Recent reports have revived interest in the active role that β-cells may play in type 1 diabetes pathogenesis at different stages of disease. In some studies, investigators suggested an initiating role and proposed that type 1 diabetes may be primarily a disease of β-cells and only secondarily a disease of autoimmunity. This scenario is possible and invites the search for environmental triggers damaging β-cells. Another major contribution of β-cells may be to amplify autoimmune vulnerability and to eventually drive it into an intrinsic, self-detrimental state that turns the T cell–mediated homicide into a β-cell suicide. On the other hand, protective mechanisms are also mounted by β-cells and may provide novel therapeutic targets to combine immunomodulatory and β-cell protective agents. This integrated view of autoimmunity as a disease of T-cell/β-cell cross talk will ultimately advance our understanding of type 1 diabetes pathogenesis and improve our chances of preventing or reversing disease progression.

β-Cell failure in type 1 diabetes creeps forward at a pace that is defined by a dynamic relationship between immune cells and β-cell intrinsic responses (1,2). This relationship is affected by host factors such as genetics, age, and by poorly defined environmental triggers. In some individuals, autoimmunity is initiated but does not progress further, notably in the presence of a single autoantibody (aAb) (3). This may reflect an aborted immune process or responses of β-cells, or other cells, that prevent progression to the next stage of autoimmune amplification and β-cell destruction (4). However, in others, particularly young individuals, β-cell destruction is more rapid once autoimmunity (stage 1) or hyperglycemia (stage 3) appears (5,6), with little evidence of remission phases.

The precise molecular and cellular mechanisms that account for β-cell death remain unclear. Direct cytotoxic lysis by CD8+ T cells reactive to β-cell antigens such as preproinsulin is postulated. However, immune infiltrates in the pancreas are sparse at the time of diagnosis. While the late timing of these histological studies, i.e., years after autoimmune initiation and even after β-cell destruction and clinical onset, may account for these findings (2), other mechanisms capable of mediating β-cell killing at a distance have been suggested. Inflammatory molecules are produced by several cell types, including β-cells themselves (7), and may cause dysfunction or killing. Findings of shrinkage of the exocrine pancreas before type 1 diabetes diagnosis suggest that inflammation of the whole organ may occur and that β-cells are perhaps killed by soluble mediators produced by cells that are not present in the islets. Indeed, findings of a recent trial of anti–TNF-α antibody showed how neutralizing this cytokine may arrest progression in patients with new-onset type 1 diabetes (8). However, the way in which these acute inflammatory mediators affect the chronic progression remains undefined.

A number of investigators have described, as part of this immune/β-cell dynamics, how β-cells may respond to the autoimmune attack in ways that may promote their demise or protect them (9). The question of whether β-cells die by homicide versus suicide was originally proposed by G. F. Bottazzo (10), but newer studies call for revisiting this question. Clearly, both T cells and β-cells are active disease players, but is one the initiator and the other the victim or do both accelerate β-cell death? We here review this dynamic interplay in the light of recent studies.

The increasing evidence that T cells, together with other immune players, are not the sole culprits in type 1 diabetes pathogenesis has recently been reviewed (2). Briefly, arguments for their nonexclusive involvement include the following. First, experimental autoimmune diabetes cannot be induced in animal models by immunization with islet extracts or β-cell antigens alone (2), as is the case for other autoimmune diseases such as experimental autoimmune encephalomyelitis (a model of multiple sclerosis). Second, human T cells adoptively transferred into immunodeficient mice can cause insulitis but are not diabetogenic in the absence of strong priming conditions, e.g., streptozotocin administration to induce some β-cell death and self-antigen release (11,12). Third, histopathological, age-related endotypes (13,14) mirroring clinical endotypes defined by age and aAb targets (5,6) indicate that a number of other factors may determine rates of progression. Finally, a universal state of “benign” autoimmunity is found in the circulation of all individuals, as shown by the presence of islet-reactive (and largely naive) CD8+ T cells (1517) and islet-reactive CD4+ T cells (18,19). T cells with the same antigen specificities are enriched in the pancreas of donors who had type 1 diabetes (15,16), pointing to local factors that may affect the phenotypes of T cells and attract them into the target organ. Although two case reports of type 1 diabetes transfer, on grafting of bone marrow from a donor who had diabetes (20) and grafting of islets into a recipient with diabetes (21), argue for a central role of T cells, an alternative explanation, regarding the contribution of β-cells, is possible, as explained below.

The active role of β-cells in type 1 diabetes pathogenesis has been linked to their specialized function of insulin secretion (1)—a paradigm that may apply more generally to other endocrine cells. Namely, the high endoplasmic reticulum (ER) and oxidative stress imposed by the requirements for rapid and considerable insulin biosynthesis, the rich islet vascularization—which exposes cells to inflammatory soluble mediators and cells, and the secretion of insulin and other antigenic granule contents directly in the bloodstream with high amplitude (22) are all features that may render β-cells more vulnerable to autoimmunity.

Importantly, new emerging data extend this picture to alterations of the exocrine pancreas (23). A reduced pancreas organ weight was reported in donors positive for a single aAb (24) and even in aAb-negative first-degree relatives (25). Investigators of other studies reported reduced serum trypsinogen and lipase levels starting at stage 1 (two or more aAb) type 1 diabetes (26,27) and enriched exocrine immune infiltration (28) and HLA class II expression in ductal cells from donors who had type 1 diabetes (29). In another notable recent single-cell epigenomics study (30), high expression was documented of novel type 1 diabetes risk gene variants (e.g., chymotrypsinogens, lipase, and cystic fibrosis transmembrane conductance regulator) in acinar and ductal cells, which is postulated to promote subclinical pancreas inflammation favoring later immune infiltration.

The pathogenic role of β-cells could theoretically be called into question at three sequential stages (Fig. 1).

Figure 1

Pathogenic role of β-cells in type 1 diabetes. The three possible stages of initiation, amplification, and finalization of the pathogenic cascade where β-cells can come into play are depicted.

Figure 1

Pathogenic role of β-cells in type 1 diabetes. The three possible stages of initiation, amplification, and finalization of the pathogenic cascade where β-cells can come into play are depicted.

Close modal
  1. Self-initiated homicide: are β-cells the original drivers of T-cell autoimmunity?

  2. Self-amplified homicide: do β-cells enhance T cell–mediated killing?

  3. Self-finalized suicide: do β-cells under attack eventually die by suicide?

Self-initiated Homicide

Studies by Larger, Boitard, and colleagues (31) nearly 30 years ago showed that β-cells are needed to initiate autoimmune diabetes in NOD mice. These results support a primary role of β-cells in developing an autoimmune repertoire endowed with β-cell specificity, since splenocytes from β-cell–deficient mice could not transfer diabetes but the mice were otherwise immune competent and developed other autoimmune manifestations, i.e., sialitis. This observation pointed to an early critical time window in which T cells are primed by β-cell antigens and trigger diabetes. Of further note, this priming occurs primarily in pancreatic lymph nodes and is an early event, as pancreatic lymphadenectomy at 3 weeks, but not at 10 weeks of age, led to near-complete diabetes protection in NOD mice (32). Pancreatic lymph nodes may also harbor stem-like CD8+ T cells that can proliferate and generate effector T cells that invade islets (33).

Several lines of experimental evidence suggest that β-cell aberrancies, which may include increased antigen exposure and neoantigen expression, may be the first drivers of islet autoimmunity. Islet autoimmunity does not develop if β-cells are not visible to the immune system due to reduced antigen expression, i.e., if immune ignorance is maintained. An emblematic example is provided by NOD mice carrying an insulin B16 mutation that abolishes the immunogenicity of the initiating epitope insulin B9–23, which are completely protected from diabetes (34). Serreze et al. (35) found that initiation of autoimmune diabetes in NOD mice is dependent on MHC class I. β2-microglobulin (β2m)−/− mice, which are MHC class I deficient, did not develop insulitis or diabetes, whereas splenocytes from diabetic NOD donors transferred diabetes into both wild-type and β2m−/− NOD/scid recipients. In contrast, splenocytes from young prediabetic NOD donors transferred diabetes only to wild-type NOD/scid recipients, while β2m−/− NOD/scid recipients required prior grafting with wild-type islets to become diabetic. The authors concluded that islet expression of MHC class I (i.e., β-cell visibility) was needed to prime T cells, which could then mediate β-cell destruction independent of MHC class I (35). Other adoptive T-cell transfer models suggest that, in addition to antigen presentation, β-cell alterations such as those imprinted in the NOD background or induced by streptozotocin are needed to enable antigen-driven T-cell priming and β-cell killing (2). On the same lines, T-cell receptor transgenic models in which T cells recognize a foreign antigen transgenically expressed in β-cells show that provision of costimulatory or inflammatory signals is required to activate these T cells (36). These mouse studies may provide an interpretation for some observations in patients. In a case report of identical twin-to-twin transplantation of a nondiabetic pancreas into a recipient with type 1 diabetes, the transplantation led to rapid autoimmune relapse (21); conversely, bone marrow transplantation from a sibling with type 1 diabetes into an HLA-identical recipient without diabetes triggered type 1 diabetes (20). In both cases, the previous priming of the autoimmune T-cell repertoire prior to transplantation may have bypassed the need for a β-cell trigger.

Results of recent elegant studies have revived interest in this hypothesis of self-initiated homicide in showing that the induction of an early and transient β-cell dedifferentiation protects NOD mice from diabetes. A first model of NOD mice with high β-cell proliferation rates and decreased antigen expression (37) was obtained either with a liver-specific insulin receptor genetic knockout or by pharmacological treatment with an insulin receptor antagonist from 4 to 6 weeks of age. In a second model (38), a conditional knockout of the inositol-requiring enzyme 1α, which activates the unfolded protein response (UPR) in β-cells, was induced by tamoxifen treatment during the neonatal period (1–5 days of life), resulting in a similar dedifferentiated β-cell phenotype. Both NOD models are protected from diabetes.

Can all these extreme murine models that “blind” the immune system from β-cells be taken as evidence for an active role of β-cells in initiating autoimmunity? Other arguments from human genetic studies need to be considered. First, the contribution of type 1 diabetes–associated gene variants expressed in β-cells to genetic susceptibility is marginal in comparison with HLA class II polymorphisms (2)—a feature that type 1 diabetes shares with several other autoimmune diseases. Second, the major influence of HLA class II haplotypes is on the antigen specificity and rate of aAb seroconversion (i.e., autoimmune initiation) rather than on the rate of actual β-cell destruction (i.e., clinical progression) (2,39,40), suggesting a driving effect on pathology. This may further suggest that β-cell–related non-HLA gene variants may act as modulators of this later clinical progression rather than drivers. Third, monogenic forms of neonatal diabetes causing β-cell defects, including mutations (e.g., in INS and EIF2AK3 genes) that induce severe ER stress, show little evidence of islet autoimmunity, even in the context of high-risk HLA class II haplotypes, with only 5.4% positivity for islet aAb, mostly (92%) for a single aAb (41). This is observed in spite of a dysregulated expression of genes (and proteins) associated with monogenic forms of diabetes resulting from ER stress in the pancreas of donors who had type 1 diabetes and were aAb+ (42), suggesting a role of primary β-cell alterations as disease contributors rather than drivers.

However, an alternative explanation could be that the β-cell contribution to type 1 diabetes pathogenesis may simply be less genetically imprinted and more environmentally driven than the immune contribution. This hypothesis may invite the search for environmental triggers that may compromise β-cell health rather than immune tolerance per se, e.g., islet-tropic viral infections and xenotoxic agents.

Altogether, this literature suggests that β-cell stress/dysfunction may play a role at initiating islet autoimmunity, and then significantly contribute to its amplification, as discussed in the next section.

Self-Amplified Homicide

A key observation suggesting the role of β-cell–driven initiation and amplification mechanisms is the heterogeneity of insulitis across islets, which do not appear to be attacked and destroyed at the same time (43). There are multiple mechanisms by which β-cells amplify the autoimmune attack (Fig. 2). The inflammatory environment of insulitis, particularly interferons, stimulates several active responses in β-cells that go beyond the passive effect of inducing ER stress, dysfunction, and apoptosis. First, inflammation drives β-cells themselves to release chemokines (e.g., CCL5, CXCL2, CXCL9, CXCL10) and cytokines (e.g., IL-1β, IL-6) (7), which further attract immune cells to the pancreas and contribute to their activation in situ.

Figure 2

β-Cell mechanisms of self-amplified homicide/suicide (red arrows) vs. protection (blue arrows). See text for details. DCs, dendritic cells; PLN, pancreatic lymph node.

Figure 2

β-Cell mechanisms of self-amplified homicide/suicide (red arrows) vs. protection (blue arrows). See text for details. DCs, dendritic cells; PLN, pancreatic lymph node.

Close modal

The second major outcome of islet inflammation is HLA class I upregulation, which results in the exposure of a larger and more diversified repertoire of antigenic peptides (16), thus increasing the visibility and vulnerability of β-cells to infiltrating T cells. Not surprisingly, peptides derived from proteins of secretory granules (e.g., insulin, chromogranin A, secretogranin-5, urocortin-3, proconvertase-2) are highly represented across different human and murine MHC restrictions (16,17) and are targeted by diabetogenic CD8+ T cells, which induce disease when transferred into NOD/scid mice (17). This pathogenic role may rely on features that granule proteins share with (pro)insulin (44). All of these antigens are synthesized as proprotein precursors, which are subsequently enzymatically cleaved, mostly by proconvertases, to generate their bioactive products. Hence, the impaired proinsulin processing by proconvertases described in β-cells from patients with type 1 diabetes (44,45) may extend to all of these proteins, possibly diverting their degradation toward the MHC class I presentation pathway. In addition, all of these proteins are released in the bloodstream with insulin exocytosis, both in intact form and as antigenic degradation products (22). They can thus be taken up by extrapancreatic antigen-presenting cells and prime T cells at a distance on uptake by extrapancreatic antigen-presenting cells (1).

The MHC peptide repertoire displayed by β-cells further includes neoantigens, i.e., peptide sequences that are not templated in the genome and may therefore be regarded as nonself and trigger autoimmunity, since they have not participated in the physiological mechanisms of immune tolerance. These neoantigens are produced through different mechanisms: alternative mRNA splicing (16,17), out-of-frame protein translation due to defective ribosomal scanning under high insulin demand (46), posttranslational modifications (47), and transpeptidation (16,48), i.e., the fusion of noncontiguous peptide fragments from the same protein or from two distinct partners.

Other amplification mechanisms may be related to β-cell sensitivity to stress-induced senescence. In a recent study (49), the accumulation of senescent β-cells in NOD mice was documented during a critical early time window that marks the transition from peri-insulitis to destructive insulitis. Although these senescent β-cells increase the expression of prosurvival factors such as Bcl2, they also have a key role in amplifying the autoimmune response because they produce a specific secretome (the so-called SASP [senescence-associated secretory phenotype]) enriched in factors (e.g., IGFBP3, serpin E1) that propagate this senescent phenotype to neighboring β-cells, as well as in cytokines/chemokines (e.g., IL-6, CXCL10) that attract and activate immune cells in the pancreas. Senescence markers (CDKN1A, serpin E1, IL-6) were also observed in the pancreas of patients with type 1 diabetes (49). Importantly, when senescence was inhibited by an anti-Bcl2 small molecule that drives apoptosis specifically in these cells, diabetes was prevented, thus documenting the pathogenic role of the senescent phenotype. This observation identifies a novel therapeutic target involving a rather counterintuitive strategy of inducing apoptosis of selected β-cell subsets. This also suggests that the physiologic process of clearing senescent β-cells, or hypersecreting mutants (adenomas) (50), is misdirected in autoimmunity. It will be important to clarify whether the relationship of β-cell dysfunction, senescence, and apoptosis is sequential, mutually exclusive, and/or naturally reversible.

Besides inciting autoimmunity via all these immunogenic signals, β-cells may further amplify these mechanisms by ceasing to secrete protective antiapoptotic factors. A notable example is growth/differentiation factor (GDF)15, a transforming growth factor-β superfamily member that was found to be downregulated in β-cells from individuals with type 1 diabetes and on in vitro exposure to IL-1β and IFN-γ (7). Although its mechanisms of action remain unclear, GDF15 protects NOD mice from diabetes (7), presumably through anti-inflammatory effects, and exerts additional effects in the hindbrain (reducing food intake and promoting weight loss) and in peripheral tissues (hepatocytes, myocytes, and adipocytes), by increasing insulin sensitivity.

Self-Finalized Suicide Versus Protection

Further compensatory mechanisms mounted by β-cells may be self-detrimental (“suicidal”) in the long-term, acting through intrinsic mechanisms that do not require the participation of other cells (Fig. 2).

Some cytokines (e.g., IL-1β, IL-6) that are secreted by β-cells themselves (7) can be proapoptotic in an autocrine/paracrine loop. Their secretion may be particularly important during the extended period of time—as long as 5 years—when insulin secretion may be impaired but clinically silent. The ability to compensate for metabolic demands becomes critical during this time for β-cell survival. As metabolic demands increase, reactive oxygen species are needed for glucose-induced insulin secretion (51) and mass expansion (52). However, the low expression of antioxidant enzymes such as catalase and superoxide dismutase exposes β-cells to cell-intrinsic oxidative damage, particularly in the ER (53). Even β-cell identity may be affected by this oxidative stress, since exposure to hydrogen peroxide can induce loss of nuclear localization of MafA, a transcription factor associated with mature β-cells that may enhance the β-cell antioxidant defenses, protecting them against deterioration during hyperglycemia (5456). Increased metabolic demands also upregulate the UPR, which is constitutively active in β-cells due to their basal ER stress from high protein (mostly insulin) biosynthesis rates. This increased UPR can drive apoptosis when reaching a critical threshold (1). Indeed, UPR upregulation was observed in β-cells from two type 1 diabetes mouse models and from patients (57), as identified by the expression of activating transcription factor 6 (ATF6) and spliced X-box binding protein 1 (sXBP1). The deleterious effect of what is initially a compensatory response is suggested by the observation that surviving β-cells display lower ATF6 and sXBP1 expression and that relieving UPR with tauroursodeoxycholic acid protects mice from diabetes (57).

We recently showed that Tet2 is increased in β-cells from NOD mice and in human insulitis (58). Its deleterious β-cell intrinsic outcomes were documented in several ways. In β-cells that survive autoimmune attack, Tet2 expression was reduced. Despite unchanged insulitis and T-cell diabetogenicity, Tet2−/− NOD mice were protected from diabetes, even after grafting of Tet2+/+ bone marrow or transferring diabetogenic splenocytes. Tet2 affects the epigenetic control of signaling molecules such as STAT1, nuclear factor-κB, and IRF2. While its loss was associated with CXCL10 downregulation by β-cells, there was also reduced expression of the CXCR3 homing receptor on β-cell–reactive T cells in pancreatic lymph nodes.

Protective mechanisms are also mounted by β-cells (Fig. 2). During progression of autoimmunity in NOD mice, we described degranulation, loss of Ins1/Ins2 gene expression, and acquisition of a more undifferentiated phenotype, with expression of other islet hormones (e.g., Gcg, Sst) and stem cell markers (Sox9, L-Myc, Oct-4) in residual β-cells (59). In addition to insulin, there was reduced expression of other diabetes autoantigens (GAD, ZnT8, IA-2). In line with these results, Damond et al. (60) compared β-cells of patients with new-onset or long-standing type 1 diabetes with those of individuals without diabetes by image cytometry pseudo-timing using Network for Pancreatic Organ donors with Diabetes (nPOD) specimens. β-Cell destruction was preceded by downregulation of identity markers (INS, proinsulin, IAPP, PTPRN) with (pseudo)time, without changes in β-cell transcription factors PDX1 and NKX6-1. Loss of lineage-specific markers may thus be an early event, before further disease progression driven by β-cell death. In our study (59), dedifferentiated β-cells also expressed immunoregulatory ligands (PD-L1, Qa-2) and resisted immune killing by islet infiltrates or cytokines—unlike their mature counterparts. PD-L1 is induced on human and murine β-cells by IFN-γ and IFN-α (61). β-Cells also secrete antimicrobial peptides (e.g., CRAMP), a process triggered by in-vitro exposure to IL-1β or LPS (62) that is impaired in NOD mice (63). Besides inducing tolerogenic antigen-presenting cells and regulatory T cells (63), these peptides reduce β-cell apoptosis and production of inflammatory prostaglandin E2 (62). These mechanisms, while protective against killing in specific experimental settings, clearly fail to arrest the autoimmune response in most clinical settings.

Altogether, the available literature suggests that, following autoimmune insult, β-cells may also contribute in a direct way to their own killing. This concept of self-detrimental versus protective β-cell responses has clinical relevance. Successful immunotherapies (e.g., teplizumab, antithymocyte globulin, alefacept, rituximab) share a mechanism of modulation/deletion of inflammatory cells. While treatment is often followed by C-peptide stabilization, a decline subsequently occurs in many patients (64), suggesting either that the conditions that led to clinical onset have recurred or that β-cell intrinsic mechanisms may drive this decline. As an example, even continuous administration of CTLA-4 Ig at type 1 diabetes onset did not avoid a decline in β-cell function after an initial improvement (65). This suggests that maintaining β-cell intrinsic protective mechanisms together with immunomodulation may limit this later decline.

The concepts of β-cell responsiveness to autoimmune attack also suggest ways in which autoimmunity might be arrested with a combination of agents that target immune cells and β-cells. Evidence (33,66) for stem-like features of islet-reactive CD8+ T cells that can proliferate and are the source of short-lived effector cells that migrate to the pancreas highlights the challenge in purging the repertoire of effector T cells sufficiently to prevent disease (33). Intriguingly, results from teplizumab trials (67) suggest that immunotherapies may be most effective when β-cell stress is occurring, i.e., at stage 2 of rapid C-peptide decline, consistent with preclinical data in NOD mice. Although the β-cell changes occurring during this period of rapid progression may expose new and more antigen targets, the T cells that recognize them may represent the more differentiated ones that are amenable to treatment.

Modulating the β-cell and antigen targets of autoimmune T cells may thus be needed in addition to immunotherapies. Clinical studies are ongoing with agents that are postulated to limit β-cell stress responses, such as verapamil (clinical trial reg. no. NCT04545151, ClinicalTrials.gov), a calcium channel blocker that inhibits TXNIP, a mediator of oxidative stress (68), and tauroursodeoxycholic acid (NCT02218619), a chemical chaperone that mitigates ER stress (57). In a previous study of the tyrosine kinase inhibitor imatinib as a single agent, a significant effect on C-peptide responses was not sustained (69), but ongoing studies are evaluating effects on markers of β-cell stress. Other studies with agents such as JAK1/2 inhibitors, that can block critical signaling pathways in β-cells in addition to blunting T-cell activation, are under development. Finally, in a randomized four-arm placebo-controlled trial, anti–IL-21 antibodies and the GLP-1 receptor agonist liraglutide significantly reduced C-peptide decline at 54 weeks in patients with recently diagnosed type 1 diabetes compared with placebo (70). The finding that this decline was lower than with either agent alone suggests that immune and β-cell combination therapies may be synergistic in achieving clinical outcomes.

These studies highlight the dynamic relationships between immune effector cells and their β-cell targets. The findings have shown how both parties contribute to killing but also suggest that therapies that are directed only at one arm may be insufficient to completely arrest the ongoing process. Much work has been done to characterize the autoimmune repertoire, including the targets and features of T and B cells that can mediate disease. Attention to the attributes of β-cells that can instill protection from or participation in autoimmunity are now needed.

Acknowledgments. The authors apologize to colleagues whose publications could not be referenced due to space constraints. Figs. 1 and 2 were created with elements from Servier Medical ART (SMART) (https://smart.servier.com).

Funding. Work performed in the laboratory of R.M. is supported by The Leona M. and Harry B. Helmsley Charitable Trust (no. 1901-03689), Agence Nationale de la Recherche (ANR-19-CE15-0014-01), and Fondation pour la Recherche Medicale (EQU20193007831) and by the Innovative Medicines Initiative 2 Joint Undertaking under grant agreements 115797 and 945268, INNODIA and INNODIA HARVEST, which receive support from the European Union Horizon 2020 program, the European Federation of Pharmaceutical Industries and Associations, JDRF, and The Leona M. and Harry B. Helmsley Charitable Trust. C.H. was funded by an Année Recherche fellowship of University of Paris-Saclay. Work performed in the laboratory of K.C.H. is supported by National Institutes of Health grants DK057846, DK101122, DK104205, and DK045735; JDRF grant SRA2014-142-S-B; a Pilot Award from the Yale Diabetes Research Center, P30DK116577; and a gift from the Howalt family.

Duality of Interest. K.C.H. is co-inventor on a patent application for use of teplizumab. No other potential conflicts of interest relevant to this article were reported.

1.
Mallone
R
,
Eizirik
DL
.
Presumption of innocence for beta cells: why are they vulnerable autoimmune targets in type 1 diabetes?
Diabetologia
2020
;
63
:
1999
2006
2.
Carré
A
,
Richardson
SJ
,
Larger
E
,
Mallone
R
.
Presumption of guilt for T cells in type 1 diabetes: lead culprits or partners in crime depending on age of onset?
Diabetologia
2021
;
64
:
15
25
3.
Insel
RA
,
Dunne
JL
,
Atkinson
MA
, et al
.
Staging presymptomatic type 1 diabetes: a scientific statement of JDRF, the Endocrine Society, and the American Diabetes Association
.
Diabetes Care
2015
;
38
:
1964
1974
4.
Faucher
P
,
Beuvon
F
,
Fignani
D
, et al
.
Immunoregulated insulitis and slow-progressing type 1 diabetes after duodenopancreatectomy
.
Diabetologia
2021
;
64
:
2731
2740
5.
Frohnert
BI
,
Ide
L
,
Dong
F
, et al
.
Late-onset islet autoimmunity in childhood: the Diabetes Autoimmunity Study in the Young (DAISY)
.
Diabetologia
2017
;
60
:
998
1006
6.
Hao
W
,
Gitelman
S
,
DiMeglio
LA
,
Boulware
D
;
Type 1 Diabetes TrialNet Study Group
.
Fall in C-peptide during first 4 years from diagnosis of type 1 diabetes: variable relation to age, HbA1c, and insulin dose
.
Diabetes Care
2016
;
39
:
1664
1670
7.
Nakayasu
ES
,
Syed
F
,
Tersey
SA
, et al
.
Comprehensive proteomics analysis of stressed human islets identifies GDF15 as a target for type 1 diabetes intervention
.
Cell Metab
2020
;
31
:
363
374.e6
8.
Quattrin
T
,
Haller
MJ
,
Steck
AK
, et al.;
T1GER Study Investigators
.
Golimumab and beta-cell function in youth with new-onset type 1 diabetes
.
N Engl J Med
2020
;
383
:
2007
2017
9.
Atkinson
MA
,
Bluestone
JA
,
Eisenbarth
GS
, et al
.
How does type 1 diabetes develop?: the notion of homicide or β-cell suicide revisited
.
Diabetes
2011
;
60
:
1370
1379
10.
Bottazzo
GF
.
Lawrence lecture. Death of a beta cell: homicide or suicide?
Diabet Med
1986
;
3
:
119
130
11.
Tan
S
,
Li
Y
,
Xia
J
, et al
.
Type 1 diabetes induction in humanized mice
.
Proc Natl Acad Sci U S A
2017
;
114
:
10954
10959
12.
Viehmann Milam
AA
,
Maher
SE
,
Gibson
JA
, et al
.
A humanized mouse model of autoimmune insulitis
.
Diabetes
2014
;
63
:
1712
1724
13.
Leete
P
,
Mallone
R
,
Richardson
SJ
,
Sosenko
JM
,
Redondo
MJ
,
Evans-Molina
C
.
The effect of age on the progression and severity of type 1 diabetes: potential effects on disease mechanisms
.
Curr Diab Rep
2018
;
18
:
115
14.
Leete
P
,
Oram
RA
,
McDonald
TJ
, et al
.
Studies of insulin and proinsulin in pancreas and serum support the existence of aetiopathological endotypes of type 1 diabetes associated with age at diagnosis
.
Diabetologia
2020
;
63
:
1258
1267
15.
Culina
S
,
Lalanne
AI
,
Afonso
G
, et al.;
ImMaDiab Study Group
.
Islet-reactive CD8+ T cell frequencies in the pancreas, but not in blood, distinguish type 1 diabetic patients from healthy donors
.
Sci Immunol
2018
;
3
:
eaao4013
16.
Gonzalez-Duque
S
,
Azoury
ME
,
Colli
ML
, et al
.
Conventional and neo-antigenic peptides presented by β cells are targeted by circulating naïve CD8+ T cells in type 1 diabetic and healthy donors
.
Cell Metab
2018
;
28
:
946
960.e6
17.
Azoury
ME
,
Tarayrah
M
,
Afonso
G
, et al
.
Peptides derived from insulin granule proteins are targeted by CD8+ T cells across MHC class I restrictions in humans and NOD mice
.
Diabetes
2020
;
69
:
2678
2690
18.
Viglietta
V
,
Kent
SC
,
Orban
T
,
Hafler
DA
.
GAD65-reactive T cells are activated in patients with autoimmune type 1a diabetes
.
J Clin Invest
2002
;
109
:
895
903
19.
Danke
NA
,
Koelle
DM
,
Yee
C
,
Beheray
S
,
Kwok
WW
.
Autoreactive T cells in healthy individuals
.
J Immunol
2004
;
172
:
5967
5972
20.
Lampeter
EF
,
Homberg
M
,
Quabeck
K
, et al
.
Transfer of insulin-dependent diabetes between HLA-identical siblings by bone marrow transplantation
.
Lancet
1993
;
341
:
1243
1244
21.
Sutherland
DE
,
Sibley
R
,
Xu
XZ
, et al
.
Twin-to-twin pancreas transplantation: reversal and reenactment of the pathogenesis of type I diabetes
.
Trans Assoc Am Physicians
1984
;
97
:
80
87
22.
Wan
X
,
Zinselmeyer
BH
,
Zakharov
PN
, et al
.
Pancreatic islets communicate with lymphoid tissues via exocytosis of insulin peptides
.
Nature
2018
;
560
:
107
111
23.
Alexandre-Heymann
L
,
Mallone
R
,
Boitard
C
,
Scharfmann
R
,
Larger
E
.
Structure and function of the exocrine pancreas in patients with type 1 diabetes
.
Rev Endocr Metab Disord
2019
;
20
:
129
149
24.
Campbell-Thompson
M
,
Wasserfall
C
,
Montgomery
EL
,
Atkinson
MA
,
Kaddis
JS
.
Pancreas organ weight in individuals with disease-associated autoantibodies at risk for type 1 diabetes
.
JAMA
2012
;
308
:
2337
2339
25.
Campbell-Thompson
ML
,
Filipp
SL
,
Grajo
JR
, et al
.
Relative pancreas volume is reduced in first-degree relatives of patients with type 1 diabetes
.
Diabetes Care
2019
;
42
:
281
287
26.
Li
X
,
Campbell-Thompson
M
,
Wasserfall
CH
, et al
.
Serum trypsinogen levels in type 1 diabetes
.
Diabetes Care
2017
;
40
:
577
582
27.
Ross
JJ
,
Wasserfall
CH
,
Bacher
R
, et al
.
Exocrine pancreatic enzymes are a serological biomarker for type 1 diabetes staging and pancreas size
.
Diabetes
2021
;
70
:
944
954
28.
Rodriguez-Calvo
T
,
Ekwall
O
,
Amirian
N
,
Zapardiel-Gonzalo
J
,
von Herrath
MG
.
Increased immune cell infiltration of the exocrine pancreas: a possible contribution to the pathogenesis of type 1 diabetes
.
Diabetes
2014
;
63
:
3880
3890
29.
Fasolino
M
,
Schwartz
GW
,
Patil
AR
, et al.;
HPAP Consortium
.
Single-cell multi-omics analysis of human pancreatic islets reveals novel cellular states in type 1 diabetes
.
Nat Metab
2022
;
4
:
284
299
30.
Chiou
J
,
Geusz
RJ
,
Okino
ML
, et al
.
Interpreting type 1 diabetes risk with genetics and single-cell epigenomics
.
Nature
2021
;
594
:
398
402
31.
Larger
E
,
Bécourt
C
,
Bach
JF
,
Boitard
C
.
Pancreatic islet beta cells drive T cell-immune responses in the nonobese diabetic mouse model
.
J Exp Med
1995
;
181
:
1635
1642
32.
Gagnerault
MC
,
Luan
JJ
,
Lotton
C
,
Lepault
F
.
Pancreatic lymph nodes are required for priming of beta cell reactive T cells in NOD mice
.
J Exp Med
2002
;
196
:
369
377
33.
Gearty
SV
,
Dündar
F
,
Zumbo
P
, et al
.
An autoimmune stem-like CD8 T cell population drives type 1 diabetes
.
Nature
2022
;
602
:
156
161
34.
Nakayama
M
,
Abiru
N
,
Moriyama
H
, et al
.
Prime role for an insulin epitope in the development of type 1 diabetes in NOD mice
.
Nature
2005
;
435
:
220
223
35.
Serreze
DV
,
Chapman
HD
,
Varnum
DS
,
Gerling
I
,
Leiter
EH
,
Shultz
LD
.
Initiation of autoimmune diabetes in NOD/Lt mice is MHC class I-dependent
.
J Immunol
1997
;
158
:
3978
3986
36.
Brezar
V
,
Carel
JC
,
Boitard
C
,
Mallone
R
.
Beyond the hormone: insulin as an autoimmune target in type 1 diabetes
.
Endocr Rev
2011
;
32
:
623
669
37.
Dirice
E
,
Kahraman
S
,
De Jesus
DF
, et al
.
Increased β-cell proliferation before immune cell invasion prevents progression of type 1 diabetes
.
Nat Metab
2019
;
1
:
509
518
38.
Lee
H
,
Lee
YS
,
Harenda
Q
, et al
.
Beta cell dedifferentiation induced by IRE1α deletion prevents type 1 diabetes
.
Cell Metab
2020
;
31
:
822
836.e5
39.
Valdes
AM
,
Erlich
HA
,
Carlson
J
,
Varney
M
,
Moonsamy
PV
,
Noble
JA
.
Use of class I and class II HLA loci for predicting age at onset of type 1 diabetes in multiple populations
.
Diabetologia
2012
;
55
:
2394
2401
40.
Dufort
MJ
,
Greenbaum
CJ
,
Speake
C
,
Linsley
PS
.
Cell type-specific immune phenotypes predict loss of insulin secretion in new-onset type 1 diabetes
.
JCI Insight
2019
;
4
:
e125556
41.
Wyatt
RC
,
Hagopian
WA
,
Roep
BO
, et al.;
EXE-T1D Consortium
.
Congenital beta-cell defects are not associated with markers of islet autoimmunity, even in the context of high genetic risk for type 1 diabetes
.
Diabetologia
2022
;
65
:
1179
1184
42.
Hiller
H
,
Beachy
DE
,
Lebowitz
JJ
, et al
.
Monogenic diabetes and integrated stress response genes display altered gene expression in type 1 diabetes
.
Diabetes
2021
;
70
:
1885
1897
43.
Campbell-Thompson
M
,
Fu
A
,
Kaddis
JS
, et al
.
Insulitis and β-cell mass in the natural history of type 1 diabetes
.
Diabetes
2016
;
65
:
719
731
44.
Carré
A
,
Mallone
R
.
Making insulin and staying out of autoimmune trouble: the beta-cell conundrum
.
Front Immunol
2021
;
12
:
639682
45.
Sims
EK
,
Chaudhry
Z
,
Watkins
R
, et al
.
Elevations in the fasting serum proinsulin–to–C-peptide ratio precede the onset of type 1 diabetes
.
Diabetes Care
2016
;
39
:
1519
1526
46.
Kracht
MJ
,
van Lummel
M
,
Nikolic
T
, et al
.
Autoimmunity against a defective ribosomal insulin gene product in type 1 diabetes
.
Nat Med
2017
;
23
:
501
507
47.
Purcell
AW
,
Sechi
S
,
DiLorenzo
TP
.
The evolving landscape of autoantigen discovery and characterization in type 1 diabetes
.
Diabetes
2019
;
68
:
879
886
48.
Delong
T
,
Wiles
TA
,
Baker
RL
, et al
.
Pathogenic CD4 T cells in type 1 diabetes recognize epitopes formed by peptide fusion
.
Science
2016
;
351
:
711
714
49.
Thompson
PJ
,
Shah
A
,
Ntranos
V
,
Van Gool
F
,
Atkinson
M
,
Bhushan
A
.
Targeted elimination of senescent beta cells prevents type 1 diabetes
.
Cell Metab
2019
;
29
:
1045
1060.e10
50.
Korem Kohanim
Y
,
Tendler
A
,
Mayo
A
,
Friedman
N
,
Alon
U
.
Endocrine autoimmune disease as a fragility of immune surveillance against hypersecreting mutants
.
Immunity
2020
;
52
:
872
884.e5
51.
Leloup
C
,
Tourrel-Cuzin
C
,
Magnan
C
, et al
.
Mitochondrial reactive oxygen species are obligatory signals for glucose-induced insulin secretion
.
Diabetes
2009
;
58
:
673
681
52.
Ahmed Alfar
E
,
Kirova
D
,
Konantz
J
,
Birke
S
,
Mansfeld
J
,
Ninov
N
.
Distinct levels of reactive oxygen species coordinate metabolic activity with beta-cell mass plasticity
.
Sci Rep
2017
;
7
:
3994
53.
Tersey
SA
,
Nishiki
Y
,
Templin
AT
, et al
.
Islet β-cell endoplasmic reticulum stress precedes the onset of type 1 diabetes in the nonobese diabetic mouse model
.
Diabetes
2012
;
61
:
818
827
54.
Guo
S
,
Dai
C
,
Guo
M
, et al
.
Inactivation of specific β cell transcription factors in type 2 diabetes
.
J Clin Invest
2013
;
123
:
3305
3316
55.
Harmon
JS
,
Bogdani
M
,
Parazzoli
SD
, et al
.
beta-Cell-specific overexpression of glutathione peroxidase preserves intranuclear MafA and reverses diabetes in db/db mice
.
Endocrinology
2009
;
150
:
4855
4862
56.
Kaneto
H
,
Kajimoto
Y
,
Miyagawa
J
, et al
.
Beneficial effects of antioxidants in diabetes: possible protection of pancreatic beta-cells against glucose toxicity
.
Diabetes
1999
;
48
:
2398
2406
57.
Engin
F
,
Yermalovich
A
,
Nguyen
T
, et al
.
Restoration of the unfolded protein response in pancreatic β cells protects mice against type 1 diabetes
.
Sci Transl Med
2013
;
5
:
211ra156
58.
Rui
J
,
Deng
S
,
Perdigoto
AL
, et al
.
Tet2 controls the responses of β cells to inflammation in autoimmune diabetes
.
Nat Commun
2021
;
12
:
5074
59.
Rui
J
,
Deng
S
,
Arazi
A
,
Perdigoto
AL
,
Liu
Z
,
Herold
KC
.
β cells that resist immunological attack develop during progression of autoimmune diabetes in NOD mice
.
Cell Metab
2017
;
25
:
727
738
60.
Damond
N
,
Engler
S
,
Zanotelli
VRT
, et al
.
A map of human type 1 diabetes progression by imaging mass cytometry
.
Cell Metab
2019
;
29
:
755
768.e5
61.
Osum
KC
,
Burrack
AL
,
Martinov
T
, et al
.
Interferon-gamma drives programmed death-ligand 1 expression on islet β cells to limit T cell function during autoimmune diabetes
.
Sci Rep
2018
;
8
:
8295
62.
Sun
J
,
Xu
M
,
Ortsäter
H
, et al
.
Cathelicidins positively regulate pancreatic β-cell functions
.
FASEB J
2016
;
30
:
884
894
63.
Sun
J
,
Furio
L
,
Mecheri
R
, et al
.
Pancreatic β-cells limit autoimmune diabetes via an immunoregulatory antimicrobial peptide expressed under the influence of the gut microbiota
.
Immunity
2015
;
43
:
304
317
64.
Greenbaum
CJ
,
Schatz
DA
,
Haller
MJ
,
Sanda
S
.
Through the fog: recent clinical trials to preserve β-cell function in type 1 diabetes
.
Diabetes
2012
;
61
:
1323
1330
65.
Orban
T
,
Bundy
B
,
Becker
DJ
, et al.;
Type 1 Diabetes TrialNet Abatacept Study Group
.
Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial
.
Lancet
2011
;
378
:
412
419
66.
Abdelsamed
HA
,
Zebley
CC
,
Nguyen
H
, et al
.
Beta cell-specific CD8+ T cells maintain stem cell memory-associated epigenetic programs during type 1 diabetes
.
Nat Immunol
2020
;
21
:
578
587
67.
Herold
KC
,
Bundy
BN
,
Long
SA
, et al.;
Type 1 Diabetes TrialNet Study Group
.
An anti-CD3 antibody, teplizumab, in relatives at risk for type 1 diabetes
.
N Engl J Med
2019
;
381
:
603
613
68.
Ovalle
F
,
Grimes
T
,
Xu
G
, et al
.
Verapamil and beta cell function in adults with recent-onset type 1 diabetes
.
Nat Med
2018
;
24
:
1108
1112
69.
Gitelman
SE
,
Bundy
BN
,
Ferrannini
E
, et al.;
Gleevec Trial Study Group
.
Imatinib therapy for patients with recent-onset type 1 diabetes: a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial
.
Lancet Diabetes Endocrinol
2021
;
9
:
502
514
70.
von Herrath
M
,
Bain
SC
,
Bode
B
, et al.;
Anti-IL-21–liraglutide Study Group investigators and contributors
.
Anti-interleukin-21 antibody and liraglutide for the preservation of β-cell function in adults with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled, phase 2 trial
.
Lancet Diabetes Endocrinol
2021
;
9
:
212
224
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.