You are on page 1of 11

Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

Contents lists available at ScienceDirect

Journal of Trace Elements in Medicine and Biology


journal homepage: www.elsevier.com/locate/jtemb

Review

Antigen-specific immunotherapies in type 1 diabetes


Xuejiao Zhang a, Ying Dong b, Dianyuan Liu a, Liu Yang a, Jiayi Xu c, Qing Wang a, *
a
Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
b
Department of Radiation Oncology, Jilin Cancer Hospital, Changchun 130000, China
c
School of Public Health, Jilin University, Changchun 130000, China

A R T I C L E I N F O A B S T R A C T

Keywords: Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by the destruction of pancreatic beta cells, in
Type 1 diabetes mellitus which immune system disorder plays an important role. Finding a cure for T1DM and restoring beta cell function
Antigen-specific immunotherapies has been a long-standing goal. Research has shown that immune regulation with pancreatic islet auto-antigens
Immune tolerance
may be the most specific and safe treatment for T1DM. Immunological intervention using diabetogenic auto-
ZnT8
antigens as a target can help identify T1DM in high-risk individuals by early screening of autoantibodies
Prevention
(AAbs) before the loss of pancreatic islet function and thus achieve primary prevention of T1DM. However,
induction of self-tolerance in patients with pre-diabetes can also slow down the attack of autoimmunity, and
achieve secondary prevention. Antigen-based immune therapy opens up new avenues for the prevention and
treatment of T1DM. The zinc transporter 8 (ZnT8) protein, presents in the serum of pre-diabetic and diabetic
patients, is immunogenic and can cause T1D autoimmune responses. ZnT8 has become a potential target of
humoral autoimmunity; it is of great significance for the early diagnosis of T1D. ZnT8-specific CD8+ T cells can
be detected in most T1DM patients, and play a key role in the progression of T1D. As an immunotherapy target, it
can improve the dysfunction of beta cells in T1DM and provide new ideas for the treatment of T1D. In this
review, we summarize research surrounding antigen-specific immunotherapies (ASI) over the past 10 years and
the ZnT8 antigen as an autoimmune target to induce self-tolerance for T1DM.

1. Introduction AAbs (IAA) [7,8]. In this paper, we summarize the development and use
of ASIs over the past 10 years and the ZnT8 auto-antigen as a target to
Type 1 diabetes mellitus (T1DM) is an autoimmune metabolic dis­ induce immune tolerance for T1DM.
ease characterized by the damage of beta cells [1]. The physical, social,
and economic costs of T1DM are huge; according to statistics late 1.1. T1DM and autoimmune pathogenesis
diagnosis and lack of access to insulin are considered to be the main
causes of death from T1DM [2]. At present, the most effective treatment Although the pathogenesis of T1DM is not yet clear, it is certain that
is lifelong exogenous insulin replacement therapy, but this inevitably a disorder of the immune system plays an essential role in beta cell
leads to diabetic complications and cannot prevent the autoimmune destruction. One consequence of autoimmune attack and loss of beta cell
destruction of beta cells [3]. Allogeneic islet transplantation was suc­ function is a decrease or even lack of insulin secretion, which is manifest
cessful, but because of severe immune rejection this method was not as hyperglycemia and gradually progresses to diabetes [9,10]. An im­
widely used [4]. The development of antigen-based immunotherapy mediate need for exogenous insulin and lifelong replacement therapy
targeting the antigen-specific immune tolerance pathways is expected to are important features of T1DM. Insulin is the only hormone in the body
prevent or reverse T1DM [5]. It is aimed at regulating the potential that lowers blood glucose, and beta cells are responsible for sensing and
defects of the immune system that cause the destruction of beta cells by releasing insulin [11]. T1DM mostly occurs in children and adolescents,
auto-antigens to alleviate T1DM [6]. Furthermore, ZnT8 antibodies are but age is not a limiting factor [12]. Autoimmune-mediated diabetes can
newly discovered independent biomarkers for the diagnosis of T1DM in be divided into two types according to different ages: the classic T1DM,
addition to glutamic acid decarboxylase AAbs (GADA), insulinoma and the latent type autoimmune diabetes in adults (LADA); the latter
associated protein tyrosine phosphatase 2 AAbs (IA-2A), and insulin exhibits slow progression in adults, with the rate of beta cell failure

* Correspondence to: Department of Endocrinology, China-Japan Union Hospital of Jilin University, 126 Xiantai St, Changchun 130033, China.
E-mail address: wang_qing@jlu.edu.cn (Q. Wang).

https://doi.org/10.1016/j.jtemb.2022.127040
Received 10 January 2022; Received in revised form 18 June 2022; Accepted 14 July 2022
Available online 15 July 2022
0946-672X/© 2022 The Authors. Published by Elsevier GmbH. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

being much slower and the residual ability of beta cell function much responses to auto-antigens in normal individuals? That is because of the
stronger than that of classic T1DM [13]. existence of central immune tolerance and peripheral immune tolerance
How is the autoimmune process in beta cells implemented in the to ensure that the body responds correctly to self-antigens [24]. But in
pancreatic islets? Islet-specific cytotoxic T cells cloned from a patient some cases, when immune tolerance is lost, auto-reactive T cells are
with T1DM leads to beta cell destruction after transplantation into HLA- activated and proliferate after encountering auto-antigens and then
A2 transgenic non obese diabetic (NOD) -scid IL2Rγnull mice, indicating secrete and release inflammatory factors to induce insulitis, resulting in
that autoreactive CD8+ T cells are involved in T1DM initiation and beta the destruction and loss of beta cells [25,26]. Therefore, the fourth
cell destruction [14]. This showed that the immune process of T1DM is typical feature of T1DM is insulitis [27,28]. Autoantigen-specific path­
mediated by auto-reactive T lymphocytes. Therefore, persistent ogenic T cells and insulitis are considered to be immune factors that
auto-reactive T-cells in diabetic patients are the key to the onset of mediate the destruction of pancreatic beta cells in T1DM [29,30].
T1DM [1]. Why do T cells target the destruction of beta cells? It may Taken together, a plausible way to treat T1DM based on the etiology
depend on the recognition of diabetogenic antigen epitopes produced in and pathogenesis is to combat the destructive pathogenic T cells and
beta cells by anti-islet T-cells [15]. Another study also demonstrated this restore the function of pancreatic islet beta cells at the same time.
point, showing that auto-reactive T-cells in the pancreatic islets are Therefore, rebuilding the immune tolerance of beta cells has become the
auto-antigen specific [16]. So, the second characteristic feature of T1DM target of treatment for T1DM (Fig. 1).
is the existence of persistent beta cell specific auto-reactive CD8+ T cells
in the circulation [17,18], which play a leading role in the pathogenesis
of T1DM [19]. The destruction of beta cells leads to the release of 1.2. The development of T1DM treatment
auto-antigens and expansion of AAb-producing B lymphocytes, so the
third distinguishing feature of T1DM is the presence of pancreatic T1DM has been proven to be an autoimmune disease that damages
islet-specific AAbs against beta cell auto-antigens in the serum, which beta cells due to over activation of effector T cells. A humanized Fc re­
can be used as a reliable biological marker for the diagnosis of T1DM ceptor nonbinding anti-CD3 monoclonal antibody (mAb), teplizumab,
[20]. AAbs are key to distinguishing T1DM from type 2 diabetes mellitus was developed based on this strategy for the treatment of patients with
(T2DM), about 96 % of patients are detected to be positive for at least stage 2 and 3 T1D. Clinical trials have confirmed that teplizumab can
one type of AAb, including GADA, IAA, or IA-2A [21,22], and the ZnT8A improve the stimulated C-peptide responses [31], and can significantly
[7,23]. However, why do T cells not produce abnormal immune delay the onset of diabetes in high-risk groups [32]. Based on these
clinical trial results, the US Food and Drug Administration (FDA) is

Fig. 1. Mechanism of T cell-mediated beta cell destruction in T1D. T cells are enabled to identify self and non-self through positive selection, those with strong
binding affinity to self-peptides are removed by negative selection. Some T cells that escape the central tolerance require action from peripheral mechanisms. APCs
migrate from the pancreas take up and present diabetogenic auto-antigens and activate T cells; activated pathogenic T cells migrate to pancreatic islets and begin the
process of beta cell destruction, eventually leading to T1D. Tregs attempt to prevent beta cell damage by secreting cytokines. Autoreactive B cells are also involved in
beta cell damage. They activate autoreactive T cells, which act on islets by secreting inflammatory cytokines (IFN-γ, TNF, IL-17) and CTL-mediated killing; in turn,
auto-reactive T cells provide help signals to auto-reactive B cells, which further exacerbate T1D by promoting the differentiation of auto-reactive B cells into plasma
cells and the production of islet-specific AAbs. Bregs inhibit beta cell damage by secreting the cytokines IL-10 and TGF-γ. ZnT8 is the main autoantigen in T1D. The
secretion of insulin granules can increase the exposure of ZnT8 autoantigen to the cell surface and initiate ZnT8 epitope-specific T cell-mediated beta cell destruction,
resulting in T1D. As beta cells are destroyed, autoantigens release increases, amplifying the T cell-mediated immune response.Abbreviations: T1D: type 1 diabetes;
APCs: antigen presenting cells; Tregs: regulatory T cells; CTL: cytotoxic T lymphocytes; IFN-γ: interferon-γ; TNF: tumor necrosis factor; IL-17: interleukin-17; AAbs:
autoantibodies; Bregs: regulatory B cells; IL-10: interleukin − 10; TGF-γ: transforming growth factor-γ; ZnT8: zinc transporter 8.

2
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

currently considering approval of teplizumab as a treatment for T1D. RTX treatment [53]. In line with this notion, a 2-year clinical trial also
Other immunotherapy drugs targeting T cells in clinical trials include confirmed that the combined therapy with regulatory T cells (Tregs) and
anti-thymocyte globulin (ATG) [33]. An animal randomized controlled rituximab was generally consistently superior to monotherapy with
study showed that Low-dose ATG slowed decline of C- peptide and Tregs in controlling recent-onset T1DM in paediatric patients [54],
reduced HbA1c in new-onset T1D [34]; Abatacept, inhibits the activa­ which also reminds us that it can be personalized at least according to
tion of B cells and other antigen-presenting cells by acting on follicular the age of the patient.
helper T cells [35]; Alefacept, a second costimulatory receptor CD2 In­ Pancreatic islet transplantation has brought great hope to T1DM
hibitor [36]. These drugs have a significant effect on delaying the pro­ patients. However, due to the shortage of donors, poor revasculariza­
gression of the disease, but these effects are sustainable for not all tion, immunosuppression and many other serious problems, the failure
patients. Therefore, the treatment time window is particularly of islet transplantation has greatly affected the development of islet
important. transplantation. To address these issues, biomaterial-based strategies
Increasing evidence showed that B lymphocytes are important not are being explored. The islet encapsulation strategy encapsulates
only for autoantibody-mediated diseases, but also for those that are ul­ xenogeneic islets by semi permeable membrane encapsulate, creating a
timately T cell-mediated, such as T1DM. There is a collaborative inter­ protective environment between the graft and recipient, allowing oxy­
action between autoreactive B and T lymphocytes. B lymphocytes gen and nutrients to enter the encapsulated cells, insulin and metabolic
contribute to islet-specific autoimmunity by presenting antigens to waste products to be excreted while preventing immune cells [55]. It can
autoreactive T cells and providing costimulatory signals, acting on the promote islet transplantation without using toxic immunosuppressants
activation of pathogenic T cells [37]. In turn, activated T cells provide to prevent the host immune response. In theory, it can effectively protect
help signals to autoreactive B cells to further aggravate impaired B cell the survival of transplanted islets and improve insulin secretion.
tolerance and promote the production of autoantibodies, promoting the Therefore, this strategy has high therapeutic potential. Current Islet
occurrence and development of T1D [38,39]; in addition, regulatory B encapsule approaches include macroencapsulation, microencapsulation
cells (Bregs) play an immunoregulatory role by secreting cytokines IL-10 and nanoencapsulation. Researchers have studied from the choose of
and transforming growth factor-β (TGF-β) [40]. Immunotherapy to islet encapsulation biomaterials and coencapsulation or modification of
antibody B cell against new-onset T1DM was initially performed mainly the islet material [56], the application in islet transplantation [55],
by pan-B-cell-depletion by targeting surface antigens with monoclonal merits and demerits of different approaches to the encapsulation tech­
antibodies. rituximab (RTX), a monoclonal antibody specific to B-cell nology [55,57], immune modulation [58], alternative islet sources and
surface protein CD20. Although RTX treatment of T1DM caused signif­ implantation sites [59], and the major challenge faced by islet trans­
icant B cell depletion, the long-term efficacy has not been established yet plantation [57,59,60] and other perspectives. The goal is to minimize
[41]. One study showed efficacy in preserving beta cell function for 1 the immune system’s attack on the graft and to improve islet secretion.
year; Extended follow-up of participants, whereas such therapy showed Relevant clinical studies have confirmed that encapsulated islets can
limited effect after 2 years, the improvement in C-peptide preservation effectively reduce the demand for exogenous insulin in patients with
was limited to the early period after RTX and disappeared 30 months T1D [61,62]. Although this strategy yielded positive results over time in
later [42]. Thus, pan-B-cell-depletion is not very effective as used and the experiments, all patients returned exogenous insulin requirements to
frequent depletion might be required to minimize the presence of pre-transplant levels within a 7-year follow-up period [61], and most
autoreactive B cells. patients cannot achieve insulin independence from this treatment
Then it leads to antigen-specific treatment using micro- and nano­ strategy [62]. Further studies have found that the fibrotic response of
particles for treatment of autoimmune disease. It includes micro- and the graft is significantly associated with treatment failure [63], and
nanoparticle-based therapy, such as antigen- or peptide-conjugated interleukin-1 (IL-1) and tumor necrosis factor (TNF-α) is also contribute
nanoparticles, antigen- or peptide-conjugated microparticles and to graft damage [64]. In addition, the oxygen and nutrient delivery of
pMHC-coated nanoparticles [43–46]; cell-based immunotherapy such as the encapsulated islets are also key factors that determine the success or
antigen-coupled syngeneic lymphoid cells and autoantigen-loaded failure of transplantation. Therefore, an ideal islet encapsulation envi­
erythrocytes [47,48]. Conjugate antigens or polypeptides to the sur­ ronment should have the following points: able to provide islet struc­
face of microparticles or nanoparticles or encapsulate antigens inside tural support, able to ensure a good blood supply, biocompatible [65],
microparticles or nanoparticles, transport them to designated sites, and and minimize the level of fibrosis and inflammation to achieve optimal
release them through conformational changes when blood glucose levels efficacy. More research is still needed to optimize this treatment
fluctuate. Its tolerance effect may involve multiple mechanisms, strategy.
including clonal anergy, clonal deletion, immune deviation, production In general, immunotherapy for T1D is in a stage of rapid develop­
of regulatory cells and the release of autoantigen-loaded apoptotic ment, and the above treatments have been proven to be effective in
bodies [49]. The Antigen-polymer conjugate polymeric modular nano­ relieving disease progression in T1D patients. However, there are still
particles (acNPs) platform can precisely control the size of antigens, many challenges, such as unsustainable immune tolerance and it is
precisely control over their size, Antigen loading, Antigen release and necessary to accurately grasp the treatment time window, which brings
deliver single or multiple pathogenic antigenic epitopes with loading of great difficulties to clinical application. Based on the above consider­
single or multiple Antigens [50]. ations, ASI induces self-tolerance to β-cell self-antigens, selectively
In T1DM, antigen-specific immunotherapy (ASI) offers a good and eliminates self-reactive T cells without affecting overall immunity, and
safe therapy to induce immune tolerance. Although islet antigen mon­ can delay the time of immune tolerance to a certain extent. Currently,
otherapy has shown promise in animal models, prevention or reversal relevant research is still in progress (Table 1). ASI may be important
has not been achieved in human clinical trials. So there is still an urgent contributors to future therapeutic prospects, and their specificity may be
need to optimize those immunotherapies, and combination therapy may beneficial from a safety perspective.
be needed to further improve the therapeutic efficacy. Anti-CD20
combined with insulin has a moderate protective effect in prevention 1.3. T1DM and immune tolerance
of T1D onset, but no therapeutic efficacy in NOD mice [51]. B cell
depletion might be combined with T cell-based therapies to restore The immune system is one of the most complex networks in humans;
immune tolerance. A study reported that the combined treatment of its main function is to recognize and protect the body from foreign
intravenous anti-CD20 mAb and oral anti-CD3 mAb was more effective pathogens, while maintaining self-tolerance to avoid abnormal immu­
than anti-CD20 monotherapy in NOD mice [52]. In human, an increase nity caused by its own tissues [66]. Self-tolerance maintenance and
in T cell frequency and activity was observed in T1D patients receiving immune regulation are the two important functions of the immune

3
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

Table 1
Reports of antigen-specific immunotherapy for type 1 diabetes mellitus (T1DM) in the last decade.
Author Year Subjects Cases Autoantigen Administration route Pathogenesis Clinical Efficacy Evaluation
(n)

Casas [156] 2022 individuals aged 12 GAD-alum Intralymphatic induced secretion of IL-13 C-peptide secretion increased
12–24 y with T1DM almost 4 years after the initial
< 6 months from immune intervention and insulin
diagnosis, requirement remained stable
Assfalg 2021 islet autoantibody- 44 Insulin Oral Associated with the T cell No differences in insulin and c-
[157] negative children responses to insulin. peptide in serum
aged
6 months to
2.99 years who had
a first-degree
relative with type 1
diabetes
Larsson et al. 2018 Patients aged 4–17.9 50 GAD-Alum – Retain residual beta cell No rapid decrease in C-peptide
[158] y function and increase
regulatory T cells.
Tavira et al. 2018 Recent-onset 12 GAD-Alum Intralymphatic+subcutaneous Depend on Th2-associate C-peptide remained stable at 180
[159] patients cytokine IL-13, IL-13 days while insulin dose
suppresses Teff decreased in 24 h in Lymph
proliferation and IL-2 nodes group; An increase in
secretion. insulin requirement with a
greater loss of C-peptide in
subcutaneous group.
Krischer 2017 Nondiabetic 560 Insulin Oral – Improve short-term c-peptide
[160] relatives of probands secretion
with T1DM
Alhadj 2017 < 100 d from 27 Proinsulin C19- Intradermal injection Induce IL-10 responses and Insulin use and stimulated c-
[161] diagnosis of T1DM A3 adaptive Tregs arising peptide remained stable
peptide after treatment; and retain
residual beta cell function
and increase regulatory T
cells.
Bonifacio 2015 Children 25 Insulin Oral Induce insulin-responsive –
[162] 2–7 y and proinsulin-responsive
regulatory T cells.
Roep [19] 2013 < 5 y from diagnosis 80 An engineered Intramuscular injections Deletion of CD8 + T cells C-peptide increases
of T1DM and > 18 y DNA plasmid reactive to proinsulin.
encoding
proinsulin (BHT-
3021).
Ludvigsson 2012 Recent-onset T1DM 334 GAD-Alum Subcutaneous injections – No significant increase in c-
[163] 10–20 y peptide secretion
Wherrett[ 2011 < 100 d from 145 GAD-Alum Subcutaneous injection – No significant increase in c-
164] diagnosis of T1DM peptide secretion
Vehik [165] 2011 Subjects with a 275 Insulin Oral Induce adaptive Tregs –
projected 5 y risk of from cohorts of preexisting
diabetes of 26–50 % insulin-specific T cells.
Fourlanos 2011 Recent-onset T1DM 52 Insulin Intranasal Induce immune tolerance –
[166] to insulin.

GAD-Alum: Glutamic acid decarboxylase formulated with aluminum hydroxide; alum has been the adjuvant of choice to minimize the possibility of promoting beta-
cell-mediated destruction together with GAD65.

system [66]. When the immune system fails in certain individuals, and positive selection makes T cells able to recognize a peptide/major his­
causes an attack on its own components resulting in the overexpression tocompatibility complex (pMHC), and the interaction between T cell
of AAbs, this can lead to disease, which is termed as autoimmune dis­ receptor (TCR) and MHC/self-antigen complex. T cells that do not pass
ease, characterized by the production of AAbs and activation of the central tolerance test undergo an intrinsic suicide program called
auto-reactive T lymphocytes [67]. When an autoimmune disease occurs, negative selection to remove them and prevent auto-reactivity [24].
auto-reactive T cells become resistant to suppression during the active There is evidence that defects in thymic T cell negative selection related
phase; but if immunity is normal, the underlying auto-reactive T cells to insulin reactivity is a key factor in the genetic predisposition in T1DM
will not cause autoimmune diseases owing to the regulation of a variety [71]. The purpose of central selection is to ensure that T cells entering
of inhibitory mechanisms, collectively known as “immune tolerance” the periphery do not respond to their own antigens [24]. However, many
[68]. Immune tolerance refers to the states of nonresponse in immunity self-reactive T cells still escape from thymic negative selection, which
to foreign antigens and self-tissues [69]. It is an essential mechanism leads to the need for peripheral mechanisms to ensure that self-tolerance
used to balance physiological responses and constantly fights off is maintained to prevent autoimmune diseases [72].
harmful molecules in the human body and eliminates or inactivates Peripheral immune tolerance may delete those escaped T cells in the
auto-reactive T lymphocytes that recognize self-antigens, to prevent the peripheral lymphoid organs through various mechanisms including
immune system from destroying itself and maintain the immune balance clone deletion, clone anergy, and immune suppression of regulatory T
[70]. cells [73]. The most important mechanism is the presence of specific cell
Immune tolerance is divided into central tolerance and peripheral populations to suppress abnormal immune responses that unintention­
tolerance [24]. The former occurs in the thymus, passing through ally target their own tissues; Tregs develop in the thymus and periphery

4
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

are critical for maintaining tolerance [74]. Tregs include naturally pancreatic tissues [96] where it maintains the balance of zinc in beta
arising Treg cells (Foxp3+ CD25+ CD4+ Tregs), induced Treg cells cells by importing and exporting zinc ions [97]. Pancreatic islet beta
(iTreg), type 1 regulatory T cells (Tr1) that produce the immunosup­ cells are the only source of insulin in the human body, and insufficient
pressive cytokine interleukin-10 (IL-10), and Type 3 helper T cells (Th3) insulin secretion is the main pathogenic factor of T1DM [98]. Zinc is an
that produce transforming growth factor beta (TGF-beta) [75]. Tregs are important ion in humans, which is involved in the synthesis, storage,
essential suppressors of unwanted autoimmune responses [76]. The and secretion of insulin, and plays an important role in the regulation of
mechanism of Tregs includes contact-dependent suppression and insulin signaling and glucose homeostasis [99]. ZnT8 is mainly
contact-independent suppression mechanisms [77]. The former can responsible for zinc enrichment in insulin secretory granules, and the
inhibit costimulatory signals and cytokine secretion by connecting increase of zinc accumulation in beta cells is the result of overexpression
Cytotoxic T lymphocyte antigen 4 (CTLA-4) on Tregs to CD80/CD86 on of ZnT8 [100].
APCs. The latter contact-independent mechanisms disrupt the metabolic Peptides derived from ZnT8 have been identified as biomarkers of
state of T effector cells via the production of the immunosuppressive diabetogenic self-antigens independent of other auto-antigens that can
cytokines IL-10 and TGF-beta [77,78]. Among them, the most typical be recognized by auto-reactive CD8+ T cells; hence, it is a major CD8+ T
mechanism is the natural CD25+ CD4+ Tregs that can specifically ex­ cell target of autoimmunity [101]. Therefore, the autoimmune process
press Foxp3 transcription, which plays an extremely critical role in of ZnT8 is being studied at the level of both AAbs and T-cells. T-cell
maintaining peripheral tolerance and preventing autoimmune diseases reactivity of ZnT8 antigen has been shown to exist in both rodent models
[79]. Animal experiments have shown that a deficiency of Foxp3+ Tregs and T1DM patients [102]. The proper application of ZnT8 antibody
can activate even weak or rare autoimmune T-cell clones, and subse­ (ZnT8A) complements the use of biomarkers including IAA, GADA, and
quently cause an autoimmune disease [80]. Tregs deficiency in humans IA-2A [9]. Furthermore, ZnT8-specific responses are particularly broad:
has also been confirmed to be associated with autoimmune diseases such GAD triggers responses in 50 % of patients, while other antigens stim­
as T1DM [81,82]. In the absence of specific antigen activation, Tregs ulate around 40 %; however, ZnT8 epitopes trigger responses in more
have no effect on regulating disease [83]. Therefore, Foxp3+ CD25+ than 64 % of patients [101]. Unlike other pancreatic auto-antigens,
CD4+ natural Tregs actively participate in the maintenance of ZnT8 is located on the cell surface; it can be transported to the cell
self-tolerance, and their abnormality is one of the causes of autoimmune membrane after insulin secretion, thereby exposing the transmembrane
disease. A balance is formed between autoimmunity and self-tolerance, domain (TMD) and being recognized by serum ZnT8A [103]. This
similar to a scale. The dynamic balance between Tregs and auto-reactive feature improves the possibility of ZnT8 being directly involved in
T cells determines the risk, timing, and progression of disease. antibody-mediated cellular dysfunction and cytotoxicity [104]. After
The onset and development of T1DM is also a process of dynamic insulin, ZnT8 is the second antigen that is specifically expressed in the
evolution. Although the specific pathogenesis is not yet clear, it can be pancreatic beta-cells and is more restricted in its tissue distribution than
determined that disorder of the immune system plays an extremely other auto-antigens; there are no other cells destroyed in T1DM [104,
important role in the loss of immune tolerance and the subsequent 105]. Furthermore, the expression of ZnT8A may not occur until there is
destruction of beta cells [84]. Naive T cells first encounter sufficient beta cell damage to make this auto-antigen perceptible to the
diabetes-related auto-antigens presented by APCs in lymph nodes, the immune system [106]. The unique position and tissue specificity of
result of which is a tolerance in healthy humans; but in T1DM, naïve T ZnT8 in pancreatic beta cells makes it an attractive candidate as a
cells are activated as pathogenic auto-reactive T cells and then migrate therapeutic target.
to the pancreatic islets and begin to damage beta cells [85]. Only when a
new balance is reached can autoimmune diseases be prevented, in which 1.5. ZnT8 and T1DM
Tregs, that are responsible for peripheral tolerance and attempt to pre­
vent unwanted autoimmune responses, recognize self-polypeptides and The pathogenesis of T1DM can be divided into three stages: 1) dia­
inhibit auto-reactive pathogenic T cells. betogenic AAbs exist, blood glucose is normal and no diabetes-related
Rebuilding immune tolerance refers to inducing the body to actively symptoms; 2) diabetogenic AAbs continually exist, beginning to
suppress the immune response to a specific antigen [86]. If tolerance to induce impaired glucose tolerance (IGT) but still no symptoms; 3) The
diabetogenic auto-antigens can be effectively reinstated then the beta persistence of diabetogenic AAbs leads to T1DM and the occurrence of
cell attack is prevented. Rebuilding immune tolerance can be roughly related symptoms of diabetes [25,26]. The appearance of AAbs is asso­
divided into two parts, one is by decreasing the number of pathogenic T ciated with the decline in the number and dysfunction of beta cells [3].
cells by inducing apoptosis and functional inactivation of highly acti­ Therefore, autoimmunity persistence in the first stage is owing to the
vated effector cells [87], or inducing the deletion of pathogenic T cells destruction of beta cells; when the number of beta cells is reduced to a
[88,89]. The other is through promoting immune regulation and certain extent, the inability to compensate by the remaining beta cells
increasing the number of Tregs and enhancing the function of Tregs [90, leads to hyperglycemia, and then to the second stage. In the asymp­
91]. This may be a future strategy for the treatment and prevention of tomatic first stage, namely pre-diabetes, the emergence of AAbs is the
autoimmune disease such as T1DM through reestablishing earliest sign of the establishment of autoimmunity and destruction of
self-tolerance. beta cells; about 96 % of patients show at least one AAb positivity at the
onset of T1DM, including GADA, IAA, IA-2A, or the new ZnT8A [7].
1.4. A new strategy for inducing immune tolerance: Evaluation of ZnT8- ZnT8 is now a confirmed autoimmune target for most patients with
antigen specific immunotherapy T1DM, and exists in the serum of pre-diabetic and diabetic patients
[107–109]. ZnT8 is a dimer-membrane protein consisted of 369 amino
ZnT8 is a product encoded by the SLC30A8 gene located on chro­ acids in a form and 320 amino acids in B form [110]. Each monomer
mosome 8, and is specifically expressed on the insulin granular mem­ composed of 6 transmembrane domains and a histidine loop, about 50 %
brane of pancreatic beta cells [92,93]. The protective effects of ZnT8 of ZnT8 is located in the endoplasmic reticulum [111]. ZnT8 mainly
loss-of-function mutations are either different between mouse or maintains the balance of zinc ions in pancreatic beta cells by pumping
human [94]. The polymorphic variant rs13266634 (R325W) in the zinc ions into intracellular vesicles or extracellular space [97]. Re­
human ZnT8 gene SLC30A8 is strongly associated with diabetes risk searches have shown, a principal epitope targeted by ZnT8A is affected
[94]. In NOD mice, ZnT8 is a minor diabetogenic antigen that can by a single amino acid at position 325; This amino acid is encoded by the
participate in diabetes in conditions in which the islet is first made SLC30A8 gene as three different variants, ZnT8RA arginine 325 Zinc
receptive to immunological insults [95]. It is the zinc transporter with transporter 8 autoantibody, ZnT8WA tryptophan 325 Zinc transporter 8
the highest known expression levels and unique tissue specificity in autoantibody and ZnT8QA glutamine Zinc transporter 8 autoantibody

5
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

[112,113]. The COOH-terminal domain of the ZnT8 is the early patho­ recognized after 35 y [106]. In summary, ZnT8 can be used as an
genesis of T1DM; therefore, screening these two main ZnT8 variants can immunobiological target for T1DM in children; early detection is
help early prediction of T1DM risk [96]. ZnT8A has been positively essential in individuals with a genetic risk for T1DM but without islet
detected in about 60–80 % of newly diagnosed patients with T1DM, of AAbs to achieve the primary prevention [133,134]. Furthermore, in
which 26 % detected no other diabetogenic AAbs, making it a new in­ individuals with multiple islet AAbs but without overt hyperglycemia,
dependent biological marker for the diagnosis of T1DM [114,115]. ZnT8 can be used to delay the disease and achieve secondary prevention
Studies have also found that patients who are ZnT8A-positive at an early [128,129,135]. Detection of pancreatic islet AAbs makes T1DM a pre­
stage, are often more likely to develop T1DM than ZnT8A-negative pa­ dictable disease. However, the relationship between ZnT8A and adult
tients, but there is no correlation between antibody titers and disease patients in T1DM requires further study.
progression [116]. The combined detection of ZnT8A with GADA, LADA type diabetes is a form of autoimmune-mediated diabetes in
IA-2A, and IAA can improve the sensitivity of detection in T1DM to 98 % adults where the two vital characteristics are: the rate of beta cell failure
[117]. Andersson et al. reported that the negative rate of AAbs was is slower than that of T1DM, and the early symptoms are similar to
reduced from 10 % to 7 % in combination with the detection of ZnT8A T2DM where there is slow progress in insulin therapy and it does not
on the basis of the three antigens GADA, IA-2A, and IAA, which require insulin replacement for at least 6 months after diagnosis [136].
improved the diagnostic sensitivity of T1DM [118]. As mentioned The former feature indicates that LADA has a wider time window than
above, ZnT8-specific responses are also particularly broad: compared T1DM, and timely screening intervention may slow down beta cell
with CD8+ T-cell responses induced by other auto-antigens, ZnT8 epi­ failure. The latter feature suggests that newly diagnosed patients with
topes elicit responses in more than 64 % of patients [101]. Several LADA are initially insulin-independent and can only be identified by
studies have shown that autoimmunity may commence many years detecting diabetogenic AAbs. Early research results showed that detec­
before manifestation of T1DM [28,81,119]. Furthermore, an autopsy tion of IAA, GADA, and IA-2A is currently the only means of dis­
result showed that the number of beta cells in pancreatic islets of pa­ tinguishing LADA from phenotypic T2DM [137,138], and at least one
tients with T1DM was about 10 % of that in individuals without T1DM, auto-antigen can be tested in the serum of LADA patients [13]. Huang
which once again demonstrated that the result of autoimmunity is the et al., in a Chinese study of 3062 individuals, showed that the new ZnT8
destruction of beta cells [120]. T1DM in general has a long prodromal auto-antigen was also an independent immune marker for differenti­
stage in which only AAbs reactive with diabetogenic antigen can be ating LADA from phenotypicT2DM; the detection of ZnT8A along with
detected [81]. Sofla etal followed the fate of beta-cell-specific CD8+ T GADA and IA-2A was shown to improve the diagnostic sensitivity of
cells in non-obese diabetic mice throughout the course of T1DM, showed Chinese LADA [139,140]. In summary, LADA and phenotypic T2DM can
that antigen-specific CD8+ T cells can be found in the pancreatic be distinguished by detection of diabetogenic AAbs, and early identifi­
draining lymph node and pancreas as early as 5 weeks of age. It indi­ cation and intervention may better preserve the function of beta cells
cated that autoimmune destruction can be found prior to clinical course, and delay disease progression [141–143]. Diabetes-related AAbs
and there is an asymptomatic period before the typical presentation with detected from LADA and classic T1DM are similar, so this cannot be used
clinical type 1 diabetes, which is associated with beta-cell functional loss as a diagnostic criteria.
[121]. Thus, the early assessment of ZnT8A can allow identification
from rapid progression to clinical onset of the disease, and subsequent 1.6. ZnT8 and the treatment of T1DM
tolerogenic therapies may be most effective in the earliest disease phase.
The incidence of ZnT8A-positive is correlated with the age of diag­ The mechanism of ZnT8 inducing T1DM autoimmunity may be
nosis of T1DM in children [122]. A study showed that among newly related to the structure of ZnT8. ZnT8 is the most abundantly expressed
diagnosed T1DM children < 15 y, ZnT8A-positive individuals were zinc transporter and is considered to be the regulator of zinc concen­
generally older [123]. A longitudinal study of children with genetic risk tration in beta cells, which is critical for insulin biosynthesis and storage
after birth also indicated that ZnT8A tends to appear later and is rarely [144]. Studies have shown that proper intracellular zinc concentration
detected before 6 months [124]. Also in support, a French study can ensure the function of islet cells, but expressive ZnT8 and zinc
involving 516 children with T1DM showed that children aged 6–17 y concentrations aggravate islet cell death when exposed to different
had a higher detection rate of ZnT8A-positive compared with children cellular stressors, such as hypoxia and cytokines [145]. ZnT8 is a dimer
< 5 y [125]. Other studies have also reported that the positive rate of membrane protein with a Y-shaped molecular architecture similar to
ZnT8A is higher in children aged 5–10 y [126] and that ZnT8A tends to that of immunoglobulin [146]. It readily binds to (Fab)2 and the Fc re­
go undetected before 3 y [127]. The abnormal autoimmunity is mostly gion of the bound autoantibody is able to bind immune effector cells to
initiated by the two antibodies GADA and IA-2Am with GADA appearing trigger antibody-dependent cytotoxicity against opsonized beta cells
steadily up to 10–15 y [128,129]. The greater the age at which auto­ [147]. Less than half of the protein can be immune surveilled, which
immunity begins, the less likely it is to develop into T1DM [130], with may explain why ZnT8 is a target for autoimmune attack in T1D.
children with negative AAbs < 15 y having a lower risk of developing Secondly, the ZnT8 protein is immunogenic and causes T1DM
T1DM, at around 0.4 % [22]. Therefore, early detection of ZnT8A autoimmune responses, which may be related to the etiology of T1DM.
combined with GADA and IA-2A is sufficient to predict childhood T1DM is an autoimmune disease characterized by the autoimmune
T1DM. destruction of pancreatic beta cells mediated by specific T cells, and
However, studies on adult patients with T1DM are few and incon­ CD8+ T cells are the final mediators of islet destruction, CD8+ T-cell
sistent. One study reported a negative correlation between the age of responses against ZnT8 and anti-ZnT8 AAbs were found in T1DM pa­
T1DM and the positive rate of ZnT8A, with it decreasing significantly in tients [148]. Ehlers [149] found that ZnT8-specific CD8+ T cells were
elderly T1DM patients [131]; this is contrary to the results of T1DM in detectable in the majority of T1DM patients (> 68 %), whereas the
children. In a Chinese population study, it was also found that the pos­ frequency of ZnT8-specific CD8+ T cells was significantly lower in
itive rate of ZnT8A decreased with increasing age of diagnosis [117]. In healthy individuals and patients with T2DM (<8 % and 30 %, respec­
another Pittsburgh Epidemiology of Diabetes Complications (EDC) tively). Nayak et al. [95] also found that ZnT8-specific CD8+ T cells
study, there was a positive correlation between the age of patients with generated after ZnT8 345–359 peptide stimulation could accelerate the
T1DM and positive rate of ZnT8A. The older the patient, the greater the progression of diabetes in irradiated recipient mice (NOD or NOD.
chance of being ZnT8A-positive; this may be because of a less aggressive Rag1-/-). Studies have also confirmed the presence of more znt8-specific
and thus more persistent immune process in those with older age at CD8+ T cells in the pancreas of patients with T1DM compared with
onset [132]. It has also been noted that the autoimmune process patients with type 2 diabetes or without diabetes, suggesting that
observed in T1DM < 35 is more aggressive compared to diabetes znt8-specific lymphocytes home to the pancreas [150]. And CD8+ T cells

6
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

isolated from peripheral blood of T1DM patients can secrete more IFN-γ Funding
after stimulation with ZnT8 peptide [150]. In addition, ZnT8 has
become a potential target of humoral autoimmunity [151] and is also This work was supported by the National Natural Science Foundation
related to the insulin secretory pathway. Glucose-stimulated insulin of China [grant number 82070796].
secretion (GSIS) promotes the display of ZnT8 on the surface of beta
cells, which may also aggravate beta cell destruction [147]. GSIS is CRediT authorship contribution statement
mainly mediated through a series of events that lead to the ATP-sensitive
potassium channels are inhibited, the consequential decreased efflux of QW helped perform the analysis with constructive discussions and
K+ results in a depolarization of the beta cells plasma membrane, and revised the manuscript. XZ contributed to the conception of the review
voltage-sensitive Ca2+-channels open, facilitating Ca2+ entry, which and wrote the manuscript. YD and LY performed the analysis with
triggers insulin secretion [152]. Huang et al. [103] showed that GSIS constructive discussions, carried out the study and collected important
increased the expression levels of ZnT8 autoantigen and its exposure on background information and references. DL and JX contributed signifi­
the beta cell surface. After glucose-stimulated insulin secretion, ZnT8 cantly to analysis and manuscript preparation. All authors have read and
moves to the cell surface along the insulin secretion pathway, resulting approved the final content of the manuscript.
in an increase in GSIS/ZnT8 exposure. In turn, coupling between the
GSIS and ZnT8 surface display enhanced beta cell damage. The above Conflict of Interest
results provide a basis for ZnT8 as a surface biomarker.
The authors declare that the research was conducted in the absence
1.7. Future directions of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Great progress has been made in our understanding of T1DM
immunotherapy, but there are still many problems that need to be
References
solved. First of all, therapies based on auto-antigens have been admin­
istered as native proteins; will the same or better therapeutic effect be [1] J.A. Bluestone, K. Herold, G. Eisenbarth, Genetics, pathogenesis and clinical
achieved if proteins modified as toleragens are used? Second, pancreatic interventions in type 1 diabetes, Nature 464 (2010) 1293–1300, https://doi.org/
10.1038/nature08933.
AAbs are often triggered years before the symptoms of severe hyper­
[2] C. De Beaufort, S. Besançon, N. Balde, Management of type 1 diabetes, Med Sante
glycemia, but the relationship between humoral autoreactivity and islet Trop. 28 (2018) 359–362, https://doi.org/10.1684/mst.2018.0834.
pathology remains unclear. Next, as mentioned above, early detection of [3] M.A. Atkinson, G.S. Eisenbarth, A.W. Michels, Type 1 diabetes, Lancet 383 (2014)
ZnT8A combined with GADA and IA-2A can predict T1DM in children at 69–82, https://doi.org/10.1016/s0140-6736(13)60591-7.
[4] R.P. Robertson, Islet transplantation as a treatment for diabetes - a work in
an early stage, but whether the age of onset will affect the autoimmune progress, N. Engl. J. Med 350 (2004) 694–705, https://doi.org/10.1056/
status, severity, and manifestations in adults requires further research to NEJMra032425.
explore the existence of sensitive markers for the diagnosis of adult [5] A.W. Michels, M. von Herrath, 2011 Update: antigen-specific therapy in type 1
diabetes, Curr. Opin. Endocrinol. Diabetes Obes. 18 (2011) 235–240, https://doi.
autoimmune diabetes. As an important AAb in T1DM, is ZnT8A also org/10.1097/MED.0b013e32834803ae.
associated with T2DM? Studies have reported that polymorphism of the [6] X. Wan, F.B. Guloglu, A.M. VanMorlan, L.M. Rowland, R. Jain, C.L. Haymaker, J.
SLC30A8 gene may be a risk factor for T2DM [144,153,154]. Hunt et al. A. Cascio, M. Dhakal, C.M. Hoeman, D.M. Tartar, H. Zaghouani, Mechanisms
underlying antigen-specific tolerance of stable and convertible Th17 cells during
also collected 800 serum samples from children aged 0–12 y before the suppression of autoimmune diabetes, Diabetes 61 (2012) 2054–2065, https://
diagnosis of T2DM, and found that in about 3 % of patients a positive doi.org/10.2337/db11-1723.
AAb appeared before diagnosis, suggesting that the presence of AAbs as [7] J.M. Wenzlau, J.C. Hutton, Novel diabetes autoantibodies and prediction of type
1 diabetes, Curr. Diab Rep. 13 (2013) 608–615, https://doi.org/10.1007/s11892-
risk factors may accelerate the development of T2DM [155]. Therefore,
013-0405-9.
more studies are needed to demonstrate a possible connection between [8] W.E. Winter, D.L. Pittman, I. Jialal, Practical clinical applications of islet
ZnT8 and T2DM. Moreover, it may not be sufficient to completely autoantibody testing in type 1 diabetes, J. Appl. Lab Med 7 (2022) 197–205,
https://doi.org/10.1093/jalm/jfab113.
control the self-destruction of beta cells by single-agent immunotherapy;
[9] S.E. Regnell, Å. Lernmark, Early prediction of autoimmune (type 1) diabetes,
combination immunotherapies through multiple pathogenic pathways Diabetologia 60 (2017) 1370–1381, https://doi.org/10.1007/s00125-017-4308-
may be needed to optimize immune intervention, such as by depleting 1.
effector cell populations before induction of antigen-specific immune [10] (2) Classification and diagnosis of diabetes, Diabetes Care 38 (Suppl) (2015)
S8–s16, https://doi.org/10.2337/dc15-S005.
tolerance or at the same time. Which drugs can result in lasting relief of [11] T. Haak, S. Gölz, A. Fritsche, M. Füchtenbusch, T. Siegmund, E. Schnellbächer, H.
beta cell function? Finally, we should increase our understanding of the H. Klein, T. Uebel, D. Droßel, Therapy of type 1 diabetes, Exp. Clin. Endocrinol.
molecular mechanism of T1DM and explore more biomarkers and Diabetes 127 (2019) S27–s38, https://doi.org/10.1055/a-0984-5696.
[12] T. Lorenzen, F. Pociot, P. Hougaard, J. Nerup, Long-term risk of IDDM in first-
therapeutic targets. degree relatives of patients with IDDM, Diabetologia 37 (1994) 321–327, https://
doi.org/10.1007/bf00398061.
2. Conclusions [13] B. Liu, Y. Xiang, Z. Liu, Z. Zhou, Past, present and future of latent autoimmune
diabetes in adults, Diabetes Metab. Res. Rev. 36 (2020), e3205, https://doi.org/
10.1002/dmrr.3205.
It is necessary to intervene early in autoimmune progression, prior to [14] W.W.J. Unger, T. Pearson, J.R.F. Abreu, S. Laban, A.R. van der Slik, der Kracht
the occurrence of T1DM. Although antigen-specific immunotherapy is SM-v, M.G.D. Kester, D.V. Serreze, L.D. Shultz, M. Griffioen, J.W. Drijfhout, D.
L. Greiner, B.O. Roep, Islet-specific CTL cloned from a type 1 diabetes patient
still in its infancy, it may be a potentially powerful weapon in the future
cause beta-cell destruction after engraftment into HLA-A2 transgenic NOD/scid/
owing to its advantages of safety and effectiveness. Many islet-specific T IL2RG null mice, PloS One 7 (2012), e49213, https://doi.org/10.1371/journal.
cell antigens have been identified that contribute to diabetes develop­ pone.0049213.
[15] M.A. Atkinson, J.A. Bluestone, G.S. Eisenbarth, M. Hebrok, K.C. Herold, D. Accili,
ment. If a solution is developed that can selectively expand antigen-
M. Pietropaolo, P.R. Arvan, M. Von Herrath, D.S. Markel, C.J. Rhodes, How does
specific Tregs while reducing the pathogenic responses and reinstating type 1 diabetes develop?: the notion of homicide or β-cell suicide revisited,
a homeostatic balance for long term tolerance induction, it will raise Diabetes 60 (2011) 1370–1379, https://doi.org/10.2337/db10-1797.
hopes of the treatment for T1DM. Islet-specific expression as well as its [16] G.P. Lennon, M. Bettini, A.R. Burton, E. Vincent, P.Y. Arnold, P. Santamaria, D.
A. Vignali, T cell islet accumulation in type 1 diabetes is a tightly regulated, cell-
accessibility on the cell surface may provide new ideas for ZnT8 as an autonomous event, Immunity 31 (2009) 643–653, https://doi.org/10.1016/j.
immunotherapy target to improve the dysfunction of beta cells in T1DM. immuni.2009.07.008.
[17] D.E. Sutherland, R. Sibley, X.Z. Xu, A. Michael, A.M. Srikanta, F. Taub,
J. Najarian, F.C. Goetz, Twin-to-twin pancreas transplantation: reversal and
reenactment of the pathogenesis of type I diabetes, Trans. Assoc. Am. Physicians
97 (1984) 80–87.

7
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

[18] V.B. Gibson, T. Nikolic, V.Q. Pearce, J. Demengeot, B.O. Roep, M. Peakman, promote the development of type 1 diabetes, Mol. Med 28 (2022) 18, https://doi.
Proinsulin multi-peptide immunotherapy induces antigen-specific regulatory T org/10.1186/s10020-022-00447-y.
cells and limits autoimmunity in a humanized model, Clin. Exp. Immunol. 182 [39] G. Fousteri, E. Ippolito, R. Ahmed, A.R.A. Hamad, Beta-cell specific
(2015) 251–260, https://doi.org/10.1111/cei.12687. autoantibodies: are they just an indicator of type 1 diabetes? Curr. Diabetes Rev.
[19] B.O. Roep, N. Solvason, P.A. Gottlieb, J.R.F. Abreu, L.C. Harrison, G. 13 (2017) 322–329, https://doi.org/10.2174/1573399812666160427104157.
S. Eisenbarth, L. Yu, M. Leviten, W.A. Hagopian, J.B. Buse, M. von Herrath, [40] J. Boldison, F.S. Wong, Regulatory B cells: role in type 1 diabetes, Front.
J. Quan, R.S. King, W.H. Robinson, P.J. Utz, H. Garren, B.H.T. Investigators, Immunol. 12 (2021), 746187, https://doi.org/10.3389/fimmu.2021.746187.
L. Steinman, Plasmid-encoded proinsulin preserves C-peptide while specifically [41] J.A. Bluestone, J.H. Buckner, K.C. Herold, Immunotherapy: Building a bridge to a
reducing proinsulin-specific CD8(+) T cells in type 1 diabetes, Sci. Transl. Med 5 cure for type 1 diabetes, Sci. N. Y. 373 (2021) 510–516, https://doi.org/10.1126/
(2013) 191ra82, https://doi.org/10.1126/scitranslmed.3006103. science.abh1654.
[20] S.A. Greeley, M. Katsumata, L. Yu, G.S. Eisenbarth, D.J. Moore, H. Goodarzi, C. [42] M.D. Pescovitz, C.J. Greenbaum, B. Bundy, D.J. Becker, S.E. Gitelman, R. Goland,
F. Barker, A. Naji, H. Noorchashm, Elimination of maternally transmitted P.A. Gottlieb, J.B. Marks, A. Moran, P. Raskin, H. Rodriguez, D.A. Schatz, D.
autoantibodies prevents diabetes in nonobese diabetic mice, Nat. Med. 8 (2002) K. Wherrett, D.M. Wilson, J.P. Krischer, J.S. Skyler, B-lymphocyte depletion with
399–402, https://doi.org/10.1038/nm0402-399. rituximab and β-cell function: two-year results, Diabetes Care 37 (2014) 453–459,
[21] T. Orban, J.M. Sosenko, D. Cuthbertson, J.P. Krischer, J.S. Skyler, R. Jackson, https://doi.org/10.2337/dc13-0626.
L. Yu, J.P. Palmer, D. Schatz, G. Eisenbarth, Pancreatic islet autoantibodies as [43] P. Serra, P. Santamaria, Peptide-MHC-based nanomedicines for the treatment of
predictors of type 1 diabetes in the Diabetes Prevention Trial-Type 1, Diabetes autoimmunity: engineering, mechanisms, and diseases, Front. Immunol. 11
Care 32 (2009) 2269–2274, https://doi.org/10.2337/dc09-0934. (2020), 621774, https://doi.org/10.3389/fimmu.2020.621774.
[22] A.G. Ziegler, M. Rewers, O. Simell, T. Simell, J. Lempainen, A. Steck, C. Winkler, [44] M. Dul, T. Nikolic, M. Stefanidou, M.A. McAteer, P. Williams, J. Mous, B.O. Roep,
J. Ilonen, R. Veijola, M. Knip, E. Bonifacio, G.S. Eisenbarth, Seroconversion to E. Kochba, Y. Levin, M. Peakman, F.S. Wong, C.M. Dayan, D. Tatovic, S.
multiple islet autoantibodies and risk of progression to diabetes in children, A. Coulman, J.C. Birchall, Conjugation of a peptide autoantigen to gold
JAMA 309 (2013) 2473–2479, https://doi.org/10.1001/jama.2013.6285. nanoparticles for intradermally administered antigen specific immunotherapy,
[23] P.J. Bingley, Clinical applications of diabetes antibody testing, J. Clin. Int J. Pharm. 562 (2019) 303–312, https://doi.org/10.1016/j.
Endocrinol. Metab. 95 (2010) 25–33, https://doi.org/10.1210/jc.2009-1365. ijpharm.2019.03.041.
[24] J.A. Bluestone, M. Anderson, Tolerance in the Age of Immunotherapy, N. Engl. J. [45] J.S. Lewis, J.M. Stewart, G.P. Marshall, M.R. Carstens, Y. Zhang, N.V. Dolgova,
Med 383 (2020) 1156–1166, https://doi.org/10.1056/NEJMra1911109. C. Xia, T.M. Brusko, C.H. Wasserfall, M.J. Clare-Salzler, M.A. Atkinson, B.
[25] R.A. Insel, J.L. Dunne, M.A. Atkinson, J.L. Chiang, D. Dabelea, P.A. Gottlieb, C. G. Keselowsky, Dual-sized microparticle system for generating suppressive
J. Greenbaum, K.C. Herold, J.P. Krischer, Å. Lernmark, R.E. Ratner, M.J. Rewers, dendritic cells prevents and reverses type 1 diabetes in the nonobese diabetic
D.A. Schatz, J.S. Skyler, J.M. Sosenko, A.G. Ziegler, Staging presymptomatic type mouse model, ACS Biomater. Sci. Eng. 5 (2019) 2631–2646, https://doi.org/
1 diabetes: a scientific statement of JDRF, the Endocrine Society, and the 10.1021/acsbiomaterials.9b00332.
American Diabetes Association, Diabetes Care 38 (2015) 1964–1974, https://doi. [46] R.K. Singh, C. Malosse, J. Davies, B. Malissen, E. Kochba, Y. Levin, J.C. Birchall, S.
org/10.2337/dc15-1419. A. Coulman, J. Mous, M.A. McAteer, C.M. Dayan, S. Henri, F.S. Wong, Using gold
[26] F. Pociot, Å. Lernmark, Genetic risk factors for type 1 diabetes, Lancet 387 (2016) nanoparticles for enhanced intradermal delivery of poorly soluble auto-antigenic
2331–2339, https://doi.org/10.1016/s0140-6736(16)30582-7. peptides, Nanomedicine 32 (2021), 102321, https://doi.org/10.1016/j.
[27] M. Campbell-Thompson, A. Fu, J.S. Kaddis, C. Wasserfall, D.A. Schatz, nano.2020.102321.
A. Pugliese, M.A. Atkinson, Insulitis and β-cell mass in the natural history of type [47] N. Pishesha, A.M. Bilate, M.C. Wibowo, N.-J. Huang, Z. Li, R. Deshycka,
1 diabetes, Diabetes 65 (2016) 719–731, https://doi.org/10.2337/db15-0779. D. Bousbaine, H. Li, H.C. Patterson, S.K. Dougan, T. Maruyama, H.F. Lodish, H.
[28] V. Lampasona, D. Liberati, Islet autoantibodies, Curr. Diab Rep. 16 (2016) 53, L. Ploegh, Engineered erythrocytes covalently linked to antigenic peptides can
https://doi.org/10.1007/s11892-016-0738-2. protect against autoimmune disease, Proc. Natl. Acad. Sci. USA 114 (2017)
[29] T.L. van Belle, K.T. Coppieters, M.G. von Herrath, Type 1 diabetes: etiology, 3157–3162, https://doi.org/10.1073/pnas.1701746114.
immunology, and therapeutic strategies, Physiol. Rev. 91 (2011) 79–118, https:// [48] P.M. Glassman, E.D. Hood, L.T. Ferguson, Z. Zhao, D.L. Siegel, S. Mitragotri, J.
doi.org/10.1152/physrev.00003.2010. S. Brenner, V.R. Muzykantov, Red blood cells: the metamorphosis of a neglected
[30] A. Lehuen, J. Diana, P. Zaccone, A. Cooke, Immune cell crosstalk in type 1 carrier into the natural mothership for artificial nanocarriers, Adv. Drug Deliv.
diabetes, Nat. Rev. Immunol. 10 (2010) 501–513, https://doi.org/10.1038/ Rev. 178 (2021), 113992, https://doi.org/10.1016/j.addr.2021.113992.
nri2787. [49] C.D. Moorman, S.J. Sohn, H. Phee, Emerging therapeutics for immune tolerance:
[31] K.C. Herold, B.N. Bundy, J.P. Krischer, Type 1 diabetes TrialNet study G, tolerogenic vaccines, T cell therapy, and IL-2 therapy, Front. Immunol. 12 (2021),
Teplizumab in relatives at risk for type 1 diabetes, Reply N. Engl. J. Med 381 657768, https://doi.org/10.3389/fimmu.2021.657768.
(2019) 1880–1881, https://doi.org/10.1056/NEJMc1912500. [50] R.M. Pearson, L.M. Casey, K.R. Hughes, L.Z. Wang, M.G. North, D.R. Getts, S.
[32] E.K. Sims, B.N. Bundy, K. Stier, E. Serti, N. Lim, S.A. Long, S.M. Geyer, A. Moran, D. Miller, L.D. Shea, Controlled delivery of single or multiple antigens in
C.J. Greenbaum, C. Evans-Molina, K.C. Herold, Type 1 diabetes TrialNet Study G, tolerogenic nanoparticles using peptide-polymer bioconjugates, Mol. Ther. 25
Teplizumab improves and stabilizes beta cell function in antibody-positive high- (2017) 1655–1664, https://doi.org/10.1016/j.ymthe.2017.04.015.
risk individuals, Sci. Transl. Med 13 (2021), https://doi.org/10.1126/ [51] G. Sarikonda, S. Sachithanantham, Y. Manenkova, T. Kupfer, A. Posgai,
scitranslmed.abc8980. C. Wasserfall, P. Bernstein, L. Straub, P.P. Pagni, D. Schneider, T.R. Calvo,
[33] M.J. Haller, D.A. Schatz, J.S. Skyler, J.P. Krischer, B.N. Bundy, J.L. Miller, M. M. Coulombe, K. Herold, R.G. Gill, M. Atkinson, G. Nepom, M. Ehlers, T. Staeva,
A. Atkinson, D.J. Becker, D. Baidal, L.A. DiMeglio, S.E. Gitelman, R. Goland, P. H. Garren, L. Steinman, A.C. Chan, M. von Herrath, Transient B-cell depletion
A. Gottlieb, K.C. Herold, J.B. Marks, A. Moran, H. Rodriguez, W. Russell, D. with anti-CD20 in combination with proinsulin DNA vaccine or oral insulin:
M. Wilson, C.J. Greenbaum, Type 1 diabetes TrialNet ATGGSG, low-dose anti- immunologic effects and efficacy in NOD mice, PloS One 8 (2013), e54712,
thymocyte globulin (ATG) preserves beta-cell function and improves HbA1c in https://doi.org/10.1371/journal.pone.0054712.
new-onset type 1 diabetes, Diabetes Care 41 (2018) 1917–1925, https://doi.org/ [52] C. Hu, H. Ding, X. Zhang, F.S. Wong, L. Wen, Combination treatment with anti-
10.2337/dc18-0494. CD20 and oral anti-CD3 prevents and reverses autoimmune diabetes, Diabetes 62
[34] M.J. Haller, D.A. Schatz, J.S. Skyler, J.P. Krischer, B.N. Bundy, J.L. Miller, M. (2013) 2849–2858, https://doi.org/10.2337/db12-1175.
A. Atkinson, D.J. Becker, D. Baidal, L.A. DiMeglio, S.E. Gitelman, R. Goland, P. [53] M.J. Smith, J.C. Cambier, P.A. Gottlieb, Endotypes in T1D: B lymphocytes and
A. Gottlieb, K.C. Herold, J.B. Marks, A. Moran, H. Rodriguez, W. Russell, D. early onset, Curr. Opin. Endocrinol., Diabetes, Obes. 27 (2020) 225–230, https://
M. Wilson, C.J. Greenbaum, A.T.G. Low-Dose Anti-Thymocyte Globulin, doi.org/10.1097/MED.0000000000000547.
Preserves β-cell function and improves HbA in new-onset type 1 diabetes, [54] M. Zieliński, M. Żalińska, D. Iwaszkiewicz-Grześ, M. Gliwiński, M. Hennig,
Diabetes Care 41 (2018) 1917–1925, https://doi.org/10.2337/dc18-0494. A. Jaźwińska-Curyłło, H. Kamińska, J. Sakowska, A. Wołoszyn-Durkiewicz,
[35] N.M. Edner, F. Heuts, N. Thomas, C.J. Wang, L. Petersone, R. Kenefeck, R. Owczuk, W. Młynarski, P. Jarosz-Chobot, A. Bossowski, A. Szadkowska,
A. Kogimtzis, V. Ovcinnikovs, E.M. Ross, E. Ntavli, Y. Elfaki, M. Eichmann, J. Siebert, M. Myśliwiec, N. Marek-Trzonkowska, P. Trzonkowski, Combined
R. Baptista, P. Ambery, L. Jermutus, M. Peakman, M. Rosenthal, L.S.K. Walker, therapy with CD4 CD25highCD127 T regulatory cells and anti-CD20 antibody in
Follicular helper T cell profiles predict response to costimulation blockade in type recent-onset type 1 diabetes is superior to monotherapy: Randomized phase I/II
1 diabetes, Nat. Immunol. 21 (2020) 1244–1255, https://doi.org/10.1038/ trial, Diabetes Obes. Metab. (2022), https://doi.org/10.1111/dom.14723.
s41590-020-0744-z. [55] S. Wu, L. Wang, Y. Fang, H. Huang, X. You, J. Wu, Advances in encapsulation and
[36] M.R. Rigby, K.M. Harris, A. Pinckney, L.A. DiMeglio, M.S. Rendell, E.I. Felner, J. delivery strategies for islet transplantation, Adv. Health Mater. 10 (2021),
M. Dostou, S.E. Gitelman, K.J. Griffin, E. Tsalikian, P.A. Gottlieb, C. e2100965, https://doi.org/10.1002/adhm.202100965.
J. Greenbaum, N.A. Sherry, W.V. Moore, R. Monzavi, S.M. Willi, P. Raskin, [56] Q. Zhang, C. Gonelle-Gispert, Y. Li, Z. Geng, S. Gerber-Lemaire, Y. Wang,
L. Keyes-Elstein, S.A. Long, S. Kanaparthi, N. Lim, D. Phippard, C.L. Soppe, M. L. Buhler, Islet encapsulation: new developments for the treatment of type 1
L. Fitzgibbon, J. McNamara, G.T. Nepom, M.R. Ehlers, Alefacept provides diabetes, Front. Immunol. 13 (2022), 869984, https://doi.org/10.3389/
sustained clinical and immunological effects in new-onset type 1 diabetes fimmu.2022.869984.
patients, J. Clin. Invest 125 (2015) 3285–3296, https://doi.org/10.1172/ [57] A. Opara, A. Jost, S. Dagogo-Jack, E.C. Opara, Islet cell encapsulation -
JCI81722. application in diabetes treatment, Exp. Biol. Med. 246 (2021) 2570–2578,
[37] Y. Xiao, C. Deng, Z. Zhou, The Multiple Roles of B Lymphocytes in the onset and https://doi.org/10.1177/15353702211040503.
treatment of type 1 diabetes: interactions between B Lymphocytes and T cells, [58] M.M. Samojlik, C.L. Stabler, Designing biomaterials for the modulation of
J. Diabetes Res. 2021 (2021) 6581213, https://doi.org/10.1155/2021/6581213. allogeneic and autoimmune responses to cellular implants in Type 1 Diabetes,
[38] Q. Ling, L. Shen, W. Zhang, D. Qu, H. Wang, B. Wang, Y. Liu, J. Lu, D. Zhu, Y. Bi, Acta Biomater. 133 (2021), https://doi.org/10.1016/j.actbio.2021.05.039.
Increased plasmablasts enhance T cell-mediated beta cell destruction and

8
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

[59] M.-C. Vantyghem, E.J.P. de Koning, F. Pattou, M.R. Rickels, Advances in β-cell [84] S.A. Tersey, Y. Nishiki, A.T. Templin, S.M. Cabrera, N.D. Stull, S.C. Colvin,
replacement therapy for the treatment of type 1 diabetes, Lancet 394 (2019) C. Evans-Molina, J.L. Rickus, B. Maier, R.G. Mirmira, Islet β-cell endoplasmic
1274–1285, https://doi.org/10.1016/S0140-6736(19)31334-0. reticulum stress precedes the onset of type 1 diabetes in the nonobese diabetic
[60] T. Desai, L.D. Shea, Advances in islet encapsulation technologies, Nat. Rev. Drug mouse model, Diabetes 61 (2012) 818–827, https://doi.org/10.2337/db11-1293.
Discov. 16 (2017) 338–350, https://doi.org/10.1038/nrd.2016.232. [85] C. Scheinecker, R. McHugh, E.M. Shevach, R.N. Germain, Constitutive
[61] R.A. Valdés-González, L.M. Dorantes, G.N. Garibay, E. Bracho-Blanchet, A. presentation of a natural tissue autoantigen exclusively by dendritic cells in the
J. Mendez, R. Dávila-Pérez, R.B. Elliott, L. Terán, D.J.G. White, draining lymph node, J. Exp. Med. 196 (2002) 1079–1090, https://doi.org/
Xenotransplantation of porcine neonatal islets of Langerhans and Sertoli cells: a 10.1084/jem.20020991.
4-year study, Eur. J. Endocrinol. 153 (2005) 419–427. [86] R.N. Bone, C. Evans-Molina, Combination immunotherapy for Type 1 diabetes,
[62] R. Valdes-Gonzalez, A.L. Rodriguez-Ventura, D.J.G. White, E. Bracho-Blanchet, Curr. Diab Rep. 17 (2017) 50, https://doi.org/10.1007/s11892-017-0878-z.
A. Castillo, B. Ramírez-González, M.G. López-Santos, B.H. León-Mancilla, L. [87] M.J. Haller, S.A. Long, J.L. Blanchfield, D.A. Schatz, J.S. Skyler, J.P. Krischer, B.
M. Dorantes, Long-term follow-up of patients with type 1 diabetes transplanted N. Bundy, S.M. Geyer, M.V. Warnock, J.L. Miller, M.A. Atkinson, D.J. Becker, D.
with neonatal pig islets, Clin. Exp. Immunol. 162 (2010) 537–542, https://doi. A. Baidal, L.A. DiMeglio, S.E. Gitelman, R. Goland, P.A. Gottlieb, K.C. Herold, J.
org/10.1111/j.1365-2249.2010.04273.x. B. Marks, A. Moran, H. Rodriguez, W.E. Russell, D.M. Wilson, C.J. Greenbaum,
[63] B.E. Tuch, G.W. Keogh, L.J. Williams, W. Wu, J.L. Foster, V. Vaithilingam, Low-dose anti-thymocyte globulin preserves c-peptide, reduces HbA(1c), and
R. Philips, Safety and viability of microencapsulated human islets transplanted increases regulatory to conventional T-cell ratios in new-onset type 1, Diabetes:
into diabetic humans, Diabetes Care 32 (2009) 1887–1889, https://doi.org/ Two-Year Clin. Trial Data Diabetes 68 (2019) 1267–1276, https://doi.org/
10.2337/dc09-0744. 10.2337/db19-0057.
[64] R. Cornolti, M. Figliuzzi, A. Remuzzi, Effect of micro- and macroencapsulation on [88] C.T. Ellebrecht, V.G. Bhoj, A. Nace, E.J. Choi, X. Mao, M.J. Cho, G. Di Zenzo,
oxygen consumption by pancreatic islets, Cell Transpl. 18 (2009) 195–201. A. Lanzavecchia, J.T. Seykora, G. Cotsarelis, M.C. Milone, A.S. Payne,
[65] R. Valdes-Gonzalez, A.L. Rodriguez-Ventura, D.J. White, E. Bracho-Blanchet, Reengineering chimeric antigen receptor T cells for targeted therapy of
A. Castillo, B. Ramirez-Gonzalez, M.G. Lopez-Santos, B.H. Leon-Mancilla, L. autoimmune disease, Science 353 (2016) 179–184, https://doi.org/10.1126/
M. Dorantes, Long-term follow-up of patients with type 1 diabetes transplanted science.aaf6756.
with neonatal pig islets, Clin. Exp. Immunol. 162 (2010) 537–542, https://doi. [89] G. Flórez-Grau, I. Zubizarreta, R. Cabezón, P. Villoslada, D. Benitez-Ribas,
org/10.1111/j.1365-2249.2010.04273.x. Tolerogenic dendritic cells as a promising antigen-specific therapy in the
[66] Y.X. Wu, S.H. Jin, J. Cui, Autophagy and immune tolerance, Adv. Exp. Med. Biol. treatment of multiple sclerosis and neuromyelitis optica from preclinical to
1206 (2019) 635–665, https://doi.org/10.1007/978-981-15-0602-4_28. clinical trials, Front Immunol. 9 (2018) 1169, https://doi.org/10.3389/
[67] L. Wang, F.S. Wang, M.E. Gershwin, Human autoimmune diseases: a fimmu.2018.01169.
comprehensive update, J. Intern Med 278 (2015) 369–395, https://doi.org/ [90] J. Koreth, H.T. Kim, K.T. Jones, P.B. Lange, C.G. Reynolds, M.J. Chammas,
10.1111/joim.12395. K. Dusenbury, J. Whangbo, S. Nikiforow, E.P. Alyea 3rd, P. Armand, C.S. Cutler,
[68] F. Rayner, J.D. Isaacs, Therapeutic tolerance in autoimmune disease, Semin V.T. Ho, Y.B. Chen, D. Avigan, B.R. Blazar, J.H. Antin, J. Ritz, R.J. Soiffer,
Arthritis Rheum. 48 (2018) 558–562, https://doi.org/10.1016/j. Efficacy, durability, and response predictors of low-dose interleukin-2 therapy for
semarthrit.2018.09.008. chronic graft-versus-host disease, Blood 128 (2016) 130–137, https://doi.org/
[69] Z.A. Radi, T.A. Wynn, Opinion on immune tolerance therapeutic development, 10.1182/blood-2016-02-702852.
Toxicol. Pathol. 48 (2020) 712–717, https://doi.org/10.1177/ [91] S. Gregori, M.G. Roncarolo, Engineered T regulatory type 1 cells for clinical
0192623320948805. application, Front Immunol. 9 (2018) 233, https://doi.org/10.3389/
[70] O. Alpdogan, M.R. van den Brink, Immune tolerance and transplantation, Semin. fimmu.2018.00233.
Oncol. 39 (2012) 629–642, https://doi.org/10.1053/j.seminoncol.2012.10.001. [92] Z. Mao, H. Lin, W. Su, J. Li, M. Zhou, Z. Li, B. Zhou, Q. Yang, M. Zhou, K. Pan,
[71] P. Vafiadis, S.T. Bennett, J.A. Todd, J. Nadeau, R. Grabs, C.G. Goodyer, J. He, W. Zhang, Deficiency of ZnT8 promotes adiposity and metabolic
S. Wickramasinghe, E. Colle, C. Polychronakos, Insulin expression in human dysfunction by increasing peripheral serotonin production, Diabetes 68 (2019)
thymus is modulated by INS VNTR alleles at the IDDM2 locus, Nat. Genet 15 1197–1209, https://doi.org/10.2337/db18-1321.
(1997) 289–292, https://doi.org/10.1038/ng0397-289. [93] A. Solomou, G. Meur, E. Bellomo, D.J. Hodson, A. Tomas, S.M. Li, E. Philippe, P.
[72] A.N. Theofilopoulos, D.H. Kono, R. Baccala, The multiple pathways to L. Herrera, C. Magnan, G.A. Rutter, The zinc transporter Slc30a8/ZnT8 Is
autoimmunity, Nat. Immunol. 18 (2017) 716–724, https://doi.org/10.1038/ required in a subpopulation of pancreatic α-cells for hypoglycemia-induced
ni.3731. glucagon secretion, J. Biol. Chem. 290 (2015) 21432–21442, https://doi.org/
[73] E.E. Hamilton-Williams, A.S. Bergot, P.L. Reeves, R.J. Steptoe, Maintenance of 10.1074/jbc.M115.645291.
peripheral tolerance to islet antigens, J. Autoimmun. 72 (2016) 118–125, https:// [94] R. Lawson, W. Maret, C. Hogstrand, ZnT8 haploinsufficiency impacts MIN6 cell
doi.org/10.1016/j.jaut.2016.05.009. zinc content and β-Cell phenotype via ZIP-ZnT8 coregulation, Int. J. Mol. Sci. 20
[74] L. Göschl, C. Scheinecker, M. Bonelli, Treg cells in autoimmunity: from (2019), https://doi.org/10.3390/ijms20215485.
identification to Treg-based therapies, Semin Immunopathol. 41 (2019) 301–314, [95] D.K. Nayak, B. Calderon, A.N. Vomund, E.R. Unanue, ZnT8-reactive T cells are
https://doi.org/10.1007/s00281-019-00741-8. weakly pathogenic in NOD mice but can participate in diabetes under
[75] P.A. Savage, D.E.J. Klawon, C.H. Miller, Regulatory T cell development, Annu inflammatory conditions, Diabetes 63 (2014) 3438–3448, https://doi.org/
Rev. Immunol. 38 (2020) 421–453, https://doi.org/10.1146/annurev-immunol- 10.2337/db13-1882.
100219-020937. [96] P. Achenbach, V. Lampasona, U. Landherr, K. Koczwara, S. Krause, H. Grallert,
[76] M. Feuerer, J.A. Hill, D. Mathis, C. Benoist, Foxp3+ regulatory T cells: C. Winkler, M. Pflüger, T. Illig, E. Bonifacio, A.G. Ziegler, Autoantibodies to zinc
differentiation, specification, subphenotypes, Nat. Immunol. 10 (2009) 689–695, transporter 8 and SLC30A8 genotype stratify type 1 diabetes risk, Diabetologia 52
https://doi.org/10.1038/ni.1760. (2009) 1881–1888, https://doi.org/10.1007/s00125-009-1438-0.
[77] L.M.R. Ferreira, Y.D. Muller, J.A. Bluestone, Q. Tang, Next-generation regulatory [97] R. Thirunavukkarasu, A.J. Asirvatham, A. Chitra, M. Jayalakshmi, SLC30A8 gene
T cell therapy, Nat. Rev. Drug Disco 18 (2019) 749–769, https://doi.org/ rs13266634C/T polymorphism in children with type 1 diabetes in Tamil Nadu,
10.1038/s41573-019-0041-4. India, J. Clin. Res. Pedia Endocrinol. 11 (2019) 55–60, https://doi.org/10.4274/
[78] M.G. Roncarolo, S. Gregori, R. Bacchetta, M. Battaglia, Tr1 cells and the counter- jcrpe.galenos.2018.2018.0195.
regulation of immunity: natural mechanisms and therapeutic applications, Curr. [98] F.M. Ashcroft, P. Rorsman, Diabetes mellitus and the β cell: the last ten years, Cell
Top. Microbiol Immunol. 380 (2014) 39–68, https://doi.org/10.1007/978-3-662- 148 (2012) 1160–1171, https://doi.org/10.1016/j.cell.2012.02.010.
43492-5_3. [99] A. Fukunaka, Y. Fujitani, Role of zinc homeostasis in the pathogenesis of diabetes
[79] S. Sakaguchi, M. Ono, R. Setoguchi, H. Yagi, S. Hori, Z. Fehervari, J. Shimizu, and obesity, Int. J. Mol. Sci. 19 (2018), https://doi.org/10.3390/ijms19020476.
T. Takahashi, T. Nomura, Foxp3+ CD25+ CD4+ natural regulatory T cells in [100] S. Kleiner, D. Gomez, B. Megra, E. Na, R. Bhavsar, K. Cavino, Y. Xin, J. Rojas,
dominant self-tolerance and autoimmune disease, Immunol. Rev. 212 (2006) G. Dominguez-Gutierrez, B. Zambrowicz, G. Carrat, P. Chabosseau, M. Hu, A.
8–27, https://doi.org/10.1111/j.0105-2896.2006.00427.x. J. Murphy, G.D. Yancopoulos, G.A. Rutter, J. Gromada, Mice harboring the
[80] M. Ono, J. Shimizu, Y. Miyachi, S. Sakaguchi, Control of autoimmune myocarditis human SLC30A8 R138X loss-of-function mutation have increased insulin
and multiorgan inflammation by glucocorticoid-induced TNF receptor family- secretory capacity, Proc. Natl. Acad. Sci. USA 115 (2018) E7642–e9, https://doi.
related protein(high), Foxp3-expressing CD25+ and CD25- regulatory T cells, org/10.1073/pnas.1721418115.
J. Immunol. 176 (2006) 4748–4756, https://doi.org/10.4049/ [101] É. Énée, R. Kratzer, J.B. Arnoux, E. Barilleau, Y. Hamel, C. Marchi, J. Beltrand,
jimmunol.176.8.4748. B. Michaud, L. Chatenoud, J.J. Robert, P. van Endert, ZnT8 is a major CD8+ T
[81] R. Gianani, G.S. Eisenbarth, The stages of type 1A diabetes: 2005, Immunol. Rev. cell-recognized autoantigen in pediatric type 1, Diabetes Diabetes 61 (2012)
204 (2005) 232–249, https://doi.org/10.1111/j.0105-2896.2005.00248.x. 1779–1784, https://doi.org/10.2337/db12-0071.
[82] E. Gambineri, T.R. Torgerson, H.D. Ochs, Immune dysregulation, [102] J.M. Wenzlau, J.C. Hutton, H.W. Davidson, New antigenic targets in type 1
polyendocrinopathy, enteropathy, and X-linked inheritance (IPEX), a syndrome diabetes, Curr. Opin. Endocrinol. Diabetes Obes. 15 (2008) 315–320, https://doi.
of systemic autoimmunity caused by mutations of FOXP3, a critical regulator of T- org/10.1097/MED.0b013e328308192b.
cell homeostasis, Curr. Opin. Rheuma 15 (2003) 430–435, https://doi.org/ [103] Q. Huang, C. Merriman, H. Zhang, D. Fu, Coupling of insulin secretion and display
10.1097/00002281-200307000-00010. of a granule-resident zinc transporter ZnT8 on the surface of pancreatic beta cells,
[83] K.V. Tarbell, M. Lee, E. Ranheim, C.C. Chao, M. Sanna, S.K. Kim, P. Dickie, J. Biol. Chem. 292 (2017) 4034–4043, https://doi.org/10.1074/jbc.
L. Teyton, M. Davis, H. McDevitt, CD4(+) T cells from glutamic acid M116.772152.
decarboxylase (GAD)65-specific T cell receptor transgenic mice are not [104] C. Merriman, Q. Huang, W. Gu, L. Yu, D. Fu, A subclass of serum anti-ZnT8
diabetogenic and can delay diabetes transfer, J. Exp. Med 196 (2002) 481–492, antibodies directed to the surface of live pancreatic β-cells, J. Biol. Chem. 293
https://doi.org/10.1084/jem.20011845. (2018) 579–587, https://doi.org/10.1074/jbc.RA117.000195.

9
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

[105] A.W. Michels, M. Nakayama, The anti-insulin trimolecular complex in type 1 in the diagnosis of autoimmune diabetes in a large French cohort, Clin. Chim.
diabetes, Curr. Opin. Endocrinol. Diabetes Obes. 17 (2010) 329–334, https://doi. Acta 478 (2018) 162–165, https://doi.org/10.1016/j.cca.2017.12.043.
org/10.1097/MED.0b013e32833aba41. [126] H. Hussein, F. Ibrahim, E. Sobngwi, J.F. Gautier, P. Boudou, Zinc transporter 8
[106] A. Rogowicz-Frontczak, S. Pilacinski, K. Wyka, B. Wierusz-Wysocka, autoantibodies assessment in daily practice, Clin. Biochem 50 (2017) 94–96,
D. Zozulinska-Ziolkiewicz, Zinc transporter 8 autoantibodies (ZnT8-ab) are https://doi.org/10.1016/j.clinbiochem.2016.06.008.
associated with higher prevalence of multiple diabetes-related autoantibodies in [127] T. Kimpimäki, A. Kupila, A.M. Hämäläinen, M. Kukko, P. Kulmala, K. Savola,
adults with type 1 diabetes, Diabetes Res Clin. Pr. 146 (2018) 313–320, https:// T. Simell, P. Keskinen, J. Ilonen, O. Simell, M. Knip, The first signs of beta-cell
doi.org/10.1016/j.diabres.2018.11.007. autoimmunity appear in infancy in genetically susceptible children from the
[107] E.P. Sørgjerd, F. Skorpen, K. Kvaløy, K. Midthjell, V. Grill, Time dynamics of general population: the Finnish Type 1 diabetes prediction and prevention study,
autoantibodies are coupled to phenotypes and add to the heterogeneity of J. Clin. Endocrinol. Metab. 86 (2001) 4782–4788, https://doi.org/10.1210/
autoimmune diabetes in adults: the HUNT study, Norway, Diabetologia 55 (2012) jcem.86.10.7907.
1310–1318, https://doi.org/10.1007/s00125-012-2463-y. [128] P.M. Pöllänen, S.J. Ryhänen, J. Toppari, J. Ilonen, P. Vähäsalo, R. Veijola,
[108] M. Knip, S. Korhonen, P. Kulmala, R. Veijola, A. Reunanen, O.T. Raitakari, H. Siljander, M. Knip, Dynamics of islet autoantibodies during prospective follow-
J. Viikari, H.K. Akerblom, Prediction of type 1 diabetes in the general population, up from birth to age 15 years, J. Clin. Endocrinol. Metab. 105 (2020)
Diabetes Care 33 (2010) 1206–1212, https://doi.org/10.2337/dc09-1040. e4638–e4651, https://doi.org/10.1210/clinem/dgaa624.
[109] J.M. Howson, S. Krause, H. Stevens, D.J. Smyth, J.M. Wenzlau, E. Bonifacio, [129] K. Vehik, E. Bonifacio, Å. Lernmark, L. Yu, A. Williams, D. Schatz, M. Rewers, J.
J. Hutton, A.G. Ziegler, J.A. Todd, P. Achenbach, Genetic association of zinc X. She, J. Toppari, W. Hagopian, B. Akolkar, A.G. Ziegler, J.P. Krischer,
transporter 8 (ZnT8) autoantibodies in type 1 diabetes cases, Diabetologia 55 Hierarchical order of distinct autoantibody spreading and progression to Type 1
(2012) 1978–1984, https://doi.org/10.1007/s00125-012-2540-2. diabetes in the TEDDY study, Diabetes Care 43 (2020) 2066–2073, https://doi.
[110] D.S. Parsons, C. Hogstrand, W. Maret, The C-terminal cytosolic domain of the org/10.2337/dc19-2547.
human zinc transporter ZnT8 and its diabetes risk variant, Febs J. 285 (2018) [130] A.K. Steck, F. Dong, K. Waugh, B.I. Frohnert, L. Yu, J.M. Norris, M.J. Rewers,
1237–1250, https://doi.org/10.1111/febs.14402. Predictors of slow progression to diabetes in children with multiple islet
[111] T. Kambe, Molecular architecture and function of ZnT transporters, Curr. Top. autoantibodies, J. Autoimmun. 72 (2016) 113–117, https://doi.org/10.1016/j.
Membr. 69 (2012) 199–220, https://doi.org/10.1016/b978-0-12-394390- jaut.2016.05.010.
3.00008-2. [131] F. Vaziri-Sani, S. Oak, J. Radtke, K. Lernmark, K. Lynch, C.D. Agardh, C.M. Cilio,
[112] J.M. Wenzlau, Y. Liu, L. Yu, O. Moua, K.T. Fowler, S. Rangasamy, J. Walters, G. A.L. Lethagen, E. Ortqvist, M. Landin-Olsson, C. Törn, C.S. Hampe, ZnT8
S. Eisenbarth, H.W. Davidson, J.C. Hutton, A common nonsynonymous single autoantibody titers in type 1 diabetes patients decline rapidly after clinical onset,
nucleotide polymorphism in the SLC30A8 gene determines ZnT8 autoantibody Autoimmunity 43 (2010) 598–606, https://doi.org/10.3109/
specificity in type 1 diabetes, Diabetes 57 (2008) 2693–2697, https://doi.org/ 08916930903555927.
10.2337/db08-0522. [132] R.G. Miller, L. Yu, D.J. Becker, T.J. Orchard, T. Costacou, Older age of childhood
[113] L. Yu, D.C. Boulware, C.A. Beam, J.C. Hutton, J.M. Wenzlau, C.J. Greenbaum, P. type 1 diabetes onset is associated with islet autoantibody positivity >30 years
J. Bingley, J.P. Krischer, J.M. Sosenko, J.S. Skyler, G.S. Eisenbarth, J.L. Mahon, later: the Pittsburgh epidemiology of diabetes complications study, Diabet. Med.:
Zinc transporter-8 autoantibodies improve prediction of type 1 diabetes in a J. Br. Diabet. Assoc. 37 (2020) 1386–1394, https://doi.org/10.1111/
relatives positive for the standard biochemical autoantibodies, Diabetes Care 35 dme.14261.
(2012) 1213–1218, https://doi.org/10.2337/dc11-2081. [133] M. Knip, S.M. Virtanen, K. Seppä, J. Ilonen, E. Savilahti, O. Vaarala, A. Reunanen,
[114] A. Lounici Boudiaf, D. Bouziane, M. Smara, Y. Meddour, E.M. Haffaf, B. Oudjit, K. Teramo, A.M. Hämäläinen, J. Paronen, H.M. Dosch, T. Hakulinen, H.
S. Chaib Mamouzi, S. Aouichat Bouguerra, Could ZnT8 antibodies replace ICA, K. Akerblom, Dietary intervention in infancy and later signs of beta-cell
GAD, IA2 and insulin antibodies in the diagnosis of type 1 diabetes? Curr. Res autoimmunity, N. Engl. J. Med 363 (2010) 1900–1908, https://doi.org/10.1056/
Transl. Med 66 (2018) 1–7, https://doi.org/10.1016/j.retram.2018.01.003. NEJMoa1004809.
[115] S. Han, W. Donelan, H. Wang, W. Reeves, L.J. Yang, Novel autoantigens in type 1 [134] O. Vaarala, J. Ilonen, T. Ruohtula, J. Pesola, S.M. Virtanen, T. Härkönen,
diabetes, Am. J. Transl. Res 5 (2013) 379–392. M. Koski, H. Kallioinen, O. Tossavainen, T. Poussa, A.L. Järvenpää,
[116] F.K. Gorus, E.V. Balti, I. Vermeulen, S. Demeester, A. Van Dalem, O. Costa, J. Komulainen, R. Lounamaa, H.K. Akerblom, M. Knip, Removal of Bovine insulin
H. Dorchy, S. Tenoutasse, T. Mouraux, C. De Block, P. Gillard, K. Decochez, J. from cow’s milk formula and early initiation of beta-cell autoimmunity in the
M. Wenzlau, J.C. Hutton, D.G. Pipeleers, I. Weets, Screening for insulinoma FINDIA pilot study, Arch. Pedia Adolesc. Med 166 (2012) 608–614, https://doi.
antigen 2 and zinc transporter 8 autoantibodies: a cost-effective and age- org/10.1001/archpediatrics.2011.1559.
independent strategy to identify rapid progressors to clinical onset among [135] J.S. Skyler, J.P. Krischer, J. Wolfsdorf, C. Cowie, J.P. Palmer, C. Greenbaum,
relatives of type 1 diabetic patients, Clin. Exp. Immunol. 171 (2013) 82–90, D. Cuthbertson, L.E. Rafkin-Mervis, H.P. Chase, E. Leschek, Effects of oral insulin
https://doi.org/10.1111/j.1365-2249.2012.04675.x. in relatives of patients with type 1 diabetes: the diabetes prevention trial–type 1,
[117] L. Yang, S. Luo, G. Huang, J. Peng, X. Li, X. Yan, J. Lin, J.M. Wenzlau, H. Diabetes Care 28 (2005) 1068–1076, https://doi.org/10.2337/
W. Davidson, J.C. Hutton, Z. Zhou, The diagnostic value of zinc transporter 8 diacare.28.5.1068.
autoantibody (ZnT8A) for type 1 diabetes in Chinese, Diabetes Metab. Res Rev. 26 [136] S. Pieralice, P. Pozzilli, Latent autoimmune diabetes in adults: a review on clinical
(2010) 579–584, https://doi.org/10.1002/dmrr.1128. implications and management, Diabetes Metab. J. 42 (2018) 451–464, https://
[118] C. Andersson, F. Vaziri-Sani, A. Delli, B. Lindblad, A. Carlsson, G. Forsander, doi.org/10.4093/dmj.2018.0190.
J. Ludvigsson, C. Marcus, U. Samuelsson, S. Ivarsson, A. Lernmark, H.E. Larsson, [137] S. Fourlanos, F. Dotta, C.J. Greenbaum, J.P. Palmer, O. Rolandsson, P.G. Colman,
Triple specificity of ZnT8 autoantibodies in relation to HLA and other islet L.C. Harrison, Latent autoimmune diabetes in adults (LADA) should be less latent,
autoantibodies in childhood and adolescent type 1 diabetes, Pedia Diabetes 14 Diabetologia 48 (2005) 2206–2212, https://doi.org/10.1007/s00125-005-1960-
(2013) 97–105, https://doi.org/10.1111/j.1399-5448.2012.00916.x. 7.
[119] R. Gianani, M. Campbell-Thompson, S.A. Sarkar, C. Wasserfall, A. Pugliese, J. [138] E.P. Sørgjerd, Type 1 diabetes-related autoantibodies in different forms of
M. Solis, S.C. Kent, B.J. Hering, E. West, A. Steck, S. Bonner-Weir, M.A. Atkinson, diabetes, Curr. Diabetes Rev. 15 (2019) 199–204, https://doi.org/10.2174/
K. Coppieters, M. von Herrath, G.S. Eisenbarth, Dimorphic histopathology of long- 1573399814666180730105351.
standing childhood-onset diabetes, Diabetologia 53 (2010) 690–698, https://doi. [139] G. Huang, Y. Xiang, L. Pan, X. Li, S. Luo, Z. Zhou, Zinc transporter 8 autoantibody
org/10.1007/s00125-009-1642-y. (ZnT8A) could help differentiate latent autoimmune diabetes in adults (LADA)
[120] A.V. Matveyenko, P.C. Butler, Relationship between beta-cell mass and diabetes from phenotypic type 2 diabetes mellitus, Diabetes Metab. Res Rev. 29 (2013)
onset, Diabetes Obes. Metab. 10 (Suppl 4) (2008) 23–31, https://doi.org/ 363–368, https://doi.org/10.1002/dmrr.2396.
10.1111/j.1463-1326.2008.00939.x. [140] K. Haller-Kikkatalo, K. Pruul, K. Kisand, V. Nemvalts, K. Reimand, R. Uibo, GADA
[121] S.V. Gearty, F. Dündar, P. Zumbo, G. Espinosa-Carrasco, M. Shakiba, F.J. Sanchez- and anti-ZnT8 complicate the outcome of phenotypic type 2 diabetes of adults,
Rivera, N.D. Socci, P. Trivedi, S.W. Lowe, P. Lauer, N. Mohibullah, A. Viale, T. Eur. J. Clin. Invest 45 (2015) 255–262, https://doi.org/10.1111/eci.12404.
P. DiLorenzo, D. Betel, A. Schietinger, An autoimmune stem-like CD8 T cell [141] E. Otto-Buczkowska, Type LADA diabetes–interrogation points, Przegl Lek. 70
population drives type 1 diabetes, Nature 602 (2022) 156–161, https://doi.org/ (2013) 25–27.
10.1038/s41586-021-04248-x. [142] G. Huang, M. Yin, Y. Xiang, X. Li, W. Shen, S. Luo, J. Lin, Z. Xie, P. Zheng,
[122] C.L. Williams, A.E. Long, What has zinc transporter 8 autoimmunity taught us Z. Zhou, Persistence of glutamic acid decarboxylase antibody (GADA) is
about type 1 diabetes, Diabetologia 62 (2019) 1969–1976, https://doi.org/ associated with clinical characteristics of latent autoimmune diabetes in adults: a
10.1007/s00125-019-04975-x. prospective study with 3-year follow-up, Diabetes Metab. Res Rev. 32 (2016)
[123] M. Juusola, A. Parkkola, T. Härkönen, H. Siljander, J. Ilonen, H.K. Åkerblom, 615–622, https://doi.org/10.1002/dmrr.2779.
M. Knip, Positivity for zinc transporter 8 autoantibodies at diagnosis is [143] K. Vehik, K.F. Lynch, D.A. Schatz, B. Akolkar, W. Hagopian, M. Rewers, J.X. She,
subsequently associated with reduced β-cell function and higher exogenous O. Simell, J. Toppari, A.G. Ziegler, Å. Lernmark, E. Bonifacio, J.P. Krischer,
insulin requirement in children and adolescents with type 1 diabetes, Diabetes Reversion of β-cell autoimmunity changes risk of type 1 diabetes: TEDDY study,
Care 39 (2016) 118–121, https://doi.org/10.2337/dc15-1027. Diabetes Care 39 (2016) 1535–1542, https://doi.org/10.2337/dc16-0181.
[124] J.P. Krischer, K.F. Lynch, D.A. Schatz, J. Ilonen, Å. Lernmark, W.A. Hagopian, M. [144] P. Chabosseau, G.A. Rutter, Zinc and diabetes, Arch. Biochem. Biophys. 611
J. Rewers, J.X. She, O.G. Simell, J. Toppari, A.G. Ziegler, B. Akolkar, E. Bonifacio, (2016) 79–85, https://doi.org/10.1016/j.abb.2016.05.022.
The 6 year incidence of diabetes-associated autoantibodies in genetically at-risk [145] M. Karsai, R.A. Zuellig, R. Lehmann, F. Cuozzo, D. Nasteska, E. Luca, C. Hantel, D.
children: the TEDDY study, Diabetologia 58 (2015) 980–987, https://doi.org/ J. Hodson, G.A. Spinas, G.A. Rutter, P.A. Gerber, Lack of ZnT8 protects pancreatic
10.1007/s00125-015-3514-y. islets from hypoxia- and cytokine-induced cell death, J. Endocrinol. 253 (2022),
[125] L. Garnier, L. Marchand, M. Benoit, M. Nicolino, N. Bendelac, C. Wright, https://doi.org/10.1530/JOE-21-0271.
P. Moulin, C. Lombard, C. Thivolet, N. Fabien, Screening of ZnT8 autoantibodies

10
X. Zhang et al. Journal of Trace Elements in Medicine and Biology 73 (2022) 127040

[146] Q. Huang, J. Du, C. Merriman, Z. Gong, Genetic, Functional, and Immunological A. Weiss, M. Hippich, K. Halfter, S.M. Hauck, J. Hasford, J.F. Petrosino,
Study of ZnT8 in Diabetes, Int. J. Endocrinol. 2019 (2019) 1524905, https://doi. P. Achenbach, E. Bonifacio, A.-G. Ziegler, Oral insulin immunotherapy in
org/10.1155/2019/1524905. children at risk for type 1 diabetes in a randomised controlled trial, Diabetologia
[147] Q. Huang, C. Merriman, H. Zhang, D. Fu, Coupling of Insulin Secretion and 64 (2021) 1079–1092, https://doi.org/10.1007/s00125-020-05376-1.
Display of a Granule-resident Zinc Transporter ZnT8 on the Surface of Pancreatic [158] H. Elding Larsson, M. Lundgren, B. Jonsdottir, D. Cuthbertson, J. Krischer, A.-I.T.
Beta Cells, J. Biol. Chem. 292 (2017) 4034–4043, https://doi.org/10.1074/jbc. S.G. Di, Safety and efficacy of autoantigen-specific therapy with 2 doses of alum-
M116.772152. formulated glutamate decarboxylase in children with multiple islet
[148] C. Yu, J.C. Burns, W.H. Robinson, P.J. Utz, P.P. Ho, L. Steinman, A.B. Frey, autoantibodies and risk for type 1 diabetes: A randomized clinical trial, Pedia
Identification of candidate tolerogenic CD8(+) T cell epitopes for therapy of type Diabetes 19 (2018) 410–419, https://doi.org/10.1111/pedi.12611.
1 diabetes in the NOD mouse model, J. Diabetes Res. 2016 (2016) 9083103, [159] B. Tavira, H. Barcenilla, J. Wahlberg, P. Achenbach, J. Ludvigsson, R. Casas,
https://doi.org/10.1155/2016/9083103. Intralymphatic Glutamic Acid Decarboxylase-Alum Administration Induced Th2-
[149] M.R. Ehlers, Who let the dogs out? The ever-present threat of autoreactive T cells, Like-Specific Immunomodulation in Responder Patients: A Pilot Clinical Trial in
Sci. Immunol. 3 (2018), https://doi.org/10.1126/sciimmunol.aar6602. Type 1 Diabetes, J. Diabetes Res 2018 (2018) 9391845, https://doi.org/10.1155/
[150] S. Culina, A.I. Lalanne, G. Afonso, K. Cerosaletti, S. Pinto, G. Sebastiani, 2018/9391845.
K. Kuranda, L. Nigi, A. Eugster, T. Østerbye, A. Maugein, J.E. McLaren, K. Ladell, [160] Writing Committee, for the type 1 Diabetes TrialNet Oral Insulin Study G,
E. Larger, J.-P. Beressi, A. Lissina, V. Appay, H.W. Davidson, S. Buus, D.A. Price, Krischer JP, Schatz DA, Bundy B, Skyler JS, Greenbaum CJ, Effect of Oral insulin
M. Kuhn, E. Bonifacio, M. Battaglia, S. Caillat-Zucman, F. Dotta, R. Scharfmann, on prevention of diabetes in relatives of patients with type 1 diabetes: a
B. Kyewski, R. Mallone, Islet-reactive CD8 T cell frequencies in the pancreas, but randomized clinical trial, JAMA 318 (2017) 1891–1902, https://doi.org/
not in blood, distinguish type 1 diabetic patients from healthy donors, Sci. 10.1001/jama.2017.17070.
Immunol. 3 (2018), https://doi.org/10.1126/sciimmunol.aao4013. [161] M. Alhadj Ali, Y.F. Liu, S. Arif, D. Tatovic, H. Shariff, V.B. Gibson, N. Yusuf,
[151] Y. Gu, C. Merriman, Z. Guo, X. Jia, J. Wenzlau, H. Li, H. Li, M. Rewers, L. Yu, R. Baptista, M. Eichmann, N. Petrov, S. Heck, J.H.M. Yang, T.I.M. Tree, I. Pujol-
D. Fu, Novel autoantibodies to the beta-cell surface epitopes of ZnT8 in patients Autonell, L. Yeo, L. Baumard, R. Stenson, A. Howell, A. Clark, Z. Boult, J. Powrie,
progressing to type-1 diabetes, J. Autoimmun. 122 (2021), 102677, https://doi. L. Adams, F.S. Wong, S. Luzio, G. Dunseath, K. Green, A. O’Keefe, G. Bayly,
org/10.1016/j.jaut.2021.102677. N. Thorogood, R. Andrews, N. Leech, F. Joseph, S. Nair, S. Seal, H. Cheung,
[152] K.G. Slepchenko, N.A. Daniels, A. Guo, Y.V. Li, Autocrine effect of Zn2⁺ on the C. Beam, R. Hills, M. Peakman, C.M. Dayan, Metabolic and immune effects of
glucose-stimulated insulin secretion, Endocrine 50 (2015) 110–122, https://doi. immunotherapy with proinsulin peptide in human new-onset type 1 diabetes, Sci.
org/10.1007/s12020-015-0568-z. Transl. Med 9 (2017), https://doi.org/10.1126/scitranslmed.aaf7779.
[153] O.P. Dwivedi, M. Lehtovirta, B. Hastoy, V. Chandra, N.A.J. Krentz, S. Kleiner, [162] E. Bonifacio, A.G. Ziegler, G. Klingensmith, E. Schober, P.J. Bingley,
D. Jain, A.M. Richard, F. Abaitua, N.L. Beer, A. Grotz, R.B. Prasad, O. Hansson, M. Rottenkolber, A. Theil, A. Eugster, R. Puff, C. Peplow, F. Buettner, K. Lange,
E. Ahlqvist, U. Krus, I. Artner, A. Suoranta, D. Gomez, A. Baras, B. Champon, A. J. Hasford, P. Achenbach, P.S.G. Pre, Effects of high-dose oral insulin on immune
J. Payne, D. Moralli, S.K. Thomsen, P. Kramer, I. Spiliotis, R. Ramracheya, responses in children at high risk for type 1 diabetes: the Pre-POINT randomized
P. Chabosseau, A. Theodoulou, R. Cheung, M. van de Bunt, J. Flannick, clinical trial, JAMA 313 (2015) 1541–1549, https://doi.org/10.1001/
M. Trombetta, E. Bonora, C.B. Wolheim, L. Sarelin, R.C. Bonadonna, P. Rorsman, jama.2015.2928.
B. Davies, J. Brosnan, M.I. McCarthy, T. Otonkoski, J.O. Lagerstedt, G.A. Rutter, [163] J. Ludvigsson, D. Krisky, R. Casas, T. Battelino, L. Castano, J. Greening,
J. Gromada, A.L. Gloyn, T. Tuomi, L. Groop, Loss of ZnT8 function protects O. Kordonouri, T. Otonkoski, P. Pozzilli, J.J. Robert, H.J. Veeze, J. Palmer,
against diabetes by enhanced insulin secretion, Nat. Genet 51 (2019) 1596–1606, U. Samuelsson, H. Elding Larsson, J. Aman, G. Kardell, J. Neiderud Helsingborg,
https://doi.org/10.1038/s41588-019-0513-9. G. Lundstrom, E. Albinsson, A. Carlsson, M. Nordvall, H. Fors, C.G. Arvidsson,
[154] J. Flannick, G. Thorleifsson, N.L. Beer, S.B. Jacobs, N. Grarup, N.P. Burtt, S. Edvardson, R. Hanas, K. Larsson, B. Rathsman, H. Forsgren, H. Desaix,
A. Mahajan, C. Fuchsberger, G. Atzmon, R. Benediktsson, J. Blangero, D. G. Forsander, N.O. Nilsson, C.G. Akesson, P. Keskinen, R. Veijola, T. Talvitie,
W. Bowden, I. Brandslund, J. Brosnan, F. Burslem, J. Chambers, Y.S. Cho, K. Raile, T. Kapellen, W. Burger, A. Neu, I. Engelsberger, B. Heidtmann,
C. Christensen, D.A. Douglas, R. Duggirala, Z. Dymek, Y. Farjoun, T. Fennell, S. Bechtold, D. Leslie, F. Chiarelli, A. Cicognani, G. Chiumello, F. Cerutti, G.
P. Fontanillas, T. Forsén, S. Gabriel, B. Glaser, D.F. Gudbjartsson, C. Hanis, V. Zuccotti, A. Gomez Gila, I. Rica, R. Barrio, M. Clemente, M.J. Lopez Garcia,
T. Hansen, A.B. Hreidarsson, K. Hveem, E. Ingelsson, B. Isomaa, S. Johansson, M. Rodriguez, I. Gonzalez, J.P. Lopez, M. Oyarzabal, H.M. Reeser, R. Nuboer,
T. Jørgensen, M.E. Jørgensen, S. Kathiresan, A. Kong, J. Kooner, J. Kravic, P. Stouthart, N. Bratina, N. Bratanic, M. de Kerdanet, J. Weill, N. Ser, P. Barat, A.
M. Laakso, J.Y. Lee, L. Lind, C.M. Lindgren, A. Linneberg, G. Masson, M. Bertrand, J.C. Carel, R. Reynaud, R. Coutant, S. Baron, GAD65 antigen therapy
T. Meitinger, K.L. Mohlke, A. Molven, A.P. Morris, S. Potluri, R. Rauramaa, in recently diagnosed type 1 diabetes mellitus, N. Engl. J. Med 366 (2012)
R. Ribel-Madsen, A.M. Richard, T. Rolph, V. Salomaa, A.V. Segrè, H. Skärstrand, 433–442, https://doi.org/10.1056/NEJMoa1107096.
V. Steinthorsdottir, H.M. Stringham, P. Sulem, E.S. Tai, Y.Y. Teo, T. Teslovich, [164] D.K. Wherrett, B. Bundy, D.J. Becker, L.A. DiMeglio, S.E. Gitelman, R. Goland, P.
U. Thorsteinsdottir, J.K. Trimmer, T. Tuomi, J. Tuomilehto, F. Vaziri-Sani, B. A. Gottlieb, C.J. Greenbaum, K.C. Herold, J.B. Marks, R. Monzavi, A. Moran,
F. Voight, J.G. Wilson, M. Boehnke, M.I. McCarthy, P.R. Njølstad, O. Pedersen, T. Orban, J.P. Palmer, P. Raskin, H. Rodriguez, D. Schatz, D.M. Wilson, J.
L. Groop, D.R. Cox, K. Stefansson, D. Altshuler, Loss-of-function mutations in P. Krischer, J.S. Skyler, Type 1 Diabetes TrialNet GADSG, Antigen-based therapy
SLC30A8 protect against type 2 diabetes, Nat. Genet 46 (2014) 357–363, https:// with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset
doi.org/10.1038/ng.2915. type 1 diabetes: a randomised double-blind trial, Lancet 378 (2011) 319–327,
[155.] E.P. Sorgjerd, P.M. Thorsby, P.A. Torjesen, F. Skorpen, K. Kvaloy, V. Grill, https://doi.org/10.1016/S0140-6736(11)60895-7.
Presence of anti-GAD in a non-diabetic population of adults; time dynamics and [165] K. Vehik, D. Cuthbertson, H. Ruhlig, D.A. Schatz, M. Peakman, J.P. Krischer, Dpt,
clinical influence: results from the HUNT study, BMJ Open Diabetes Res Care 3 TrialNet Study G, Long-term outcome of individuals treated with oral insulin:
(2015), e000076, https://doi.org/10.1136/bmjdrc-2014-000076. diabetes prevention trial-type 1 (DPT-1) oral insulin trial, Diabetes Care 34
[156] R. Casas, F. Dietrich, S. Puente-Marin, H. Barcenilla, B. Tavira, J. Wahlberg, (2011) 1585–1590, https://doi.org/10.2337/dc11-0523.
P. Achenbach, J. Ludvigsson, Intra-lymphatic administration of GAD-alum in type [166] S. Fourlanos, C. Perry, S.A. Gellert, E. Martinuzzi, R. Mallone, J. Butler, P.
1 diabetes: long-term follow-up and effect of a late booster dose (the DIAGNODE G. Colman, L.C. Harrison, Evidence that nasal insulin induces immune tolerance
Extension trial, Acta Diabetol. 59 (2022) 687–696, https://doi.org/10.1007/ to insulin in adults with autoimmune diabetes, Diabetes 60 (2011) 1237–1245,
s00592-022-01852-9. https://doi.org/10.2337/db10-1360.
[157.] R. Assfalg, J. Knoop, K.L. Hoffman, M. Pfirrmann, J.M. Zapardiel-Gonzalo,
A. Hofelich, A. Eugster, M. Weigelt, C. Matzke, J. Reinhardt, Y. Fuchs, M. Bunk,

11

You might also like