You are on page 1of 10

doi:10.

1093/jmcb/mjs025 Journal of Molecular Cell Biology (2012), 4, 221– 230 | 221


Published online May 19, 2012

Article
Age- and diet-dependent requirement of DJ-1
for glucose homeostasis in mice with implications
for human type 2 diabetes
Deepak Jain1,† Ruchi Jain1,†, Daniel Eberhard1, Jan Eglinger1,2, Marco Bugliani3, Lorenzo Piemonti4,
Piero Marchetti3, and Eckhard Lammert1,2, *
1
Institute of Metabolic Physiology, Heinrich-Heine University, D-40225 Düsseldorf, Germany

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
2
Paul Langerhans Group for Beta Cell Biology, German Diabetes Center (DDZ), D-40225 Düsseldorf, Germany
3
Metabolic Unit, Department of Endocrinology and Metabolism, Cisanello Hospital, 56100 Pisa, Italy
4
Laboratory of Experimental Surgery, Surgical Department, San Raffaele Scientific Institute, 20132 Milano, Italy

These authors contributed equally to this work.
* Correspondence to: Eckhard Lammert, E-mail: lammert@uni-duesseldorf.de

Elderly patients often suffer from multiple age-related diseases. Here we show that the expression of DJ-1, an antioxidant protein
with reduced expression in the central nervous system of patients with Parkinson’s disease, is reduced in pancreatic islets of
patients with type 2 diabetes mellitus (T2DM). In contrast, under non-diabetic conditions, DJ-1 expression increases in mouse
and human islets during aging. In mouse islets, we show that DJ-1 prevents an increase in reactive oxygen species levels as the
mice age. This antioxidant function preserves mitochondrial integrity and physiology, prerequisites for glucose-stimulated
insulin secretion. Accordingly, DJ-1-deficient mice develop glucose intolerance and reduced b cell area as they age or gain
weight. Our data suggest that DJ-1 is more generally involved in age- and lifestyle-related human diseases and show for the first
time that DJ-1 plays a key role in glucose homeostasis and might serve as a novel drug target for T2DM.

Keywords: diabetes, pancreatic islets, DJ-1, mitochondria

Introduction and recently it was shown that diabetics have a 40% increased
DJ-1 is a multifunctional, highly conserved antioxidant protein risk of developing PD (Xu et al., 2011), suggesting a possible
encoded by Parkinson’s disease (PD) gene park7 (Bonifati link between type 2 diabetes mellitus (T2DM) and PD. Here, we
et al., 2003; Dawson and Dawson, 2003; Guzman et al., 2010; studied the role of DJ-1 in pancreatic islet function, glucose
Hong et al., 2010). In neurons, DJ-1 is upregulated upon oxidative homeostasis, and T2DM.
stress and protects neurons from mitochondrial dysfunction (Hao
et al., 2010; McCoy and Cookson, 2012). Interestingly, in mouse Results
pancreatic islets and mouse insulinoma cells (MIN6), which Age-dependent upregulation of DJ-1 in human pancreatic islets
have a low expression of antioxidant proteins (Tiedge et al., and reduced DJ-1 expression in islets from T2DM patients
1997; Robertson and Harmon, 2006; Wiederkehr and Wollheim, We first showed that, in the human pancreas, DJ-1 is strongly
2006; Lenzen, 2008), DJ-1 was recently shown to be specifically expressed in islets compared with the surrounding exocrine
upregulated under hyperglycemic conditions in vitro and in vivo tissue (Figure 1A – D). In addition, DJ-1 protein expression
(Waanders et al., 2009; Inberg and Linial, 2010). almost doubled in human pancreatic islets under hyperglycemic
Pancreatic islets are aggregates of endocrine cells of which b conditions (Figure 1E). When comparing islets from 38 to 56
cells form the majority (Maedler, 2008; Collombat et al., 2010). years old and 68 to 81 years old, we also discovered that the
An increase in blood glucose concentration stimulates the b mRNA expression of DJ-1 significantly increased in the islets of
cells to secrete insulin, which lowers blood glucose to normal non-diabetic humans during aging (Figure 1F and Supple-
levels. Pancreatic islets and neurons have many features in mentary Table S1). In contrast, DJ-1 expression was significantly
common (van Arensbergen et al., 2010; Soltani et al., 2011), reduced on both the mRNA and protein levels in the islets from
T2DM patients compared with islets from non-diabetic controls
(Figure 1G and H, and Supplementary Tables S1 and S2). The
Received February 9, 2012. Revised April 13, 2012. Accepted April 19, 2012. decline in DJ-1 protein expression might have been even larger
# The Author (2012). Published by Oxford University Press on behalf of Journal of
when age-matched T2DM patients and non-diabetic controls
Molecular Cell Biology, IBCB, SIBS, CAS. All rights reserved.
222 | Journal of Molecular Cell Biology Jain et al.

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
Figure 1 Age-dependent upregulation of DJ-1 in human pancreatic islets and reduced DJ-1 expression in islets from T2DM patients. (A– D) LSM
images of a section through human pancreas showing an islet (*) surrounded by exocrine pancreatic tissue, stained for cell nuclei (DAPI) (A),
DJ-1 (B), insulin (C), and both DJ-1 and insulin (D). Scale bar, 100 mm. (E) DJ-1 protein expression (normalized to b-actin) in human pancreatic
islets cultured at low (5.5 mM) and high (16.5 mM) glucose concentrations for 6 h as determined by ELISA. The islets were isolated from a male
donor (65 years, BMI 25.2, ischemic stroke). n ¼ 3. (F) Relative DJ-1 mRNA expression in human islets from non-diabetic individuals as deter-
mined by real-time RT– PCR. n ¼ 3– 4. (G) Comparison of relative DJ-1 mRNA expression in pancreatic islets from non-diabetic controls and
T2DM patients using real-time RT –PCR. n ¼ 3 –4. (H) DJ-1 protein expression (normalized to b-actin) in human pancreatic islets obtained
from non-diabetic controls and T2DM patients as determined by ELISA. n ¼ 3. **P , 0.01, *P , 0.05 (two-tailed Student’s t-test). All values
are means + SD. For further information on islet donors, see Supplementary Tables S1 and S2.

had been available for the analysis of human islet lysates. In required for lowering ROS levels in islets. In contrast, DJ-1 was ex-
summary, our data show that DJ-1 expression normally increases plicitly required for lowering ROS levels in mouse islets when the
in pancreatic islets during aging, but that, in the islets of patients mice were 12 – 13 weeks of age (Figure 2G, I, and J), suggesting
with T2DM, its expression decreases. that DJ-1 keeps ROS at constant levels as islets age. Moreover,
Age-dependent requirement of DJ-1 for reducing reactive oxygen the levels of ROS could be normalized by culturing
species in pancreatic islets DJ-1-deficient islets in the presence of the redox modulator
To investigate the role of DJ-1 in vivo, we turned to the mouse N-acetyl-L-cysteine (NAC) (Figure 2H and K).
and showed that in the mouse pancreas as in the human pan- Reduced expression of DJ-1 increases mitochondrial ROS
creas, DJ-1 is more strongly expressed in islets compared with production in mouse insulinoma cells
the surrounding exocrine tissue (Figure 2A –D). As in humans, Next, to determine the cellular compartment where ROS accu-
mouse DJ-1 expression was significantly higher in islets at both mulated in the absence of DJ-1, we transfected glucose-
the mRNA and protein levels during aging, as shown by compar- responsive mouse insulinoma cells (MIN6), a model cell line for
ing islets taken from mice at 8 and 12– 13 weeks of age (Figure 2E b cells, with a redox-sensitive variant of green fluorescent
and F). Although DJ-1 mRNA expression remained unchanged up protein (roGFP2) containing a mitochondrial matrix targeting se-
to the age of 8 weeks, the expression of DJ-1 increased from 8 to quence, or with a cytoplasmic roGFP2 (Hanson et al., 2004).
20 weeks of age (Supplementary Figure S1). In contrast, there was Comparison of untreated MIN6 cells with fully reduced [dithio-
no age-dependent change in the mRNA level for any other antioxi- threitol (DTT)-treated] and fully oxidized (Aldrithiol-treated) cells
dant protein we studied, i.e. superoxide dismutase 1 and 2 (sod1, suggested that MIN6 cells normally contain low levels of ROS in
sod2), glutathione peroxidase 1 (gpx1), and uncoupling protein 2 both mitochondria and cytosol (Figure 3A and B). We then
(ucp2) (Supplementary Figure S2). lowered DJ-1 protein expression in these cells using two different
Since the loss of DJ-1 was previously shown to increase mito- small interfering RNAs (siRNA) (Supplementary Figure S3).
chondrial reactive oxygen species (ROS) production in neurons Importantly, a reduced expression of DJ-1 protein in MIN6 cells
(Andres-Mateos et al., 2007; Guzman et al., 2010), we next significantly increased ROS levels in mitochondria as determined
studied whether DJ-1 deficiency increased ROS in mouse pancre- in living cells using Laser Scanning Microscopy (LSM) (Figure 3C,
atic islets. Surprisingly, no significant increase in ROS levels was and Figure 3E – G vs H – J and K– M). We confirmed the specificity of
observed in islets isolated from DJ-1-deficient mice at the age of 8 the knockdown by showing that normal ROS levels could be
weeks (Figure 2G), showing that DJ-1 was not constitutively restored in DJ-1-silenced cells by transfection with a DJ-1 cDNA
Role of DJ-1 in type 2 diabetes Journal of Molecular Cell Biology | 223

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
Figure 2 Age-dependent requirement of DJ-1 for reducing ROS in mouse pancreatic islets. (A –D) LSM images of sections through mouse pan-
creas showing an islet (*) surrounded by exocrine pancreatic tissue, stained for cell nuclei (DAPI) (A), DJ-1 (B), insulin (C), and both DJ-1 and
insulin (D). Scale bar, 100 mm. (E) Relative DJ-1 mRNA expression measured by real-time RT –PCR of islets isolated from C57BL/6J mice at 8
and 12– 13 weeks of age. n ¼ 3. (F) Densitometric western blot analysis of DJ-1 protein expression in islets isolated from C57BL/6J mice at
8 and 12– 13 weeks of age. Representative protein bands of western blots probed with anti-DJ-1 and -GAPDH antibodies are shown below.
n ¼ 6. (G and H) Relative levels of ROS measured by CM-H2DCFDA fluorescence intensities in islets isolated from control and DJ-1-deficient
mice at 8 and 12 –13 weeks of age in the absence (G) or presence (H) of NAC. n ¼ 3. (I – K) LSM images of optical sections through pancreatic
islets (*) isolated from control mice (I), DJ-1-deficient mice (J), and DJ-1-deficient mice after treatment of the islets with NAC for 3 days (K). ROS
were stained with CM-H2DCFDA. Scale bar, 100 mm. **P , 0.01 (two-tailed Student’s t-test). All values are means + SD.

(Figure 3C, and Figure 3K – M vs N – P). In contrast, cytoplasmic often fragmented (Figure 4A –D vs E– H). Moreover, significantly
ROS were not significantly increased (Figure 3D). The data more MIN6 cells displayed fragmented mitochondria when DJ-1
suggest that DJ-1 is specifically required for reducing ROS in expression was reduced using siRNA (Supplementary Figure
the mitochondria of insulin-secreting b cells. S4A – D and I). However, in the presence of the redox modulator
Age-dependent requirement of DJ-1 for mitochondrial integrity, NAC, mitochondrial morphology was rescued (Supplementary
function, and glucose-stimulated insulin secretion from Figure S4E – H and J), suggesting that mitochondrial fragmenta-
pancreatic islets tion arose from defects induced by ROS accumulation in b cells
Since elevated ROS levels can result in mitochondrial fragmenta- silenced for DJ-1.
tion (Mai et al., 2010; Nikic et al., 2011), we studied whether DJ-1 As a measure of mitochondrial function, we then determined
was required for normal mitochondrial morphology in pancreatic adenosine triphosphate (ATP) levels in DJ-1-deficient islets as
b cells. To this end, we compared the mitochondria of DJ-1- well as in MIN6 cells silenced for DJ-1. In b cells, ATP was
deficient b cells with those of controls in situ (Figure 4A–H). shown to be a key metabolic signal from mitochondria that trans-
A marked fragmentation was found to accompany the increased lates elevated nutrient levels into increased insulin secretion
ROS production in pancreatic b cells (Figure 4B vs F). While the (Maechler and Wollheim, 2001; Ashcroft, 2005). Interestingly,
majority of wild-type mitochondria formed networks in the ATP levels were not altered in DJ-1-deficient islets under low-
primary b cells, the mitochondria in DJ-1-deficient b cells were glucose conditions (Figure 4I, white columns). In contrast, ATP
224 | Journal of Molecular Cell Biology Jain et al.

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
Figure 3 Reduction in DJ-1 expression increases mitochondrial ROS production in mouse insulinoma cells. (A – D) Degree of oxidation of roGFP2
measured in mouse insulinoma cells (MIN6) transfected with mitochondrial roGFP2 (A), or cytoplasmic roGFP2 (B), under reducing (DTT) or
oxidizing (Aldrithiol-2) conditions. The cells were also transfected with control siRNA, DJ-1 siRNA-1, DJ-1 siRNA-2, and DJ-1 siRNA-2 + DJ-1
cDNA expression vector (rescue) (C and D). *P , 0.05, **P , 0.01 (Non-parametic test by Kruskal– Wallis analysis of variance with Dunnet’s
post hoc test for comparison of multiple groups). n ¼ 15 cells each. (E –P) LSM live cell images of MIN6 cells taken at 530 nm after excitation
at 405 and 488 nm. The ratios of the emitted fluorescence intensities (405/488 nm) are also shown, since a higher ratio (405/488 nm) indicates
an increased amount of ROS in mitochondria. MIN6 cells were transfected as indicated. Scale bar, 10 mm.

levels in response to stimulatory high-glucose concentrations old mice (Figure 4M). Glibenclamide, a sulphonylurea, is one of
were significantly lower in DJ-1-deficient islets compared with the two oral anti-diabetic drugs in the WHO list of essential med-
controls (Figure 4I, black columns), and addition of NAC partially icines for treatment of T2DM. Like other sulphonylureas, it closes
rescued the ATP production under high-glucose conditions the KATP channels of b cells independent of an increase in ATP
(Figure 4J). Similar results were obtained using MIN6 cells levels (Ashcroft, 2005). Importantly, glibenclamide was found to
silenced for DJ-1 (Supplementary Figure S4K and L). induce insulin secretion from pancreatic islets regardless of the
Since ATP is an important signal for glucose-stimulated insulin presence of DJ-1 (Figure 4M, grey columns), suggesting that
secretion (GSIS; Lowell and Shulman, 2005; Leibiger and DJ-1 acts upstream of KATP channels in pancreatic b cells. Since
Berggren, 2006; Muoio and Newgard, 2008), we studied islets harbor endocrine cells other than b cells (Thorel et al.,
whether the reduction of ATP production led to reduced GSIS 2010), we also used MIN6 cells to corroborate our results
from DJ-1-deficient islets. As shown in Figure 4K, GSIS was not obtained with pancreatic islets (Supplementary Figure S5). In
significantly changed in islets isolated from DJ-1-deficient mice these cells, DJ-1 was similarly required for glucose-stimulated
at the age of 8 weeks, showing that DJ-1 was not constitutively rather than glibenclamide-induced insulin secretion
required for GSIS. In contrast, DJ-1-deficient islets from 12 to 13 (Supplementary Figure S5A and B).
weeks old mice showed a significant reduction in GSIS compared Age- and diet-dependent glucose intolerance and reduced b cell
with controls (Figure 4K), which was not due to lower insulin area in the absence of DJ-1
content (Figure 4L). Glucose intolerance can be a consequence of b cell dysfunction
To demonstrate that DJ-1 is required for GSIS at this age and/or reduced b cell area (Ueki et al., 2006; Poy et al., 2009;
because of its effect on mitochondria and the glucose-stimulated Sachdeva et al., 2009; Liew et al., 2010; Esterhazy et al., 2011).
increase in ATP levels, we tested glibenclamide-induced insulin Since we observed reduced b cell function in DJ-1-deficient
secretion from DJ-1-deficient islets taken from 12 to 13 weeks animals, we evaluated glucose tolerance and b cell area in
Role of DJ-1 in type 2 diabetes Journal of Molecular Cell Biology | 225

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
Figure 4 Age-dependent requirement of DJ-1 for mitochondrial integrity, function and GSIS from mouse pancreatic islets. (A – H) LSM images of
islets in the pancreas of control (A – D) and DJ-1-deficient mice (E – H) stained with DAPI (A and E), anti-VDAC1 antibodies (B and F), and anti-
insulin antibodies (C, G). Merged images are shown in D and H. Scale bars, 10 mm. (I and J) ATP production in islets from control and
DJ-1-deficient mice. Data are presented as percentage of basal control in the absence (I) or presence of NAC (J). n ¼ 3. (K) Insulin secretion
(normalized to total insulin content) from mouse islets at a low (2 mM) and a high (25 mM) glucose concentration. Islets were isolated from
DJ-1-deficient mice and controls at 8 and 12– 13 weeks of age. n ¼ 3. (L) Total insulin content (normalized to total protein content) of islets
isolated from DJ-1-deficient mice and controls at 8 and 12– 13 weeks of age. n ¼ 3. (M) Insulin secretion (normalized to total insulin
content) from mouse islets at a low-glucose concentration (2 mM), a high-glucose concentration (25 mM), and at a low-glucose concentration
(2 mM) in the presence of 1 mM glibenclamide. n ¼ 3. *P , 0.05 (two-tailed Student’s t-test). All values are means + SD.

these animals (Figure 5). Consistent with the age-dependent To investigate whether glucose intolerance could also be
changes in islet physiology, DJ-1-deficient mice did not show induced in younger animals, we placed 6 weeks old mice on a
any defect in glucose-stimulated plasma insulin (Figure 5A), high-fat diet for 2 weeks. DJ-1 mRNA and protein levels were ele-
glucose tolerance (Figure 5B), and b cell area (Figure 5C) at the vated in the islets from these mice compared with islets from mice
age of 8 weeks. At 12– 13 weeks of age, however, DJ-1-deficient kept on a chow diet (Supplementary Figure S6). In contrast, there
mice had significantly lower levels of plasma insulin after was no diet-dependent change in mRNA level for any other anti-
glucose challenge (Figure 5D), displayed glucose intolerance oxidant protein we studied (Supplementary Figure S6A).
(Figure 5E), and had a small but significant reduction in b cell Importantly, plasma insulin was significantly reduced upon
area (Figure 5F). glucose challenge in the 8 weeks old mice that had been kept
226 | Journal of Molecular Cell Biology Jain et al.

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
Figure 5 Age- and diet-dependent glucose intolerance and reduced b cell area in the absence of DJ-1. (A, D, and G) Plasma insulin levels of male
DJ-1-deficient mice and controls before (0 min), and 40 min after intraperitoneal administration of glucose (2 g/kg) at 8 weeks of age (A), 12– 13
weeks of age (D) and at 8 weeks of age after 2 weeks on high-fat diet (HFD; G). n ¼ 7 –9. (B, E, and H) Glucose tolerance tests of male mice
having the indicated genotype after intraperitoneal administration of glucose (2 g/kg) at 8 weeks of age (B), 12 –13 weeks of age (E), and at 8
weeks of age after 2 weeks on HFD (H). n ¼ 6 – 10. (C, F, and I) Morphometric analyses of relative b cell mass from DJ-1-deficient mice and
controls quantified and calculated as insulin-positive area in total area of evenly spaced pancreatic sections at 8 weeks of age (C), 12– 13
weeks of age (F) and at 8 weeks of age after 2 weeks on HFD (I). n ¼ 3. *P , 0.05, **P , 0.01 (two-tailed Student’s t-test). All values are
means + SD. (J) Model. During aging and upon weight gain, DJ-1 is upregulated in pancreatic b cells to maintain ROS at constant low
levels, so they do not disturb mitochondrial morphology and function. Thus, elevated blood glucose levels accurately increase the intracellular
ATP concentration, a prerequisite for GSIS and maintenance of normal glucose tolerance (right green panel). Following a failure to increase DJ-1
expression, b cells do not efficiently reduce ROS levels upon aging and weight gain. The increased amount of ROS not only affects the morph-
ology of the mitochondria, but also compromises their function. This lowers the ability of b cells to produce ATP in response to glucose stimu-
lation and results in glucose intolerance, a first step in the development of T2DM (left red panel).

on a high-fat diet for 2 weeks (Figure 5A vs G). Additionally, conserved protein that is expressed in bacteria, plants, and
glucose intolerance was detected (Figure 5B vs H), and the b animals (Lin et al., 2011). A few different scenarios have been pro-
cell area of the fat mice was reduced to a small but significant posed for how DJ-1 reduces high ROS levels or oxidative stress in
extent compared with that of the age-matched controls that had cells. For example, DJ-1 has a conserved cysteine residue
been kept on a chow diet (Figure 5C vs I). This metabolic pheno- (Cys106), which allows DJ-1 to act both as an ROS scavenger
type was observed in male mice (Figure 5) and, to a lesser but sig- and sensor of cellular redox homeostasis (Canet-Aviles et al.,
nificant extent, in female mice (Supplementary Figure S7). The 2004). It has also been suggested that DJ-1 acts as a transcrip-
data show that DJ-1 is specifically required for GSIS, normal b tional co-activator (Inberg and Linial, 2010), stabilizes the
cell area and glucose tolerance during aging and on a high-fat NF-E2-related factor (Nrf2) (Clements et al., 2006), and is an inte-
diet. gral mitochondrial protein that maintains the activity of mitochon-
drial complex I in mammalian cells (Hayashi et al., 2009). In plant
Discussion cells, it was recently shown that DJ-1 interacts with glutathione
DJ-1 is an antioxidant protein, and its loss leads to an early peroxidase (gpx) and superoxide dismutase 1 (SOD1), and acti-
onset of PD (Bonifati et al., 2003). In neurons, DJ-1 deficiency vates the latter (Xu et al., 2010).
was shown to increase ROS as well as mitochondrial fragmenta- We have shown previously that the DJ-1 protein is significantly
tion and dysfunction (Surmeier et al., 2011). DJ-1 is a highly upregulated in the islets of mice when they are fed with a high-fat
Role of DJ-1 in type 2 diabetes Journal of Molecular Cell Biology | 227

diet and become hyperglycemic (Waanders et al., 2009). In add- pancreatic islets than 79 + 3 years old males (Supplementary
ition, DJ-1 was found to be upregulated in mouse islets cultured Table S3), a finding that might help us to explain gender-specific
in vitro under hyperglycemic conditions (Waanders et al., 2009; differences in the incidence and phenotype of PD and T2DM, re-
Inberg and Linial, 2010). However, the role of DJ-1 in maintaining spectively (Ding et al., 2006; Wirdefeldt et al., 2011).
pancreatic islet function and glucose homeostasis in vivo Based on our findings, we now propose that DJ-1 expression
remained unknown. Furthermore, no information was available increases in pancreatic b cells upon aging and a high-fat diet to
on the role of DJ-1 in islets during aging and on a high-fat diet. prevent a pathological increase in mitochondrial ROS levels
The data of this study now suggest that DJ-1 is required to main- (Figure 5J, right panel). This function helps in preserving mito-
tain physiological ROS levels in an age- and diet-dependent chondrial morphology and physiology during aging and weight
manner (Figure 5J). gain, thus maintaining normal ATP-dependent GSIS and glucose
Physiological levels of ROS are important to maintain glucose tolerance (Figure 5J, right panel). In contrast, when DJ-1 expres-
homeostasis, whereas prolonged exposure to high ROS levels sion fails to increase, ROS levels begin to increase beyond a
results in chronic oxidative stress and impaired glucose homeo- normal level, thereby altering mitochondrial morphology and
stasis (Robertson, 2004; Wang and Hai, 2011). Importantly, pan- function in pancreatic islets. This results in glucose intolerance,

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
creatic islets are particularly vulnerable to oxidative stress as they a first step in the development of T2DM (Figure 5J, left panel).
express low levels of antioxidant proteins (Lenzen et al., 1996; Since we identified T2DM as a human disease where DJ-1 expres-
Tiedge et al., 1997). In addition, increased ROS levels are sion is significantly reduced in elderly humans, similar to the situ-
observed in animal models of T2DM (i.e. db/db mice, GK rats, ation observed in PD, our data point to a more general role of the
and ZDF rats) and human T2DM patients (Ihara et al., 1999; oxidative stress modulator DJ-1 in age- and lifestyle-related
Kaneto et al., 1999; Tanaka et al., 1999). human diseases.
Mitochondria are a major source of ROS, and mutations in mito- Finally, our findings suggest that DJ-1 expression could be tar-
chondrial DNA in humans and b-cell-specific deletion of mito- geted for the treatment of T2DM. Along this line, it has recently
chondrial genes in animal models cause T2DM in humans and been shown that the histone deacetylase inhibitor, phenylbuty-
mice, respectively (Silva et al., 2000; Maassen et al., 2004). Our rate increases the expression of DJ-1 by 300% in the N27 dopa-
data point to mitochondria as a main site of elevated ROS produc- mine cell line and that it is useful for the treatment of both PD
tion in pancreatic b cells during aging, and we show that DJ-1 is and glucose intolerance (Ozcan et al., 2006; Xiao et al., 2011;
required to reduce mitochondrial fragmentation in pancreatic b Zhou et al., 2011). Besides phenylbutyrate, substances have
cells during aging and on a high-fat diet. This fragmentation been developed to specifically increase the activity of DJ-1 by pre-
might contribute to the reduced ATP levels and impaired GSIS venting its hyperoxidation and inactivation (Inden et al., 2011;
observed in DJ-1-deficient islets, as mitochondrial fission proteins Kitamura et al., 2011). Since we showed that upregulation of
have been shown to influence ATP levels and GSIS in pancreatic DJ-1 in pancreatic islets is key to maintain GSIS and blood
islets and b cell lines (Park et al., 2008; Twig et al., 2008). glucose levels during aging and weight gain, our study now war-
Oxidative stress and mitochondrial dysfunction are also the rants investigations on compounds that increase DJ-1 expression
hallmarks of PD (Surmeier et al., 2011), and a recent report indi- or activity for the treatment of T2DM, the most common metabolic
cates an 40% increase in prevalence of PD in T2DM patients (Xu disease worldwide.
et al., 2011). In turn, several studies reveal glucose intolerance in
patients with PD (Aviles-Olmos et al., 2012). The link between PD Materials and methods
and T2DM might lie in the disruption of common molecular Human islet isolation and analysis of DJ-1 expression
mechanisms. Alternatively, PD may simply result from hypergly- Human pancreatic islets were prepared with the approval of the
cemia, hyperinsulinemia, or both. In support of the first scenario, local ethics committees from the pancreases of multi-organ
we showed that DJ-1 is not only expressed in the substantia nigra, donors by collagenase digestion followed by density gradient
which gets functionally impaired during PD (Surmeier et al., purification. Then, the islets were maintained in M199 culture
2011), but is also expressed in human pancreatic islets, which medium, containing 5.5 mM glucose, supplemented with 10%
become dysfunctional during T2DM. Similar to the situation in (v/v) serum and 100 U/ml penicillin, 100 mg/ml streptomycin,
PD where the concentration of DJ-1 decreases in the cerebrospinal 50 mg/ml gentamicin, and 750 ng/ml amphotericin B
fluid (Hong et al., 2010), we found that in the islets of elderly (Sigma-Aldrich). Total RNA was extracted, quantified, and its in-
T2DM patients, DJ-1 expression is decreased compared with age- tegrity was assessed using an Agilent 2100 Bioanalyzer (Agilent
matched controls. A possible reason for a lower expression of DJ-1 Technologies). For semiquantitative RT– PCR experiments, 1 mg
during T2DM might be an epigenetic silencing, since several total RNA was reverse transcribed with iScript cDNA Synthesis
genes have been shown to be epigenetically downregulated in Kit (Bio-Rad Laboratories), and the following primers were used
the islets from T2DM patients (Ling et al., 2008; Yang et al., 2011). for PCR: DJ-qPCR_huRP1 (5′ -GACCACATCACGGCTACACT-3′ ) and
Since DJ-1 regulates the androgen receptor (Niki et al., 2003) DJ-1-qPCR_huFP1 (5′ -GAGCTGGGATTAAGGTCACCG-3′ ). The level
that binds to the tyrosine hydroxylase (TH) promoter, active in of mRNA for DJ-1 was quantified using an Applied Biosystems
both the neurons of the substantia nigra and the beta cells of pan- 7700 Bioanalyzer (Applied Biosystems) and normalized to the
creatic islets, we also studied whether gender-specific differences housekeeping gene hypoxanthine-guanine phosphoribosyltrans-
in DJ-1 gene expression could be detected. Interestingly, 74 + 7 ferase. DJ-1 and b-actin protein levels were measured in human
years old females expressed significantly more DJ-1 in their islet lysates using specific enzyme-linked immunosorbent assay
228 | Journal of Molecular Cell Biology Jain et al.

(ELISA) kits (MBL international cooperation; Cell signaling). sample at the excitation wavelengths of 405 and 488 nm. I488ox
Mouse model and glucose tolerance test and I488red are the fluorescence intensities at the excitation
Control (C57BL/6J) and DJ-1-deficient (B6.Cg-Park7tm1Shn/J) wavelength of 488 nm of the fully oxidized and fully reduced
mice were purchased from Jackson Laboratory (Goldberg et al., sample, respectively.
2005), and fed with standard laboratory chow or a high-fat diet Immunohistochemistry, immunocytochemistry, morphometric,
(Research Diets INC), and drinking water ad libitum. All experi- and western blot analyses
ments were approved by the local Animal Ethics Committee. If Immunofluorescence studies were performed with antibodies
not mentioned, the age of mice was always more than 12 against DJ-1 (Santa Cruz) and insulin (DAKO) on 4%
weeks. Glucose tolerance tests were carried out after the mice paraformaldehyde fixed pancreas sections. Secondary antibodies
had been fasted overnight. were conjugated with AlexaFluor488 (Molecular Probes) and with
Cell culture and RNA interference Cy5 (Dianova). DAPI (Sigma) was used to stain cell nuclei.
MIN6 cells (Miyazaki et al., 1990) were used from passage 29– Mitochondria of cells in culture were stained using MitoTracker
35 and were electroporated via nucleofection (Lonza) according Deep Red 633 (Molecular Probes). In addition, the mitochondria
to the manufacturer’s instructions with either pEGFP-DJ-1 or of b cells in pancreatic sections were stained using an antibody

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
pc-DNA3-FLAG-DJ-1 plasmid (kindly provided by Dr Hiroyoshi against VDAC 1 (Abcam; voltage-dependent anion channel).
Ariga at the University of Hokkaido, Japan) and/or siRNA. siRNA Pancreatic sections and MIN6 cells were imaged using a Carl
against DJ-1 was prepared as previously described (Nikolova Zeiss LSM 710 confocal microscope and quantified using the soft-
et al., 2006; Konstantinova et al., 2007), and knockdown efficien- ware ImageJ/Fiji. Rabbit anti-DJ-1 (Santa Cruz) and rabbit
cies were tested using western blots. anti-GAPDH antibodies (Abcam) were used for western blots.
Insulin secretion and content The intensities of DJ-1 signal were normalized to the signal of
Islets and MIN6 cells were maintained at 378C for 1 h in 2 mM GAPDH and presented as percent of basal control.
glucose, 25 mM glucose, or 2 mM glucose with 1 mM glibencla- Measurement of cellular ATP levels
mide in Krebs-Ringer HEPES buffer. The amount of insulin Cellular ATP levels were measured using the ApoSENSOR ATP
secreted into the buffer was measured using an ultrasensitive assay kit (Biovision) according to the manufacturer’s instruc-
rat insulin ELISA (Crystal Chem.). Total insulin content was deter- tions. Luminescence was determined using an LB9507 lumin-
mined in lysates of islets or MIN6 cells using the same ELISA kit. ometer (Berthold Technologies) and normalized to total protein
Measurement of ROS levels levels.
ROS levels were measured in islets using the ROS-sensitive
fluorescent dye CM-H2DCFDA (Invitrogen). After starvation in Supplementary material
2 mM glucose, islets were stimulated with 25 mM glucose con- Supplementary material is available at Journal of Molecular Cell
taining 10 mM CM-H2DCFDA. Islets were homogenized and after Biology online.
a centrifugation step, the fluorescence was measured in the
supernatant using a microplate reader (Tecan, infinite M1000).
Acknowledgements
The fluorescence intensity was also detected in intact pancreatic
We thank all our colleagues in Düsseldorf, in particular
islets using an LSM 710 confocal microscope with an excitation
B. Bartosinska for the islet isolation, and T. Dick in Heidelberg
wavelength of 488 nm and emission wavelength at 520 nm. In
for helpful advice and discussions.
addition, ROS levels were measured in MIN6 cells co-transfected
with roGFP2 plasmid (kindly provided by the University of Oregon,
Eugene, OR, USA) and siRNA against DJ-1 or control siRNA. Cells Funding
were starved for 1 h in KRH buffer containing 2 mM glucose and This work was supported by the Collaborative Research Center
then treated with 25 mM glucose, 25 mM glucose containing 590 (SFB 590, B11 to R.J., D.E., E.L.), the European Foundation for
10 mM DTT to reduce or 500 mM Aldrithiol-2 (Ald) to oxidize the the Study of Diabetes and Lilly Research Fellowship (EFSD to D.J.),
samples. The fluorescence intensities (I) of cells were measured and the German Center for Diabetes Research of the Federal
at excitation wavelengths of 405 and 488 nm and emission wave- Ministry for Education and Research (DZD to J.E., E.L.).
lengths of 530 nm using an LSM 710 confocal microscope. The
Conflict of interest: none declared.
fluorescence intensities were determined using ImageJ, and the
degree of oxidation of roGFP2 was determined using fluorescence
measurements after fully reducing the samples with DTT and then References
Andres-Mateos, E., Perier, C., Zhang, L., et al. (2007). DJ-1 gene deletion
fully oxidizing with Aldrithiol-2 using
reveals that DJ-1 is an atypical peroxiredoxin-like peroxidase. Proc. Natl
Acad. Sci. USA 104, 14807 – 14812.
OxDroGFP2 = R − Rred /[I488 ox /I488 red (Rox − R) + (R − Rred )] Ashcroft, F.M. (2005). ATP-sensitive potassium channelopathies: focus on
insulin secretion. J. Clin. Invest. 115, 2047 – 2058.
(Meyer and Dick, 2010), where R is the ratio of the fluorescence Aviles-Olmos, I., Limousin, P., Lees, A., et al. (2012). Parkinson’s disease,
insulin resistance and novel agents of neuroprotection. Brain, doi:
intensities of the sample at the excitation wavelengths of 405
10.1093/brain/aws009.
and 488 nm, Rred is the ratio of the intensities of the fully Bonifati, V., Rizzu, P., van Baren, M.J., et al. (2003). Mutations in the DJ-1 gene
reduced sample at the excitation wavelengths of 405 and associated with autosomal recessive early-onset parkinsonism. Science
488 nm, and Rox is the ratio of the intensities of the fully oxidized 299, 256 – 259.
Role of DJ-1 in type 2 diabetes Journal of Molecular Cell Biology | 229

Canet-Aviles, R.M., Wilson, M.A., Miller, D.W., et al. (2004). The Parkinson’s Lowell, B.B., and Shulman, G.I. (2005). Mitochondrial dysfunction and type 2
disease protein DJ-1 is neuroprotective due to cysteine-sulfinic acid-driven diabetes. Science 307, 384 – 387.
mitochondrial localization. Proc. Natl Acad. Sci. USA 101, 9103 –9108. Maassen, J.A., ‘t Hart, L.M., Van Essen, E., et al. (2004). Mitochondrial dia-
Clements, C.M., McNally, R.S., Conti, B.J., et al. (2006). DJ-1, a cancer- and betes: molecular mechanisms and clinical presentation. Diabetes
Parkinson’s disease-associated protein, stabilizes the antioxidant transcrip- 53(Suppl 1), S103 – S109.
tional master regulator Nrf2. Proc. Natl Acad. Sci. USA 103, 15091 –15096. Maechler, P., and Wollheim, C.B. (2001). Mitochondrial function in normal and
Collombat, P., Xu, X., Heimberg, H., et al. (2010). Pancreatic beta-cells: from diabetic beta-cells. Nature 414, 807 –812.
generation to regeneration. Semin. Cell Dev. Biol. 21, 838 – 844. Maedler, K. (2008). Beta cells in type 2 diabetes—a crucial contribution to
Dawson, T.M., and Dawson, V.L. (2003). Molecular pathways of neurodegen- pathogenesis. Diabetes Obes. Metab. 10, 408– 420.
eration in Parkinson’s disease. Science 302, 819 – 822. Mai, S., Klinkenberg, M., Auburger, G., et al. (2010). Decreased expression of
Ding, E.L., Song, Y., Malik, V.S., et al. (2006). Sex differences of endogenous Drp1 and Fis1 mediates mitochondrial elongation in senescent cells and
sex hormones and risk of type 2 diabetes: a systematic review and enhances resistance to oxidative stress through PINK1. J. Cell Sci. 123,
meta-analysis. J. Am. Med. Assoc. 295, 1288– 1299. 917 – 926.
Esterhazy, D., Stutzer, I., Wang, H., et al. (2011). Bace2 is a beta cell-enriched McCoy, M.K., and Cookson, M.R. (2012). Mitochondrial quality control and dy-
protease that regulates pancreatic beta cell function and mass. Cell Metab. namics in Parkinson’s disease. Antioxid. Redox Signal. 16, 869 –882.
14, 365 –377. Meyer, A.J., and Dick, T.P. (2010). Fluorescent protein-based redox probes.

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
Goldberg, M.S., Pisani, A., Haburcak, M., et al. (2005). Nigrostriatal dopamin- Antioxid. Redox Signal. 13, 621 –650.
ergic deficits and hypokinesia caused by inactivation of the familial Miyazaki, J., Araki, K., Yamato, E., et al. (1990). Establishment of a pancreatic
Parkinsonism-linked gene DJ-1. Neuron 45, 489 –496. beta cell line that retains glucose-inducible insulin secretion: special refer-
Guzman, J.N., Sanchez-Padilla, J., Wokosin, D., et al. (2010). Oxidant stress ence to expression of glucose transporter isoforms. Endocrinology 127,
evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. 126 – 132.
Nature 468, 696 – 700. Muoio, D.M., and Newgard, C.B. (2008). Mechanisms of disease: molecular
Hanson, G.T., Aggeler, R., Oglesbee, D., et al. (2004). Investigating mitochon- and metabolic mechanisms of insulin resistance and beta-cell failure in
drial redox potential with redox-sensitive green fluorescent protein indica- type 2 diabetes. Nat. Rev. Mol. Cell Biol. 9, 193 – 205.
tors. J. Biol. Chem. 279, 13044 –13053. Niki, T., Takahashi-Niki, K., Taira, T., et al. (2003). DJBP: a novel DJ-1-binding
Hao, L.Y., Giasson, B.I., and Bonini, N.M. (2010). DJ-1 is critical for mitochon- protein, negatively regulates the androgen receptor by recruiting histone
drial function and rescues PINK1 loss of function. Proc. Natl Acad. Sci. deacetylase complex, and DJ-1 antagonizes this inhibition by abrogation
USA 107, 9747 – 9752. of this complex. Mol. Cancer Res. 1, 247– 261.
Hayashi, T., Ishimori, C., Takahashi-Niki, K., et al. (2009). DJ-1 binds to mito- Nikic, I., Merkler, D., Sorbara, C., et al. (2011). A reversible form of axon
chondrial complex I and maintains its activity. Biochem. Biophys. Res. damage in experimental autoimmune encephalomyelitis and multiple scler-
Commun. 390, 667 – 672. osis. Nat. Med. 17, 495 –499.
Hong, Z., Shi, M., Chung, K.A., et al. (2010). DJ-1 and alpha-synuclein in human Nikolova, G., Jabs, N., Konstantinova, I., et al. (2006). The vascular basement
cerebrospinal fluid as biomarkers of Parkinson’s disease. Brain 133, membrane: a niche for insulin gene expression and beta cell proliferation.
713 – 726. Dev. Cell 10, 397 – 405.
Ihara, Y., Toyokuni, S., Uchida, K., et al. (1999). Hyperglycemia causes oxida- Ozcan, U., Yilmaz, E., Ozcan, L., et al. (2006). Chemical chaperones reduce ER
tive stress in pancreatic beta-cells of GK rats, a model of type 2 diabetes. stress and restore glucose homeostasis in a mouse model of type 2 dia-
Diabetes 48, 927 – 932. betes. Science 313, 1137– 1140.
Inberg, A., and Linial, M. (2010). Protection of pancreatic beta-cells from Park, K.S., Wiederkehr, A., Kirkpatrick, C., et al. (2008). Selective actions of
various stress conditions is mediated by DJ-1. J. Biol. Chem. 285, mitochondrial fission/fusion genes on metabolism-secretion coupling in
25686 – 25698. insulin-releasing cells. J. Biol. Chem. 283, 33347 – 33356.
Inden, M., Kitamura, Y., Takahashi, K., et al. (2011). Protection against dopa- Poy, M.N., Hausser, J., Trajkovski, M., et al. (2009). miR-375 maintains normal
minergic neurodegeneration in Parkinson’s disease-model animals by a pancreatic alpha- and beta-cell mass. Proc. Natl Acad. Sci. USA 106,
modulator of the oxidized form of DJ-1, a wild-type of familial Parkinson’s 5813 –5818.
disease-linked PARK7. J. Pharmacol. Sci. 117, 189 – 203. Robertson, R.P. (2004). Chronic oxidative stress as a central mechanism for
Kaneto, H., Kajimoto, Y., Miyagawa, J., et al. (1999). Beneficial effects of anti- glucose toxicity in pancreatic islet beta cells in diabetes. J. Biol. Chem.
oxidants in diabetes: possible protection of pancreatic beta-cells against 279, 42351 – 42354.
glucose toxicity. Diabetes 48, 2398 –2406. Robertson, R.P., and Harmon, J.S. (2006). Diabetes, glucose toxicity, and oxi-
Kitamura, Y., Watanabe, S., Taguchi, M., et al. (2011). Neuroprotective effect of dative stress: a case of double jeopardy for the pancreatic islet beta cell.
a new DJ-1-binding compound against neurodegeneration in Parkinson’s Free Radic. Biol. Med. 41, 177 – 184.
disease and stroke model rats. Mol. Neurodegener. 6, 1– 19. Sachdeva, M.M., Claiborn, K.C., Khoo, C., et al. (2009). Pdx1 (MODY4) regu-
Konstantinova, I., Nikolova, G., Ohara-Imaizumi, M., et al. (2007). lates pancreatic beta cell susceptibility to ER stress. Proc. Natl Acad. Sci.
EphA-Ephrin-A-mediated beta cell communication regulates insulin secre- USA 106, 19090 –19095.
tion from pancreatic islets. Cell 129, 359 – 370. Silva, J.P., Kohler, M., Graff, C., et al. (2000). Impaired insulin secretion and
Leibiger, I.B., and Berggren, P.O. (2006). Sirt1: a metabolic master switch that beta-cell loss in tissue-specific knockout mice with mitochondrial diabetes.
modulates lifespan. Nat. Med. 12, 34– 36. Nat. Genet. 26, 336 – 340.
Lenzen, S. (2008). Oxidative stress: the vulnerable beta-cell. Biochem. Soc. Soltani, N., Qiu, H., Aleksic, M., et al. (2011). GABA exerts protective and regen-
Trans. 36, 343 – 347. erative effects on islet beta cells and reverses diabetes. Proc. Natl Acad. Sci.
Lenzen, S., Drinkgern, J., and Tiedge, M. (1996). Low antioxidant enzyme gene USA 108, 11692 –11697.
expression in pancreatic islets compared with various other mouse tissues. Surmeier, D.J., Guzman, J.N., Sanchez-Padilla, J., et al. (2011). The role of
Free Radic. Biol. Med. 20, 463 – 466. calcium and mitochondrial oxidant stress in the loss of substantia nigra
Liew, C.W., Bochenski, J., Kawamori, D., et al. (2010). The pseudokinase trib- pars compacta dopaminergic neurons in Parkinson’s disease.
bles homolog 3 interacts with ATF4 to negatively regulate insulin exocytosis Neuroscience 198, 221 –231.
in human and mouse beta cells. J. Clin. Invest. 120, 2876 – 2888. Tanaka, Y., Gleason, C.E., Tran, P.O., et al. (1999). Prevention of glucose tox-
Lin, J., Nazarenus, T.J., Frey, J.L., et al. (2011). A plant DJ-1 homolog is essential icity in HIT-T15 cells and Zucker diabetic fatty rats by antioxidants. Proc.
for Arabidopsis thaliana chloroplast development. PLoS One 6, e23731. Natl Acad. Sci. USA 96, 10857 – 10862.
Ling, C., Del Guerra, S., Lupi, R., et al. (2008). Epigenetic regulation of Thorel, F., Nepote, V., Avril, I., et al. (2010). Conversion of adult pancreatic
PPARGC1A in human type 2 diabetic islets and effect on insulin secretion. alpha-cells to beta-cells after extreme beta-cell loss. Nature 464,
Diabetologia 51, 615 – 622. 1149 – 1154.
230 | Journal of Molecular Cell Biology Jain et al.

Tiedge, M., Lortz, S., Drinkgern, J., et al. (1997). Relation between antioxidant Wirdefeldt, K., Adami, H.O., Cole, P., et al. (2011). Epidemiology and etiology of
enzyme gene expression and antioxidative defense status of insulin- Parkinson’s disease: a review of the evidence. Eur. J. Epidemiol. 26(Suppl 1),
producing cells. Diabetes 46, 1733– 1742. S1– S58.
Twig, G., Elorza, A., Molina, A.J., et al. (2008). Fission and selective fusion Xiao, C., Giacca, A., and Lewis, G.F. (2011). Sodium phenylbutyrate, a drug with
govern mitochondrial segregation and elimination by autophagy. EMBO J. known capacity to reduce endoplasmic reticulum stress, partially alleviates
27, 433 – 446. lipid-induced insulin resistance and beta-cell dysfunction in humans.
Ueki, K., Okada, T., Hu, J., et al. (2006). Total insulin and IGF-I resistance in pan- Diabetes 60, 918 – 924.
creatic beta cells causes overt diabetes. Nat. Genet. 38, 583 – 588. Xu, X.M., Lin, H., Maple, J., et al. (2010). The Arabidopsis DJ-1a protein confers
van Arensbergen, J., Garcia-Hurtado, J., Moran, I., et al. (2010). Derepression of stress protection through cytosolic SOD activation. J. Cell Sci. 123,
Polycomb targets during pancreatic organogenesis allows insulin-producing 1644 – 1651.
beta-cells to adopt a neural gene activity program. Genome Res. 20, 722–732. Xu, Q., Park, Y., Huang, X., et al. (2011). Diabetes and risk of Parkinson’s
Waanders, L.F., Chwalek, K., Monetti, M., et al. (2009). Quantitative proteomic ana- disease. Diabetes Care 34, 910 –915.
lysis of single pancreatic islets. Proc. Natl Acad. Sci. USA 106, 18902–18907. Yang, B.T., Dayeh, T.A., Kirkpatrick, C.L., et al. (2011). Insulin promoter DNA
Wang, X., and Hai, C.X. (2011). ROS acts as a double-edged sword in the patho- methylation correlates negatively with insulin gene expression and positive-
genesis of type 2 diabetes mellitus: is Nrf2 a potential target for the treat- ly with HbA(1c) levels in human pancreatic islets. Diabetologia 54, 360 – 367.
ment? Mini Rev. Med. Chem. 11, 1082 – 1092. Zhou, W., Bercury, K., Cummiskey, J., et al. (2011). Phenylbutyrate up-regulates

Downloaded from http://jmcb.oxfordjournals.org/ at Lund University Libraries, Head Office on August 18, 2015
Wiederkehr, A., and Wollheim, C.B. (2006). Minireview: implication of mito- the DJ-1 protein and protects neurons in cell culture and in animal models of
chondria in insulin secretion and action. Endocrinology 147, 2643 –2649. Parkinson disease. J. Biol. Chem. 286, 14941 – 14951.

You might also like