Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Curr Diab Rep (2016) 16: 53

DOI 10.1007/s11892-016-0738-2

IMMUNOLOGY AND TRANSPLANTATION (L PIEMONTI AND V SORDI, SECTION EDITORS)

Islet Autoantibodies
Vito Lampasona 1,2 & Daniela Liberati 1,2

Published online: 25 April 2016


# Springer Science+Business Media New York 2016

Abstract Islet autoantibodies are the main markers of Introduction


pancreatic autoimmunity in type 1 diabetes (T1D). Islet
autoantibodies recognize insulin (IAA), glutamic acid Islet autoantibodies have been fundamental in the recognition
decarboxylase (GADA), protein phosphatase-like IA-2 of type 1 diabetes (T1D) as an autoimmune disease. A clear
(IA-2A), and ZnT8 (ZnT8A), all antigens that are found role of the immune system in the pathogenesis of insulin-
on secretory granules within pancreatic beta cells. Islet dependent diabetes was widely recognized after Willy
antibodies, measured by sensitive and specific liquid Gepts’ systematic description of insulitis in pancreatic islets
phase assays, are the key parameters of the autoimmune at onset of diabetes [1]. However, the autoimmune nature of
response monitored for diagnostics or prognostics in this lymphocytic infiltration became fully established only in
patients with T1D or for disease prediction in at-risk 1974, when Franco Bottazzo and Deborah Doniach [2] report-
individuals before T1D onset. Islet autoantibodies have ed the presence of islets cell antibodies (ICA) against pancreatic
been the main tool used to explore the natural history of islets in patients with insulin-dependent diabetes.
T1D; this review summarizes the current knowledge The detection of ICA, in combination with metabolic
about the autoantigens and the phenotype of islets auto- studies of islet function in the pre-clinical phase of diabetes,
antibodies acquired in large prospective studies from was key in laying the foundations of the current conceptual mod-
birth in children at risk of developing T1D. el of progressive disease stages in T1D pathogenesis, that was
elegantly formulated by George Eisenbarth in his 1986 seminal
paper [3].
Keywords Autoantibodies . T1D . GAD65 . Insulin . IA-2 . Today, islet autoantibodies are key markers of T1D associ-
ZnT8 ated autoimmunity and are used in variety of clinical and re-
search settings that include the diagnostics of T1D [4], the
prediction of future T1D development [5], the use as
prognostic markers in the context of pancreas [6] or islet trans-
This article is part of the Topical Collection on Immunology and
Transplantation plantation [7], and as surrogate markers of efficacy in the con-
text of experimental therapies aimed at T1D prevention [8].
* Vito Lampasona
lampasona.vito@hsr.it
Islet Autoantibody Standardization Program
Daniela Liberati
liberati.daniela@hsr.it
After the discovery of ICA, the discrepancies that soon
emerged across results from laboratories using different pro-
1
Division of Genetics and Cell Biology, IRCCS San Raffaele tocols for ICA testing, led to the establishment of the immu-
Scientific Institute, via Olgettina 60, 20132 Milano, Italy nology of diabetes workshops, a grassroots effort by the sci-
2
Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, entific community aimed at the standardization of islet auto-
via Olgettina 60, 20132 Milano, Italy antibody assays and the evaluation of laboratory performance.
53 Page 2 of 10 Curr Diab Rep (2016) 16: 53

The workshops proved remarkably successful for solving sci- in the central nervous system. The GAD65 structure includes a
entific controversy and fostered the achievement of high qual- NH2 terminal domain, a middle domain containing a pyridoxal
itative standards in islet autoantibody measurement [9]. 5′-phosphate (PLP) cofactor binding site, and a COOH terminal
Today, the program continues under the name Islet catalytic domain [25, 26]. The GAD67 isoform is encoded by a
Autoantibody Standardization Program, previously known distinct gene and is overall 76 % homologous to GAD65. Both
as DASP [10], supervised by the Immunology of Diabetes isoforms are obligate functional dimers that can form both
Society, supported by the NIDDK branch of the NIH, and homodimers and heterodimers, and GAD65 is generally found
organized by the TrialNet Islet Cell Autoantibody Core in a multi-protein complex associated with the cytoplasmic side
Laboratory at the Department of Pathology of the University membrane of SLMV, where GABA is stored upon synthesis.
of Florida. GAD65 undergoes several post-translational modifications like
Overall, the major contribution of workshops has been the phosphorylation, palmitoylation, N-terminal cleavage, and re-
clarification that assays in which antibody binding occurs at versible association with PLP [27] which are critical for its
the interface of a solid surface coated with antigen, like direct association with SLMV and the control of enzymatic activity.
ELISA, do not allow for sensitive and specific detection of
islet autoantibodies [11–15]. Consequently, the de facto gold
standard formats for islet autoantibodies measurement GAD65 Tissue Expression
are based on radio binding, bridge-ELISA, and ECL
assays [12, 16, 17], in which the interaction of antibodies with GAD65 expression is not exclusive of pancreatic beta cells but
antigen happens completely or partially in fluid phase. is present at the mRNA and protein levels in several tissues
where GABAergic systems exist [28]. In addition to neurons
in the central and peripheral nervous systems, GABA exert
Islet Autoantibody Antigens signaling functions that only partially understood in several
peripheral tissues and cells of the intestine, stomach, pancreas,
Over the years, numerous protein and non-protein targets in fallopian tube, uterus, ovary, testis, kidney, urinary bladder,
pancreatic islets were reported as antigens of the autoantibody lung, eye lens, and liver. However, while expression of
response associated with diabetes. However, only a handful of glutamic acid decarboxylase(s) is therefore expected to take
these proposed autoantigens survived the test of time and the place in selected cells within those tissues, exhaustive
close scrutiny of standardization workshops. Currently, four studies of GAD65 localization outside the brain are still
proteins are confirmed autoantigens of islet autoantibodies: lacking in humans.
insulin [18], glutamic acid decarboxylase 2 (GAD65) [19], In the adult human pancreas, GAD65 is highly expressed
the protein tyrosine phosphatase like protein IA-2 [20], and exclusively in pancreatic islets, where it is found mostly in β
the zinc transporter 8 (ZnT8) [21]. In addition, autoantibodies cells and in a minority of α cells [29]. Expression of the
can be found in T1D patients that recognize also two closely GAD67 isoform is instead absent at the protein level, suggest-
homologous isoforms GAD67 [22] and IA-2β [23]. ing that the regulation of GABA production in pancreatic
Very recently, Christie et al. [24] reported the identification islets must be at least partially different from the brain [30].
of the tetraspanin protein family member TSPAN7 as Finally, expression of GAD67, but not GAD65, has been
glima38, a pancreatic islet glycosylated membrane- reported in human thymic medullary epithelial cells, that are
associated protein of 38 kDa, previously reported to be the in charge of presenting self-antigen during thymic
target of autoantibodies in >30 % of T1D patients. education of immature lymphocytes [31], suggesting a
A common thread linking antigens recognized by islet auto- potential mechanism of escape from negative selection
antibodies is their involvement in the secretory machinery of of autoreactive lymphocytes recognizing GAD65.
pancreatic islet beta cells. In fact, IA-2, ZnT8, and TSPAN7 are
found on the membrane of secretory granules (SG), in addition
to insulin contained within, while GAD65 is instead found on GADA Epitopes
the cytoplasmic site of synaptic-like micro-vesicles (SLMV).
GAD65 autoantibodies (GADA) have been extensively
characterized in terms of their epitope recognition on the
GAD65 Autoantibodies antigen in T1D and other diseases. Three broad categories of
GADA reactivities against GAD65 can be identified:
The Autoantigen of GADA GADA directed against epitopes in the NH2 terminal
domain; GADA to epitopes contained within the middle
GAD65 is a key enzyme for the biosynthesis of gamma- PLP binding domain [32]; and GADA to epitopes in the
aminobutyric acid (GABA), a major inhibitory neurotransmitter COOH terminal domain [33].
Curr Diab Rep (2016) 16: 53 Page 3 of 10 53

GADA in Other Diseases producing D cells, and selected cells in the basalis of the
gastric glands [40, 45]. Within the pancreas, IA-2 is absent
GADA have been described in several severe autoimmune from the exocrine tissue and exclusively found in β, α, and δ
pathologies of the central nervous system like classical stiff cells of pancreatic islets [40]. Additionally, IA-2 expression
person syndrome [34], cerebellar ataxia, Batten disease, and has been also described, at both the mRNA and protein levels,
others [35, 36]. In addition, GADA are relatively common in in a subset of splenocytes and in the thymus, where it is
patients affected by autoimmune polyendocrinopathy (APS) thought to be implicated in the negative selection of self-
[37] or by the IPEX syndrome [38]. While development of reactive immune cells [46].
T1D is relatively frequent in these patients, GADA are often
present in the absence of diabetes. In particular, in APS,
presence of GADA has been associated more strongly with IA-2A Epitopes
gastrointestinal dysfunction than with diabetes [37]. Finally,
GADA are the hallmark of patients with latent autoimmune IA-2A at onset of disease can be divided in two major
diabetes of the adult (LADA) a slowly progressing form of categories: those directed against multiple short mostly, but
autoimmune diabetes [39]. not exclusively, linear, epitopes within the cytoplasmic JM
domain [47, 48] and those against conformational epitopes
within the PTP-like domains [49, 50].
IA-2 Autoantibodies Recently, a close structural interaction between the two
domains and the modulation of proteolytic processing of both
The Autoantigen of IA-2A IA-2 cytoplasmic domains by autoantibodies bound to JM
epitopes was described [51], suggesting a model mechanism
IA-2 is a receptor type protein tyrosine phosphatase (PTP) like through which epitope spreading might occur.
protein intrinsic to the membrane of SG [40]. IA-2 includes a
large ectodomain, localized in the SG lumen, followed by a IA-2A in Other Diseases
single transmembrane, a short cytoplasmic juxtamembrane
(JM), and PTP-like cytoplasmic domains. IA-2β is an IA-2A have been reported in few patients with APS [37, 52],
isoform, encoded by a distinct gene, that is 88 % homologous Stiff-man syndrome, and other neurological diseases [36, 53].
at the level of the PTP-like domain. The biological role However, unlike for GADA, presence of IA-2A in these
of IA-2 is only partially known: no ligand has been identified patients is in the majority of the cases a marker of actual or
that binds the IA-2 ectodomain, and the IA-2 PTP-like domain future presence of T1D.
does not possess phosphatase activity. In pancreatic beta cells,
in addition to form homodimers and heterodimers with IA-2β,
IA-2 can establish interactions with a range of diverse proteins
[41] indirectly interacting with actin microfilaments. In ZnT8 Autoantibodies
pancreatic islets, upon Ca++ stimulated insulin exocytosis,
the IA-2 cytosolic domain can be cleaved, promoting the mo- The Autoantigen of ZnT8A
bilization of additional SG towards the plasma membrane
[42]. Furthermore, the cleaved cytosolic fragment of IA-2 ZnT8 is a multi-pass protein with six transmembrane domains
can translocate to the nucleus, where it promotes the up- and a histidine-rich loop between transmembrane domains IV
regulation of genes associated with SG function in beta cells, and V, a classical feature of ZnT proteins, a family of trans-
including insulin and IA-2 themselves [43], and cell prolifer- membrane proteins involved in the regulation of cellular zinc
ation [44]. IA-2 can therefore influence insulin secretion, SG content [54]. ZnT8 forms homodimers that are inserted in the
biogenesis and homeostasis, and β-cell expansion. membrane of SG, with NH2 and COOH terminal domains
facing the cytoplasm while the histidine-rich loop is directed
IA-2 Tissue Expression towards the lumen. Other ZnT transporters are expressed in β
cells, but ZnT8 appears to be critical for the accumulation of
IA-2 expression is not exclusive of pancreatic beta cells zinc into SG and the maintenance of stored insulin as a tightly
but is present at the mRNA and protein levels in several packed hexamers [55].
neuroendocrine cells, i.e., enriched for dense core peptide Polymorphic variants of ZnT8 at the level of amino acid
secretory vesicles. In rodents, it has been demonstrated in 325 in the cytoplasmic COOH terminal domain have been
the pituitary gland, amygdala, and hypothalamus in the brain, linked to increased type 2 diabetes risk and impaired glucose
the adrenal medulla, autonomic nerve fibers and ganglia, homeostasis [56, 57] and altered rate of proinsulin conversion
thyroid C cells, lung Kulchitsky cells, stomach somatostatin- to mature hormone [58].
53 Page 4 of 10 Curr Diab Rep (2016) 16: 53

ZnT8 Tissue Expression IAA Epitopes

Expression of ZnT8 in humans is largely restricted to pancreatic IAA epitopes have been only indirectly characterized using
islets at the mRNA level [55]. While few other tissue have been different recombinant insulins that form other species, ana-
reported to contain minimal amounts the ZnT8 gene mRNA, logs, and proinsulin as competitors in binding experiment
like fetal retinal pigment epithelium, adipocytes, and a subset of [69]. IAA are directed against conformational epitopes and
lymphocytes, so far the presence of ZnT8 at the protein level is do not react with denatured or reduced antigen. Key amino
confirmed only in pancreatic islets and possibly in cells of the acid residues for binding of high affinity IAA have been
retina [59, 60]. Within islets, ZnT8 is highly enriched in β cells mapped within residues 8 to 13 of insulin A chain, while most
[59] but apparently present at lower levels in other cell types low affinity IAA are dependent on residues 28–30 of the B
like α cells [61]. chain. However, in some subjects, IAA also exist which are
not influenced by either of these A or B chain residues.
Competition experiments suggest that only IAA depending
ZnT8A Epitopes for binding on A chain residues 8–13, but not B chain residues
28–30, can bind also proinsulin.
ZnT8A epitopes are present in the NH2 or the COOH terminal
cytoplasmic domains [21]. Less than 10 % of ZnT8A-positive IAA in Other Diseases
patients have antibodies to the NH2 terminal domain, while
almost all recognize the COOH terminal domain. IAA have been described in other autoimmune diseases like
Interestingly, ZnT8A have been identified that are specific APS [37, 52] and Stiff-man syndrome [36]. Unlike for GADA,
for either the arginine or tryptophan polymorphic variants at and similar to IA-2A and ZnT8A, IAA in these patients are rare
amino acid residue 325 in the COOH cytoplasmic domain and associated with co-occurrence or future development of
[62]. These ZnT8A appear to be restricted by the presence T1D. High-titer insulin antibodies can develop in patients after
of the corresponding polymorphism in the genome of patients insulin treatment but their binding characteristics are not those
[63]. However, additional reactivities against the COOH of IAA [70]. Insulin autoantibodies with binding characteris-
domain exist that are not affected by the residue encoded at tics different from IAA also develop in patients affected by
amino acid 325 [64]. immune hypoglycemia syndrome (IAS) [71].

ZnT8A in Other Diseases Natural History and Phenotype of Islet Autoantibodies

ZnT8A are rare in the absence of T1D and found only in a Several large prospective studies have focused on the devel-
minor fraction of APS patients, subjects with antibodies opment of autoimmunity in subjects at risk of T1D, either first
to 21-hydroxylase without polyendocrinopathy, and in degree relatives of patients with T1D or subjects with HLA
few relatives of T1D patients with antibodies to tissue haplotypes predisposing to T1D. In all of these studies, islet
transglutaminase [21]. autoantibody appearance and phenotype were the key param-
eter used to track autoimmunity in sequential samples taken
from birth over a time span that, by now, for some individuals
covers more than two decades.
Insulin Autoantibodies Ziegler et al. [72•] recently reported a combined analysis of
islet autoantibodies development in 13,377 at-risk children
Insulin Tissue Expression enrolled in three prospective studies from Germany, Finland,
and the USA. During follow-up, 1059 children (7.9 %) con-
Insulin in the human adult body is expressed almost verted to islet autoantibody positive. Risk of developing T1D
exclusively in pancreatic beta cells, and the claim of local was clearly associated with presence of islet autoantibodies; in
expression of insulin in extra-pancreatic tissues in humans fact, 403 of 428 (94.1 %) children who developed diabetes
has raised controversy in the past [65, 66]. However, in tested positive for islet autoantibodies before disease onset.
humans, extra-pancreatic expression of insulin has been clearly
demonstrated at both the mRNA and protein levels in thymic The Genetics of Islet Autoantibodies
medullary epithelial cells [67, 68]. Potentially relevant for the
negative selection of autoreactive lymphocytes is the observed A clear association between specific HLA haplotypes and
association between specific polymorphic variants the insulin development of islet autoantibodies has been long known. In
gene, known to confer different genetic risk for T1D, and a particular, GADA are associated more strongly with the HLA
diminished amount of insulin mRNA in the thymus. DR3-DQ2 haplotype, while IAA and IA-2A are more
Curr Diab Rep (2016) 16: 53 Page 5 of 10 53

associated with the HLA DR4-DQ8 haplotype [73]. Islet Islet Autoantibody Phenotype and T1D Risk
autoantibody development is further modulated by the
interaction of these HLA haplotypes with other polymor- Several studies analyzed the phenotype of islet autoantibodies
phisms outside the HLA, like those in the PTPN22, ERBB3, at seroconversion measuring characteristics like combinations
SH2B3, and INS genes [74••]. of different autoantibodies, titer, isotype, IgG sub-class,
epitope specificity, and affinity [33, 73, 81–88].
Isolated IAA or GADA are the most frequent single
Age at Seroconversion to Islet Autoantibody Positive reactivities found in the first positive samples of children at
risk but multiple autoantibodies can be present already
The age at which de novo seroconversion to islet autoanti- at seroconversion in 60 % of samples children who will
bodies positive occurs has been carefully analyzed in children remain persistently autoantibody positive during follow-
at risk of T1D, taking into account the transplacental transmis- up [73, 77, 80••]. IAA and GADA are also the key
sion in utero of islet autoantibodies, in children of mothers components of multiple autoantibodies at seroconversion, as
with T1D, and the persistence of maternal autoantibodies after an example in the study of Streck et al. [80••] the autoantibody
birth for several months [75, 76]. combinations found were as follows: IAA-GADA-IA-2A in
Islet autoantibodies develop very rarely before the age of 32 %, IAA-GADA in 17 %, IAA-IA-2A in 7 %, and GADA-
6 months, and the peak of incidence of seroconversion takes IA-2A in 5 % of the cases. However, the contribution to T1D
place between 9 and 24 months of age [73, 77]. In their paper, risk of other islet autoantibodies like IA-2A, IA-2β
Giannopoulu et al. [73] calculated the rate of seroconversion antibodies, and ZnT8A should not be discounted. In
at 9 and 24 months at 18.5 and 21 new cases per 1000 high- fact, development of autoantibodies to these targets, that
risk children per year, respectively. Incidence then dropped to all reside on SG in beta cells, was clearly associated in
9.1, 5.1, and 6.9 seroconverters per 1000 high-risk children- other studies with a faster progression to T1D and a
years at 5, 11, and 14 years of age. In the same report, the risk of developing disease within 5 years from their
incidence of IAA at seroconversion peaked earlier and was appearance greater than 45 % [63, 89, 90].
significantly higher than that of GADA, IA-2A, and ZnT8A. The titer of islet autoantibodies at seroconversion is also
Simultaneous development of multiple islet autoantibodies significantly associated with T1D risk. Although islet autoan-
is also highest at 9 and 24 months of age compared to later tibody titer can fluctuate during follow-up, children with IAA
time points, while seroconversion to single autoantibody re- or IA-2A, but not GADA, in the upper quartile of the range of
activities, other than IAA, is less pronounced at the same time antibody titer, have a two to fourfold increase in risk of devel-
points and less variable over time [78]. In children with a oping T1D within 10 years compared to children with titers of
single autoantibody at seroconversion that during follow-up the same antibodies in the lower quartile [85].
will become multiple autoantibody positive, the development Islet autoantibodies show a predominance of immunoglob-
of additional reactivities occurs within 2 years in around 70 % ulins mostly of the IgG1 class for GADA, IA-2A, and in
of the cases [79]. Multiple islet autoantibodies confer in- addition also of the IgG3 and IgG4 class for IAA, already at
creased risk of developing T1D by age 15 years, with children seroconversion [69, 81]. This is unlike the sequential appear-
having two or more autoantibodies showing a diabetes risk of ance of different immunoglobulin classes from IgM to IgG of
61.6 and 79.1 % [72•], respectively. Seroconversion at a a classical de novo immunization process.
young age is a further risk factor, and children who developed Already at seroconversion, islet autoantibodies can be
multiple autoantibodies between 1 and 2 years of age directed against a variety of epitopes on their target antigens.
progress to diabetes in about 75 % of the cases within However, there is also clear evidence of epitope spreading
6 years [72•, 80••]. Overall, seroconversion before age during follow-up [33, 91]. Within children who are single
of 3 is linked to an increase risk of developing T1D GADA positive, the analysis of epitope reactivity suggests
risk within 10 years (74.9 vs 60.9 %) [72•]. that progression to T1D rarely, if ever, occurs when GADA
Children instead who had antibodies to just one islet are directed exclusively to epitopes within the NH2 terminal
antigen at seroconversion and remain single autoanti- domain of GAD65 [92, 93]. Conversely, epitope spreading
body positive during follow-up have a lower T1D risk that leads to the appearance of autoantibodies cross-reactive
by age 15 of 12.7 % [72•]. These children are slightly between IA-2 and IA-2β or IA-2β specific is associated with a
older at seroconversion compared to multiple autoanti- more aggressive autoimmunity and faster progression to
body positive, but antibodies appear nonetheless in clinical onset of T1D [89, 90].
75 % of the cases before 8 years of age. When occurs, Finally, the affinity of islet autoantibodies, i.e., the strength
progression to T1D can be relatively rapid also in these with which they bind to their antigens, in at-risk children, can
children, in the majority of the cases within 4 years be already high at seroconversion and generally show little
from seroconversion [72•]. changes during follow-up in most cases [92–95]. Again, this
53 Page 6 of 10 Curr Diab Rep (2016) 16: 53

is unlike the progressive increase over time, due to affinity Islet Autoantibodies Upon Intervention Therapies
maturation, of a classical humoral response upon immuniza-
tion. The affinity of IAA and GADA at seroconversion is Allogeneic pancreas or pancreatic islet transplantation are
particularly heterogeneous, with high affinity mostly limited established therapies aimed at the restoration of insulin pro-
to subjects who are multiple autoantibody positive or will duction and secretion in T1D patients. Despite the invariable
develop multiple antibodies during follow-up, while low administration of an immunosuppressive regimen, transplant
affinity is usually associated with persistent positivity for a rejection is frequently preceded by recurrence of autoimmuni-
single antibody [92, 94, 95]. The affinity of IA-2A instead, ty. In fact, in several studies that evaluated islet autoantibodies
while being particularly variable across IA-2A positive before and after pancreas or islet transplantation, the reappear-
children, is almost always confined to a high range [95]. All ance of islet autoantibodies, or an increase in their titers, pre-
these evidences indicate that high affinity is an additional ceded graft failure [104–106]. Usually, GADA are the earliest
feature of islet autoantibodies which is associated with autoantibodies to reappear in these patients but the recurrence,
increased risk of T1D. or the increase in titers, of IA-2A and ZnT8A are the strongest
After the analysis of islet autoantibody phenotypes at predictors of graft function loss [7]. However, in several pa-
seroconversion, some conclusions can be drawn [73]: tients in which graft function was maintained, islet autoanti-
children positive for multiple autoantibodies, or for just bodies were already present at the time of transplant and
IAA with high affinity that later convert to multiple remained stable during follow-up. This observation suggests
antibody positive, are those at the greatest risk of that in transplanted patients with long-standing T1D, similarly
T1D; early development of autoimmunity, around or to new onset patients, islet autoantibodies are not mediators of
within the first year of life, is clearly associated with direct cytopathic islet or graft damage. Furthermore, B cell
an antibody response to insulin; the GADA antibody depletion performed in T1D patients as a treatment for co-
response occurs somewhat later in life, between 2 and occurring diseases does not invariably lead to a relevant
3 years of age, and can be directed against diverse change in islet autoantibody titers, despite a clear positive
epitopes, only some of which are associated with T1D clinical response [107]. Similarly, GADA, IA-2A, and
risk. ZnT8A autoantibody levels were unaffected by an experimen-
Overall, the characteristics of islet autoantibodies found at tal treatment of new onset T1D patients with an anti-CD3
seroconversion appear to be those of a mature humoral monoclonal, known to be associated not only with T cell but
response with a particularly rapid development in at also partial B cell depletion [108]. Intriguingly, in patients that
least some at-risk children. These findings are consistent had high IAA titers and greater beta cell function at T1D
with the emerging picture of the existence of T1D subtypes, in onset, treatment with an anti-CD3 monoclonal resulted in bet-
some of which B cells likely play a major role in disease ter preservation of beta cell function and a reduction of the
pathogenesis [96••, 97, 98]. further rise of insulin antibodies that follows the start of insu-
lin therapy [108].These findings suggest that, both in new
onset and long-standing T1D patients, islet autoantibodies
Islet Autoantibodies at and After T1D Onset might be produced either by long-lived plasmacells, usually
unaffected by common immunosuppressive therapies, or by
Frequency of autoantibody positivity at onset of disease range short-lived plasmablasts, following T cell-dependent B cell
in decreasing order from >80 % for GADA, ≥70 % for IA-2A activation and differentiation, or both.
[99], ≥65 % for ZnT8A, and >50 % for IAA. Confirming the
prevalent concept that GAD65 and IA-2 are the primary
targets of autoimmunity in T1D [100], autoantibodies to the Conclusions
GAD67 and IA-2β isoforms are less frequent and essentially
always coexist with antibodies to GAD65 or IA-2, The study of islet autoantibodies has provided fundament in-
respectively. sights on the natural history of T1D. Islet autoantibodies will
Studies in long-standing patients with T1D from the continue to be the main marker of T1D autoimmunity in the
BGolden years^ and BJoslin medalist study^ cohorts demon- near future both in diagnostics and in clinical research. Further
strate that autoantibodies can persist more than 50 years after technical advancements are emerging aimed at improving islet
onset of disease in up to 50 % of patients [101, 102]. Persisting autoantibody assays and the accuracy of T1D develop-
islet autoantibodies are in most cases GADA and less ment predictions based on autoantibodies. Nevertheless,
frequently ZnT8A and IA-2A [102]. Overall, the number fundamental questions about the basis for development
and titer of islet autoantibodies decline after progression to of islet autoantibodies in T1D and their role in disease
T1D, and the type of persistent antibodies can be influenced pathogenesis remain unanswered and will likely be the
by parameters like age at onset and disease duration [103]. focus of future research efforts.
Curr Diab Rep (2016) 16: 53 Page 7 of 10 53

Compliance with Ethical Standards Decarboxylase Antibody Workshop. Diabetes. 1994;43:1005–9.


doi:10.2337/diabetes.43.8.1005.
Conflict of Interest Vito Lampasona and Daniela Liberati declare that 13. Verge CF, Stenger D, Bonifacio E, Colman PG, Pilcher C, Bingley
they have no conflict of interest. PJ, et al. Combined use of autoantibodies (IA-2 autoantibody,
GAD autoantibody, insulin autoantibody, cytoplasmic islet cell
Human and Animal Rights and Informed Consent This article does antibodies) in type 1 diabetes: Combinatorial Islet Autoantibody
not contain any studies with human or animal subjects performed by any Workshop. Diabetes. 1998;47:1857–66.
of the authors. 14. Liu E, Eisenbarth GS. Accepting clocks that tell time poorly: fluid-
phase versus standard ELISA autoantibody assays. Clin Immunol.
2007;125:120–6. doi:10.1016/j.clim.2007.08.005.
15. Lampasona V, Schlosser M, Mueller PW, Williams AJK, Wenzlau
JM, Hutton JC, et al. Diabetes antibody standardization program:
first proficiency evaluation of assays for autoantibodies to zinc
References transporter 8. Clin Chem. 2011;57:1693–702. doi:10.1373/
clinchem.2011.170662.
16. Brooking H, Ananieva-Jordanova R, Arnold C, Amoroso M,
Papers of particular interest, published recently, have been Powell M, Betterle C, et al. A sensitive non-isotopic assay for
highlighted as: GAD65 autoantibodies. Clin Chim Acta. 2003;331:55–9. doi:10.
1016/S0009-8981(03)00088-3.
• Of importance
17. Miao D, Guyer KM, Dong F, Jiang L, Steck AK, Rewers M, et al.
•• Of major importance GAD65 autoantibodies detected by electrochemiluminescence
assay identify high risk for type 1 diabetes. Diabetes. 2013;62:
1. Gepts W. Pathologic anatomy of the pancreas in juvenile diabetes 4174–8. doi:10.2337/db13-0534.
mellitus. Diabetes. 1965;14:619–33. doi:10.2337/diab.14.10.619. 18. Palmer J, Asplin C, Clemons P, Lyen K, Tatpati O, Raghu P, et al.
2. Bottazzo GF, Florin-Christensen A, Doniach D. Islet-cell antibodies Insulin antibodies in insulin-dependent diabetics before insulin
in diabetes mellitus with autoimmune polyendocrine deficiencies. treatment. Science (80- ). 1983;222:1337–9. doi:10.1126/
Lancet. 1974;2:1279–83. science.6362005.
3. Eisenbarth GS. Type I diabetes mellitus. A chronic autoimmune 19. Baekkeskov S, Aanstoot HJ, Christgau S, Reetz A, Solimena M,
disease. N Engl J Med. 1986;314:1360–8. doi:10.1056/ Cascalho M, et al. Identification of the 64K autoantigen in insulin-
NEJM198605223142106. dependent diabetes as the GABA-synthesizing enzyme glutamic
4. Winter WE, Pittman D. The clinical application of islet autoanti- acid decarboxylase. Nature. 1990;347:151–6.
body testing for the diagnosis of autoimmune diabetes. MLO. 20. Rabin DU, Pleasic SM, Palmer-Crocker R, Shapiro JA. Cloning
Med Lab Obs. 2013;45:16, 20, 22 passim. and expression of IDDM-specific human autoantigens. Diabetes.
5. Bonifacio E. Predicting type 1 diabetes using biomarkers. 1992;41:183–6. doi:10.2337/diabetes.41.2.183.
Diabetes Care. 2015;38:989–96. doi:10.2337/dc15-0101. 21. Wenzlau JM, Juhl K, Yu L, Moua O, Sarkar SA, Gottlieb P, et al.
The cation efflux transporter ZnT8 (Slc30A8) is a major
6. Mujtaba MA, Fridell J, Book B, Faiz S, Sharfuddin A, Wiebke E,
autoantigen in human type 1 diabetes. Proc Natl Acad Sci U S A.
et al. Re-exposure to beta cell autoantigens in pancreatic allograft
2007;104:17040–5. doi:10.1073/pnas.0705894104.
recipients with preexisting beta cell autoantibodies. Clin
22. Kaufman DL, Erlander MG, Clare-Salzler M, Atkinson MA,
Transplant. 2015;29:991–6. doi:10.1111/ctr.12619.
Maclaren NK, Tobin AJ. Autoimmunity to two forms of
7. Piemonti L, Everly MJ, Maffi P, Scavini M, Poli F, Nano R, et al.
glutamate decarboxylase in insulin-dependent diabetes mellitus.
Alloantibody and autoantibody monitoring predicts islet
J Clin Invest. 1992;89:283–92. doi:10.1172/JCI115573.
transplantation outcome in human type 1 diabetes. Diabetes.
23. Wasmeier C, Hutton JC. Molecular cloning of phogrin, a protein-
2013;62:1656–64. doi:10.2337/db12-1258.
tyrosine phosphatase homologue localized to insulin secretory
8. Knip M, Akerblom HK, Becker D, Dosch H-M, Dupre J, Fraser W, granule membranes. J Biol Chem. 1996;271:18161–70. doi:10.
et al. Hydrolyzed infant formula and early β-cell autoimmunity: a 1074/jbc.271.30.18161.
randomized clinical trial. JAMA. 2014;311:2279–87. doi:10.1001/ 24. McLaughlin K, Richardson CC, Ravishankar A, Feltbower R,
jama.2014.5610. Morgan D, Christie M. Identification of tetraspanin-7 as a target
9. Lernmark A, Molenaar JL, van Beers WA, Yamaguchi Y, Nagataki of autoantibodies in type 1 diabetes. Diabetes. 2016. doi:10.2337/
S, Ludvigsson J, et al. The Fourth International Serum Exchange db15-1058.
Workshop to standardize cytoplasmic islet cell antibodies. The 25. Fenalti G, Law RHP, Buckle AM, Langendorf C, Tuck K, Rosado
Immunology and Diabetes Workshops and Participating CJ, et al. GABA production by glutamic acid decarboxylase is
Laboratories. Diabetologia. 1991;34:534–5. regulated by a dynamic catalytic loop. Nat Struct Mol Biol.
10. Bingley PJ, Bonifacio E, Mueller PW. Diabetes Antibody 2007;14:280–6. doi:10.1038/nsmb1228.
Standardization Program: first assay proficiency evaluation. 26. Kass I, Hoke DE, Costa MGS, Reboul CF, Porebski BT,
Diabetes. 2003;52:1128–36. Cowieson NP, et al. Cofactor-dependent conformational
11. Greenbaum C, Palmer J, Kuglin B, Kolb H. Insulin autoantibodies heterogeneity of GAD65 and its role in autoimmunity and neuro-
measured by radioimmunoassay methodology are more related to transmitter homeostasis. Proc Natl Acad Sci. 2014;111:E2524–9.
insulin-dependent diabetes mellitus than those measured by doi:10.1073/pnas.1403182111.
enzyme-linked immunosorbent assay: results of the Fourth 27. Wei J, Jin Y, Wu H, Sha D, Wu J-Y. Identification and functional
International Workshop on the Standardization of Insulin analysis of truncated human glutamic acid decarboxylase
Autoantibody Measurement. J Clin Endocrinol Metab. 1992;74: 65. J Biomed Sci. 2003;10:617–24.
1040–4. doi:10.1210/jc.74.5.1040. 28. Erdö SL. GABA outside the CNS. Springer Berlin Heidelberg; 2012.
12. Schmidli RS, Colman PG, Bonifacio E, Bottazzo GF, Harrison 29. Mally MI, Cirulli V, Otonkoski T, Soto G, Hayek A. Ontogeny
LC. High level of concordance between assays for glutamic acid and tissue distribution of human GAD expression. Diabetes.
decarboxylase antibodies. The First International Glutamic Acid 1996;45:496–501. doi:10.2337/diabetes.45.4.496.
53 Page 8 of 10 Curr Diab Rep (2016) 16: 53

30. Kanaani J, Cianciaruso C, Phelps EA, Pasquier M, Brioudes E, neuroendocrine tissues using two novel monoclonal antibodies.
Billestrup N, et al. Compartmentalization of GABA synthesis by Life Sci. 2009;84:678–87. doi:10.1016/j.lfs.2009.02.012.
GAD67 differs between pancreatic beta cells and neurons. PLoS 46. Diez J, Park Y, Zeller M, Brown D, Garza D, Ricordi C, et al.
One. 2015;10:e0117130. doi:10.1371/journal.pone.0117130. Differential splicing of the IA-2 mRNA in pancreas and lymphoid
31. Gotter J, Brors B, Hergenhahn M, Kyewski B. Medullary epithe- organs as a permissive genetic mechanism for autoimmunity
lial cells of the human thymus express a highly diverse selection of against the IA-2 type 1 diabetes autoantigen. Diabetes.
tissue-specific genes colocalized in chromosomal clusters. J Exp 2001;50:895–900.
Med. 2004;199:155–66. doi:10.1084/jem.20031677. 47. Lampasona V, Bearzatto M, Genovese S, Bosi E, Ferrari M,
32. Piquer S, Belloni C, Lampasona V, Bazzigaluppi E, Vianello M, Bonifacio E. Autoantibodies in insulin-dependent diabetes
Giometto B, et al. Humoral autoimmune responses to glutamic recognize distinct cytoplasmic domains of the protein tyro-
acid decarboxylase have similar target epitopes and subclass that sine phosphatase-like IA-2 autoantigen. J Immunol.
show titer-dependent disease association. Clin Immunol. 1996;157:2707–11.
2005;117:31–5. doi:10.1016/j.clim.2005.06.009. 48. Bearzatto M, Naserke H, Piquer S, Koczwara K, Lampasona V,
33. Bonifacio E, Lampasona V, Bernasconi L, Ziegler AG. Maturation Williams A, et al. Two distinctly HLA-associated contiguous
of the humoral autoimmune response to epitopes of GAD in pre- linear epitopes uniquely expressed within the islet antigen 2 mole-
clinical childhood type 1 diabetes. Diabetes. 2000;49:202–8. cule are major autoantibody epitopes of the diabetes-specific
34. Solimena M, Folli F, Denis-Donini S, Comi GC, Pozza G, De tyrosine phosphatase-like protein autoantigens. J Immunol.
Camilli P, et al. Autoantibodies to glutamic acid decarboxylase 2002;168:4202–8.
in a patient with stiff-man syndrome, epilepsy, and type I diabetes 49. Bearzatto M, Lampasona V, Belloni C, Bonifacio E. Fine mapping
mellitus. N Engl J Med. 1988;318:1012–20. doi:10.1056/ of diabetes-associated IA-2 specific autoantibodies. J Autoimmun.
NEJM198804213181602. 2003;21:377–82.
35. Saiz A, Blanco Y, Sabater L, González F, Bataller L, Casamitjana 50. Dromey JA, Weenink SM, Peters GH, Endl J, Tighe PJ, Todd I, et
R, et al. Spectrum of neurological syndromes associated with al. Mapping of epitopes for autoantibodies to the type 1 diabetes
glutamic acid decarboxylase antibodies: diagnostic clues for this autoantigen IA-2 by peptide phage display and molecular model-
association. Brain. 2008;131:2553–63. doi:10.1093/brain/ ing: overlap of antibody and T cell determinants. J Immunol.
awn183. 2004;172:4084–90.
36. McKeon A, Robinson MT, McEvoy KM, Matsumoto JY, Lennon
51. McLaughlin KA, Richardson CC, Williams S, Bonifacio E,
VA, Ahlskog JE, et al. Stiff-man syndrome and variants: clinical
Morgan D, Feltbower RG, et al. Relationships between major
course, treatments, and outcomes. Arch Neurol. 2012;69:230–8.
epitopes of the IA-2 autoantigen in type 1 diabetes: implications
doi:10.1001/archneurol.2011.991.
for determinant spreading. Clin Immunol. 2015;160:226–36. doi:
37. Soderbergh A, Myhre AG, Ekwall O, Gebre-Medhin G,
10.1016/j.clim.2015.06.002.
Hedstrand H, Landgren E, et al. Prevalence and Clinical associa-
52. Gylling M, Tuomi T, Björses P, Kontiainen S, Partanen J, Christie
tions of 10 defined autoantibodies in autoimmune polyendocrine
MR, et al. beta-cell autoantibodies, human leukocyte antigen II
syndrome type I. J Clin Endocrinol Metab. 2004;89:557–62. doi:
alleles, and type 1 diabetes in autoimmune polyendocrinopathy-
10.1210/jc.2003-030279.
candidiasis-ectodermal dystrophy. J Clin Endocrinol Metab.
38. Lampasona V, Passerini L, Barzaghi F, Lombardoni C,
2000;85:4434–40. doi:10.1210/jcem.85.12.7120.
Bazzigaluppi E, Brigatti C, et al. Autoantibodies to harmonin
53. Martino G, Grimaldi LM, Bazzigaluppi E, Passini N, Sinigaglia F,
and villin are diagnostic markers in children with IPEX syndrome.
Rogge L. The insulin-dependent diabetes mellitus-associated ICA
PLoS One. 2013;8:e78664. doi:10.1371/journal.pone.0078664.
105 autoantigen in stiff-man syndrome patients. J Neuroimmunol.
39. Buzzetti R, Di Pietro S, Giaccari A, Petrone A, Locatelli M, Suraci
1996;69:129–34. doi:10.1016/0165-5728(96)00104-X.
C, et al. High titer of autoantibodies to GAD identifies a specific
phenotype of adult-onset autoimmune diabetes. Diabetes Care. 54. Chimienti F, Devergnas S, Favier A, Seve M. Identification and
2007;30:932–8. doi:10.2337/dc06-1696. cloning of a beta-cell-specific zinc transporter, ZnT-8, localized
40. Solimena M, Dirkx R, Hermel JM, Pleasic-Williams S, Shapiro into insulin secretory granules. Diabetes. 2004;53:2330–7.
JA, Caron L, et al. ICA 512, an autoantigen of type I diabetes, is an 55. Lemaire K, Chimienti F, Schuit F. Zinc transporters and their role
intrinsic membrane protein of neurosecretory granules. EMBO J. in the pancreatic β-cell. J Diabetes Investig. 2012;3:202–11. doi:
1996;15:2102–14. 10.1111/j.2040-1124.2012.00199.x.
41. Hu YF, Zhang HL, Cai T, Harashima S, Notkins AL. The IA-2 56. Xu K, Zha M, Wu X, Yu Z, Yu R, Xu X, et al. Association
interactome. Diabetologia. 2005;48:2576–81. doi:10.1007/ between rs13266634 C/T polymorphisms of solute carrier family
s00125-005-0037-y. 30 member 8 (SLC30A8) and type 2 diabetes, impaired glucose
42. Trajkovski M, Mziaut H, Schubert S, Kalaidzidis Y, Altkrüger A, tolerance, type 1 diabetes—a meta-analysis. Diabetes Res Clin
Solimena M. Regulation of insulin granule turnover in pancreatic Pract. 2011;91:195–202. doi:10.1016/j.diabres.2010.11.012.
beta-cells by cleaved ICA512. J Biol Chem. 2008;283:33719–29. 57. Shan Z, Bao W, Zhang Y, Rong Y, Wang X, Jin Y, et al.
doi:10.1074/jbc.M804928200. Interactions between zinc transporter-8 gene (SLC30A8) and
43. Trajkovski M, Mziaut H, Altkrüger A, Ouwendijk J, Knoch K-P, plasma zinc concentrations for impaired glucose regulation
Müller S, et al. Nuclear translocation of an ICA512 cytosolic and type 2 diabetes. Diabetes. 2014;63:1796–803. doi:10.
fragment couples granule exocytosis and insulin expression in 2337/db13-0606.
{beta}-cells. J Cell Biol. 2004;167:1063–74. doi:10.1083/jcb. 58. Kirchhoff K, Machicao F, Haupt A, Schäfer SA, Tschritter O,
200408172. Staiger H, et al. Polymorphisms in the TCF7L2, CDKAL1 and
44. Mziaut H, Kersting S, Knoch K-P, Fan W-H, Trajkovski M, SLC30A8 genes are associated with impaired proinsulin
Erdmann K, et al. ICA512 signaling enhances pancreatic beta- conversion. Diabetologia. 2008;51:597–601. doi:10.1007/
cell proliferation by regulating cyclins D through STATs. Proc s00125-008-0926-y.
Natl Acad Sci U S A. 2008;105:674–9. doi:10.1073/pnas. 59. Chimienti F, Devergnas S, Pattou F, Schuit F, Garcia-Cuenca R,
0710931105. Vandewalle B, et al. In vivo expression and functional character-
45. Takeyama N, Ano Y, Wu G, Kubota N, Saeki K, Sakudo A, et al. ization of the zinc transporter ZnT8 in glucose-induced insulin
Localization of insulinoma associated protein 2, IA-2 in mouse secretion. J Cell Sci. 2006;119:4199–206. doi:10.1242/jcs.03164.
Curr Diab Rep (2016) 16: 53 Page 9 of 10 53

60. Deniro M, Al-Mohanna FA. Zinc transporter 8 (ZnT8) expression analysis of genetic contribution to development of islet
is reduced by ischemic insults: a potential therapeutic target to autoantibodies.
prevent ischemic retinopathy. PLoS One. 2012;7:e50360. 75. Hämäläinen AM, Ronkainen MS, Akerblom HK, Knip M.
doi:10.1371/journal.pone.0050360. Postnatal elimination of transplacentally acquired disease-
61. Mohanasundaram D, Drogemuller C, Brealey J, Jessup CF, Milner associated antibodies in infants born to families with type 1
C, Murgia C, et al. Ultrastructural analysis, zinc transporters, glu- diabetes. The Finnish TRIGR Study Group. Trial to reduce
cose transporters and hormones expression in New world primate IDDM in the genetically at risk. J Clin Endocrinol Metab.
(Callithrix jacchus) and human pancreatic islets. Gen Comp 2000;85:4249–53. doi:10.1210/jcem.85.11.6987.
Endocrinol. 2011;174:71–9. doi:10.1016/j.ygcen.2011.07.004. 76. Naserke HE, Bonifacio E, Ziegler AG. Prevalence, characteristics
62. Wenzlau JM, Liu Y, Yu L, Moua O, Fowler KT, Rangasamy S, et and diabetes risk associated with transient maternally acquired
al. A common nonsynonymous single nucleotide polymorphism in islet antibodies and persistent islet antibodies in offspring of
the SLC30A8 gene determines ZnT8 autoantibody parents with type 1 diabetes. J Clin Endocrinol Metab. 2001;86:
specificity in type 1 diabetes. Diabetes. 2008;57:2693–7. 4826–33. doi:10.1210/jcem.86.10.7931.
doi:10.2337/db08-0522. 77. Krischer JP, Lynch KF, Schatz DA, Ilonen J, Lernmark Å,
63. Achenbach P, Lampasona V, Landherr U, Koczwara K, Krause S, Hagopian WA, et al. The 6 year incidence of diabetes-associated
Grallert H, et al. Autoantibodies to zinc transporter 8 and autoantibodies in genetically at-risk children: the TEDDY study.
SLC30A8 genotype stratify type 1 diabetes risk. Diabetologia. Diabetologia. 2015;58:980–7. doi:10.1007/s00125-015-3514-y.
2009;52:1881–8. doi:10.1007/s00125-009-1438-0. 78. Ziegler A-G, Bonifacio E. Age-related islet autoantibody inci-
64. Andersson C, Larsson K, Vaziri-Sani F, Lynch K, Carlsson A, dence in offspring of patients with type 1 diabetes. Diabetologia.
Cedervall E, et al. The three ZNT8 autoantibody variants together 2012;55:1937–43. doi:10.1007/s00125-012-2472-x.
improve the diagnostic sensitivity of childhood and adolescent 79. Chmiel R, Giannopoulou EZ, Winkler C, Achenbach P,
type 1 diabetes. Autoimmunity. 2011;44:394–405. doi:10.3109/ Ziegler A-G, Bonifacio E. Progression from single to multiple islet
08916934.2010.540604. autoantibodies often occurs soon after seroconversion: implica-
65. Rosenzweig JL, Havrankova J, Lesniak MA, Brownstein M, Roth J. tions for early screening. Diabetologia. 2015;58:411–3. doi:10.
Insulin is ubiquitous in extrapancreatic tissues of rats and 1007/s00125-014-3443-1.
humans. Proc Natl Acad Sci U S A. 1980;77:572–6. doi: 80.•• Steck AK, Vehik K, Bonifacio E, Lernmark A, Ziegler A-G,
10.1073/pnas.77.1.572. Hagopian WA, et al. Predictors of progression from the appearance
66. Eng J, Yalow RS. Evidence against extrapancreatic insulin of islet autoantibodies to early childhood diabetes: The
synthesis. Proc Natl Acad Sci U S A. 1981;78:4576–8. doi:10. Environmental Determinants of Diabetes in the Young (TEDDY).
1073/pnas.78.7.4576. Diabetes Care. 2015;38:808–13. doi:10.2337/dc14-2426. This
67. Pugliese A, Zeller M, Fernandez A, Zalcberg LJ, Bartlett RJ, study presents an updated analysis of the contribution of islet
Ricordi C, et al. The insulin gene is transcribed in the human autoantibody combinations and titers to T1D risk.
thymus and transcription levels correlated with allelic variation 81. Bonifacio E, Scirpoli M, Kredel K, Füchtenbusch M, Ziegler AG.
at the INS VNTR-IDDM2 susceptibility locus for type 1 diabetes. Early autoantibody responses in prediabetes are IgG1 dom-
Nat Genet. 1997;15:293–7. doi:10.1038/ng0397-293. inated and suggest antigen-specific regulation. J Immunol.
68. Vafiadis P, Bennett ST, Todd JA, Nadeau J, Grabs R, Goodyer CG, 1999;163:525–32.
et al. Insulin expression in human thymus is modulated by INS 82. Hawa MI, Fava D, Medici F, Deng YJ, Notkins AL, De Mattia G,
VNTR alleles at the IDDM2 locus. Nat Genet. 1997;15:289–92. et al. Antibodies to IA-2 and GAD65 in type 1 and type 2 diabetes:
doi:10.1038/ng0397-289. isotype restriction and polyclonality. Diabetes Care. 2000;23:228–
69. Achenbach P, Koczwara K, Knopff A, Naserke H, Ziegler A-G, 33. doi:10.1128/AAC.03728-14.
Bonifacio E. Mature high-affinity immune responses to 83. Seissler J, Eikamp K, Schott M, Scherbaum WA, DENIS Study
(pro)insulin anticipate the autoimmune cascade that leads to type Group. IA-2 autoantibodies restricted to the IgG4 subclass are
1 diabetes. J Clin Invest. 2004;114:589–97. doi:10.1172/ associated with protection from type 1 diabetes. Horm Metab
JCI21307. Res. 2002;34:186–91. doi:10.1055/s-2002-26708.
70. Hall TR, Thomas JW, Padoa CJ, Torn C, Landin-Olsson M, 84. Hoppu S, Ronkainen MS, Kimpimäki T, Simell S, Korhonen S,
Ortqvist E, et al. Longitudinal epitope analysis of insulin- Ilonen J, et al. Insulin autoantibody isotypes during the prediabetic
binding antibodies in type 1 diabetes. Clin Exp Immunol. process in young children with increased genetic risk of type 1
2006;146:9–14. doi:10.1111/j.1365-2249.2006.03178.x. diabetes. Pediatr Res. 2004;55:236–42. doi:10.1203/01.PDR.
71. Wong SL, Priestman A, Holmes DT. Recurrent hypoglycemia 0000100905.41131.3F.
from insulin autoimmune syndrome. J Gen Intern Med. 85. Achenbach P, Warncke K, Reiter J, Naserke HE, Williams AJK,
2014;29:250–4. doi:10.1007/s11606-013-2588-9. Bingley PJ, et al. Stratification of type 1 diabetes risk on the basis
72.• Ziegler AG, Rewers M, Simell O, Simell T, Lempainen J, Steck A, of islet autoantibody characteristics. Diabetes. 2004;53:384–92.
et al. Seroconversion to multiple islet autoantibodies and risk of doi:10.2337/diabetes.53.2.384.
progression to diabetes in children. JAMA. 2013;309:2473–9. 86. A.J.K. Williams, V. Lampasona, M. Schlosser, P.W. Mueller, D.
doi:10.1001/jama.2013.6285. This study presents a Pittman, W.E. Winter RW, B. Akolkar, P.J. Bingley1 PA.
combined analysis of islet autoimmunity markers and N-terminally truncated GAD discriminates progression in
disease risk in all of the major ongoing prospective GAD antibody positive relatives of patients with type 1
studies worldwide. diabetes. Diabetologia. 2013;65:56.
73. Giannopoulou EZ, Winkler C, Chmiel R, Matzke C, Scholz M, 87. Williams AJK, Lampasona V, Wyatt R, Brigatti C, Gillespie KM,
Beyerlein A, et al. Islet autoantibody phenotypes and incidence in Bingley PJ, et al. Reactivity to N-terminally truncated
children at increased risk for type 1 diabetes. Diabetologia. GAD65(96-585) identifies GAD autoantibodies that are
2015;58:2317–23. doi:10.1007/s00125-015-3672-y. more closely associated with diabetes progression in rela-
74.•• Törn C, Hadley D, Lee H-S, Hagopian W, Lernmark Å, Simell O, tives of patients with type 1 diabetes. Diabetes. 2015;64:
et al. Role of type 1 diabetes-associated SNPs on risk of autoanti- 3247–52. doi:10.2337/db14-1694.
body positivity in the TEDDY Study. Diabetes. 2015;64:1818–29. 88. Williams AJK, Lampasona V, Schlosser M, Mueller PW, Pittman
doi:10.2337/db14-1497. This study presents a comprehensive DL, Winter WE, et al. Detection of antibodies directed to the N-
53 Page 10 of 10 Curr Diab Rep (2016) 16: 53

terminal region of GAD is dependent on assay format and human type 1 diabetes. Clin Exp Immunol. 2014;177:571–85.
contributes to differences in the specificity of GAD autoantibody doi:10.1111/cei.12362.
assays for type 1 diabetes. Diabetes. 2015;64:3239–46. doi:10. 99. Kawasaki E, Yu L, Rewers M, Hutton J, Eisenbarth G. Definition
2337/db14-1693. of multiple ICA512/phogrin autoantibody epitopes and
89. Achenbach P, Bonifacio E, Williams AJK, Ziegler AG, Gale detection of intramolecular epitope spreading in relatives
EAM, Bingley PJ, et al. Autoantibodies to IA-2beta improve of patients with type 1 diabetes. Diabetes. 1998;47:733–42. doi:
diabetes risk assessment in high-risk relatives. Diabetologia. 10.2337/diabetes.47.5.733.
2008;51:488–92. doi:10.1007/s00125-007-0912-9. 100. Jayakrishnan B, Hoke DE, Langendorf CG, Buckle AM, Rowley
90. De Grijse J, Asanghanwa M, Nouthe B, Albrecher N, Goubert P, MJ. An analysis of the cross-reactivity of autoantibodies to
Vermeulen I, et al. Predictive power of screening for antibodies GAD65 and GAD67 in diabetes. PLoS One. 2011;6:e18411.
against insulinoma-associated protein 2 beta (IA-2beta) and zinc doi:10.1371/journal.pone.0018411.
transporter-8 to select first-degree relatives of type 1 diabetic pa- 101. Keenan HA, Sun JK, Levine J, Doria A, Aiello LP, Eisenbarth G,
tients with risk of rapid progression to clinical onset of the disease: et al. Residual insulin production and pancreatic ß-cell turnover
implications for prevention trials. Diabetologia. 2010;53:517–24. after 50 years of diabetes: Joslin Medalist Study. Diabetes.
doi:10.1007/s00125-009-1618-y. 2010;59:2846–53. doi:10.2337/db10-0676.
91. Naserke HE, Ziegler AG, Lampasona V, Bonifacio E. Early 102. Richardson CC, Dromey JA, McLaughlin KA, Morgan D,
development and spreading of autoantibodies to epitopes Bodansky HJ, Feltbower RG, et al. High frequency of
of IA-2 and their association with progression to type 1 autoantibodies in patients with long duration type 1 diabe-
diabetes. J Immunol. 1998;161:6963–9. tes. Diabetologia. 2013;14:2538–40. doi:10.1007/s00125-
92. Mayr A, Schlosser M, Grober N, Kenk H, Ziegler AG, Bonifacio 013-3017-7.
E, et al. GAD autoantibody affinity and epitope specificity identify 103. Tridgell DM, Spiekerman C, Wang RS, Greenbaum CJ.
distinct immunization profiles in children at risk for type 1 Interaction of onset and duration of diabetes on the percent of
diabetes. Diabetes. 2007;56:1527–33. doi:10.2337/db06-1715. GAD antibody and insulinoma-antigen 2 antibodies-
positive subjects in the Type 1 Diabetes Genetic
93. Bender C, Schlosser M, Christen U, Ziegler AG, Achenbach P.
Consortium Database. Diabetes Care. 2011. doi:10.2337/
GAD autoantibody affinity in schoolchildren from the general
dc10-1903.
population. Diabetologia. 2014;57:1911–8. doi:10.1007/s00125-
104. Roep BO, Stobbe I, Duinkerken G, van Rood JJ, Lernmark A,
014-3294-9.
Keymeulen B, et al. Auto- and alloimmune reactivity to human
94. Achenbach P, Schlosser M, Williams AJK, Yu L, Mueller PW, islet allografts transplanted into type 1 diabetic patients. Diabetes.
Bingley PJ, et al. Combined testing of antibody titer and affinity 1999;48:484–90.
improves insulin autoantibody measurement: Diabetes Antibody 105. Braghi S, Bonifacio E, Secchi A, Di Carlo V, Pozza G, Bosi E.
Standardization Program. Clin Immunol. 2007;122:85–90. doi:10. Modulation of humoral islet autoimmunity by pancreas allotrans-
1016/j.clim.2006.09.004. plantation influences allograft outcome in patients with type 1
95. Krause S, Chmiel R, Bonifacio E, Scholz M, Powell M, diabetes. Diabetes. 2000;49:218–24.
Furmaniak J, et al. IA-2 autoantibody affinity in children at risk 106. Bosi E, Braghi S, Maffi P, Scirpoli M, Bertuzzi F, Pozza G, et al.
for type 1 diabetes. Clin Immunol. 2012;145:224–9. doi:10.1016/ Autoantibody response to islet transplantation in type 1 diabetes.
j.clim.2012.09.010. Diabetes. 2001;50:2464–71.
96.•• Arif S, Leete P, Nguyen V, Marks K, Nor NM, Estorninho M, et al. 107. Madaschi S, Rossini A, Formenti I, Lampasona V, Marzoli SB,
Blood and islet phenotypes indicate immunological heterogeneity Cammarata G, et al. Treatment of thyroid-associated orbitopathy
in type 1 diabetes. Diabetes. 2014;63:3835–45. doi:10.2337/db14- with rituximab—a novel therapy for an old disease: case report
0365. This paper present striking data regarding the existence and literature review. Endocr Pract. 2010;16:677–85. doi:10.4158/
of diverse sub-types of islet autoimmunity in T1D. EP09385.RA.
97. Skyler JS. Characterizing subgroups of type 1 diabetes. Diabetes. 108. Demeester S, Keymeulen B, Kaufman L, Van Dalem A, Balti EV,
2014;63:3578–80. doi:10.2337/db14-1160. Van de Velde U, et al. Preexisting insulin autoantibodies predict
98. Thompson WS, Pekalski ML, Simons HZ, Smyth DJ, Castro- efficacy of otelixizumab in preserving residual β-cell function in
Dopico X, Guo H, et al. Multi-parametric flow cytometric and recent-onset type 1 diabetes. Diabetes Care. 2015;38:644–51.
genetic investigation of the peripheral B cell compartment in doi:10.2337/dc14-1575.

You might also like