ArticlePDF Available

Functional and morphological alterations of mitochondria in pancreatic beta cells from type 2 diabetic patients

Authors:

Abstract and Figures

Little information is available on the insulin release properties of pancreatic islets isolated from type 2 diabetic subjects. Since mitochondria represent the site where important metabolites that regulate insulin secretion are generated, we studied insulin release as well as mitochondrial function and morphology directly in pancreatic islets isolated from type 2 diabetic patients. Islets were prepared by collagenase digestion and density gradient purification, and insulin secretion in response to glucose and arginine was assessed by the batch incubation method. Adenine nucleotides, mitochondrial membrane potential, the expression of UCP-2, complex I and complex V of the respiratory chain, and nitrotyrosine levels were evaluated and correlated with insulin secretion. Compared to control islets, diabetic islets showed reduced insulin secretion in response to glucose, and this defect was associated with lower ATP levels, a lower ATP/ADP ratio and impaired hyperpolarization of the mitochondrial membrane. Increased protein expression of UCP-2, complex I and complex V of the respiratory chain, and a higher level of nitrotyrosine were also found in type 2 diabetic islets. Morphology studies showed that control and diabetic beta cells had a similar number of mitochondria; however, mitochondrial density volume was significantly higher in type 2 diabetic beta cells. In pancreatic beta cells from type 2 diabetic subjects, the impaired secretory response to glucose is associated with a marked alteration of mitochondrial function and morphology. In particular, UCP-2 expression is increased (probably due to a condition of fuel overload), which leads to lower ATP, decreased ATP/ADP ratio, with consequent reduction of insulin release.
Content may be subject to copyright.
Diabetologia (2005) 48: 282289
DOI 10.1007/s00125-004-1627-9
ARTICLE
M. Anello
.
R. Lupi
.
D. Spampinato
.
S. Piro
.
M. Masini
.
U. Boggi
.
S. Del Prato
.
A. M. Rabuazzo
.
F. Purrello
.
P. Marchetti
Functional and morphological alterations of mitochondria
in pancreatic beta cells from type 2 diabetic patients
Received: 27 April 2004 / Accepted: 4 September 2004 / Published online: 15 January 2005
# Springer-Verlag 2005
Abstract Aims/hypothesis: Little information is available
on the insulin release properties of pancreatic islets iso-
lated from type 2 diabetic subjects. Since mitochondria
represent the site where important metabolites that regulate
insulin secretion are generated, we studied insulin release
as well as mitochondrial function and morphology directly
in pancreatic islets isolated from type 2 diabetic patients.
Methods: Islets were prepared by collagenase digestion
and density gradient purification, and insulin secretion in
response to glucose and arginine was assessed by the batch
incubation method. Adenine nucleotides, mitochondrial
membrane potential, the expression of UCP-2, complex I
and complex V of the respiratory chain, and nitrotyrosine
levels were evaluated and correlated with insulin secretion.
Results: Compared to control islets, diabetic islets showed
reduced insulin secretion in response to glucose, and this
defect was associated with lower ATP levels, a lower
ATP/ADP ratio and impaired hyperpolarization of the
mitochondrial membrane. Increased protein expression of
UCP-2, complex I and complex V of the respiratory chain,
and a higher level of nitrotyrosine were also found in type
2 diabetic islets. Morphology studies showed that control
and diabetic beta cells had a similar number of mitochon-
dria; however, mitochondrial density volume was signif-
icantly higher in type 2 diabetic beta cells. Conclusions/
interpretation: In pancreatic beta cells from type 2 dia-
betic subjects, the impaired secretory response to glucose
is associated with a marked alteration of mitochondrial
function and morphology. In particular, UCP-2 expression
is increased (probably due to a condition of fuel overload),
which leads to lower ATP, decreased ATP/ADP ratio, with
consequent reduction of insulin release.
Keywords Adenine nucleotides
.
Insulin secretion
.
Mitochondria
.
Type 2 diabetes
.
UCP-2
Abbreviations ADP: adenosine diphosphate
.
ATP:
adenosine triphosphate
.
BMI: body mass index
.
BSA:
bovine serum albumin
.
FCCP: carbonylcyanide p-
trifluoromethoxyphenylhydrazone
.
KRB: krebsRinger
bicarbonate solution
.
ΔΨ
m
: mitochondrial membrane
potential
.
NEFA: non-esterified fatty acid
.
Rh123:
rhodamine-123
.
SI: stimulation index
.
TCA:
trichloracetic acid
.
TMB: tetramethyl-benzidine
.
UCP-2:
uncoupling protein-2
Introduction
Type 2 diabetes mellitus is a metabolic and vascular
disease that has reached epidemic proportions, and rep-
resents a serious health concern. Its prevalence worldwide
is set to increase from its present level of 150 million to
225 million by the end of the decade [1]. Moreover, its
incidence is increasing at an alarming rate also in children
and adolescents [2]. The long-term complications of this
disease carry a crushing burden of morbidity and mortal-
ity, and most type 2 diabetic patients die prematurely from
a cardiovascular event [35].
Type 2 diabetes is characterized by defective pancreatic
beta cell insulin release in response to glucose and by
impaired insulin action on its target tissues. The relative
importance of the secretory defects has been recently
outlined by several clinical observations. Insulin resistance
alone is not sufficient to lead to type 2 diabetes in the
absence of a beta cell defect [68]. Patients with impaired
M. Anello
.
D. Spampinato
.
S. Piro
.
A. M. Rabuazzo
.
F. Purrello
Internal Medicine, Department of Internal and Specialistic
Medicine, University of Catania, Ospedale Cannizzaro,
Catania, Italy
R. Lupi
.
M. Masini
.
U. Boggi
.
S. Del Prato
.
P. Marchetti
Department of Endocrinology and Metabolism, Metabolic Unit,
University of Pisa,
Pisa, Italy
F. Purrello (*)
Clinica Medica, Ospedale Cannizzaro,
Via Messina 829,
95126 Catania, Italy
e-mail: fpurrello@virgilio.it
Tel.: +39-095-7262053
Fax: +39-095-7262582
glucose tolerance or in the early stages of type 2 diabetes
always present with defects of beta cell secretion [9].
Clinical diabetes develops only when the compensatory
hypersecretion of insulin by the pancreatic beta cell de-
clines [8]. Moreover, as demonstrated in the UKPDS study
[10], type 2 diabetic patients are characterized by a pro-
gressive decline of insulin secretion that becomes more
severe with the increasing duration of the disease.
Conceivably, a more direct assessment of the functional
characteristics of the diabetic beta cell would represent a
better tool for identification of alterations associated with
impaired insulin secretion. However, little information is
available on the insulin release properties of islets isolated
from type 2 diabetic subjects. Pancreatic islets were stud-
ied from seven type 2 diabetic patients (obtained by intra-
operative biopsy) [11]. The authors reported that despite a
marked reduction of glucose-stimulated insulin secretion
in vivo, a normal insulin release was induced by glucose
from the isolated islets, suggesting that extrapancreatic
factors influence beta cell reaction to glucose in type 2
diabetes. Another study investigated insulin secretion func-
tion in pancreatic islets from two type 2 diabetic organ
donors, and found a marked decrease of insulin secretion
during glucose stimulation, although the secretory response
to a combination of leucine and glutamine was less se-
verely affected [12]. In a recent report, islets from diabetic
donors secreted less insulin and exhibited an elevated
threshold for glucose-induced insulin release [13].
The altered insulin secretory pattern might depend on
genetic and/or acquired abnormalities, including the neg-
ative influence of chronic high glucose [1417] and/or
high non-esterified fatty acids (NEFA) [1821] plasma
concentrations (gluco- or lipo-toxicity). In normal beta
cells glucose regulates insulin release through its metab-
olism, and mitochondria represent the site where important
metabolites that regulate insulin secretion are generated
[2224]. Several studies have focused the attention on the
adenine nucleotides as regulators of insulin secretion. In
particular, the increase of ATP/ADP ratio tightly associates
to glucose-induced insulin granule release [25, 26]. In
addition to mitochondrial glucose oxidation, ATP synthe-
sis and ATP/ADP ratio are regulated by uncoupling
protein-2 (UCP-2) expression [2730]. UCP-2 is a mem-
ber of a family of proteins located in the mitochondrial
inner membrane, which act as proton channels uncoupling
mitochondrial oxidative phosphorylation. By this mecha-
nism, energy is wasted through heat and cellular ATP
synthesis is decreased [31].
The aim of this work was, therefore, to investigate in-
sulin secretion and mitochondrial function and morphology
in human islets from type 2 diabetic patients. We measured
adenine nucleotides, mitochondrial membrane potential,
the expression of UCP-2, complex I and complex V of the
respiratory chain, nitrotyrosine levels, and correlated them
with insulin secretion. Moreover, we studied mitochondrial
ultrastructure. We found distinct differences between dia-
betic and non-diabetic subjects.
Materials and methods
Human islet preparation Pancreatic islets were prepared
by collagenase digestion and density gradient purification,
as previously reported [32, 33]. All protocols were ap-
proved by the local Ethics Committee. For this study, islets
were obtained from 11 non-diabetic human multiorgan do-
nors (age 58±5.4 years, BMI 24.6±1.4 kg/m
2
, mean±SEM),
and from seven type 2 diabetic patients (age 65±6 years,
BMI 27.4±2.2 kg/m
2
, mean±SEM). Mean duration of
clinical diabetes was 5.6±0.6 years; plasma glucose con-
centration, at the time of admission, was 273.3±38.5 mg/dl.
Four diabetic donors were treated with only diet restriction;
two with sulphonylurea treatment; one with both sulpho-
nylurea and metformin. Three diabetic subjects and three
controls were also screened for GAD antibodies, which
resulted negative. Digestion time was similar in control
(38±3 min) and diabetic (36±4 min) islet isolations. At the
end of the isolation procedure, islets were resuspended in
M199 culture medium (containing 5.5 mmol/l glucose),
supplemented with 10% adult bovine serum, antibiotics
(penicillin, 100 U/ml; streptomycin, 100 μg/ml; gentami-
cin, 50 μg/ml; and amphotericin B, 0.25 μg/ml), and cul-
tured at 37°C in 5% CO
2
.
Insulin secretion Insulin secretion studies were performed
by the batch incubation technique as previously described
[33, 34]. Following a 45-min period of incubation at 37°C
in medium containing 3.3 mmol/l glucose, groups of ap-
proximately 30 islets of comparable size were kept at 37°C
for 45 min in KrebsRinger bicarbonate solution (KRB),
0.5% albumin, pH 7.4, containing 3.3 mmol/l glucose. At
the end of this period, medium was completely removed,
assayed to measure basal insulin secretion, and replaced
with KRB containing either 16.7 mmol/l glucose, or 3.3
mmol/l glucose plus 20 mmol/l arginine. After additional
45-min incubation, medium was removed, and insulin
levels were measured to assess stimulated insulin re-
lease. Insulin secretion was expressed as absolute value, as
percent of islet insulin content, and as stimulation index
(SI), i.e. the ratio of stimulated over basal insulin secretion
[35]. Insulin concentrations were measured by a commer-
cially available immunoradiometric assay (Pantec For-
niture Biomediche, Turin, Italy).
Adenine nucleotide measurement Adenine nucleotides were
measured as previously reported [36]. Following islet in-
cubation with 3.3 or 16.7 mmol/l glucose, the experiments
were stopped by the addition of 0.125 ml of trichloracetic
acid (TCA) (Sigma, St. Louis, MO, USA). The extracts were
frozen at 80°C until the day of the assay, which started with
an appropriate further dilution. ATP and ADP were assayed
in triplicate by a luminometric method [37]. To measure total
ATP+ADP, ADP was first converted into ATP. Samples, with
known concentrations of ADP, without ATP, were run in
parallel to check that the transformation was complete. ATP
was measured by the addition of a reagent containing lu-
ciferase and luciferin (Sigma, St. Louis, MO, USA). The
emitted light was measured in a luminometer (Junior LB
283
9509-Berthold Technologies, Germany). To measure only
ATP, the same previously described procedure was fol-
lowed, except that in the first incubation step pyruvate
kinase was lacking. ADP levels were then calculated by
subtracting ATP from the total ATP+ADP. Blanks and ATP
standards were run through the entire procedure, including
the extraction steps.
Mitochondrial membrane potential (ΔΨ
m
) ΔΨ
m
was mea-
sured using rhodamine-123 (Rh123) (Sigma, St. Louis,
MO, USA) as an indicator of mitochondrial membrane po-
tential changes in an islet cell suspension under glucose
stimulation (16.7 mmol/l). Cell were prepared from 3,000
human pancreatic islets according to the method described
before [16]. Briefly, islets were transferred to Ca
2+
-free
KRB at 30°C with 1 mmol/l EGTA, 16.5 μg/ml trypsin,
2 μ g/ml DNAse (Boehringer, Mannheim, Germany), and
were gently resuspended with a Pasteur pipette. Cell dis-
sociation was monitored, by observing the suspension with
a microscope. Single cell suspension was then cultured in
M199 medium overnight at 37°C in a 95% O
2
/5% CO
2
atmosphere. Islet cells were then loaded in KRB buffer
containing 3.3 mmol/l glucose and 10 μg/ml Rh123 for 30
min at 37°C. Cells were resuspended in the same buffer
without Rh123 and transferred to a fluorometer (Hitachi
F-2000) cuvette, and the fluorescence excited at 490 nm
was measured at 530 nm, at 37°C with gentle stirring. Re-
sults are expressed as percentage of basal fluorescence (at
3.3 mmol/l glucose).
Determination of nitrotyrosine Nitrotyrosine concentra-
tions were determined in islet cell lysates by an ELISA
method as reported [38]. Briefly, 96-well plates were
coated with 200 μl of standard curve samples (0.16615
nmol/l) and 1 μg/μl of lysate (65 μl/well) in 0.1 mol/l
carbonatebicarbonate buffer (135 μl), pH 9.6, overnight
at 4°C. Afterwards, non-specific binding sites were blocked
with 1% BSA in PBS-T (PBS plus 0.05% Tween 20), for
1 h at 37°C and washed with PBS-T. Plates were then
incubated with purified monoclonal anti-nitrotyrosine mouse
IgG for 1 h at 37°C, washed and incubated with peroxi-
dase-conjugated goat anti-mouse IgG secondary antibody
for 45 min at 37°C. Peroxidase reaction product was gen-
erated using tetramethyl-benzidine (TMB) Microwell Per-
oxidase Substrate (Sigma, St. Louis, MO, USA). Plates
were then incubated 510 min at room temperature and the
reaction was stopped with 0.5 mol/l H
2
SO
4
, and optical
density read at 492 nm in a microplate reader.
Western blot analysis Uncoupling protein-2 (UCP-2),
NADH-ubiquinone oxidoreductase (complex I), F
1
-ATP-
synthase (complex V) and SREBP-1c protein levels were
measured by western blot analysis. Briefly, groups of 300
human islets were homogenized by sonication in SDS-
PAGE sample buffer and equivalent amounts of proteins
were separated on SDS-polyacrylamide gel (Mini-Protean,
Bio-Rad, Hercules, CA, USA) and electrophoretically trans-
ferred onto nitrocellulose membrane (Amersham Pharmacia
Biotech, England). Blotting efficiency as well as the posi-
tion of protein standards was assessed by Ponceau staining.
After blocking, the membranes were incubated with a rabbit
polyclonal anti-UCP-2 antibody (Alpha Diagnostic Inter-
national, San Antonio, TX, USA) at 1:2,000 dilution in
blocking solution, or with a monoclonal anti-NADH-ubi-
quinone oxidoreductase (Molecular Probes, Eugene, OR,
USA) 1:1,000, or with a goat polyclonal anti-F
1
-ATP-
synthase antibody (Santa Cruz Biotechnology, Inc., USA)
1:1,000, or with a monoclonal anti-SREBP-1c (2A4) anti-
body (Santa Cruz Biotechnology, Inc., USA) 1:1,000 dilu-
tion at 4°C, overnight. The membranes were then blotted
with an anti-rabbit (1:2,000) or an anti-mouse (1:5,000) IgG
peroxidase-linked whole antibody (Pierce, Rockford, IL,
USA), or with a monoclonal anti-goat IgG peroxidase con-
jugate (Sigma, St. Louis, MO, USA) diluted 1:10,000, 1 h
at room temperature. Peroxidase activity was detected
using ECL (Amersham Pharmacia Biotech, England).
Electron microscopy evaluation Electron microscopy stud-
ies were performed as previously described [33, 34, 39].
Pancreatic samples were fixed with 2.5% glutaraldehyde in
0.1 mol/l cacodylate buffer, pH 7.4 for 1 h at 4°C. After
rinsing in cacodylate buffer, the tissue was postfixed in 1%
cacodylate buffered osmium tetroxide for 2 h at room tem-
perature, then dehydrated in a graded series of ethanol,
briefly transferred to propylene oxide and embedded in
Epon-Araldite. Ultrathin sections (6080 nm thick) were
cut with a diamond knife, placed on formvar-carbon coated
copper grids (200 mesh), and stained with uranyl acetate
and lead citrate.
Statistical analysis Data are presented as the mean±SEM.
Statistical significance was assessed by Students t-test, or
one-way ANOVA followed by NewmanKeuls test when
more than two groups were compared. p Values of less
than 0.05 were considered statistically significant.
Results
Insulin secretion As shown in Table 1, glucose (16.7 mmol/
l)-induced insulin release was significantly lower from the
diabetic as compared to non-diabetic islets. Since islets
from diabetic subjects contained 34% less insulin than
control islets (78±4.7 vs. 118±4.2 μU/islet, p<0.01), we
also expressed our data as percent of islet insulin content
(Table 2). Using this method, glucose-induced insulin
secretion was again lower in diabetic islets. Using both
methods, arginine-stimulated insulin release did not differ
significantly between the two groups (Tables 1 and 2).
These experiments, therefore, showed a selective defect of
type 2 diabetes beta cells to release insulin in response to
glucose stimulation.
Measurements of adenine nucleotides content The ATP-
to-ADP ratio, also in human islets, plays a critical role in
glucose-induced beta cell insulin secretion [25]. There-
fore, we measured adenine nucleotide content in pan-
creatic islets from diabetic and non-diabetic subjects, in
284
the presence of basal (3.3 mmol/l) and stimulating (16.7
mmol/l) glucose concentrations (Fig. 1). Islets from dia-
betic subjects had a higher ATP content in basal condition
(14.22±1.58 vs. 9.82±0.31 pmol/μg of islet DNA, n=30
replicates from type 2 diabetic subjects vs. 45 from
controls, p<0.01) and a higher ATP/ADP ratio (Fig. 1).
In response to glucose stimulation, ATP levels signifi-
cantly increased in control islets (from 9.82±0.31 to
16.11±0.27 pmol/μg of islet DNA, p<0.001, but not in
diabetic islets (from 14.22±1.58 to 13.22±1.31 pmol/μg
of islet DNA), the latter being significantly lower than
control (p=0.01). As a consequence, in response to glu-
cose stimulation, the ATP/ADP ratio was lower in dia-
betic subjects than in control group (15.85±0.98 vs. 24.14±
1.77, p<0.001) (Fig. 1).
Mitochondrial membrane potential measurements Since
the energy to drive ATP formation is provided by a proton
gradient across the inner mitochondrial membrane, we
measured glucose-induced changes in mitochondrial mem-
brane potential (ΔΨ
m
)[40]. The Rh123 fluorescence was
recorded in a cell suspension from islets of diabetic and
non-diabetic subjects, as indicated on a graph section. In
control cells when glucose concentration was increased to
16.7 mmol/l, fluorescence decreased (9.6±0.1%, mean±
SEM, n=5), indicating the glucose-induced hyperpolari-
zation of ΔΨ
m
(Fig. 2a). Cells from diabetic subjects
showed a decreased hyperpolarization of ΔΨ
m
when glu-
cose was raised to 16.7 mmol/l (6.5±0.54%, mean±SEM,
n=4, p<0.001) (Fig. 2b). The addition of the uncoupler
carbonylcyanide p-trifluoromethoxyphenylhydrazone (FCCP)
1 μmol/l readily depolarised ΔΨ
m
in both the experimental
conditions (Fig. 2).
Mitochondrial protein expression To analyse the expres-
sion of mitochondrial proteins involved in pancreatic beta
cell energy production, we studied, by Western blot, the
levels of the uncoupling protein-2 (UCP-2), and of com-
plex I (NADH-ubiquinone oxidoreductase) and complex
V(F
1
-ATP synthase) of the respiratory chain. We found
that UCP-2 protein levels were significantly increased in
islets from diabetic subjects (+24±6%, mean±SEM, n=4,
p<0.01) compared to control subjects (Fig. 3). We also
Fig. 1 Adenine nucleotide concentrations (ATP in panel a, ADP in
panel b, ATP-to-ADP ratio in panel c). Batches of five islets from
control (white bar) or type 2 diabetic subjects (black bar) were
incubated in 1 ml of KRB medium containing the indicated glucose
concentrations. At the end of incubation (1 h), islets were processed
for measuring adenine nucleotide content. Results are means±SEM
of 45 replicates from five control and 30 replicates from four type 2
diabetic subjects. ***p<0.001 vs 3.3 mmol/l glucose,
§
p<0.05 vs
control,
§§
p<0.01 vs control,
§§§
p<0.001 vs control
Table 2 Insulin secretion as percentage of islet insulin content/45
min, under glucose (Glu) or arginine (Arg) stimulation in pancreatic
islets from control and type 2 diabetic subjects
Glu (3.3
mmol/l)
Glu (16.7
mmol/l)
Arg (20
mmol/l)
Controls (n=11) 1.98±0.14 4.77±0.38a
b
4.82±0.46a
b
Type 2 diabetes
(n=7)
2.8±0.34b
a
3.29±0.41b
a
5.52±0.71a
b
Data are means ± SEM
a
p<0.05 or less vs. 3.3 mmol/l glucose
b
p<0.05 or less vs. controls
Table 1 Insulin secretion (μU/islet/45 min), under glucose (Glu) or arginine (Arg) stimulation in pancreatic islets from control and type 2
diabetic subjects
Glu (3.3 mmol/l) Glu (16.7 mmol/l) SI-Glu Arg (20 mmol/l) SI-Arg
Controls (n=11) 2.34±0.17 5.63±0.45
a
2.5±0.2 5.71±0.54
a
2.4±0.26
Type 2 diabetes (n=7) 2.18±0.26 2.57±0.32
b
1.26±0.19
b
4.3±0.55
a
2.0±0.14
Data are means ± SEM
a
p<0.05 or less vs. 3.3 mmol/l glucose
b
p<0.05 or less vs. controls
SI-Glu Ratio of glucose-stimulated over basal insulin secretion, SI-Arg ratio of arginine-stimulated over basal insulin secretion
285
found that protein expression of both complex I and V of
the respiratory chain were increased in islets from diabetic
subjects (+14±4.5% and +31±13%, n=4, p<0.01) com-
pared to control (Fig. 3). In order to elucidate if up-
regulation of UCP-2 was due to a higher expression of
SREBP-1c, we also measured the levels of this latter
protein and found no differences between diabetic and
control groups (Fig. 4).
Nitrotyrosine levels Respiratory chain activity leads to the
formation of reactive oxygen species. To investigate if the
increased levels of the respiratory chain enzymes in islets
from diabetic subjects might lead to increased oxidative
stress, we measured nitrotyrosine levels. This compound
derives from the reaction of superoxide and nitric oxide,
and is considered a reliable marker of oxidative stress.
Nitrotyrosine levels were respectively 7.2±0.4 and 9.9±0.4
nmol/l in control islets (n=7) and type 2 diabetes islets
(n=6) (p<0.05).
Fig. 2 Glucose-induced mitochondrial membrane potential changes
(ΔΨ
m
). Glucose (16.7 mmol/l) induced hyperpolarisation in human
pancreatic islet cell suspension, from non-diabetic (a) and diabetic
subjects (b). The depolarizing effect of the protonophore FCCP at
the end of each trace is used as control of the ΔΨ
m
integrity. Results
are percentage of basal fluorescence (at 3.3 mmol/l glucose). The
traces represent mean of five separate experiments for control and
four separate experiments for diabetic subjects. c Summary of ΔΨ
m
changes over the basal level in glucose-induced hyperpolarization of
mitochondrial membrane potential in control (white bar) and in type
2 diabetic subjects (black bar). Results are percentage of decrement
under basal fluorescence (at 3.3 mmol/l glucose). ***p<0.001
Fig. 3 A representative Western blot for UCP-2, F
1
-F
0
ATPsynthase
(complex V) and NADH-ubiquinol oxidoreductase (complex I) in
human pancreatic islets from non-diabetic (lane 1) and type 2 dia-
betic subjects (lane 2). Results are mean±SEM of scanning den-
sitometry relative to islet actin of four separate western blots.
*p<0.05, **p<0.01 vs. non-diabetic subjects
Fig. 4 Representative western blot for SREBP-1c in human pan-
creatic islets from non-diabetic (lane 1) and type 2 diabetic subjects
(lane 2)
286
Electron microscopy studies Endocrine cellular composi-
tion from pancreas preparation was 69±4, 22±4 and 9±2%
in controls and 61±3, 25±7 and 14±5% in diabetic patients
(mean±SEM, n=4), for beta, alpha and delta cells, respec-
tively. Cell viability, measured as trypan blue exclusion,
was higher than 90% in both controls (n=5) and diabetic
patients (n=3). As shown in Fig. 5, mitochondria in type 2
diabetes beta cells appeared round-shaped and hypertro-
phic. Compared to control beta cells (n=112 cells, from
three pancreases), type 2 diabetes beta cells (n=108 cells,
from three pancreases) had a similar number of mito-
chondria (12.0±0.9 vs. 12.4±0.6 per microscopy field).
However, the mitochondrial density volume from type 2
diabetic subjects was significantly (p<0.01) higher (4.7
±0.3 ml %) than control beta cells (3.1±0.4 ml %).
Discussion
In islets isolated from the pancreas of seven multiorgan
donors who were affected by type 2 diabetes, we observed
a clearly reduced insulin release in response to glucose,
whereas the secretion of the hormone during stimulation
with the non-fuel secretagogue arginine was only slightly
affected. In order to investigate the basis of this selective
defect, since mitochondrial metabolism, and the subse-
quent rise of ATP and of ATP/ADP ratio plays a central
role in glucose-induced insulin release, we measured
several key steps of the mitochondrial events that lead to
ATP synthesis and correlated them with insulin secretion.
The energy for ATP production is provided by oxidation
of reducing equivalents via the electron-transport chain.
The enzyme complexes I to V are located at the inner
mitochondrial membrane and the flux of electrons along
the respiratory chain establishes the proton gradient, which
generates the membrane potential. Glucose stimulation
results in the transfer of reducing equivalents to the res-
piratory chain, leading to hyperpolarization of the mito-
chondrial membrane (ΔΨ
m
) and generation of ATP. In
islets from diabetic subjects we found that glucose-in-
duced hyperpolarization of the mitochondrial membrane
was reduced. We also found that ATP levels were lower,
at high glucose, and the ATP/ADP ratio was blunted, in
response to glucose stimulation. These defects could con-
ceivably be due to a reduced electron flux through the res-
piratory chain, or to an over-expression of proteins (such
as UCP-2) that tends to diminish the proton gradient gen-
erated by the respiratory chain. To test the first possibility
we measured the protein expression of complex I and
complex V of the respiratory chain and we found an in-
creased expression that makes this hypothesis unlikely. To
test the second possibility, we measured the protein ex-
pression of UCP-2, and we found, indeed, an increased
expression of this protein. UCP-2 is a member of a family
of proteins located in the mitochondrial inner membrane,
which uncouples mitochondrial oxidative phosphoryla-
tion. By this mechanism, energy is wasted through heat
and cellular ATP synthesis is decreased. UCP-2 protein
expression could be activated by an increased formation of
reactive oxygen species [41]. In agreement with this in-
terpretation, in our model we found increased levels of
nitrotyrosine (a marker of oxidative stress) in diabetic is-
lets. According to these data, therefore, it is possible to
suppose that in beta cells from diabetic patients the in-
creased expression of UCP-2 is responsible of the reduced
hyperpolarization of the mitochondrial membrane, lower
ATP levels, ATP/ADP ratio, and eventually, of the reduced
insulin release in response to glucose.
This sequence of events is coherent with several data
obtained in vitro or in animal models, and recently put in
perspective [42]. Increased UCP-2 levels in beta cells are
associated with decreased insulin secretion [43, 44], and
UCP-2 overexpression in rat pancreatic islets has been
shown to inhibit glucose-stimulated insulin secretion by de-
creasing ATP formation [45]. Moreover, in rodent pan-
creatic islets chronically exposed to high glucose or NEFA
glucose-induced impairment of insulin secretion is asso-
ciated with altered mitochondrial function, including over-
expression of the UCP-2 protein and a consequent decrease
of ATP production [46]. In islets from hyperglycaemic
90% pancreatectomized rats [47] or in human islets ex-
posed to high glucose [48], UCP-2 mRNA or protein ex-
pression was increased, in accordance with a decrease of
glucose-induced insulin release. In a tumoral beta cell line,
chronic exposure to high NEFA both reduced insulin
secretion and increased UCP-2 levels by regulating glu-
cose-induced ATP formation [49, 50]. In other reports,
UCP-2 overexpression by enhancing ATP/ADP ratio re-
stores insulin secretion in islets from ZDF rat [51].
Fig. 5 Mitochondrial structure
in pancreatic islets beta cell
from normal (a) and diabetic
subjects (b). White arrow indi-
cates mitochondria. Hashed
arrow indicates insulin granules.
Mitochondrial density volume
was significantly higher in beta
cells from type 2 diabetic sub-
jects compared to control sub-
jects (4.7±0.3 ml % vs. 3.1±0.4
ml %, p<0.01). Magnification
×16,000
287
Further support to the concept that mitochondria in the
diabetic beta cell are in an altered state comes from the
morphological studies we performed. In fact, by electron
microscopy examination, we found that the density vol-
ume of these organelles was significantly higher in type 2
diabetes beta cells than in control cells. Mitochondria
undergo structural changes that parallel their functional
state in both physiological as well as pathological con-
ditions [52], and their enlargement, which can be induced
by various pathological conditions, can be classified into
two categories: the swelling and the formation of mega-
mitochondria [53]. Both situations are considered to be
adaptive processes at the subcellular level to unfavourable
environments. For example, it has been demonstrated that
when cells are exposed to excess amount of free radicals,
the mitochondria become enlarged decreasing the rate of
oxygen consumption and reducing ROS production [54].
These mechanisms are likely to play a role also in diabetes
and in beta cell function. Indeed, evidence has been re-
cently reported of swelling of mitochondria in sural nerve
biopsies from patients with diabetic neuropathy [55]. In
addition, loss of glucose-stimulated insulin secretion from
isolated rat islets was associated with mitochondrial en-
largement [56]. Finally, we have shown that exposure of
human pancreatic islets to cytotoxic cytokines induces
functional and survival beta cell damage, which is ac-
companied by mitochondrial swelling and enlargement
[57, 58].
We observed a selective secretory defect in response to
glucose, whereas the secretion of the hormone during stim-
ulation with the non-fuel secretagogue arginine was only
slightly affected, demonstrating an intrinsic and selective
functional defect of beta cell function in type 2 diabetes. It is
noteworthy that arginine-induced insulin release is largely
independent on ATP synthesis, since this amino acid di-
rectly affects beta cell membrane potential and ion flux.
The present data, the first to our knowledge, indicate
that in pancreatic beta cells from type 2 diabetic subjects,
the impaired secretory response to glucose is associated
with a marked impairment of mitochondrial function. The
presence of excessive fuel availability increases substrate
influx through the metabolic mitochondria pathways,
leading to the generation of large amounts of high-energy
metabolites and reactive oxygen species. Our novel results
suggest that in the presence of such a situation, pancreatic
beta cell UCP-2 expression is increased in humans, which
leads to a lower ATP level and reduced ATP/ADP ratio in
response to glucose, with consequent impairment of insu-
lin release. A better understanding of these mechanisms,
and the discovering of specific molecular targets would
greatly enhance our clinical efficacy in preserving beta cell
function in type 2 diabetic patients.
References
1. King H, Aubert RE, Herman WH (1998) Global burden of
diabetes, 19952025: prevalence, numerical estimates, and
projections. Diabetes Care 21:14141431
2. American Diabetes Association (2000) Type 2 diabetes in
children and adolescents (consensus statement). Diabetes Care
23:381389
3. Adler AI, Boyko EJ, Ahroni JH, Smith DG (1999) Hypergly-
cemia and hyperinsulinemia at diagnosis of diabetes and their
association with cardiovascular disease in the United Kingdom
Prospective Diabetes Study (UKPDS 47). Am Heart J 138:
S353S359
4. Adler AI, Stratton IM, Neil HA et al (2000) Association of
systolic blood pressure with macrovascular and microvascular
complications of type 2 diabetes (UKPDS 36): prospective
observational study. BMJ 321:412419
5. U.K. Prospective Diabetes Study Group (1998) Tight blood
pressure control and risk of macrovascular and microvascular
complications in type 2 diabetes (UKPDS 38). BMJ 317:703
713
6. Polonsky KS, Sturis J, Bell G (1996) Non-insulin-dependent
diabetes mellitus: a genetically programmed failure of the beta
cell to compensate for insulin resistance. N Engl J Med 334:777
783
7. Weyer C, Bogardus C, Mott DM, Pratley RE (1999) The
natural history of insulin secretory dysfunction and insulin
resistance in the pathogenesis of type 2 diabetes mellitus. J Clin
Invest 104:787794
8. DeFronzo RA, Bonadonna RC, Ferrannini E (1992) Pathogen-
esis of NIDDM. A balanced overview. Diabetes Care 15:318
368
9. Ferrannini E, Gastaldelli A, Miyazaki Y et al (2003)
Predominant role of reduced beta-cell sensitivity to glucose
over insulin resistance in impaired glucose tolerance. Diabet-
ologia 46:12111219
10. UK Prospective Diabetes Study (UKPDS) Group (1998) In-
tensive blood-glucose control with sulphonylureas or insulin
compared with conventional treatment and risk of complica-
tions in patients with type 2 diabetes (UKPDS 33). Lancet
352:837853
11. Lohmann D, Jahr H, Verlohren HJ et al (1980) Insulin secretion
in maturity-onset-diabetes. Function of isolated islets. Horm
Metab Res 12:349 353
12. Fernandez-Alvarez J, Conget I, Rasschaert J, Sener A, Gomis
R, Malaisse WJ (1994) Enzymatic, metabolic and secretory
patterns in human islets of type 2 (non-insulin-dependent)
diabetic patients. Diabetologia 37:177181
13. Deng S, Vatamaniuk M, Huang X et al (2004) Structural and
functional abnormalities in the islets isolated from type 2
diabetic subjects. Diabetes 53:624632
14. Grodsky GM, Bolaffi JL (1992) Desensitization of the insulin-
secreting beta cell. J Cell Biochem 48:311
15. Leahy JL, Bonner-Weir S, Weir GC (1992) Beta-cell dysfunc-
tion induced by chronic hyperglycemia. Current ideas on
mechanism of impaired glucose-induced insulin secretion. Dia-
betes Care 15:442455
16. Purrello F, Vetri M, Gatta C, Gullo D, Vigneri R (1989) Effects
of high glucose on insulin secretion by isolated rat islets and
purified beta-cells and possible role of glycosylation. Diabetes
38:14171422
17. Robertson RP, Harmon J, Tran PO, Poitout V (2004) Beta-cell
glucose toxicity, lipotoxicity, and chronic oxidative stress in
type 2 diabetes. Diabetes 53 (Suppl 1):S119S124
18. Kashyap S, Belfort R, Gastaldelli A et al (2003) A sustained
increase in plasma free fatty acids impairs insulin secretion in
nondiabetic subjects genetically predisposed to develop type 2
diabetes. Diabetes 52:24612474
19. Unger RH, Zhou YT (2001) Lipotoxicity of beta-cells in
obesity and in other causes of fatty acid spillover. Diabetes 50
(Suppl 1):S118S121
20. Wyne KL (2003) Free fatty acids and type 2 diabetes mellitus.
Am J Med 115 (Suppl 8A):29S36S
21. Poitout V, Robertson RP (2002) Secondary beta-cell failure in
type 2 diabetes: a convergence of glucotoxicity and lipotoxicity.
Endocrinology 143:339342
288
22. Maechler P (2002) Mitochondria as the conductor of metabolic
signals for insulin exocytosis in pancreatic beta-cells. Cell Mol
Life Sci 59:18031818
23. Wollheim CB, Maechler P (2002) Beta-cell mitochondria and
insulin secretion: messenger role of nucleotides and metabo-
lites. Diabetes 51 (Suppl 1):S37S42
24. Maechler P, Wollheim CB (2001) Mitochondrial function in
normal and diabetic beta-cells. Nature 13:807812
25. Detimary P, Dejonghe S, Ling Z, Pipeleers D, Schuit F, Henquin
JC (1998) The changes in adenine nucleotides measured in
glucose-stimulated rodent islets occur in beta cells but not in
alpha cells and are also observed in human islets. J Biol Chem
273:3390533908
26. Henquin JC (2000) Triggering and amplifying pathways of
regulation of insulin secretion by glucose. Diabetes 49:1751
1760
27. Chan CB, Saleh MC, Koshkin V, Wheeler MB (2004) Un-
coupling protein 2 and islet function. Diabetes 53 (Suppl 1):
S136S142
28. Rousset S, Alves-Guerra MC, Mozo J et al (2004) The biology
of mitochondrial uncoupling proteins. Diabetes 53 (Suppl 1):
S130S135
29. Saleh MC, Wheeler MB, Chan CB (2002) Uncoupling protein-
2: evidence for its function as a metabolic regulator. Diabeto-
logia 45:174187
30. Zhang CY, Baffy G, Perret P et al (2001) Uncoupling protein-2
negatively regulates insulin secretion and is a major link
between obesity, beta cell dysfunction, and type 2 diabetes. Cell
15:745755
31. Chan CB, De Leo D, Joseph JW et al (2001) Increased
uncoupling protein-2 levels in beta-cells are associated with
impaired glucose-stimulated insulin secretion: mechanism of
action. Diabetes 50:13021310
32. Marchetti P, Dotta F, Ling Z et al (2000) Function of pancreatic
islets isolated from Type 1 diabetic patient. Diabetes Care
23:701703
33. Lupi R, Dotta F, Marselli L et al (2002) Prolonged exposure to
free fatty acids has cytostatic and pro-apoptotic effects on
human pancreatic islets. Evidence that β-cell death is caspase-
mediated, partially dependent on ceramide pathway, and Bcl-2
regulated. Diabetes 51:14371442
34. Marchetti P, Lupi R, Federici M et al (2002) Insulin secretory
function is impaired in isolated human islets carrying the Gly
(972)Arg IRS-1 polymorphism. Diabetes 51:14191424
35. Gatto C, Callegari M, Folin M et al (2003) Effects of cryo-
preservation and coculture with pancreatic ductal epithelial cells
on insulin secretion from human pancreatic islets. Int J Mol Med
12:851854
36. Anello M, Ucciardello V, Piro S et al (2001) Chronic exposure
to high leucine impairs glucose-induced insulin release by
lowering the ATP/ADP ratio. Am J Physiol Endocrinol Metab
281:E1082E1087
37. Hampp R (1986) Luminometric method. In: Bergmeyer HU
(ed) Methods of enzymatic analysis. Verlagsgesellschaft,
Weinheim, Germany, pp 370379
38. Quagliaro L, Piconi L, Assaloni R, Martinelli L, Motz E,
Ceriello A (2003) Intermittent high glucose enhances apoptosis
related to oxidative stress in human umbilical vein endothelial
cells: the role of protein kinase C and NAD(P)H-oxidase
activation. Diabetes 52:27952804
39. Weibel ER (1969) Stereological principles for morphometry in
electron microscopic cytology. Int Rev Cytol 26:235302
40. Duchen MR, Smith PA, and Ashcroft FM (1993) Substrate-
dependent changes in mitochondrial function, intracellular free
calcium concentration and membrane channels in pancreatic β-
cells. Biochem J 294:3542
41. Krauss S, Zhang CY, Scorrano L et al (2003) Superoxide-
mediated activation of uncoupling protein 2 causes pancreatic
beta cell dysfunction. J Clin Invest 112:18311842
42. Brownlee M (2003) A radical explanation for glucose-induced
beta cell dysfunction. J Clin Invest 112:17881790
43. Polonsky KS, Semenkovich CF (2001) The pancreatic beta cell
heats up: UCP2 and insulin secretion in diabetes. Cell 105:705
707
44. Joseph JW, Koshkin V, Zhang CY (2002) Uncoupling protein
2 knockout mice have enhanced insulin secretory capacity
after a high-fat diet. Diabetes 51:32113219
45. Chan CB, MacDonald PE, Saleh MC, Johns DC, Marban E,
Wheeler MB (1999) Overexpression of uncoupling protein 2
inhibits glucose-stimulated insulin secretion from rat islets.
Diabetes 48:14821486
46. Patane G, Anello M, Piro S, Vigneri R, Purrello F, Rabuazzo
AM (2002) Role of ATP production and uncoupling protein-2
in the insulin secretory defect induced by chronic exposure to
high glucose or free fatty acids and effects of peroxisome
proliferator-activated receptor-gamma inhibition. Diabetes
51:27492756
47. Laybutt DR, Sharma A, Sgroi DC, Gaudet J, Bonner-Weir S,
Weir GC (2002) Genetic regulation of metabolic pathways in
beta-cells disrupted by hyperglycemia. J Biol Chem 277:10912
10921
48. Brown JE, Thomas S, Digby JE, Dunmore SJ (2002) Glucose
induces and leptin decreases expression of uncoupling protein-
2 mRNA in human islets. FEBS Lett 27:189192
49. Medvedev AV, Robidoux J, Bai X et al (2002) Regulation of
the uncoupling protein-2 gene in INS-1 beta-cells by oleic acid.
J Biol Chem 277:4263942644
50. Lameloise N, Muzzin P, Prentki M, Assimacopoulos-Jeannet F
(2001) Uncoupling protein 2: a possible link between fatty acid
excess and impaired glucose-induced insulin secretion? Diabe-
tes 50:803809
51. Wang MY, Shimabukuro M, Lee Y et al (1999) Adenovirus-
mediated overexpression of uncoupling protein-2 in pancreatic
islets of Zucker diabetic rats increases oxidative activity and
improves beta-cell function. Diabetes 48:10201025
52. Wakabayashi T (2002) Megamitochondria formationphysi-
ology and pathology. J Cell Mol Med 6:497538
53. Wakabayashi T, Karbowski M (2001) Structural changes of
mitochondria related to apoptosis. Biol Signals Recept 10:26
56
54. Karbowski M, Kurono C, Wozniak M et al (1999) Free radical-
induced megamitochondria formation and apoptosis. Free Radic
Biol Med 26:396409
55. Vincent AM, Brownlee M, Russell JW (2002) Oxidative stress
and programmed cell death in diabetic neuropathy. Ann NY
Acad Sci 959:368383
56. Pai GM, Slavin BG, Tung P (1993) Morphologic basis for loss
of regulated insulin secretion by isolated rat pancreatic islets.
Anat Rec 237:498505
57. Marselli L, Dotta F, Piro S et al (2001) Th2 cytokines have a
partial, direct protective effect on the function and survival of
isolated human islets exposed to combined proinflammatory
and Th1 cytokines. J Clin Endocrinol Metab 86:49744978
58. Trincavelli ML, Marselli L, Falleni A et al (2002) Upregulation
of mitochondrial peripheral benzodiazepine receptor expression
by cytokine-induced damage of human pancreatic islets. J Cell
Biochem 84:636644
289
... β-cell failure has been proposed to be a central feature of T2D pathogenesis and may include changes in oxidative or endoplasmic reticulum (ER) stress (Swisa et al. 2017), mitochondrial network fragmentation and interruption of cell-cycle progression (Anello et al. 2005, Montemurro et al. 2019, maladaptation in insulin secretion and prohormone processing, including hyperinsulinemia, hyperproinsulinemia, and increased proinsulin-to-insulin ratio (Mykkänen et al. 1997, Pfutzner et al. 2004, Mezza et al. 2018, cellular exhaustion from glucotoxicity causing dedifferentiation and loss of mature identity (Spijker et al. 2015, Swisa et al. 2017, among others. β-cell identity loss can present with endocrine hormone colocalization, such as co-expression of insulin and glucagon (Talchai et al. 2012, Brereton et al. 2014, Spijker et al. 2015, Beamish et al. 2019, Beamish et al. 2022, as well as alterations or loss of nuclear location of key transcription factors including PDX1 and NKX6.1 (Talchai et al., 2012, Guo et al. 2013, Brereton et al. 2014, Spijker et al. 2015. ...
... Mitochondrial function and morphology have been shown to be altered in β cells from T2D human pancreas (Anello et al. 2005), shown by Tomm20 fluorescent area per β cell being significantly reduced in T2D vs nondiabetic islets (Montemurro et al. 2019). We therefore similarly examined mitochondrial fluorescence intensity using Tomm20 immunofluorescence, with results demonstrating a reduction of Tomm20 ( Fig. 6A and C, red) fluorescent intensity in β cells (insulin, green, A) in LDLr −/− HFD + PBS relative to chow-fed mice (Fig. 6A, lower right, 6B). ...
Article
Metabolic syndrome (MetS) is an increasing global health threat and strong risk factor for type 2 diabetes (T2D). MetS causes both hyperinsulinemia and islet size overexpansion, and pancreatic beta (β)-cell failure impacts insulin and proinsulin secretion, mitochondrial density, and cellular identity loss. The low-density lipoprotein receptor knockout (LDLr-/-) model combined with high fat diet (HFD) has been used to study alterations in multiple organs, but little is known about changes to β-cell identity resulting from MetS. Osteocalcin (OC), an insulin-sensitizing protein secreted by bone, shows promising impact on β-cell identity and function. LDLr-/- mice at 12mo were fed chow or HFD for 3 months ± 4.5 ng/h osteocalcin. Islets were examined by immunofluorescence for alterations in nuclear Nkx6.1 and PDX1 presence, insulin-glucagon colocalization, islet size and %β-cell and islet area by insulin and synaptophysin, and mitochondria fluorescence intensity by Tomm20. Bone mineral density (BMD) and %fat changes were examined by Piximus Dexa scanning. HFD-fed mice showed fasting hyperglycemia by 15mo, increased weight gain, %fat, and fasting serum insulin and proinsulin; concurrent OC treatment mitigated weight increase and showed lower proinsulin/insulin ratio, and higher BMD. HFD increased %β and %islet area, while simultaneous osteocalcin-treatment with HFD was comparable to chow-fed mice. Significant reductions in nuclear PDX1 and Nkx6.1 expression, increased insulin-glucagon colocalization, and reduction in β-cell mitochondria fluorescence intensity were noted with HFD, but largely prevented with OC administration. Osteocalcin supplementation here suggests a benefit to β-cell identity in LDLr-/- mice and offers intriguing clinical implications for countering metabolic syndrome.
... [83][84][85] The alterations in mitochondrial morphology and functional phenotypes resulting from genetic manipulation of multiple mitochondrial fusion and fission proteins in various βcell models have been thoroughly outlined. 86 There is evidence of perturbed mitochondrial morphology with swollen and enlarged mitochondria in pancreatic islets of patients with T2D, 87 as well as in β cells of glucose intolerant high-fat diet-fed obese mice. 88 This suggests altered mitochondrial dynamics in diabetic conditions. ...
Article
Full-text available
Pancreatic β cells play an essential role in the control of systemic glucose homeostasis as they sense blood glucose levels and respond by secreting insulin. Upon stimulating glucose uptake in insulin‐sensitive tissues post‐prandially, this anabolic hormone restores blood glucose levels to pre‐prandial levels. Maintaining physiological glucose levels thus relies on proper β‐cell function. To fulfill this highly specialized nutrient sensor role, β cells have evolved a unique genetic program that shapes its distinct cellular metabolism. In this review, the unique genetic and metabolic features of β cells will be outlined, including their alterations in type 2 diabetes (T2D). β cells selectively express a set of genes in a cell type‐specific manner; for instance, the glucose activating hexokinase IV enzyme or Glucokinase ( GCK ), whereas other genes are selectively “disallowed”, including lactate dehydrogenase A ( LDHA ) and monocarboxylate transporter 1 ( MCT1 ). This selective gene program equips β cells with a unique metabolic apparatus to ensure that nutrient metabolism is coupled to appropriate insulin secretion, thereby avoiding hyperglycemia, as well as life‐threatening hypoglycemia. Unlike most cell types, β cells exhibit specialized bioenergetic features, including supply‐driven rather than demand‐driven metabolism and a high basal mitochondrial proton leak respiration. The understanding of these unique genetically programmed metabolic features and their alterations that lead to β‐cell dysfunction is crucial for a comprehensive understanding of T2D pathophysiology and the development of innovative therapeutic approaches for T2D patients.
... 60 When cells misplaced, a large amount of ROS contributed to apoptosis condition of pancreatic βcells, resulting in limited regeneration ability. Thus, in the presence of increased β-cell loss, either by apoptosis or dedifferentiation, a scenario would arise in which β-cells were lost with insufficient replacement, resulting in inadequate insulin secretion to maintain proper glucose homeostasis, 61,62 and contributing to T2DM progression. 63 ...
Article
Full-text available
Background Mutations in mitochondrial tRNA (mt-tRNA) could be the origin of some type 2 diabetes mellitus (T2DM) cases, but the mechanism remained largely unknown. Aim The aim of this study was to assess the impact of a novel mitochondrial tRNACys/tRNATyr A5826G mutation on the development and progression of T2DM. Methods A four-generation Han Chinese family with maternally inherited diabetes underwent clinical, genetic and biochemical analyses. The mitochondrial DNA (mtDNA) mutations of three matrilineal relatives were screened by PCR-Sanger sequencing. Furthermore, to see whether m.A5826G mutations affected mitochondrial functions, the cybrid cell lines were derived from three subjects with m.A5826G mutation and three controls without this mutation. ATP was evaluated by luminescent cell viability assay, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) were determined by flow cytometry. The student’s two-tailed, unpaired t-test was used to assess the statistical significance between the control and mutant results. Results The age at onset of diabetes in this pedigree varied from 40 to 63 years, with an average of 54 years. Mutational analysis of mitochondrial genomes revealed the presence of a novel m.A5826G mutation. Interestingly, the m.A5826G mutation occurred at the conjunction between tRNACys and tRNATyr, a very conserved position that was critical for tRNAs processing and functions. Using trans-mitochondrial cybrid cells, we found that mutant cells carrying the m.A5826G showed approximately 36.5% and 22.4% reductions in ATP and MMP, respectively. By contrast, mitochondrial ROS levels increased approximately 33.3%, as compared with the wild type cells. Conclusion A novel m.A5826G mutation was identified in a pedigree with T2DM, and this mutation would lead to mitochondrial dysfunction. Thus, the genetic spectrum of mitochondrial diabetes was expanded by including m.A5826G mutation in tRNACys/tRNATyr, our study provided novel insight into the molecular pathogenesis, early diagnosis, prevention and clinical treatment for mitochondrial diabetes.
... Therefore, an impaired function and morphology of mitochondria in β cells may be key factors in the pathophysiology of T2D, and this has been verified in patients with manifestations of the disease. In patients with T2D, an increase in the expression of ETC complexes I and V is reported in pancreatic β cells [127]. This increase in the ETC could be the cause of excess ROS production, generating a state of oxidative stress. ...
Article
Full-text available
Type 2 diabetes (T2D) is characterized by a state of hyperglycemia in the blood due to insulin resistance developed by organs such as muscle, liver, and adipose tissue. A common factor in individuals with T2D is mitochondrial dysfunction. Mitochondria are dynamic organelles responsible for energy and antioxidant metabolism in the cells. Estrogens, such as 17β-estradiol (E2), are steroid hormones that have shown a great capacity to regulate mitochondrial function and dynamics through estrogen receptors (ERs), modulating the expression of mitochondrial biogenesis-related genes and cell signaling mechanisms. The accumulation of reactive oxygen species, the low capacity for ATP synthesis, and morphological alterations are some of the mitochondrial processes impaired in T2D. Insulin signaling and secretion by pancreatic β-cells, ATP-dependent processes, are also altered in T2D. In this review, mitochondria were exposed as the central axis for the action of estrogens in individuals with T2D. Estrogens increased glucose uptake, insulin signaling, and mitochondrial bioenergetics, and decreased ectopic lipid accumulation in non-adipose tissues and oxidative stress, among other processes, in various preclinical and clinical models of diabetes. The development of strategies to target compounds to mitochondria could represent a novel therapeutic alternative to potentiate the effects of estrogens on this organelle in patients with insulin resistance and T2D.
Article
Full-text available
Mitochondrial dysfunction in pancreatic β-cells leads to impaired glucose-stimulated insulin secretion (GSIS) and type 2 diabetes (T2D), highlighting the importance of autophagic elimination of dysfunctional mitochondria (mitophagy) in mitochondrial quality control (mQC). Imeglimin, a new oral anti-diabetic drug that improves hyperglycemia and GSIS, may enhance mitochondrial activity. However, chronic imeglimin treatment’s effects on mQC in diabetic β-cells are unknown. Here, we compared imeglimin, structurally similar anti-diabetic drug metformin, and insulin for their effects on clearance of dysfunctional mitochondria through mitophagy in pancreatic β-cells from diabetic model db/db mice and mitophagy reporter (CMMR) mice. Pancreatic islets from db/db mice showed aberrant accumulation of dysfunctional mitochondria and excessive production of reactive oxygen species (ROS) along with markedly elevated mitophagy, suggesting that the generation of dysfunctional mitochondria overwhelmed the mitophagic capacity in db/db β-cells. Treatment with imeglimin or insulin, but not metformin, reduced ROS production and the numbers of dysfunctional mitochondria, and normalized mitophagic activity in db/db β-cells. Concomitantly, imeglimin and insulin, but not metformin, restored the secreted insulin level and reduced β-cell apoptosis in db/db mice. In conclusion, imeglimin mitigated accumulation of dysfunctional mitochondria through mitophagy in diabetic mice, and may contribute to preserving β-cell function and effective glycemic control in T2D.
Article
Full-text available
In Type 1 and Type 2 diabetes, pancreatic β-cell survival and function are impaired. Additional etiologies of diabetes include dysfunction in insulin-sensing hepatic, muscle, and adipose tissues as well as immune cells. An important determinant of metabolic health across these various tissues is mitochondria function and structure. This review focuses on the role of mitochondria in diabetes pathogenesis, with a specific emphasis on pancreatic β-cells. These dynamic organelles are obligate for β-cell survival, function, replication, insulin production, and control over insulin release. Therefore, it is not surprising that mitochondria are severely defective in diabetic contexts. Mitochondrial dysfunction poses challenges to assess in cause-effect studies, prompting us to assemble and deliberate the evidence for mitochondria dysfunction as a cause or consequence of diabetes. Understanding the precise molecular mechanisms underlying mitochondrial dysfunction in diabetes and identifying therapeutic strategies to restore mitochondrial homeostasis and enhance β-cell function are active and expanding areas of research. In summary, this review examines the multidimensional role of mitochondria in diabetes, focusing on pancreatic β-cells and highlighting the significance of mitochondrial metabolism, bioenergetics, calcium, dynamics, and mitophagy in the pathophysiology of diabetes. We describe the effects of diabetes-related gluco/lipotoxic, oxidative and inflammation stress on β-cell mitochondria, as well as the role played by mitochondria on the pathologic outcomes of these stress paradigms. By examining these aspects, we provide updated insights and highlight areas where further research is required for a deeper molecular understanding of the role of mitochondria in β-cells and diabetes.
Article
Type 1 diabetes is a disease of the endocrine pancreas; however, it also affects exocrine function. Although most studies have examined the effects of diabetes on acinar cells, much less is known regarding ductal cells, despite their important protective function in the pancreas. Therefore, we investigated the effect of diabetes on ductal function. Diabetes was induced in wild‐type and cystic fibrosis transmembrane conductance regulator (CFTR) knockout mice following an i.p. administration of streptozotocin. Pancreatic ductal fluid and HCO 3 ⁻ secretion were determined using fluid secretion measurements and fluorescence microscopy, respectively. The expression of ion transporters was measured by real‐time PCR and immunohistochemistry. Transmission electron microscopy was used for the morphological characterization of the pancreas. Serum secretin and cholecystokinin levels were measured by an enzyme‐linked immunosorbent assay. Ductal fluid and HCO 3 ⁻ secretion, CFTR activity, and the expression of CFTR, Na ⁺ /H ⁺ exchanger‐1, anoctamine‐1 and aquaporin‐1 were significantly elevated in diabetic mice. Acute or chronic glucose treatment did not affect HCO 3 ⁻ secretion, but increased alkalizing transporter activity. Inhibition of CFTR significantly reduced HCO 3 ⁻ secretion in both normal and diabetic mice. Serum levels of secretin and cholecystokinin were unchanged, but the expression of secretin receptors significantly increased in diabetic mice. Diabetes increases fluid and HCO 3 ⁻ secretion in pancreatic ductal cells, which is associated with the increased function of ion and water transporters, particularly CFTR. image Key points There is a lively interaction between the exocrine and endocrine pancreas not only under physiological conditions, but also under pathophysiological conditions The most common disease affecting the endocrine part is type‐1 diabetes mellitus (T1DM), which is often associated with pancreatic exocrine insufficiency Compared with acinar cells, there is considerably less information regarding the effect of diabetes on pancreatic ductal epithelial cells, despite the fact that the large amount of fluid and HCO 3 ⁻ produced by ductal cells is essential for maintaining normal pancreatic functions Ductal fluid and HCO 3 ⁻ secretion increase in T1DM, in which increased cystic fibrosis transmembrane conductance regulator activation plays a central role. We have identified a novel interaction between T1DM and ductal cells. Presumably, the increased ductal secretion represents a defence mechanism in the prevention of diabetes, but further studies are needed to clarify this issue.
Article
Impeded autophagy can impair pancreatic β cell function by causing apoptosis, of which DAP-related apoptosis-inducing kinase-2 (DRAK2) is a critical regulator. Here, we identified a marked up-regulation of DRAK2 in pancreatic tissue across humans, macaques, and mice with type 2 diabetes (T2D). Further studies in mice showed that conditional knockout (cKO) of DRAK2 in pancreatic β cells protected β cell function against high-fat diet feeding along with sustained autophagy and mitochondrial function. Phosphoproteome analysis in isolated mouse primary islets revealed that DRAK2 directly phosphorylated unc-51–like autophagy activating kinase 1 (ULK1) at Ser ⁵⁶ , which was subsequently found to induce ULK1 ubiquitylation and suppress autophagy. ULK1-S56A mutation or pharmacological inhibition of DRAK2 preserved mitochondrial function and insulin secretion against lipotoxicity in mouse primary islets, Min6 cells, or INS-1E cells. In conclusion, these findings together indicate an indispensable role of the DRAK2-ULK1 axis in pancreatic β cells upon metabolic challenge, which offers a potential target to protect β cell function in T2D.
Article
Full-text available
Background Improved blood-glucose control decreases the progression of diabetic microvascular disease, but the effect on macrovascular complications is unknown. There is concern that sulphonylureas may increase cardiovascular mortality in patients with type 2 diabetes and that high insulin concentrations may enhance atheroma formation. We compared the effects of intensive blood-glucose control with either sulphonylurea or insulin and conventional treatment on the risk of microvascular and macrovascular complications in patients with type 2 diabetes in a randomised controlled trial. Methods 3867 newly diagnosed patients with type 2 diabetes, median age 54 years (IQR 48-60 years), who after 3 months' diet treatment had a mean of two fasting plasma glucose (FPG) concentrations of 6.1-15.0 mmol/L were randomly assigned intensive policy with a sulphonylurea (chlorpropamide, glibenclamide, or. glipizide) or with insulin, or conventional policy with diet. The aim in the intensive group was FPG less than 6 mmol/L. in the conventional group, the aim was the best achievable FPG with diet atone; drugs were added only if there were hyperglycaemic symptoms or FPG greater than 15 mmol/L. Three aggregate endpoints were used to assess differences between conventional and intensive treatment: any diabetes-related endpoint (sudden death, death from hyperglycaemia or hypoglycaemia, fatal or non-fatal myocardial infarction, angina, heart failure, stroke, renal failure, amputation [of at least one digit], vitreous haemorrhage, retinopathy requiring photocoagulation, blindness in one eye,or cataract extraction); diabetes-related death (death from myocardial infarction, stroke, peripheral vascular disease, renal disease, hyperglycaemia or hypoglycaemia, and sudden death); all-cause mortality. Single clinical endpoints and surrogate subclinical endpoints were also assessed. All analyses were by intention to treat and frequency of hypoglycaemia was also analysed by actual therapy. Findings Over 10 years, haemoglobin A(1c) (HbA(1c)) was 7.0% (6.2-8.2) in the intensive group compared with 7.9% (6.9-8.8) in the conventional group-an 11% reduction. There was no difference in HbA(1c) among agents in the intensive group. Compared with the conventional group, the risk in the intensive group was 12% lower (95% CI 1-21, p=0.029) for any diabetes-related endpoint; 10% lower (-11 to 27, p=0.34) for any diabetes-related death; and 6% lower (-10 to 20, p=0.44) for all-cause mortality. Most of the risk reduction in the any diabetes-related aggregate endpoint was due to a 25% risk reduction (7-40, p=0.0099) in microvascular endpoints, including the need for retinal photocoagulation. There was no difference for any of the three aggregate endpoints the three intensive agents (chlorpropamide, glibenclamide, or insulin). Patients in the intensive group had more hypoglycaemic episodes than those in the conventional group on both types of analysis (both p<0.0001). The rates of major hypoglycaemic episodes per year were 0.7% with conventional treatment, 1.0% with chlorpropamide, 1.4% with glibenclamide, and 1.8% with insulin. Weight gain was significantly higher in the intensive group (mean 2.9 kg) than in the conventional group (p<0.001), and patients assigned insulin had a greater gain in weight (4.0 kg) than those assigned chlorpropamide (2.6 kg) or glibenclamide (1.7 kg). Interpretation Intensive blood-glucose control by either sulphonylureas or insulin substantially decreases the risk of microvascular complications, but not macrovascular disease, in patients with type 2 diabetes. None of the individual drugs had an adverse effect on cardiovascular outcomes. All intensive treatment increased the risk of hypoglycaemia.
Article
Full-text available
The beta-cell mitochondria are known to generate metabolic coupling factors, or messengers, that mediate plasma membrane depolarization and the increase in cytosolic Ca2+, the triggering event in glucose-stimulated insulin secretion. Accordingly, ATP closes nucleotide-sensitive K+ channels necessary for the opening of voltage-gated Ca2+ channels. ATP also exerts a permissive action on insulin exocytosis. In contrast, GTP directly stimulates the exocytotic process. CAMP is considered to have a dual function: on the one hand, it renders the beta-cell more responsive to glucose; on the other, it mediates the effect of glucagon and other hormones that potentiate insulin secretion. Mitochondrial shuttles contribute to the formation of pyridine nucleotides, which may also participate in insulin exocytosis. Among the metabolic factors generated by glucose, citrate-derived malonyl-CoA has been endorsed, but recent results have questioned its role. We have proposed that glutamate, which is also formed by mitochondrial metabolism, stimulates insulin exocytosis in conditions of permissive, clamped cytosolic Ca2+ concentrations. The evidence for the implication of these and other putative messengers in metabolism-secretion coupling is discussed in this review.
Article
Acute elevation of plasma free fatty acids (FFAs) is necessary for insulin secretion. Sustained elevation, however, leads to apoptosis of pancreatic beta-cells and is a major risk factor for cardiovascular disease and sudden death in patients with insulin resistance or a family history of diabetes mellitus, as well as in individuals with normal glucose tolerance. Data suggest that reduction of FFA plasma levels may reduce the incidence of cardiovascular disease in these at-risk patients. Thiazolidinediones have been shown not only to improve insulin sensitivity but also to reduce FFA plasma levels. Consequently, endothelial function is maintained, vascular smooth muscle cell proliferation and migration are minimized, elevated blood pressure and microalbuminuria are reduced, and high-density lipoprotein and low-density lipoprotein cholesterol particle sizes are improved. (C) 2003 by Excerpta Medica, Inc.
Article
This Statement of Endorsement was retired January 2014
Article
Guidelines for submitting commentsPolicy: Comments that contribute to the discussion of the article will be posted within approximately three business days. We do not accept anonymous comments. Please include your email address; the address will not be displayed in the posted comment. Cell Press Editors will screen the comments to ensure that they are relevant and appropriate but comments will not be edited. The ultimate decision on publication of an online comment is at the Editors' discretion. Formatting: Please include a title for the comment and your affiliation. Note that symbols (e.g. Greek letters) may not transmit properly in this form due to potential software compatibility issues. Please spell out the words in place of the symbols (e.g. replace “α” with “alpha”). Comments should be no more than 8,000 characters (including spaces ) in length. References may be included when necessary but should be kept to a minimum. Be careful if copying and pasting from a Word document. Smart quotes can cause problems in the form. If you experience difficulties, please convert to a plain text file and then copy and paste into the form.