ArticlePDF AvailableLiterature Review

Abstract

Inflammation-induced inhibition of the insulin signalling pathway can lead to insulin resistance and contribute to the development of type 2 diabetes mellitus (T2DM). Obesity and insulin resistance are associated with a chronic but subclinical inflammatory process that impairs insulin action in most tissues and could also hamper pancreatic β-cell function. The involvement of monocytic cells and the profiles of the chemokines and cytokines induced by this inflammation suggest an innate immune response. However, emerging data indicate that elements of the adaptive immune system could also be involved. As activation of an adaptive response requires antigen specificity, some researchers have hypothesized that T2DM evolves from an innate immune response to an autoimmune condition. In this Perspectives article, we present the arguments for and against this hypothesis and discuss which mechanisms could be involved in a putative switch from innate immunity to autoimmunity.
NATURE REVIEWS
|
ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION
|
1
Introduction
Type2 diabetes mellitus (T2DM) is among
the most prevalent diseases in modern soci-
eties, affecting over 340 million people in
the world.
1
Obesity is by far the most impor-
tant predisposing factor for T2DM, and the
rapid increase in the prevalence of obesity
is expected to lead to a similar rise in the
prevalence of T2DM.
The consumption of energy-dense and
fat-rich diets is the main environmen-
tal factor leading to obesity.
2
Clinical and
molecular studies from the past 20years
have provided undisputed evidence for the
role of diet-induced subclinical inflamma-
tion as an important link between obesity
and T2DM.
3
Dietary fats activate signal
transduction through Toll-like receptors
(TLRs) 2 and 4, and induce endoplasmic
reticulum stress (ERS) in adipose tissue,
liver and the hypothalamus.
4–7
TLR and
ERS signalling induce inflammatory activ-
ity, which activates intracellular serine–
threonine kinases that inhibit insulin
signal transduction.
8,9
In addition, TLR and
ERS signalling induce inflammatory gene
transcription, resulting in the production
and secretion of cytokines such as tumour
necrosis factor (TNF) and IL-1β that,
through an extracellular feed-forward loop,
boost intracellular inflammatory signalling
and insulin resistance.
8
Induction of ERS by
saturated fats contributes to activation of
inflammatory mediators
10
and triggers the
dysfunction and death of pancreatic βcells
in patients with T2DM.
9,10
Macrophages and TNF are important
for both induction and perpetuation of the
inflammatory activity linked to obesity,
which led to the concept that insulin resist-
ance results from an anomalous activation
of the innate immune system.
3,8,9,11
However,
other studies suggest that components of the
adaptive immune response also participate
in progression of the inflammatory response
associated with obesity.
12–14
Activation of an
adaptive immune response requires pro-
cessing and presentation of speci fic anti-
gens through interaction between the major
histo compatibility com plex (MHC) and the
T-cell receptor (TCR). No infectious agents
are known to trigger inflammation in either
obesity or T2DM. Thus, endogenous anti-
gens or exogenous molecules (originat ing,
for example, from dietary components)
might be presented, act as antigens or
anoma lously modulate the immune response
and thereby activate subsets of lympho cytes
that target host cells and tissues. If evidence
of such a mechanism could be obtained,
obesity (and perhaps T2DM) might have an
autoimmune component.
Our objective in this Perspectives article
is to discuss the nature of the immune
response in T2DM. We also compare the
inflammatory processes occurring in T2DM
with those of the well-defined autoimmune
disease type1 diabetes mellitus (T1DM).
Autoimmunity in T1DM and T2DM
The autoimmune diseases are a diverse
group of chronic illnesses characterized
by an immune response directed against
specific antigens in the body. Well-known
autoimmune diseases include rheumatoid
arthritis, multiple sclerosis, systemic lupus
erythematosus and T1DM. Our understand-
ing of autoimmunity has evolved consider-
ably since the first attempt was made to
define this term in 1957,
15
and continues
to be debated. In our view, a disease is con-
sidered to have an autoimmune aetiology
if most of the following criteria are met:
pathology is associated with loss of tolerance
to specific antigens; transfer of pathogenic
immune cells and/or anti bodies replicates
the disease in previously unaffected indivi-
duals; immunosuppression or immuno-
modulation modifies the natural history
of the disease; animal models point to an
autoimmune aetiology (including disease
transfer by immune cells); and the disease is
associated with genes that regulate the
immune system, such as HLA.
The evidence suggesting an autoimmune
mechanism of disease in T1DM includes
loss of tolerance to several β-cell antigens,
such as insulin, glutamate decarb oxylase2
(also known as GAD-65), receptor-type
tyrosine-protein phosphatase N2 (also
known as phogrin) and zinc transporter8.
16
More over, animal models of T1DM, such
as nonobese diabetic (NOD) mice and bio-
breeding (BB) rats, have a clear autoimmune
cause of diabetes mellitus,
17
and T1DM can
be ‘transferred’ into immune-deficient
animals by T-cell transfer. Furthermore,
in humans, bone marrow transplantation
between HLA-identical siblings can transfer
OPINION
Type2 diabetes mellitus—an autoimmune
disease?
Lício A.Velloso, Decio L.Eizirik and Miriam Cnop
Abstract | Inflammation-induced inhibition of the insulin signalling pathway can
lead to insulin resistance and contribute to the development of type2 diabetes
mellitus (T2DM). Obesity and insulin resistance are associated with a chronic but
subclinical inflammatory process that impairs insulin action in most tissues and
could also hamper pancreatic β-cell function. The involvement of monocytic cells
and the profiles of the chemokines and cytokines induced by this inflammation
suggest an innate immune response. However, emerging data indicate that
elements of the adaptive immune system could also be involved. As activation of an
adaptive response requires antigen specificity, some researchers have hypothesized
that T2DM evolves from an innate immune response to an autoimmune condition. In
this Perspectives article, we present the arguments for and against this hypothesis
and discuss which mechanisms could be involved in a putative switch from innate
immunity to autoimmunity.
Velloso, L. A. etal. Nat. Rev. Endocrinol. advance online publication 9 July 2013;
doi:10.1038/nrendo.2013.131
Competing interests
The authors declare no competing interests.
PERSPECTIVES
© 2013 Macmillan Publishers Limited. All rights reserved
2
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrendo
T1DM, and specific HLA variants can either
protect against or predispose to T1DM.
18
Finally, therapies that inhibit or modulate
T-lymphocyte activity delay the progres-
sive loss of β-cell mass, although they do
not prevent it.
19
T1DM also has a strong
inflammatory component, and inflamma-
tion could contribute to early stages of the
induction and amplification of the immune
reaction against βcells. Furthermore, in
later stages of this immune response, cross-
talk between invading immune cells and the
target β cells could lead to the progressive
loss of β cells.
20
Whether the pathogenesis of T2DM has
an autoimmune component is less clear.
So far, no potential autoantigenic target
for T lymphocytes has been identified in
this setting, but several reports describe
potential targets for IgG antibodies asso-
ciated with insulin resistance
21
and auto-
antibodies against pancreatic islet antigens
in patients with T2DM.
22
The insulin
resistance phenotype is not reported to be
transferred following bone marrow trans-
plantation, although glucose intolerance
and insulin resistance develop in recipients
of IgG antibody transfers in rodents.
21
In
humans, T-lymphocyte inhibition has no
proven therapeutic or preventive role in
T2DM; however, in mouse models, inhibi-
tion of either B cells
21
or Tlymphocytes
14
can
attenuate the progression of obesity-related
insulin resistance. Although no clear evi-
dence of autoimmunity has been described
in animal models of T2DM, targeting com-
ponents of the adaptive immune system,
such as IFN-γ-expressing type 1 Thelper
cells and B lymphocytes, can improve
insulin resistance.
14,21
Of note, the overlap
between candidate genes for T1DM and
T2DM is very limited; only GLIS3 (which
encodes a zinc finger protein) is associated
with both diseases, among over 50 candidate
genes identified.
23,24
Moreover, none of the
known candidate genes for other inflam-
matory or autoimmune diseases, such as,
IL23R, IL2RA, PTPN2 and a number of HLA
alleles, are associated with T2DM.
25,26
Most
genes linked to obesity are postulated to act
on neuroendocrine circuits that regulate
energy balance and have not been impli-
cated in either innate or adaptive immu-
nity.
27
While these genetic variants explain
only a small fraction of the heritability of
obesity and T2DM, they do not provide evi-
dence for a role of autoimmunity in these
two metabolic conditions.
T1DM (in common with several other
autoimmune conditions) is associated with
an increased risk of other autoimmune dis-
eases, but the same cannot be said of T2DM.
How ever, antibodies against G-protein-
coupled receptors have been detected in
sera from a subgroup of T2DM patients
with an increased risk of hypertension and
cardio vascular complications.
28
In addition,
Rho-kinase-activating auto antibodies are
present in sera from T2DM patients with
maculopathy and macro albuminuria,
29
and autoantibodies against IL-6 have
been detected in sera from 2.5% of Danish
patients with T2DM.
30
Adiposity and inflammation
The stromal vascular fraction of the adipose
tissue is populated by macrophages and
other cells of the immune system, includ-
ing T and B lymphocytes.
31,32
In lean
indivi duals, immune cells residing in the
perivascular space of the adipose tissue
provide a first line of defence against poten-
tial pathogens. However, as adipose tissue
mass increases in individuals with obesity,
these macrophages and adaptive immune
cells become activated even in the absence
of a pathogenic threat.
31,33
Increased adipo-
sity is accompanied by increased numbers
of both CD4
+
and CD8
+
T lymphocytes
in the adipose tissue,
31,33
recruited partly
by C-C motif chemokine 5 (also known
as T-cell-specific protein RANTES).
31
In
obese mice, the increase in numbers of
CD8
+
cells precedes the increase in CD4
+
cells and macrophages.
34
In addition,
depletion of CD8
+
lymphocytes protects
mice from diet-induced insulin resistance,
but not from obesity, whereas adoptive
transfer of CD8
+
lymphocytes can induce
adipose tissue inflammation and insulin
resistance in lean, CD8
+
-depleted mice.
34
Upon stimulation, CD8
+
lymphocytes
from obese mice produce more IFN-γ than
do CD8
+
lymphocytes from lean mice, and
adipocytes from obese mice express more
MHC class I molecules than do those of
lean mice.
33
These data suggest that, during
the early development of obesity, antigens
in the adipose tissue could be presented in
an anomalous way and trigger an adaptive
immune response, preferentially through
CD8
+
lymphocytes.
In line with this apparent key role for
CD8
+
cells in the inflammatory adaptive
immune response in hypertrophic adipose
tissue, one study has shown a reduction in
the FOXP3
+
CD4
+
subpopulation in the vis-
ceral fat depots of obese mice and humans.
13
If a similar subset of cells is experimentally
activated in leptin-deficient Lep
ob/ob
mice,
inflammatory infiltration of the adipose
tissue is reduced and hyper glycaemia is
partially corrected.
35
Regulatory Tcells have
an important role in the control of inflam-
matory responses, and down regulation of
these cells might drive the unrestrained
inflammatory response that is driven by
CD8
+
Tcells in obesity.
13
In lean mice, the
proportion of regulatory T lymphocytes
is high; however, the diversity in their
TCR speci ficity is restricted.
36
During the
develop ment of obesity, the number of regu-
latory Tcells in adipose tissue is reduced,
and their TCR diversity is further restricted,
suggesting the presence of potential adipose
tissue autoantigens.
13,36
Anomalous activation of B lympho-
cytes is also involved in the patho genesis of
obesity-linked insulin resistance.
14
Increased
numbers of B lymphocytes infiltrate the
adipose tissue of obese mice, a change
accompanied by an increased blood concen-
tration of IgG
2c
,
14
and depletion of Blympho-
cytes protects mice from obesity-linked
insulin resistance without affecting body
fat mass. Moreover, IgG anti bodies puri-
fied from obese, insulin-resistant mice can
tr ansfer the glucose-intolerance phenotype.
14
Taken together, these findings indicate
that cells and molecules of the adaptive
immune system have a role in the inflam-
matory activity linking obesity and insulin
resistance. The characterization of this phe-
nomenon has potential clinical relevance,
as disorders related to innate immunity
might respond best to cytokine antago-
nism, whereas disorders related to auto-
immunity might preferentially respond to
therapies that target T and Bcells.
37
Indeed,
treatment with anti-inflammatory drugs
(including TNF blockers, IL-1 receptor
antagonists and salicylate) is beneficial in
animal models of T2DM,
3,38
but resulted in
no or modest reductions in HbA
1c
(≤0.5%)
in patients with T2DM.
39,40
Metabolic inflammation in T2DM
Obesity-associated inflammation is not
restricted to adipose tissue; inflammatory
pro cesses also affect other tissues involved in
whole-body energy homeo stasis, and could
promote the development of glucose intoler-
ance and insulin resistance.
8
For example,
insulin-resistant individuals with obesity
can have inflammatory activity in the liver
41
and skeletal muscle.
42
In the hypothalamus,
signs of inflammation are detected as early
as 1day after introducing a high-fat diet
to rodents.
43
Genetic or pharmaco logical
targeting of this inflammatory process
PERSPECTIVES
© 2013 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS
|
ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION
|
3
ameliorates not only the obese phenotype
(reviewed elsewhere
44
) but also the defec-
tive regulation of hepatic glucose produc-
tion,
45
the impairment of pancreatic β-cell
f unction
46
and hypertension.
47
The presence of low levels of cytokines
and mild tissue infiltration by immune cells
indicates that inflammation is chronic but
low-level, characteristics that are impor-
tant aspects of the inflammatory process in
metabolic tissues. In contrast to the classic
inflammatory process occurring in most
autoimmune conditions, metabolic inflam-
mation is not associated with the typical
signs of inflammatory activity, such as heat,
oedema, redness and pain, in individuals
with obesity and/or T2DM.
48
More over,
instead of increased energy expenditure,
metabolic inflammation results in reduced
whole-body energy expenditure.
48
Despite
these major differences from classic inflam-
matory processes, however, cryptic anti-
gens could still be exposed as a result of
chronic low-level inflammation and lead
to an adaptive autoimmune response, as
proposedelsewhere.
49
β cells, inflammation and T2DM
The inflammatory process in adipose
tissue seems to be similar in all insulin-
resistant individuals with obesity, regard-
less of whether or not they go on to develop
T2DM.
50
Individuals with obesity will only
develop diabetes mellitus when their pan-
creatic β cells fail to compensate for insulin
resistance. Evidence for a role of innate
and adaptive immunity in β-cell failure in
patients who develop T2DM remains con-
troversial. The presence of macrophages
in pancreatic islets (determined by count-
ing the number of CD68
+
cells per islet)
increased from an average of 0.5 cells
in nondiabetic individuals to 1.5 cells in
people with T2DM.
51,52
The proportion
of islets containing at least five immune
cells increased from 0.6% in nondiabetic
control individuals to 5.6% in patients with
T2DM.
52
The presence of these macro-
phages has been linked to viral infection
of islets
52
and expression of human islet
amyloid polypeptide;
53
however, their role
remains unclear. Microarray analysis of
laser-capture microdissected β cells showed
a minor increase (twofold to threefold) in
levels of the chemokines CCL2, CCL11,
CCL13, and CXCL1 and in the cytokines
IL-1β and IL-8 (but not IFN-γ) in cells
from patients with T2DM, compared with
those from nondiabetic controls.
54
These
cytokine and chemokine profiles indicate
mild inflammation, which is consistent
with an innate but not an adaptive immune
response. A similar response was elicited
invitro by exposing human islets to the
saturated fatty acid palmitate or synthetic
ERS inducers, and this process was depend-
ent on IL-1β production.
54
Blocking IL-1β
with an IL-1 receptor antagonist prevented
palmitate-induced inflammation but not
β-cell apoptosis.
54
Consistent with these
findings, the results of microarray analy-
ses of islet cells from patients with T2DM
linked a group of coexpressed genes (among
which many were IL-1-related genes) to
reduced insulin secretion.
55
An autoimmune response to pan creatic
β cells is present in adults with latent
autoimmune diabetes mellitus, a condi-
tion that shares many features with T1DM
and is often misdiagnosed as T2DM.
56
In
some autoantibody-negative patients with
T2DM, responses to islet-reactive Tcells
were observed by measuring mono nuclear
cell responses to human islet proteins
CD1
+
fatty acid
T lymphocytes
Nonautoimmune adaptive response
against fatty acids
Dietary lipids
Lipid raft
TLR4
a
Dietary
lipids
Dead
adipocyte
Cryptic
antigens
Loss of tolerance to self-antigens
and autoimmunity
Regulatory T lymphocytes
Effector T lymphocytes
Loss of tolerance to self-antigens
and autoimmunity
b
d
Changes in
gut microbiota
LPS and fatty acids
Innate response followed by
adaptive response against
fatty acids leads to
no autoimmunity
T
H
17
autoimmunity
c
TCR
Dietary
lipids
Figure 1 | Putative pathways of progression from innate to adaptive immune responses in obesity
and type2 diabetes mellitus. a | Dietary lipids could act as immunomodulators through various
mechanisms: changing the composition of lipid rafts or activating signal transduction through
TLR2 and TLR4. As a result of immunomodulation, inhibition of regulatory Tlymphocytes
and stimulation of effector T lymphocytes could result in loss of tolerance to self-antigens and
autoimmunity. b | Cell death, induced by innate inflammation in response to fatty acids, could
lead to release of cryptic antigens followed by loss of tolerance to self-antigens and
autoimmunity. c | Dietary lipids could be presented to T lymphocytes in the context of CD1–TCR
signal transduction, leading to a nonautoimmune adaptive response. d | Dietary lipids might
induce changes in gut microbiota, resulting in increased blood levels of LPS and fatty acids. This
mechanism could either modulate the immune response through an anomalous T
H
17 response,
contributing in some individuals to autoimmunity, or induce an innate response followed by a
nonautoimmune adaptive response against fatty acids. Abbreviations: TLR, Toll-like receptor;
TCR, Tcell receptor; LPS, lipopolysaccharide; T
H
17, type 17 Thelper cells.
PERSPECTIVES
© 2013 Macmillan Publishers Limited. All rights reserved
4
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrendo
blotted onto nitrocellulose.
57
This method
of evaluating T-cell activation, however, is
under debate, as it might reflect unspeci-
fic inflammation and not autoimmunity,
owing to lack of control tissues or specific
identification of target antigens. The find-
ings suggesting T-cell responses against islet
proteins in patients with T2DM remain to
be confirmed by other research groups
using conventional assays based on speci-
fic β-cell antigens. In contrast to T1DM,
in which nearly all β cells are eventually
destroyed by the auto immune response,
the decrease in β-cell mass in patients with
T2DM is modest (around 30–60%). This
crucial finding suggests that the mecha-
nisms of β-cell loss are different in T1DM
and T2DM.
58
Possible pathways to T2DM
Accumulating evidence suggests that innate
(and, perhaps, adaptive) immunity have a
role in the development of obesity-related
insulin resistance. For example, several
tissues involved in the control of metabolic
functions are affected by complex sub-
clinical inflammatory activity. Moreover,
the insulin-resistance phenotype can be
partly or completely rescued (depending
on the stage of obesity) by targeting either
the innate or the adaptive components of
inflammation.
8,13,14,21,36,44,48,59
In this Perspectives article we raise the
questions of whether autoimmunity has
a pathogenic role in the onset or progres-
sion of the inflammation associated with
obesity and insulin resistance and whether
autoimmunity influences the development
of T2DM. Although current available data
are not sufficient to give definitive answers
to these questions, we propose four possible
pathways that could lead to the onset and/or
progression of the inflammatory response
in both obesity and T2DM (Figure1).
Dietary fatty acids could modulate
immune activity and lead to activation
of an anomalous adaptive immune res-
ponse against self-antigens by, for example,
chang ing the composition of lipid rafts
60
or
by activating signal transduction through
TLR2 and TLR4.
4,6
In this pathway, fatty
acids could negatively modulate regulatory
T lymphocytes and/or positively modulate
effec tor Tlymphocytes in metabolically rele-
vant tissues, resulting in loss of tolerance to
self-antigens, thus leading to au to immunity
(Figure1a).
Alternatively, fatty acids could trigger an
innate immune response and lead to cell
death in relevant tissues, such as adipose
tissue, exposing cryptic antigens and activat-
ing an anomalous adaptive immune response
against self-antigens (Figure1b). Dietary
lipids activate the innate immune response in
most metabolically relevant tissues,
8–10,44
and
chronic inflammation can result in the death
of key cell types, including adipocytes and
neurons, in at least some of these tissues.
8,44
In this pathway, the cryptic antigens released
as a consequence of i nflammation-induced
cell death could activate an adaptive immune
response against self-antigens and thereby
lead to autoimmunity.
Another possibility is that dietary fatty
acids activate an anomalous adaptive
immune response without the development
of autoimmunity (Figure1c). Endogenous
and exogenous lipids can be presented to
T lymphocytes, and the CD1–TCR signal-
ling system would have a central role in this
process.
61,62
In this pathway, dietary fatty
acids could trigger T-lymphocyte activation
through CD1-mediated antigen presen-
tation, and the ensuing adaptive immune
response would target dietary fats without
leading to autoimmune activity.
Finally, changes in gut microbiota
could result in increased blood levels of
lipo polysaccharide and/or modulation
of immune activity, which might lead
to activation of either nonauto immune,
adap tive res ponses against fatty acids or
of auto immune responses against cryptic
antigens (Figure1d). Changes in the gut
microbiota influence both obesity-linked
systemic inflammation and insulin resist-
ance.
63
The inflammation associated with
diet-induced changes in microbiota can
result from increased gut transposition of
lipo polysaccharides as well as changes in
the absorption of nutrients, particularly
lipids.
63–65
More over, changes in micro-More over, changes in micro-
biota can modify IL-17 production in the
gut, which modulates systemic immune
activity.
66
This pathway can have two pos-
sible outcomes: in the first, changes in lipo-
polysaccharide and lipid absorption in the
gut could result in increased innate immune
activity, followed by the activation of an
adaptive immune response against dietary
lipids, with no autoimmune activity; in
the second, changes in immune regulation
caused by upregulation of type17 Thelper
cells (which increases IL-17 production)
favours the development of autoimmunity.
67
Another interesting possibility is that
multiple pathways might be involved: for
example, the direct activation of an innate
immune response by fatty acids (as pro-
posed in Figure1b), could occur in parallel
with lipid presentation through CD1–TCR,
resulting in an innate immune response as
well as nonautoimmune adaptive immune
activity. Other pathways could possibly also
explain the immune response in obesity
and T2DM. The results of well-controlled
experiments in the future will indicate
whether autoimmunity is indeed activated
in the pathogenesis of T2DM.
Conclusions
The presence of autoimmune activity in the
inflammation linked to obesity and insulin
resistance remains elusive. Even if adaptive
immunity is indeed present, it might not
necessarily have a pathogenic role in the
development of T2DM. Failure of the βcell
is central to the pathogenesis of T2DM;
however, although considerable evidence
supports a pathogenic role for islet inflam-
mation and autoimmunity in T1DM, no
conclusive data suggest that auto immunity
has any role in β-cell death in T2DM. The
autoimmune response in T1DM targets
mainly the pancreatic β cells. By contrast,
the chronic, systemic low-grade inflam-
mation in T2DM affects a variety of tissues,
including adipose tissue, liver, the hypo-
thalamus, β cells and the cardio vascular
system. This broad spectrum of activity
suggests the presence of a metabolically
driven innate immune response rather than
an auto immune disease. Although an innate
immune response evolve into an adap-
tive immune response in adipose tissue, it
has not been shown to do so in β cells in
models of obesity and T2DM.
54,58
Thus, cur-
rently, T2DM cannot be characterized as
an autoimmune condition. Further studies
are required to clarify this important and
therapeutical ly relevant question.
Laboratory of Cell Signalling, Obesity and
Comorbidities Research Centre, University of
Campinas, DCM-FCM UNICAMP, 13,084-970
Campinas, São Paulo, Brazil (L. A. Velloso).
Laboratory of Experimental Medicine, Medical
Faculty, Campus Erasme, 808 Route de Lennik
(D. L. Eizirik), Division of Endocrinology, Erasmus
Hospital, Campus Erasme, 806 Route de Lennik
(M.Cnop), Université Libre de Bruxelles, B-1070
Brussels, Belgium.
Correspondence to: L. A. Velloso
lavelloso.unicamp@gmail.com
1. Danaei, G. etal. National, regional, and global
trends in fasting plasma glucose and diabetes
prevalence since 1980: systematic analysis of
health examination surveys and
epidemiological studies with 370 country-years
and 2.7 million participants. Lancet 378,
31–40 (2011).
2. Schulz, M. etal. Food groups as predictors for
short-term weight changes in men and women
PERSPECTIVES
© 2013 Macmillan Publishers Limited. All rights reserved
NATURE REVIEWS
|
ENDOCRINOLOGY ADVANCE ONLINE PUBLICATION
|
5
of the EPIC-Potsdam cohort. J. Nutr. 132,
1335–1340 (2002).
3. Shoelson, S.E., Lee, J. & Goldfine, A.B.
Inflammation and insulin resistance. J. Clin.
Invest. 116, 1793–1801 (2006).
4. Tsukumo, D.M. etal. Loss-of-function mutation
in Toll-like receptor 4 prevents diet-induced
obesity and insulin resistance. Diabetes 56,
1986–1998 (2007).
5. Ozcan, U. etal. Endoplasmic reticulum stress
links obesity, insulin action, and type2
diabetes. Science 306, 457–461 (2004).
6. Milanski, M. etal. Saturated fatty acids
produce an inflammatory response
predominantly through the activation of TLR4
signaling in hypothalamus: implications for the
pathogenesis of obesity. J. Neurosci. 29,
359–370 (2009).
7. Pal, D. etal. Fetuin-A acts as an endogenous
ligand of TLR4 to promote lipid-induced insulin
resistance. Nat. Med. 18, 1279–1285 (2012).
8. Gregor, M.F. & Hotamisligil, G.S. Inflammatory
mechanisms in obesity. Annu. Rev. Immunol.
29, 415–445 (2011).
9. Cnop, M., Foufelle, F. & Velloso, L.A.
Endoplasmic reticulum stress, obesity and
diabetes. Trends Mol. Med. 18, 59–68 (2012).
10. Eizirik, D.L., Miani, M. & Cardozo, A.K.
Signalling danger: endoplasmic reticulum
stress and the unfolded protein response in
pancreatic islet inflammation. Diabetologia 56,
234–241 (2013).
11. Osborn, O. & Olefsky, J.M. The cellular and
signaling networks linking the immune system
and metabolism in disease. Nat. Med. 18,
363–374 (2012).
12. Guarda, G. etal. Tcells dampen innate immune
responses through inhibition of NLRP1 and
NLRP3 inflammasomes. Nature 460, 269–273
(2009).
13. Feuerer, M. etal. Lean, but not obese, fat is
enriched for a unique population of regulatory
Tcells that affect metabolic parameters. Nat.
Med. 15, 930–939 (2009).
14. Winer, S. etal. Normalization of obesity-
associated insulin resistance through
immunotherapy. Nat. Med. 15, 921–929
(2009).
15. Witebsky, E., Rose, N.R., Terplan, K.,
Paine,J.R. & Egan, R.W. Chronic thyroiditis
and autoimmunization. J. Am. Med. Assoc. 164,
1439–1447 (1957).
16. Ziegler, A.G. & Nepom, G.T. Prediction and
pathogenesis in type1 diabetes. Immunity 32,
468–478 (2010).
17. Yang, Y. & Santamaria, P. Lessons on
autoimmune diabetes from animal models.
Clin. Sci. (Lond.) 110, 627–639 (2006).
18. Roep, B.O. & Peakman, M. Diabetogenic
Tlymphocytes in human type1 diabetes. Curr.
Opin. Immunol. 23, 746–753 (2011).
19. von Herrath, M., Peakman, M. & Roep, B.
Progress in immune-based therapies for type1
diabetes. Clin. Exp. Immunol. 172, 186–202
(2013).
20. Eizirik, D.L., Colli, M.L. & Ortis, F. The role of
inflammation in insulitis and β-cell loss in
type1 diabetes. Nat. Rev. Endocrinol. 5,
219–226 (2009).
21. Winer, D.A. etal. Bcells promote insulin
resistance through modulation of Tcells and
production of pathogenic IgG antibodies. Nat.
Med. 17, 610–617 (2011).
22. Brooks-Worrell, B.M., Juneja, R.,
Minokadeh,A., Greenbaum, C.J. & Palmer, J.P.
Cellular immune responses to human islet
proteins in antibody-positive type2 diabetic
patients. Diabetes 48, 983–988 (1999).
23. Barrett, J.C. etal. Genome-wide association
study and meta-analysis find that over 40 loci
affect risk of type1 diabetes. Nat. Genet. 41,
703–707 (2009).
24. Dupuis, J. etal. New genetic loci implicated in
fasting glucose homeostasis and their impact
on type2 diabetes risk. Nat. Genet. 42,
105–116 (2010).
25. Rafiq, S. etal. Gene variants influencing
measures of inflammation or predisposing to
autoimmune and inflammatory diseases are
not associated with the risk of type2 diabetes.
Diabetologia 51, 2205–2213 (2008).
26. Winkler, C., Raab, J., Grallert, H. & Ziegler, A.G.
Lack of association of type2 diabetes
susceptibility genotypes and body weight on
the development of islet autoimmunity and
type1 diabetes. PLoS ONE 7, e35410 (2012).
27. Speliotes, E.K. etal. Association analyses of
249,796 individuals reveal 18 new loci
associated with body mass index. Nat. Genet.
42, 937–948 (2010).
28. Hempel, P. etal. Sera from patients with type2
diabetes contain agonistic autoantibodies
against G protein-coupled receptors. Scand. J.
Immunol. 70, 159–160 (2009).
29. Zimering, M.B. & Pan, Z. Autoantibodies
in type2 diabetes induce stress fiber
formation and apoptosis in endothelial cells.
J.Clin. Endocrinol. Metab. 94, 2171–2177
(2009).
30. Fosgerau, K. etal. Interleukin-6 autoantibodies
are involved in the pathogenesis of a subset of
type2 diabetes. J. Endocrinol. 204, 265–273
(2010).
31. Wu, H. etal. T-cell accumulation and regulated
on activation, normal Tcell expressed and
secreted upregulation in adipose tissue in
obesity. Circulation 115, 1029–1038 (2007).
32. Corre, J. etal. Human subcutaneous adipose
cells support complete differentiation but not
self-renewal of hematopoietic progenitors.
J.Cell Physiol. 208, 282–288 (2006).
33. Rocha, V.Z. etal. Interferon-gamma, a Th1
cytokine, regulates fat inflammation: a role for
adaptive immunity in obesity. Circ. Res. 103,
467–476 (2008).
34. Nishimura, S. etal. CD8+ effector Tcells
contribute to macrophage recruitment and
adipose tissue inflammation in obesity. Nat.
Med. 15, 914–920 (2009).
35. Ilan, Y. etal. Induction of regulatory Tcells
decreases adipose inflammation and alleviates
insulin resistance in ob/ob mice. Proc. Natl
Acad. Sci. USA 107, 9765–9770 (2010).
36. Yang, H. etal. Obesity increases the production
of proinflammatory mediators from adipose
tissue Tcells and compromises TCR repertoire
diversity: implications for systemic
inflammation and insulin resistance.
J.Immunol. 185, 1836–1845 (2010).
37. Savic, S., Dickie, L.J., Wittmann, M.
& McDermott, M.F. Autoinflammatory
syndromes and cellular responses to stress:
pathophysiology, diagnosis and new treatment
perspectives. Best Pract. Res. Clin. Rheumatol.
26, 505–533 (2012).
38. Araujo, E.P. etal. Infliximab restores glucose
homeostasis in an animal model of diet-
induced obesity and diabetes. Endocrinology
148, 5991–5997 (2007).
39. Larsen, C.M. etal. Interleukin-1-receptor
antagonist in type2 diabetes mellitus. N. Engl.
J. Med. 356, 1517–1526 (2007).
40. Goldfine, A.B. etal. The effects of salsalate on
glycemic control in patients with type2
diabetes: a randomized trial. Ann. Intern. Med.
152, 346–357 (2010).
41. Tuncman, G. etal. Functional invivo
interactions between JNK1 and JNK2
isoforms in obesity and insulin resistance.
Proc. Natl Acad. Sci. USA 103, 10741–10746
(2006).
42. Perreault, M. & Marette, A. Targeted
disruption of inducible nitric oxide synthase
protects against obesity-linked insulin
resistance in muscle. Nat. Med. 7,
1138–1143 (2001).
43. Thaler, J.P. etal. Obesity is associated with
hypothalamic injury in rodents and humans.
J.Clin. Invest. 122, 153–162 (2012).
44. Velloso, L.A. & Schwartz, M.W. Altered
hypothalamic function in diet-induced obesity.
Int. J. Obes (Lond.) 35, 1455–1465 (2011).
45. Milanski, M. etal. Inhibition of hypothalamic
inflammation reverses diet-induced insulin
resistance in the liver. Diabetes 61,
1455–1462 (2012).
46. Calegari, V.C. etal. Inflammation of the
hypothalamus leads to defective pancreatic
islet function. J. Biol. Chem. 286,
12870–12880 (2011).
47. Purkayastha, S., Zhang, G. & Cai, D.
Uncoupling the mechanisms of obesity
and hypertension by targeting hypothalamic
IKK-β and NF-κB. Nat. Med. 17, 883–887
(2011).
48. Calay, E.S. & Hotamisligil, G.S. Turning off the
inflammatory, but not the metabolic, flames.
Nat. Med. 19, 265–267 (2013).
49. Brooks-Worrell, B., Narla, R. & Palmer, J.P.
Biomarkers and immune-modulating therapies
for type2 diabetes. Trends Immunol. 33,
546–553 (2012).
50. Barbarroja, N. etal. Progression from high
insulin resistance to type2 diabetes does not
entail additional visceral adipose tissue
inflammation. PLoS ONE 7, e48155 (2012).
51. Ehses, J.A. etal. Increased number of islet-
associated macrophages in type2 diabetes.
Diabetes 56, 2356–2370 (2007).
52. Richardson, S.J., Willcox, A., Bone, A.J.,
Foulis,A.K. & Morgan, N.G. Islet-associated
macrophages in type2 diabetes. Diabetologia
52, 1686–1688 (2009).
53. Masters, S.L. etal. Activation of the NLRP3
inflammasome by islet amyloid polypeptide
provides a mechanism for enhanced IL-1β in
type2 diabetes. Nat. Immunol. 11, 897–904
(2010).
54. Igoillo-Esteve, M. etal. Palmitate induces a pro-
inflammatory response in human pancreatic
islets that mimics CCL2 expression by β cells
in type2 diabetes. Diabetologia 53,
1395–1405 (2010).
55. Mahdi, T. etal. Secreted frizzled-related
protein4 reduces insulin secretion and is
overexpressed in type2 diabetes. Cell Metab.
16, 625–633 (2012).
56. Tuomi, T. etal. Antibodies to glutamic acid
decarboxylase reveal latent autoimmune
diabetes mellitus in adults with a
non-insulin-dependent onset of disease.
Diabetes 42, 359–362 (1993).
57. Brooks-Worrell, B. & Palmer, J.P. Immunology
in the Clinic Review Series; focus on metabolic
diseases: development of islet autoimmune
disease in type2 diabetes patients: potential
sequelae of chronic inflammation. Clin. Exp.
Immunol. 167, 40–46 (2012).
58. Cnop, M. etal. Mechanisms of pancreatic β-cell
death in type1 and type2 diabetes:
manydifferences, few similarities. Diabetes 54
(Suppl. 2), S97–S107 (2005).
59. Reilly, S.M. etal. An inhibitor of the protein
kinases TBK1 and IKK-ε improves obesity-related
PERSPECTIVES
© 2013 Macmillan Publishers Limited. All rights reserved
6
|
ADVANCE ONLINE PUBLICATION www.nature.com/nrendo
metabolic dysfunctions in mice. Nat. Med. 19,
313–321 (2013).
60. Brassard, P. etal. Modulation of T-cell signalling
by non-esterified fatty acids. Prostaglandins
Leukot. Essent. Fatty Acids 77, 337–343 (2007).
61. Beckman, E.M. etal. Recognition of a lipid
antigen by CD1-restricted αβ
+
Tcells. Nature
372, 691–694 (1994).
62. Cohen, N.R., Garg, S. & Brenner, M.B. Antigen
presentation by CD1 lipids, T cells, and
NKTcells in microbial immunity. Adv. Immunol.
102, 1–94 (2009).
63. Cani, P.D. etal. Metabolic endotoxemia
initiates obesity and insulin resistance.
Diabetes 56, 1761–1772 (2007).
64. Turnbaugh, P.J. etal. An obesity-associated
gut microbiome with increased capacity for
energy harvest. Nature 444, 1027–1031
(2006).
65. Caricilli, A.M. etal. Gut microbiota is a key
modulator of insulin resistance in TLR 2
knockout mice. PLoS Biol. 9, e1001212
(2011).
66. Geddes, K. etal. Identification of an innate
Thelper type17 response to intestinal
bacterial pathogens. Nat. Med. 17, 837–844
(2011).
67. Atarashi, K. & Honda, K. Microbiota in
autoimmunity and tolerance. Curr. Opin.
Immunol. 23, 761–768 (2011).
Acknowledgements
The authors’ research is supported by Fundação de
Amparo à Pesquisa do Estado de São Paulo
(Brazil), the Communauté Française de Belgique
—Actions de Recherche Concertées, and the
European Union projects Naimit and BetaBat,
in the Framework Programme 7 of the European
Community. The authors are also grateful to
MarkPeakman of King’s College London, UK, and
Bart Roep of Leiden University Medical Centre,
TheNetherlands, for helpful discussions.
Author contributions
The authors contributed equally to all aspects of
thisarticle.
PERSPECTIVES
© 2013 Macmillan Publishers Limited. All rights reserved
... Pancreatic β cell failure is the cornerstone of diabetes development, and β cells may not be innocent bystanders in this process (1). Rather, β cells may exhibit maladaptive responses to inflammatory stress in different forms of diabetes, such as prolonged exposure to systemic elevations in proinflammatory cytokines and saturated free fatty acids in type 2 diabetes (T2D) or autoimmune attack of β cells and cytokine exposure in type 1 diabetes (T1D) (2,3). ...
Preprint
β cell extracellular vesicles (EVs) play a role as paracrine effectors in islet health, yet mechanisms connecting β cell stress to changes in EV cargo and potential impacts on diabetes remain poorly defined. We hypothesized that β cell inflammatory stress engages neutral sphingomyelinase 2 (nSMase2)-dependent EV formation pathways, generating ceramide-enriched EVs that could impact surrounding β cells. Consistent with this, proinflammatory cytokine treatment of INS-1 β cells and human islets concurrently increased β cell nSMase2 and ceramide expression, as well as EV ceramide staining. Direct chemical activation or genetic knockdown of nSMase2, or treatment with a GLP-1 receptor agonist also modulated cellular and EV ceramide. Small RNA sequencing of ceramide-enriched EVs identified a distinct set of miRNAs linked to β cell function and identity. Coculture experiments using CD9-GFP tagged INS-1 cell EVs demonstrated that either cytokine treatment or chemical nSMase2 activation increased EV transfer to recipient cells. Children with recent-onset T1D showed no abnormalities in circulating ceramide-enriched EVs, suggesting a localized, rather than systemic phenomenon. These findings highlight nSMase2 as a regulator of β cell EV cargo and identify ceramide-enriched EV populations as a contributor to EV-related paracrine signaling under conditions of β cell inflammatory stress.
... Низька ефективність ендоскопічної ендоназальної дакріоцисториностомії у хворих на цукровий діабет 2 типу може бути пов'язана зі фіброзуванням отвору риностоми рубцевою чи грануляційною тканиною [1,2,3,4,5,6]. Це може відбуватися за рахунок діабетичних змін мікросудинного русла слизової оболонки носа [4], зниження імунної відповіді, подовження терміну загоєння ран та патології запального процесу, характерних для цукрового діабету [11,12,13]. ...
Article
Full-text available
Pathological changes in the lacrimal system such as dry eye syndrome in patients with diabetes are well described in modern literature, but no sufficient attention is paid to the problem of the lacrimal system in this group of patients. Currently, the established protocol for treating chronic dacryocystitis involves collaborative endoscopic endonasal dacryocystorhinostomy conducted by ophthalmic surgeons and ENT specialists. However, in patients with type 2 diabetes, there is a need to investigate the clinical effectiveness of this surgical approach. Purpose of this study is to evaluate the effectiveness of conservative and surgical treatment in patients with chronic dacryocystitis depending on the presence of type 2 diabetes and to determine the features of the postoperative period. Materials and methods. The study included 30 patients with chronic dacryocystitis and type 2 diabetes (main group) and 30 patients with chronic dacryocystitis without diabetes (control group), who underwent endoscopic endonasal dacryocystorhinostomy according to the standard technique. The results of the operation were evaluated in 2 months, 6 months and 2 years following the surgical interventions. Endoscopic endonasal dacryocystorhinostomy was considered as an effective technique for restoring unobstructed lacrimal drainage when it lead to the alleviation of tearing, and preventing re-infection of the lacrimal sac. Alongside conventional ophthalmological examinations, the assessment involved nasopharyngeal and lacrimal suction tests, reflux tests, lacrimal duct lavage, lacrimal canal probing, computer tomography, and endoscopic examination of the nasal cavity. Results and discussion. The effectiveness of surgical treatment in 2 months after the operation in patients of the main group (70% of patients) was 1.3 times, after 6 months (63.3% of patients) was 1.4 times, and in the remote period in 2 years (50% of patients) was 1.6 times lower than in patients of the control group (р<0.05). Analyzing the presence of the main symptoms of chronic dacryocystitis in the dynamics of observation, it was noted that after endoscopic endonasal dacryocystorhinostomy the most frequent symptom was lacrimation, but its frequency in patients of the main group after 2 months (30% of patients) was 4.5 times, after 6 months (36.7% of patients) - 2.8 times, after 2 years (50% of patients) - 2.5 times higher than in patients of the control group (р<0.05 ). The efficacy of surgical treatment's in 2 month following the operation in the main group patients (70%) was 1.3 times, after 6 months (63.3%) was 1.4 times, and in the long-term, 2 years post-operation (50%), was 1.6 times lower than in the control group patients (p<0.05). Analyzing the dynamics of chronic dacryocystitis symptoms during observation, it was observed that following endoscopic endonasal dacryocystorhinostomy, lacrimation emerged as the most common symptom. However, its frequency in the main group patients was notably higher after 2 months (30%), being 4.5 times, after 6 months (36.7%) it was 2.8 times higher, and after 2 years (50%) it was 2.5 times higher compared to the control group (p<0.05). Conclusions. In patients with type 2 diabetes, the clinical effectiveness of conservative and surgical treatment for chronic dacryocystitis is lower than in patients without diabetes, and the postoperative period has its own characteristics.
... DM is a leading cause of retinopathy, neuropathy, nephropathy, and macrovascular complications [1,2]. Although T1DM and T2DM have distinct aetiologies [1,3], their main clinical hallmark is hyperglycaemia, i.e., a chronic increase in the blood glucose level [1]. Numerous studies have indicated that hyperglycaemia plays a pivotal role in cell death and diabetic complications. ...
Article
Full-text available
Chronic hyperglycaemia is a devastating factor that causes diabetes-induced damage to the retina and kidney. However, the precise mechanism by which hyperglycaemia drives apoptotic cell death is incompletely known. Herein, we found that FOXD1, a FOX family transcription factor specifically expressed in the retina and kidney, regulated the transcription of BCL-2, a master regulator of cell survival. Intriguingly, the protein level of FOXD1, which responded negatively to hyperglycaemic conditions, was controlled by the TRIM21-mediated K48-linked polyubiquitination and subsequent proteasomal degradation. The TRIM21-FOXD1-BCL-2 signalling axis was notably active during diabetes-induced damage to murine retinal and renal tissues. Furthermore, we found that tartary buckwheat flavonoids effectively reversed the downregulation of FOXD1 protein expression and thus restored BCL-2 expression and facilitated the survival of retinal and renal tissues. In summary, we identified a transcription factor responsible for BCL-2 expression, a signalling axis (TRM21-FOXD1-BCL-2) underlying hyperglycaemia-triggered apoptosis, and a potential treatment for deleterious diabetic complications.
... Based on the timing of disease onset, genetic predisposition and clinical phenotype, diabetes has been traditionally classified into two main groups: type 1 diabetes, featured by an autoimmune-mediated targeting of pancreatic beta cells leading to a deficiency in insulin secretion, and type 2 diabetes, tightly associated with obesity and ageing, featured by insulin resistance and chronic inflammation in insulin-sensitive tissues [2]. However, several phenotypic and mechanistic factors are shared between the two conditions, which has led to the hypothesis that they may in part share underlying mechanisms [3][4][5][6]. ...
Article
Full-text available
We assessed whether HOMA-IR and the Matsuda Index are associated with transitions through stages of type 1 diabetes. Autoantibody (AAb)-positive relatives of individuals with type 1 diabetes (n=6256) from the TrialNet Pathway to Prevention were studied. Associations of indicators of insulin resistance (HOMA-IR) and insulin sensitivity (Matsuda Index) with BMI percentile (BMIp) and age were assessed with adjustments for measures of insulin secretion, Index60 and insulinogenic index (IGI). Cox regression was used to determine if tertiles of HOMA-IR and Matsuda Index predicted transitions from Not Staged (<2 AAbs) to Stage 1 (≥2 AAbs and normoglycaemia), from Stage 1 to Stage 2 (≥2 AAbs with dysglycaemia), and progression to Stage 3 (diabetes as defined by WHO/ADA criteria). There were strong associations of HOMA-IR (positive) and Matsuda Index (inverse) with baseline age and BMIp (p<0.0001). After adjustments for Index60, transitioning from Stage 1 to Stage 2 was associated with higher HOMA-IR and lower Matsuda Index (HOMA-IR: HR=1.71, p<0.0001; Matsuda Index, HR=0.40, p<0.0001), as with progressing from Stages 1 or 2 to Stage 3 (HOMA-IR: HR=1.98, p<0.0001; Matsuda Index: HR=0.46, p<0.0001). Without adjustments, associations of progression to Stage 3 were inverse for HOMA-IR and positive for Matsuda Index, opposite in directionality with adjustments. When IGI was used in place of Index60, the findings were similar. Progression to Stages 2 and 3 of type 1 diabetes increases with HOMA-IR and decreases with the Matsuda Index after adjustments for insulin secretion. Indicators of insulin secretion appear helpful for interpreting associations of progression to type 1 diabetes with HOMA-IR or the Matsuda Index in AAb-positive relatives.
Article
Diabetes is a rising global metabolic disorder and leads to long-term consequences. As a multifactorial disease, the gene-associated mechanisms are important to know. This study applied a bioinformatics approach to explore the molecular underpinning of type 2 diabetes mellitus through differential gene expression analysis. We used microarray datasets GSE16415 and GSE29226 to identify differentially expressed genes between type 2 diabetes and normal samples using R software. Following that, using the STRING database, the protein-protein interaction network was constructed and further analyzed by Cytoscape software. The EnrichR database was used for Gene Ontology and pathway enrichment analysis to explore key pathways and functional annotations of hub genes. We also used miRTarBase and TargetScan databases to predict miRNAs targeting hub genes. We identified 21 hub genes in type 2 diabetes, some showing more significant changes in the PPI network. Our results revealed that GLUL, SLC32A1, PC, MAPK10, MAPT, and POSTN genes are more important in the PPI network and can be experimentally investigated as therapeutic targets. Hsa-miR-492 and hsa-miR-16-5p are suggested for diagnosis and prognosis by targeting GLUL, SLC32A1, PC, MAPK10, and MAPT genes involved in the insulin signaling pathway. Insight: Type 2 diabetes, as a rising global and multifactorial disorder, is important to know the gene-associated mechanisms. In an integrative bioinformatics analysis, we integrated different finding datasets to put together and find valuable diagnostic and prognostic hub genes and miRNAs. In contrast, genes, RNAs, and enzymes interact systematically in pathways. Using multiple databases and software, we identified differential expression between hub genes of diabetes and normal samples. We explored different protein-protein interaction networks, gene ontology, key pathway analysis, and predicted miRNAs that target hub genes. This study reported 21 significant hub genes and some miRNAs in the insulin signaling pathway for innovative and potential diagnostic and therapeutic purposes.
Article
Full-text available
Diabetes mellitus (DM) is a serious chronic metabolic disease that can lead to many serious complications, such as cardiovascular disease, retinopathy, neuropathy, and kidney disease. Once diagnosed with diabetes, patients need to take oral hypoglycemic drugs or use insulin to control blood sugar and slow down the progression of the disease. This has a significant impact on the daily life of patients, requiring constant monitoring of the side effects of medication. It also imposes a heavy financial burden on individuals, their families, and even society as a whole. Adipose-derived stem cells (ADSCs) have recently become an emerging therapeutic modality for DM and its complications. ADSCs can improve insulin sensitivity and enhance insulin secretion through various pathways, thereby alleviating diabetes and its complications. Additionally, ADSCs can promote tissue regeneration, inhibit inflammatory reactions, and reduce tissue damage and cell apoptosis. The potential mechanisms of ADSC therapy for DM and its complications are numerous, and its extensive regenerative and differentiation ability, as well as its role in regulating the immune system and metabolic function, make it a powerful tool in the treatment of DM. Although this technology is still in the early stages, many studies have already proven its safety and effectiveness, providing new treatment options for patients with DM or its complications. Although based on current research, ADSCs have achieved some results in animal experiments and clinical trials for the treatment of DM, further clinical trials are still needed before they can be applied in a clinical setting.
Article
Full-text available
Activation of the innate immune system in obesity is a risk factor for the development of type 2 diabetes. The aim of the current study was to investigate the notion that increased numbers of macrophages exist in the islets of type 2 diabetes patients and that this may be explained by a dysregulation of islet-derived inflammatory factors. Increased islet-associated immune cells were observed in human type 2 diabetic patients, high-fat-fed C57BL/6J mice, the GK rat, and the db/db mouse. When cultured islets were exposed to a type 2 diabetic milieu or when islets were isolated from high-fat-fed mice, increased islet-derived inflammatory factors were produced and released, including interleukin (IL)-6, IL-8, chemokine KC, granulocyte colony-stimulating factor, and macrophage inflammatory protein 1alpha. The specificity of this response was investigated by direct comparison to nonislet pancreatic tissue and beta-cell lines and was not mimicked by the induction of islet cell death. Further, this inflammatory response was found to be biologically functional, as conditioned medium from human islets exposed to a type 2 diabetic milieu could induce increased migration of monocytes and neutrophils. This migration was blocked by IL-8 neutralization, and IL-8 was localized to the human pancreatic alpha-cell. Therefore, islet-derived inflammatory factors are regulated by a type 2 diabetic milieu and may contribute to the macrophage infiltration of pancreatic islets that we observe in type 2 diabetes.
Article
Full-text available
Interleukin-6 (IL6) is critically involved in inflammation and metabolism. About 1% of people produce IL6 autoantibodies (aAb-IL6) that impair IL6 signaling in vivo. We tested the hypothesis that the prevalence of such aAb-IL6 is increased in type 2 diabetic patients and that aAb-IL6 plays a direct role in causing hyperglycemia. In humans, the prevalence of circulating high-affinity neutralizing aAb-IL6 was 2.5% in the type 2 diabetic patients and 1% in the controls (odds ratio 2.5, 95% confidence interval 1.2-4.9, P=0.01). To test for the role of aAb-IL6 in causing hyperglycemia, such aAb-IL6 were induced in mice by a validated vaccination procedure. Mice with plasma levels of aAb-IL6 similar to the 2.5% type 2 diabetic patients developed obesity and impaired glucose tolerance (area under the curve (AUC) glucose, 2056+/-62 vs 1793+/-62, P=0.05) as compared with sham-vaccinated mice, when challenged with a high-fat diet. Mice with very high plasma levels of aAb-IL6 developed elevated fasting plasma glucose (mM, 4.8+/-0.4 vs 3.3+/-0.1, P<0.001) and impaired glucose tolerance (AUC glucose, 1340+/-38 vs 916+/-25, P<0.001) as compared with sham-control mice on normal chow. In conclusion, the prevalence of plasma aAb-IL6 at levels known to impair IL6 signaling in vivo is increased 2.5-fold in people with type 2 diabetes. In mice, matching levels of aAb-IL6 cause obesity and hyperglycemia. These data suggest that a small subset of type 2 diabetes may in part evolve from an autoimmune attack against IL6.
Article
Full-text available
Emerging evidence suggests that inflammation provides a link between obesity and insulin resistance. The noncanonical IκB kinases IKK-ɛ and TANK-binding kinase 1 (TBK1) are induced in liver and fat by NF-κB activation upon high-fat diet feeding and in turn initiate a program of counterinflammation that preserves energy storage. Here we report that amlexanox, an approved small-molecule therapeutic presently used in the clinic to treat aphthous ulcers and asthma, is an inhibitor of these kinases. Treatment of obese mice with amlexanox elevates energy expenditure through increased thermogenesis, producing weight loss, improved insulin sensitivity and decreased steatosis. Because of its record of safety in patients, amlexanox may be an interesting candidate for clinical evaluation in the treatment of obesity and related disorders.
Article
Diabetes and obesity are two metabolic diseases characterized by insulin resistance and a low-grade inflammation. Seeking an inflammatory factor causative of the onset of insulin resistance, obesity, and diabetes, we have identified bacterial lipopolysaccharide (LPS) as a triggering factor. We found that normal endotoxemia increased or decreased during the fed or fasted state, respectively, on a nutritional basis and that a 4-week high-fat diet chronically increased plasma LPS concentration two to three times, a threshold that we have defined as metabolic endotoxemia. Importantly , a high-fat diet increased the proportion of an LPS-containing microbiota in the gut. When metabolic endotoxemia was induced for 4 weeks in mice through continuous subcutaneous infusion of LPS, fasted glycemia and insulinemia and whole-body, liver, and adipose tissue weight gain were increased to a similar extent as in high-fat–fed mice. In addition, adipose tissue F4/80-positive cells and markers of inflammation, and liver triglyceride content, were increased. Furthermore, liver, but not whole-body, insulin resistance was detected in LPS-infused mice. CD14 mutant mice resisted most of the LPS and high-fat diet–induced features of metabolic diseases. This new finding demonstrates that metabolic endotoxemia dysregulates the inflammatory tone and triggers body weight gain and diabetes. We conclude that the LPS/CD14 system sets the tone of insulin sensitivity and the onset of diabetes and obesity. Lowering plasma LPS concentration could be a potent strategy for the control of metabolic diseases.
Article
Case reports published more than a century ago (1, 2) suggested that high-dose sodium salicylate could diminish glycosuria in older diabetic patients. More recently, inflammation and innate immunity have been implicated in the pathogenesis of insulin resistance and type 2 diabetes (3, 4). Obesity activates the transcription factor nuclear factor–κB (NF-κB), which promotes insulin resistance and risk for both type 2 diabetes and cardiovascular disease (5–7). High-dose sodium salicylate inhibits NF-κB (8–10). These findings may explain the original observations and provide potential new avenues for intervention in type 2 diabetes (5). A pilot trial that used aspirin, approximately 7 g/d (11), also demonstrated decreases in glucose concentrations (12, 13). However, aspirin at high doses is associated with risk for bleeding, which limits clinical utility. Sodium salicylate does not irreversibly inhibit cyclooxygenase-1 and -2 (COX-1 and COX-2) (14, 15) and is thus not antithrombotic, but it also irritates the gastrointestinal tract. We therefore initiated pilot studies of salsalate, a prodrug of salicylate that is well tolerated and considered safe after years of use for arthritis. Salsalate reduced blood glucose, triglyceride, free fatty acid and C-reactive protein concentrations; improved glucose utilization; and increased circulating insulin and adiponectin concentrations in small proof-of-concept studies (16, 17). The TINSAL-T2D (Targeting Inflammation Using Salsalate in Type 2 Diabetes) trial evaluates whether this generic and inexpensive drug is safe, tolerated, and efficacious in patients with type 2 diabetes.
Article
Results At 13 weeks, in the anakinra group, the glycated hemoglobin level was 0.46 per- centage point lower than in the placebo group (P=0.03); C-peptide secretion was enhanced (P=0.05), and there were reductions in the ratio of proinsulin to insulin (P = 0.005) and in levels of interleukin-6 (P<0.001) and C-reactive protein (P = 0.002). Insulin resistance, insulin-regulated gene expression in skeletal muscle, serum adipokine levels, and the body-mass index were similar in the two study groups. Symptomatic hypoglycemia was not observed, and there were no apparent drug- related serious adverse events.
Article
MAJOR histocompatibility complex (MHC) class I and class II molecules bind immunogenic peptides and present them to lymphocytes bearing the αβ T-cell antigen receptor (TCR)1–4. An analogous antigen-presenting function also has been proposed for the non-MHC-encoded GDI molecules5, a family of non-polymorphic, β2-microglobulin-associated glycoproteins5–8 expressed on most professional antigen-presenting cells9–11. In support of this hypothesis, CD1 molecules are recognized by selected CD4−CD8− αβ or γδ8TCR+ T-cell clones12–14, and we have recently shown that GDI molecules restrict the recognition of foreign microbial antigens by αβTGR+ T cells10. But the substantial structural divergence of GDI from MHC class I and class II molecules7, raises the possibility that the antigens presented by the GDI system may differ fundamentally from those presented by MHC-encoded molecules. Here we report that a purified CDlb-restricted antigen of Mycobacterium tuberculosis presented to αβTCR+ T cells is mycolic acid, a family of α-branched, β-hydroxy, long-chain fatty acids found in mycobacteria15,16. This example of non-protein microbial antigen recognition suggests that αβTCR+ T cells recognize a broader range of antigens than previously appreciated and that at least one member of the GDI family has evolved the ability to present lipid antigens.
Article
• An explanation of chronic thyroiditis in man was sought in observations made on rabbits, dogs, guinea pigs, and human subjects. The rabbits received injections of saline extracts of rabbit thyroid glands. The tests for circulating autoantibodies utilized the phenomena of precipitation, complement fixation, and tanned-cell hemagglutination. The third of these, based upon the principle of altering the surface of the red blood cells by dilute tannic acid so that they absorb proteins, was particularly sensitive. The three tests were applied to serums from 35 rabbits injected with rabbit thyroid extract, and thyroid autoantibodies were found in 32. Structural damage was found in the thyroid roughly in proportion to the autoantibody titer in the serum. Similar studies were carried out in dogs and guinea pigs with canine and guinea pig thyroid extracts respectively. The three tests were then applied to serums from patients with chronic thyroiditis. Twelve such patients were found whose serums contained circulating antibodies specifically directed against extracts of human thyroid glands. Three typical case histories are given, with histological findings on thyroid tissue removed during surgery. Six other patients with chronic thyroiditis, proved histologically, were not found to have autoantibodies in their serums at the time of study. These findings lead to the hypotheses that some types of chronic thyroiditis are related to an autoimmunization process within the patient against his own thyroid tissue, and that thyroid autoantibodies are at once indicators and links in the chain of pathological processes.
Article
Immune-based therapies that prevent type 1 diabetes or preserve metabolic function remaining at diagnosis have become a major objective for funding agencies and international trial consortia, and receive backing from notable patient advocate groups. The development of immune-based therapeutic strategies in this arena requires a careful balancing of the risks of the therapy against the potential benefits, because many individuals are diagnosed or identified as being at increased risk of disease in early childhood, a period when manipulation of the developing immune system should be undertaken with caution. In addition, a therapy exists (daily insulin injection) that is life-saving in the acute stages of disease and can be used effectively over a lifetime as maintenance. Conversely, the disease is increasing in incidence; is peaking in ever-younger age groups; carries significant risk of increased morbidity and early mortality; and remains difficult to manage effectively in many settings. With these issues in mind, in this article we review progress towards immune-based strategies for this chronic autoimmune disease.
Article
Metabolically driven, chronic, low-grade inflammation has a crucial role in the pathogenesis of obesity and type 2 diabetes, and multiple stress-signaling cascades link insulin resistance and the immune response. An approved immunomodulatory drug is now shown to inhibit the kinases IKK epsilon and TKB1, suppress inflammation and improve metabolic homeostasis in mouse models of obesity (pages 313-321).