livers of fasted HcB-19
mice is 30% more reduced (Fig. 5) may explain the decreased
flux of carbon units through FBPase and increased production
of fatty acids and glycerolipids. As a result, carbon units are
secreted as triglycerides as a component of VLDL (12, 13). The
data showing increased hepatic content of mRNAs encoding the
lipogenic transcription factor SREBP-1c and two of its tran-
scriptional targets (ACC and FAS) (Table I) in fasted HcB-19
mice provides an additional mechanism through which insulin
enhances the flow of carbon units from glucose export into
hepatic VLDL secretion.
Our findings suggest a role for Txnip in the pancreatic
-cells. The increased insulin secretion in fasted HcB-19 mice
could be due to increased secretion by individual
-cells or
increased
-cells mass. We suspect the latter is more likely.
Txnip has been shown to block the thioredoxin-sulfhydryl
reduction by NADPH (36). This finding predicts that the
absence of Txnip in HcB-19 mice would lead to increased
thioredoxin reduction activity (50). Pancreatic
-cells contain
abundant levels of thioredoxin (51), which may contribute to
the intracellular processing and secretion of insulin (52). It is
believed that thioredoxin facilitates the processing and mat-
uration of insulin (36) and, thus, may prevent unfolded pro-
tein response-induced apoptosis of pancreatic
-cells (53, 54).
Transgenic overexpression of thioredoxin in pancreatic
-cells protects mice from STZ-induced and autoimmune di-
abetes (51). Txnip-thioredoxin interactions may be an effec-
tive target for promoting insulin secretion without causing
pancreatic
-cell apoptosis.
Glucose causes a large increase in Txnip expression in pan-
creatic
-cells (55) and in cultured fibroblasts (56). This re-
sponse links nutritional state (e.g. glucose uptake) with redox
state and subsequent metabolic regulation. Our findings fur-
ther suggest that the cellular redox state provides a unifying
parameter linking cellular response to nutritional state. This
response is tuned via Txnip-mediated changes in sulfhydryl
redox potential.
REFERENCES
1. Liang, G., Yang, J., Horton, J. D., Hammer, R. E., Goldstein, J. L., and Brown,
M. S. (2002) J. Biol. Chem. 277, 9520 9528
2. Reshef, L., Olswang, Y., Cassuto, H., Blum, B., Croniger, C. M., Kalhan, S. C.,
Tilghman, S. M., and Hanson, R. W. (2003) J. Biol. Chem. 278,
3041330416
3. Morris, M. D., Zilversmit, D. B., and Hintz, H. F. (1972) J. Lipid Res. 13,
383389
4. Hanson, R. W., and Reshef, L. (1997) Annu. Rev. Biochem. 66, 581611
5. Pilkis, S. J., Claus, T. H., Kurland, I. J., and Lange, A. J. (1995) Annu. Rev.
Biochem. 64, 799 835
6. Chou, J. Y. (2001) Curr. Mol. Med. 1, 2544
7. Wu, C., Okar, D. A., Newgard, C. B., and Lange, A. J. (2001) J. Clin. Investig.
107, 9198
8. Kissebah, A. H., Alfarsi, S., and Adams, P. W. (1981) Metabolism 30, 856 868
9. Siri, P., Candela, N., Zhang, Y. L., Ko, C., Eusufzai, S., Ginsberg, H. N., and
Huang, L. S. (2001) J. Biol. Chem. 276, 46064 46072
10. Brunzell, J. D., Albers, J. J., Chait, A., Grundy, S. M., Groszek, E., and
McDonald, G. B. (1983) J. Lipid Res. 24, 147155
11. Ito, Y., Azrolan, N., OConnell, A., Walsh, A., and Breslow, J. L. (1990) Science
249, 790 793
12. Castellani, L. W., Weinreb, A., Bodnar, J., Goto, A. M., Doolittle, M.,
TABLE I
Quantitation of gene expression in C3H and HcB-19 mice by real-time PCR.
Livers were obtained at 1000 h (lights on from 0600 to 1800 h) from 12-week-old male C3H and HcB-19 mice that had been fasted for 18 h or
were non-fasted. Total RNA and cDNA were prepared from these samples as described under ‘‘Experimental Procedures.’’ Expression levels of
mRNA relative to the constitutively expressed apoB mRNA are indicated and were determined by quantitative real-time PCR. Results from four
mice in each group are presented as -fold increase relative to that of fasted C3H controls. Statistical differences were determined by Studentst
test, and the two-tailed pvalues are listed.
Relative mRNA expression levels
Fasted Fed
C3H HcB-19 pvalue (C3H vs. HcB-19) C3H HcB-19 pvalue (C3H vs.HcB-19)
-Fold of increase vs. fasted C3H control
SREBP-1a 1.00 1.62 0.183 1.19 1.04 0.790
SREBP-1c 1.00 3.28 0.012 11.44 9.46 0.308
SREBP-2 1.00 1.92 0.004 3.28 2.96 0.533
ACC 1.00 2.00 0.005 4.24 2.86 0.116
FAS 1.00 2.17 0.003 5.98 3.30 0.063
PEPCK 1.00 1.05 0.711 0.21 0.47 0.040
Txnip Loss Disrupts Appropriate Metabolic Response to Fasting24392
by guest on May 12, 2018http://www.jbc.org/Downloaded from
Mehrabian, M., Demant, P., and Lusis, A. J. (1998) Nat. Genet. 18, 374 377
13. Bodnar, J. S., Chatterjee, A., Castellani, L. W., Ross, D. A., Ohmen, J.,
Cavalcoli, J., Wu, C., Dains, K. M., Catanese, J., Chu, M., Sheth, S. S.,
Charugundla, K., Demant, P., West, D. B., de Jong, P., and Lusis, A. J.
(2002) Nat. Genet. 30, 110 116
14. Gething, M. J., and Sambrook, J. (1992) Nature 355, 3345
15. Follmann, H., and Haberlein, I. (1995) Biofactors 5, 147156
16. Arrigo, A. P. (1999) Free Radic. Biol. Med. 27, 936 944
17. Demant, P., and Hart, A. A. (1986) Immunogenetics 24, 416 422
18. Groot, P. C., Moen, C. J., Dietrich, W., Stoye, J. P., Lander, E. S., and Demant,
P. (1992) FASEB J. 6, 2826 2835
19. Gaja, G., Ragnotti, G., Cajone, F., and Bernelli-Zazzera, A. (1968) Biochem. J.
109, 867875
20. Clark, M. G., Bloxham, D. P., Holland, P. C., and Lardy, H. A. (1974) J. Biol.
Chem. 249, 279 290
21. Anderson, M. E. (1985) Methods Enzymol. 113, 548 555
22. Yang, J., Goldstein, J. L., Hammer, R. E., Moon, Y. A., Brown, M. S., and
Horton, J. D. (2001) Proc. Natl. Acad. Sci. U. S. A. 98, 1360713612
23. Davis, R. A. (1999) Biochim. Biophys. Acta 1440, 131
24. Davis, R. A., Boogaerts, J. R., Borchardt, R. A., Malone-McNeal, M., and
Archambault-Schexnayder, J. (1985) J. Biol. Chem. 260, 1413714144
25. Leighton, J. K., Joyner, J., Zamarripa, J., Deines, M., and Davis, R. A. (1990)
J. Lipid Res. 31, 16631668
26. Lei, K. J., Chen, H., Pan, C. J., Ward, J. M., Mosinger, B., Jr., Lee, E. J.,
Westphal, H., Mansfield, B. C., and Chou, J. Y. (1996) Nat. Genet. 13,
203209
27. Annabi, B., Hiraiwa, H., Mansfield, B. C., Lei, K. J., Ubagai, T.,
Polymeropoulos, M. H., Moses, S. W., Parvari, R., Hershkovitz, E., Mandel,
H., Fryman, M., and Chou, J. Y. (1998) Am. J. Hum. Genet. 62, 400 405
28. Hawkins, R. A., Kamath, K. R., Scott, H. M., and Burchell, A. (1995) J. Inher-
ited Metab. Dis. 18, 558 566
29. Reyes, A., Burgos, M. E., Hubert, E., and Slebe, J. C. (1987) J. Biol. Chem. 262,
84518454
30. Winiarska, K., Drozak, J., Wegrzynowicz, M., Jagielski, A. K., and Bryla, J.
(2003) Metabolism 52, 739 746
31. Steinberg, D., Vaughan, M., Margolis, S., Price, H., and Pittman, R. (1961)
J. Biol. Chem. 236, 16311637
32. Spiegelman, B. M., and Green, H. (1981) Cell 24, 503510
33. Sen, C. K. (2000) Curr. Top. Cell. Regul. 36, 130
34. Sheehan, D., Meade, G., Foley, V. M., and Dowd, C. A. (2001) Biochem. J. 360,
116
35. Tanaka, T., Nakamura, H., Nishiyama, A., Hosoi, F., Masutani, H., Wada, H.,
and Yodoi, J. (2001) Free Radic. Res. 33, 851855
36. Nishiyama, A., Matsui, M., Iwata, S., Hirota, K., Masutani, H., Nakamura, H.,
Takagi, Y., Sono, H., Gon, Y., and Yodoi, J. (1999) J. Biol. Chem. 274,
2164521650
37. Nishiyama, A., Masutani, H., Nakamura, H., Nishinaka, Y., and Yodoi, J.
(2001) IUBMB Life 52, 29 33
38. Kim, J. B., and Spiegelman, B. M. (1996) Genes Dev. 10, 1096 1107
39. Shimomura, I., Bashmakov, Y., Ikemoto, S., Horton, J. D., Brown, M. S., and
Goldstein, J. L. (1999) Proc. Natl. Acad. Sci. U. S. A. 96, 1365613661
40. Isaacs, J., and Binkley, F. (1977) Biochim. Biophys. Acta 497, 192204
41. Shimomura, I., Hammer, R. E., Richardson, J. A., Ikemoto, S., Bashmakov, Y.,
Goldstein, J. L., and Brown, M. S. (1998) Genes Dev. 12, 31823194
42. Gilbert, H. F. (1984) Methods Enzymol. 107, 330 351
43. Pontremoli, S., Luppis, B., Traniello, S., Rippa, M., and Horecker, B. L. (1965)
Arch. Biochem. Biophys. 112, 715
44. Gilbert, H. F. (1982) J. Biol. Chem. 257, 12086 12091
45. Cottam, G. L., and Srere, P. A. (1969) Biochem. Biophys. Res. Commun. 35,
895900
46. Osterlund, B. R., Packer, M. K., and Bridger, W. A. (1980) Arch. Biochem.
Biophys. 205, 489 498
47. Walters, D. W., and Gilbert, H. F. (1986) J. Biol. Chem. 261, 1313513143
48. Cho, Y. W., Kim, D., Park, E. H., and Lim, C. J. (2002) Mol. Cell 13, 315321
49. Jamdar, S. C., Soo, E., and Cao, W. F. (1998) Biochim. Biophys. Acta 1393,
4148
50. Junn, E., Han, S. H., Im, J. Y., Yang, Y., Cho, E. W., Um, H. D., Kim, D. K.,
Lee, K. W., Han, P. L., Rhee, S. G., and Choi, I. (2000) J. Immunol. 164,
62876295
51. Hotta, M., Tashiro, F., Ikegami, H., Niwa, H., Ogihara, T., Yodoi, J., and
Miyazaki, J. (1998) J. Exp. Med. 188, 14451451
52. Taljedal, I. B. (1981) Diabetologia 21, 117
53. Ron, D. (2002) J. Clin. Investig. 109, 443445
54. Kaufman, R. J. (2002) J. Clin. Investig. 110, 1389 1398
55. Shalev, A., Pise-Masison, C. A., Radonovich, M., Hoffmann, S. C., Hirshberg,
B., Brady, J. N., and Harlan, D. M. (2002) Endocrinology 143, 36953698
56. Hirota, T., Okano, T., Kokame, K., Shirotani-Ikejima, H., Miyata, T., and
Fukada, Y. (2002) J. Biol. Chem. 277, 44244 44251
57. Horton, J. D., Shah, N. A., Warrington, J. A., Anderson, N. N., Park, S. W.,
Brown, M. S., and Goldstein, J. L. (2003) Proc. Natl. Acad. Sci. U. S. A. 100,
1202712032
Txnip Loss Disrupts Appropriate Metabolic Response to Fasting 24393
by guest on May 12, 2018http://www.jbc.org/Downloaded from
Alan D. Attie and Roger A. Davis
To Yuen Hui, Sonal S. Sheth, J. Matthew Diffley, Douglas W. Potter, Aldons J. Lusis,
Redox State to Appropriate Response to Nutritional Signals
Mice Lacking Thioredoxin-interacting Protein Provide Evidence Linking Cellular
doi: 10.1074/jbc.M401280200 originally published online March 26, 2004
2004, 279:24387-24393.J. Biol. Chem.
10.1074/jbc.M401280200Access the most updated version of this article at doi:
Alerts:
When a correction for this article is posted When this article is cited
to choose from all of JBC's e-mail alertsClick here
http://www.jbc.org/content/279/23/24387.full.html#ref-list-1
This article cites 54 references, 24 of which can be accessed free at
by guest on May 12, 2018http://www.jbc.org/Downloaded from
... Thioredoxin interacting protein (TXNIP) is an α-arrestin protein with several anti-proliferative functions, predominant among these are activities as a negative regulator of glucose uptake and as a suppressor of multiple progrowth signaling pathways [1][2][3][4][5]. Consistent with a role in restricting cell growth, TXNIP expression is suppressed by multiple cancer-associated progrowth signaling pathways [6][7][8][9], and its expression is typically low in tumors compared to adjacent normal tissue. ...
... Therefore, while the mechanistic connection remains to be clarified, we favor a model where TXNIP loss drives transcriptional "repression" by an indirect mechanism rather than by increasing interactions between Myc and Miz/ZBTB17. This study and others demonstrate and establish that TXNIP is a repressor of at least 2 common features of the transformed state: Myc transcriptional activity and glucose uptake [1,5,55,56]. TXNIP expression is exquisitely dependent on the transcription factor MondoA [12,57]. ...
Article
Full-text available
The c-Myc protooncogene places a demand on glucose uptake to drive glucose-dependent biosynthetic pathways. To meet this demand, c-Myc protein (Myc henceforth) drives the expression of glucose transporters, glycolytic enzymes, and represses the expression of thioredoxin interacting protein (TXNIP), which is a potent negative regulator of glucose uptake. A Mychigh/TXNIPlow gene signature is clinically significant as it correlates with poor clinical prognosis in triple-negative breast cancer (TNBC) but not in other subtypes of breast cancer, suggesting a functional relationship between Myc and TXNIP. To better understand how TXNIP contributes to the aggressive behavior of TNBC, we generated TXNIP null MDA-MB-231 (231:TKO) cells for our study. We show that TXNIP loss drives a transcriptional program that resembles those driven by Myc and increases global Myc genome occupancy. TXNIP loss allows Myc to invade the promoters and enhancers of target genes that are potentially relevant to cell transformation. Together, these findings suggest that TXNIP is a broad repressor of Myc genomic binding. The increase in Myc genomic binding in the 231:TKO cells expands the Myc-dependent transcriptome we identified in parental MDA-MB-231 cells. This expansion of Myc-dependent transcription following TXNIP loss occurs without an apparent increase in Myc's intrinsic capacity to activate transcription and without increasing Myc levels. Together, our findings suggest that TXNIP loss mimics Myc overexpression, connecting Myc genomic binding and transcriptional programs to the nutrient and progrowth signals that control TXNIP expression.
... Txnip is closely associated with glucose and lipid metabolism and is a major regulator of glucose homeostasis (33,34). Consistently, several studies have demonstrated that Txnip expression is increased in DM and that it plays an important role in the regulation of insulin sensitivity (35,36,37). Txnip-deficient HcB-19 (HcB) mice have been shown to have a three-fold increase in insulin levels (37), increased pancreatic β-cell mass and enhanced insulin secretion and sensitivity (36,38). ...
... Consistently, several studies have demonstrated that Txnip expression is increased in DM and that it plays an important role in the regulation of insulin sensitivity (35,36,37). Txnip-deficient HcB-19 (HcB) mice have been shown to have a three-fold increase in insulin levels (37), increased pancreatic β-cell mass and enhanced insulin secretion and sensitivity (36,38). However, the associated mechanisms for Txnip in DM, including its potential role in β-cell proliferation, have not been fully characterized. ...
Article
Full-text available
Elucidating the mechanisms of regulation of β cell proliferation is key to understanding the pathogenesis of diabetes mellitus. Txnip is a tumor suppressor that is upregulated in diabetes and plays an important role in the regulation of insulin sensitivity; however, its potential effect on pancreatic β cell proliferation remains unclear. Here, we evaluated the role of Txnip in pancreatic β cell compensatory proliferation by subjecting wild-type (WT) and Txnip knockout (KO) mice to a high fat diet (HFD). Our results demonstrate that Txnip deficiency improves glucose tolerance and increases insulin sensitivity in HFD-induced obesity. The antidiabetogenic effect of Txnip deficiency was accompanied by increased β cell proliferation and enhanced β mass expansion. Furthermore, Txnip deficiency modulated the expression of a set of transcription factors with key roles in β cell proliferation and cell cycle regulation. Txnip knockout in HFD mice also led to activated levels of p-P13K, p-AKT, p-mTOR and p-GSK3β, suggesting that Txnip may act via PI3K/AKT signaling to suppress β cell proliferation. Thus, our work provides a theoretical basis for Txnip as a new therapeutic target for the treatment of diabetes mellitus.
... In doing so, TXNIP is reported to influence cardiac metabolism, including mitochondrial function and glucose uptake (9). Researchers have also demonstrated that TXNIP modulates cellular glucose utilization and mitochondrial oxidation of metabolic fuels (5,(10)(11)(12). On the other hand, TXNIPnull mice cannot survive prolonged fasting, exhibiting dysglycemia and dyslipidemia (11). ...
Article
Full-text available
Endothelial cells are highly sensitive to hemodynamic shear stresses, which act in the blood flow’s direction on the blood vessel’s luminal surface. Thus, endothelial cells on that surface are exposed to various physiological and pathological stimuli, such as disturbed flow-induced shear stress, which may exert effects on adaptive vascular diameter or structural wall remodeling. Here we showed that plasma thioredoxin-interactive protein (TXNIP) and malondialdehyde levels were significantly increased in patients with slow coronary flow. In addition, human endothelial cells exposed to disturbed flow exhibited increased levels of TXNIP in vitro. On the other hand, deletion of human endothelial TXNIP increased capillary formation, nitric oxide production and mitochondrial function, as well as lessened oxidative stress response and endothelial cell inflammation. Additional beneficial impacts from TXNIP deletion were also seen in a glucose utilization study, as reflected by augmented glucose uptake, lactate secretion and extracellular acidification rate. Taken together, our results suggested that TXNIP is a key component involved in mediating shear stress-induced inflammation, energy homeostasis, and glucose utilization, and that TXNIP may serve as a potentially novel endothelial dysfunction regulator.
... Glucose tolerance tests and insulin tolerance tests were performed as previously described (9,13,18,19). Briefly, mice were fasted for 4 hours and given glucose (3 g/kg) or regular insulin (1 IU/kg) by intraperitoneal injections. ...
Article
Thioredoxin-interacting protein (Txnip) has emerged as a key factor in pancreatic beta cell biology and its upregulation by glucose and diabetes contributes to the impairment in functional beta cell mass and glucose homeostasis. In addition, beta cell deletion of Txnip protects against diabetes in different mouse models. However, while Txnip is ubiquitously expressed, its role in pancreatic alpha cells has remained elusive. We therefore now generated an alpha cell Txnip knockout (aTKO) mouse and assessed the effects on glucose homeostasis. While no significant changes were observed on regular chow, after a 30-week high-fat diet, aTKO animals showed improvement in glucose tolerance and lower blood glucose levels compared to their control littermates. Moreover, in the context of streptozotocin (STZ)-induced diabetes, aTKO mice showed significantly lower blood glucose levels compared to controls. While serum insulin levels were reduced in both control and aTKO mice, STZ-diabetes significantly increased glucagon levels in control mice, but this effect was blunted in aTKO mice. Moreover, glucagon secretion from aTKO islets was >2-fold lower than from control islets, while insulin secretion was unchanged in aTKO islets. At the same time, no change in alpha cell or beta cell numbers or mass was observed and glucagon and insulin expression and content were comparable in isolated islets from aTKO and control mice. Thus, together the current studies suggest that downregulation of alpha cell Txnip is associated with reduced glucagon secretion and that this may contribute to the glucose-lowering effects observed in diabetic aTKO mice.
... As a result, blood glucose levels are markedly lower in total TXNIP − /− mice compared to wild type controls after either prolonged high fat feeding or STZ induction (Myers et al., 2016;Chutkow et al., 2010). On the other hand, TXNIP − /− mice become hypoglycemic when subjected to prolonged fasting (Hui et al., 2004). Since hyperglycemia is not achievable in these TXNIP − /− mice, the mechanism whereby TXNIP alters diabetes-related vascular inflammation, angiogenesis or cardiac dysfunction in TXNIP − /− mice (Elshaer et al., 2017;Myers et al., 2016) has not been specifically examined. ...
Article
Endothelial dysfunction, hallmarked by an imbalance between vasoconstriction and vasorelaxation, is associated with diabetes. Thioredoxin Interacting protein (TXNIP), controlled by an exquisitely glucose sensitive gene, is increasingly recognized for its role in diabetes. However, the role of TXNIP in modulating diabetes-related endothelial dysfunction remains unclear. To elucidate the role of TXNIP, we generated two novel mouse strains; endothelial-specific TXNIP knockout (EKO) and a Tet-O inducible, endothelial-specific TXNIP overexpression (EKI). Hyperglycemia was induced by streptozotocin (STZ) treatment in floxed control (fl/fl) and EKO mice. Doxycycline (DOX) was given to EKI mice to induce endothelial TXNIP overexpression. The ablation of endothelial TXNIP improved glucose tolerance in EKO mice. Acetylcholine-induced, endothelium-dependent vasorelaxation was impaired in STZ-treated fl/fl mice while this STZ impaired vasorelaxation was attenuated in EKO mice. Hyperglycemia induction of NLRP3 and reductions in Akt and eNOS phosphorylation were also mitigated in EKO mice. Overexpression of endothelial TXNIP did not impair glucose tolerance in DOX-treated EKI mice, however induction of endothelial TXNIP led to impaired vasorelaxation in EKI mice. This was associated with increased NLRP3 and reduced Akt and eNOS activation. In conclusion, deletion of endothelial TXNIP is protective against and overexpression of endothelial TXNIP induces endothelial dysfunction; thus, endothelial TXNIP plays a critical role in modulating endothelial dysfunction.
Article
Full-text available
Thioredoxin-interacting protein (TXNIP) is commonly considered a master regulator of cellular oxidation, regulating the expression and function of Thioredoxin (Trx). Recent work has identified that TXNIP has a far wider range of additional roles: from regulating glucose and lipid metabolism, to cell cycle arrest and inflammation. Its expression is increased by stressors commonly found in neoplastic cells and the wider tumor microenvironment (TME), and, as such, TXNIP has been extensively studied in cancers. In this review, we evaluate the current literature regarding the regulation and the function of TXNIP, highlighting its emerging role in modulating signaling between different cell types within the TME. We then assess current and future translational opportunities and the associated challenges in this area. An improved understanding of the functions and mechanisms of TXNIP in cancers may enhance its suitability as a therapeutic target.
Article
Background: Carotenoids have been reported to exert protective effects against age-related diseases via changes in DNA methylation. Although lower thioredoxin-interacting protein (TXNIP) DNA methylation is associated with age-related diseases, only a few studies have investigated the factors influencing TXNIP DNA methylation. Carotenoids may be a factor linking TXNIP to specific pathophysiological functions. The aim of this study was to examine whether serum carotenoid levels are associated with TXNIP DNA methylation levels. Methods: We conducted a cross-sectional study using 376 health examination participants (169 men). DNA methylation levels were determined using a pyrosequencing assay. Serum carotenoid levels were determined by high-performance liquid chromatography. Multivariable regression analyses were performed to examine the associations between TXNIP DNA methylation levels and serum carotenoid levels with adjustment for age, BMI, HbA1c, CRP, smoking habits, alcohol consumption, exercise habits, and percentage of neutrophils. Results: Multiple linear regression analyses showed that TXNIP DNA methylation levels were positively associated with serum levels of zeaxanthin/lutein ( β [95%CI]: 1.935 [0.184, 3.685]), β-cryptoxanthin (1.447 [0.324, 2.570]), α-carotene (1.061 [0.044, 2.077]), β-carotene (1.272 [0.319, 2.226]), total carotenes (1.255 [0.040, 2.469]), total xanthophylls (2.133 [0.315, 3.951]), provitamin A (1.460 [0.402, 2.519]), and total carotenoids (1.972 [0.261, 3.683]) in men (all p<0.05). Of these, provitamin A showed the stronger association (standardized β=0.216). No significant association of TXNIP DNA methylation and serum carotenoid was observed in women. Conclusions: The findings of this study suggest that carotenoid intake may protect against age-related diseases by altering TXNIP DNA methylation status in men.
Article
Full-text available
This study was conducted to compare the efficacy of the mouse hepatic and renal antioxidant systems against inflammation-induced oxidative stress. Increased Il-1 and Il-6 expressions, markers of inflammation, were represented by inflammation models in mouse liver and kidney tissues injected intraperitoneally with LPS. After establishing the model, the GSH level and the GSH/GSSG ratio, which are oxidative stress markers, were investigated in both tissues treated with LPS and the control group. The expression of Trx1, TrxR, and Txnip genes increased in the liver tissues of LPS-treated mice. In the kidney tissue, while Trx1 expression decreased, no change was observed in TrxR1 expression, and Txnip expression increased. In the kidneys, TRXR1 and GR activities decreased, whereas GPx activity increased. In both tissues, the TRXR1 protein expression decreased significantly, while TXNIP expression increased. In conclusion, different behaviors of antioxidant system members were observed during acute inflammation in both tissues. Additionally, it can be said that the kidney tissue is more sensitive and takes earlier measures than the liver tissue against cellular damage caused by inflammation and inflammation-induced oxidative stress.
Article
Significance: Thioredoxin interacting protein (TXNIP) is a member of the arrestin foldsuperfamily with important cellular functions, including cellular transport, mitochondrial energy generation and protein cycling. It is the only arrestin-domain protein known to covalently bind to thioredoxin and plays roles in glucose metabolism, inflammation, apoptosis, and cancer. Recent advances: The crystal structure of the TXNIP-thioredoxin complex provided details about this fascinating interaction. Recent studies showed that TXNIP is induced by endoplasmic reticulum stress, activates NLR family pyrin domain containing 3 (NLRP3) inflammasomes, and can regulate glucose transport into cells. The tumor suppressor role of TXNIP in various cancer types and the role of TXNIP in fructose absorption are now described. Critical issues: The influence of TXNIP on redox state is more complex than its interaction with thioredoxin. Future directions: It is incompletely understood which functions of TXNIP are thioredoxin-dependent. It is also unclear whether TXNIP binding can inhibit glucose transporters without endocytosis. TXNIP-regulated control of endoplasmic reticulum stress should also be investigated further.
Article
Full-text available
Transcription of UPR-responsive genes is induced when the cleaved form of ATF6 activates the XBP1 promoter. Therefore, signaling through ATF6 and IRE1 merges to induce XBP1 transcription and mRNA splicing, respectively (Figure 1, a and b). ATF6 increases XBP1 transcription to produce more substrate for IRE1-mediated splicing that generates more active XBP1, providing a positive feedback for UPR activation. However, cells that lack either IRE1α or ATF6 cleavage can induce XBP1 mRNA (20). These two pathways may thus provide parallel signaling pathways for XBP1 transcriptional induction. Alternatively, another pathway — possibly mediated by the ER-localized protein kinase PERK (see Ron, this Perspective series, ref. 2) — may also contribute to induction of XBP1 mRNA. The binding specificities of XBP1 and ATF6 are similar, although ATF6 binding requires CBF binding to an adjacent site, whereas XBP1 binds independently (17, 20, 21, 34). These binding specificities provide another avenue for complementary interaction between the IRE1-XBP1 and ATF6 pathways at the level of transcriptional activation. In addition, these transcription factors might regulate transcription from a second ERSE (ERSEII), which also contains a CCACG motif (35). During the terminal differentiation of certain secretory cells, such as those in the pancreas or liver, membrane expansion is accompanied by a dramatic increase in protein secretion. Likewise, upon B cell maturation into high-level antibody-secreting plasma cells, the ER compartment expands approximately fivefold to accommodate the large increase in Ig synthesis. The requirement for the UPR in this latter process has been demonstrated in XBP1–/– cells. Since deletion of XBP1 produces an embryonic-lethal phenotype at day E14.5, the role of XBP1 in B and T cell development had to be studied in immunoincompetent RAG1–/– mice reconstituted with XBP1–/– embryonic stem cells (73). Work in these chimeric mice demonstrated that XBP1 is required for high-level Ig production. Interestingly, the induction of Ig heavy-chain and light-chain gene rearrangement and the assembly and transport of Igμ to the surface of the B cells occurred normally. However, plasma cells were not detected, suggesting a role for XBP1 in plasma cell differentiation or survival.
Article
Full-text available
The glutathione transferases (GSTs; also known as glutathione S-transferases) are major phase II detoxification enzymes found mainly in the cytosol. In addition to their role in catalysing the conjugation of electrophilic substrates to glutathione (GSH), these enzymes also carry out a range of other functions. They have peroxidase and isomerase activities, they can inhibit the Jun N-terminal kinase (thus protecting cells against H(2)O(2)-induced cell death), and they are able to bind non-catalytically a wide range of endogenous and exogenous ligands. Cytosolic GSTs of mammals have been particularly well characterized, and were originally classified into Alpha, Mu, Pi and Theta classes on the basis of a combination of criteria such as substrate/inhibitor specificity, primary and tertiary structure similarities and immunological identity. Non-mammalian GSTs have been much less well characterized, but have provided a disproportionately large number of three-dimensional structures, thus extending our structure-function knowledge of the superfamily as a whole. Moreover, several novel classes identified in non-mammalian species have been subsequently identified in mammals, sometimes carrying out functions not previously associated with GSTs. These studies have revealed that the GSTs comprise a widespread and highly versatile superfamily which show similarities to non-GST stress-related proteins. Independent classification systems have arisen for groups of organisms such as plants and insects. This review surveys the classification of GSTs in non-mammalian sources, such as bacteria, fungi, plants, insects and helminths, and attempts to relate them to the more mainstream classification system for mammalian enzymes. The implications of this classification with regard to the evolution of GSTs are discussed.
Article
Full-text available
Summary The cytotoxicity of reactive oxygen intermediates (ROIs) has been implicated in the destruc- tion of pancreatic b cells in insulin-dependent diabetes mellitus (IDDM). Thioredoxin (TRX), a redox (reduction/oxidation)-active protein, has recently been shown to protect cells from oxidative stress and apoptosis. To elucidate the roles of oxidative stress in the development of autoimmune diabetes in vivo, we produced nonobese diabetic transgenic mice that overexpress TRX in their pancreatic b cells. In these transgenic mice, the incidence of diabetes was mark- edly reduced, whereas the development of insulitis was not prevented. Moreover, induction of diabetes by streptozotocin, an ROI-generating agent, was also attenuated by TRX overexpres- sion in b cells. This is the first direct demonstration that an antioxidative and antiapoptotic pro- tein protects b cells in vivo against both autoimmune and drug-induced diabetes. Our results strongly suggest that oxidative stress plays an essential role in the destruction of b cells by infil- trating inflammatory cells in IDDM.
Article
Full-text available
Familial combined hyperlipidaemia (FCHL) is a common, multifactorial disorder associated with elevated levels of plasma triglyceride, cholesterol, or both. A characteristic feature is increased secretion of very low density lipoproteins (VLDL) and apolipoprotein B (apoB). Although FCHL is the most common cause of premature coronary artery disease (CAD), accounting for over 10% of cases, its aetiology remains largely unknown. One powerful approach to the dissection of complex genetic traits involves the use of animal models. We have identified a mouse strain, HcB-19/Dem (HcB-19), which exhibits hypertriglyceridaemia, hypercholesterolaemia and elevated levels of plasma apoB. Like FCHL patients, HcB-19 mice also exhibit increased secretion of triglyceride-rich lipoproteins, and their hyperlipidaemia becomes progressively more severe with age. It is likely that the hyperlipidaemia results from a mutation of a novel gene that arose during development of strain HcB-19. We mapped the hyperlipidaemia gene (Hyplip1) to the distal portion of mouse chromosome 3. This region is syntenic to human chromosome 1q21-q23, which has recently been shown to harbour a gene associated with FCHL in families from a Finnish isolate.
Article
Full-text available
Hepatic 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase is an important regulatory enzyme of glucose metabolism. By controlling the level of fructose-2,6-bisphosphate, an allosteric activator of the glycolytic enzyme 6-phosphofructo-1-kinase and an inhibitor of the gluconeogenic enzyme fructose-1,6-bisphosphatase, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase regulates hepatic glucose output. We studied the effects of adenovirus-mediated overexpression of this enzyme on hepatic glucose metabolism in normal or diabetic mice. These animals were treated with virus encoding either wild-type or bisphosphatase activity–deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase. Seven days after virus injection, hepatic fructose-2,6-bisphosphate levels increased significantly in both normal and diabetic mice, with larger increases observed in animals with overexpression of the mutant enzyme. Blood glucose levels in normal mice overexpressing either enzyme were lowered, accompanied by increased plasma lactate, triglycerides, and FFAs. Blood glucose levels were markedly reduced in diabetic mice overexpressing the wild-type enzyme, and still more so in mice overexpressing the mutant form of the enzyme. The lower blood glucose levels in diabetic mice were accompanied by partially normalized plasma triglycerides and FFAs, increased plasma lactate, and increased liver glycogen levels, relative to diabetic mice treated with a control adenovirus. Our findings underscore the critical role played by hepatic 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase in control of fuel homeostasis and suggest that this enzyme may be considered as a therapeutic target in diabetes.
Article
Full-text available
The genetic control of susceptibility to many common diseases, including cancer, is multigenic both in humans and in animals. This genetic complexity has presented a major obstacle in mapping the relevant genes. As a consequence, most geneticists and molecular biologists presently focus on "single gene" diseases. To make the multigenic diseases accessible to genetic and molecular analysis, we developed a novel genetic tool, the recombinant congenic strains (RCS) in the mouse (4). The RC strains are produced by inbreeding of mice of the second backcross generation between two inbred strains, one of which serves as the "donor" and the other as the "background" strain. A series of RCS consists of approximately 20 strains, each carrying a different set of genes: approximately 12.5% genes from the common donor inbred strain, the remaining 87.5% from the common background inbred strain. As the set of donor strain genes in each RC strain is different, the nonlinked genes of the donor strain involved in the control of a multigenic trait, e.g., cancer susceptibility, become distributed into different RC strains where they can be analyzed one by one. Hence, the RCS system transforms a multigenic trait into a series of single gene traits, where each gene contributing to the multigenic control can be mapped and studied separately. Recently we demonstrated that the RCS system is indeed capable of resolving multigenic traits, which are hardly analyzable otherwise, by mapping four new colon tumor susceptibility loci (8; P. C. Groot, C. J. A. Moen, W. Dietrich, L. F. M. van Zutphen, E. S. Lander, and P. Demant, unpublished results). For successful application of the RCS system, extensive genetic characterization of the individual recombinant congenic strains is essential. In this paper we present detailed information about the genetic composition of three series of RC strains on the basis of typing of 120-180 markers distributed along all autosomes. The data indicate that the relative representation of the donor strain genes in the RC strains does not deviate from the theoretical expectation, and that the RC strains achieved a very high degree of genetic homogeneity and for all practical purposes can be considered inbred strains. The density and distribution of markers reported here permits an effective mapping of unknown genes of donor strain origin at almost all autosomal locations. Much of this information has been obtained using the new class of genetic markers, the simple sequence repeat polymorphisms.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Full-text available
Two different molecular weight forms of apoB are produced from a common initial transcript via editing of a Gln codon (CAA) to a stop codon (UAA), leading to a truncated translation product (apo BS) that consists of the amino terminal half of the larger form (apoBL). Previous studies have shown that fasting coordinately decreases lipogenesis and the secretion of very low density lipoprotein (VLDL) lipids and apoBS. Secretion of the apoBL is unaffected by fasting. We studied whether editing of apoB RNA is repressed by fasting, thus accounting for the selective decreased secretion of apoBS. Column chromatography of [35S]methionine-labeled lipoproteins secreted by hepatocytes from fed rats showed that essentially all of apoBL is secreted in the VLDL fraction, whereas a significant amount (15%) of apoBS is secreted associated as lipoproteins eluting in the HDL fractions. Fasting decreased the relative amount of apoBS that eluted in the VLDL fractions and increased the amount secreted in the HDL fractions. Consistent with previous results, hepatocytes from fasted rats show a selective twofold decrease in apoBS secretion. Fasting did not affect the relative abundance of apoB RNA, determined by slot blot hybridization assays using two different 32P-labeled cDNA probes coding either for both molecular weight forms or for only the large molecular weight form. However, quantitative of the editing of apoB RNA showed that fasting caused a 60% decrease in the amount of apoB RNA possessing the stop codon. These data show that the editing of apoB RNA is sensitive to metabolic state (i.e., fasting) resulting in a selective decrease in the secretion of apoBS. However, since the total secretion of apoB was decreased by fasting, while apoB mRNA levels remained constant, additional (post-transcriptional) mechanisms play a role in regulating apoB secretion.
Article
Citrate cleavage enzyme was phosphorylated with γ-32P-ATP. A 32P-compound was isolated by Dowex chromatography of an alkaline hydrolysate of the 32P-phosphoenzyme. The isolated compound behaved chromatographically and electrophoretically like phosphohistidine.
Article
The disulfide-sulfhydryl (SS/SH) ratios of subcellular fractions of rat hepatic tissue were found to vary diurnally with the ratio lowest in the early morning and highest in the early evening. These changes were found in the nuclear, microsomal and cytosol fractions. The primary reaction is the reversible formation of mixed disulfides of glutathione with proteins. This formation is controlled by the activity of thiol transferase and the level of oxidized glutathione (GSSG) as substrate. Several enzymes including mitochondrial and microsomal oxidases, glutathione reductase and peroxidase and glucose-6-phosphate dehydrogenase were found to control the levels of GSSG. An NADPH-dependent microsomal oxidase system, inhibited by GSSG, was found to produce activated oxygen which served as substrate for flutathione peroxidase. Evidence is presented for the concept that the formation of mixed disulfides of proteins with glutathione is a mechanism for maintenance of a disulfide-sulfhydryl ratio such that the integrity of particulate membranes is maintaine during oxidative and reductive stresses on the hepatic cells.