It may be considered unusual to consider a period of three decades “historical.” Yet, the evolution for our understanding of the natural history and pathogenesis of type 1 diabetes has been greatly advanced by a vast number of studies aimed at validating a model (1), proposed by the late Dr. George Eisenbarth in 1986 (2). As a result of this work, the majority of current conventional wisdom portrays type 1 diabetes as a T cell–mediated autoimmune disease involving the specific destruction of insulin-producing pancreatic β-cells.

In this model, persons destined to develop type 1 diabetes are assumed to begin life with a full cadre of β-cells. However, a “triggering” insult, likely environmental, initiates a process involving the recruitment of antigen-presenting cells. Antigen-presenting cells sequester self-antigens released by injured β-cells, followed by their transport to pancreatic lymph nodes where they are subsequently presented to autoreactive T cells. These T cells, rogue constituents brought to life due to genetically driven failures of thymic deletion (i.e., central tolerance) combined with defects in mechanisms designed to induce peripheral immune tolerance, come into play (3). This toxic duo, imparting lack-of-tolerance formation, again in the context of genetic susceptibility, allows for migration of self-reactive T cells to islets, mediating β-cell killing and promoting further inflammation (4). When 85–90% of pancreatic β-cells meet their demise, symptoms of the disease occur. In the final stage of the model, the autoimmune process ends with the complete elimination of β-cells.

While this concept still forms the prevailing intellectual dogma for the majority of individuals associated with diabetes care and research today, a series of recent observations has challenged multiple aspects of this long-standing model (5). Many of these evolving concepts will be presented in this Perspective, with a discussion of how our understanding of models of type 1 diabetes pathogenesis has and will likely continue to evolve as it relates to attempts seeking to prevent and/or reverse the disorder.

The timing for introduction of the Eisenbarth model appeared therapeutically “fortuitous” in its day. Contemporaneous with positing autoimmunity as the formative cause of type 1 diabetes in the 1980s were therapeutic interventions developed for organ transplantation. This research brought forward a series of immunosuppressive agents thought clinically promising for multiple immune based–disorders, including type 1 diabetes. The earliest of the immunosuppressive-based studies in type 1 diabetes, using agents such as cyclosporine or azathioprine, provided evidence that preservation of endogenous insulin secretion was possible, even if only for a relatively short period of time, in settings of recent-onset disease. Such news brought hope that a means to prevent or cure type 1 diabetes was on the horizon. While adverse effects of these agents brought a close to their use in type 1 diabetes, they set the stage for a multidecade effort to utilize a variety of biologics to target autoimmunity for the purpose of stemming the tide of β-cell destruction.

Clinical trials seeking to meet the goal of type 1 diabetes prevention (Table 1) (recently reviewed in refs. 69) in the case of autoantibody-positive subjects with type 1 diabetes or C-peptide preservation in recent-onset subjects (Table 2) (recently reviewed in ref. 10) have been quite variable in terms of their “success.” While some trials have demonstrated the ability to either delay progression to type 1 diabetes or preserve C-peptide production in individuals with recent-onset type 1 diabetes, the vast majority of such efforts has either failed to meet the predetermined end points, or even when demonstrating early success (i.e., meeting goals of C-peptide production at 12 months for subjects with type 1 diabetes), loss in C-peptide production eventually occurs for most. Mechanistic studies affiliated with these efforts have, to a large extent, failed to identify specific mechanisms associated with therapeutic failure or success. The failures in achieving therapeutic success in humans stand in stark contrast to the results of studies in the NOD mouse, where methods capable of preventing type 1 diabetes and/or reversing overt hyperglycemia abound (1113). We would suggest that many of the failures of human studies have been the by-product of having a poor understanding of the complexity of the disorder’s pathogenesis—too many factors have historically been underappreciated, misunderstood, or unknown in considerations of the pathogenesis of type 1 diabetes (Table 3).

Table 1

Prevention trials in type 1 diabetes

Study nameIntervention1°OutcomeEnd point achievedReference or ClinicalTrials.gov identifier
Primary prevention studies     
 NIP Dietary docosahexaenoic acid Pilot study ** 60 
 Finnish TRIGR pilot Hydrolyzed casein formula Autoantibodies Yes 61 
 BABYDIET Delayed dietary gluten exposure Autoantibodies No 62 
 TRIGR Hydrolyzed casein formula Autoantibodies No 63 
 FINDIA Whey-based, insulin-free bovine milk formula Autoantibodies Yes 64 
Secondary prevention studies    
 DENIS Nicotinamide Diagnosis of type 1 diabetes No 65 
 DPT-1 Parenteral Insulin Parenteral insulin Diagnosis of type 1 diabetes No 66 
 INIT I Intranasal insulin Safety Yes# 67 
 ENDIT Nicotinamide Diagnosis of type 1 diabetes No 68 
 DPT-1 Oral Insulin Oral insulin Diagnosis of type 1 diabetes No 69 
 DIPP sibling cohort Intranasal insulin Diagnosis of type 1 diabetes No 70 
 DIPP birth cohort Intranasal insulin Diagnosis of type 1 diabetes No 70 
 Belgian Parenteral Insulin Parenteral insulin Diagnosis of type 1 diabetes No 71 
 TrialNet Oral Insulin Oral insulin Diagnosis of type 1 diabetes * NCT00419562 
 INIT II Intranasal insulin Diagnosis of type 1 diabetes * NCT00336674 
 DIAPREV-IT GAD-alum (Diamyd) Diagnosis of type 1 diabetes * NCT01122446 
 TrialNet Teplizumab Teplizumab Diagnosis of type 1 diabetes * NCT01030861 
 TrialNet Abatacept CTLA4Ig (abatacept) Diagnosis of type 1 diabetes * NCT01773707 
Study nameIntervention1°OutcomeEnd point achievedReference or ClinicalTrials.gov identifier
Primary prevention studies     
 NIP Dietary docosahexaenoic acid Pilot study ** 60 
 Finnish TRIGR pilot Hydrolyzed casein formula Autoantibodies Yes 61 
 BABYDIET Delayed dietary gluten exposure Autoantibodies No 62 
 TRIGR Hydrolyzed casein formula Autoantibodies No 63 
 FINDIA Whey-based, insulin-free bovine milk formula Autoantibodies Yes 64 
Secondary prevention studies    
 DENIS Nicotinamide Diagnosis of type 1 diabetes No 65 
 DPT-1 Parenteral Insulin Parenteral insulin Diagnosis of type 1 diabetes No 66 
 INIT I Intranasal insulin Safety Yes# 67 
 ENDIT Nicotinamide Diagnosis of type 1 diabetes No 68 
 DPT-1 Oral Insulin Oral insulin Diagnosis of type 1 diabetes No 69 
 DIPP sibling cohort Intranasal insulin Diagnosis of type 1 diabetes No 70 
 DIPP birth cohort Intranasal insulin Diagnosis of type 1 diabetes No 70 
 Belgian Parenteral Insulin Parenteral insulin Diagnosis of type 1 diabetes No 71 
 TrialNet Oral Insulin Oral insulin Diagnosis of type 1 diabetes * NCT00419562 
 INIT II Intranasal insulin Diagnosis of type 1 diabetes * NCT00336674 
 DIAPREV-IT GAD-alum (Diamyd) Diagnosis of type 1 diabetes * NCT01122446 
 TrialNet Teplizumab Teplizumab Diagnosis of type 1 diabetes * NCT01030861 
 TrialNet Abatacept CTLA4Ig (abatacept) Diagnosis of type 1 diabetes * NCT01773707 

Adapted with permission from Skyler (9). DENIS, Deutsche Nicotinamide Intervention Study; DIAPREV-IT, Diabetes Prevention–Immune Tolerance; DIPP, Type 1 Diabetes Prediction and Prevention Project; DPT-1, Diabetes Prevention Trial–Type 1; ENDIT, European Nicotinamide Diabetes Intervention Trial; FINDIA, Finnish Dietary Intervention Trial for the Prevention of Type 1 Diabetes; INIT, Intranasal Insulin Trial; NIP, Nutritional Intervention to Prevent Diabetes; TRIGR, Trial to Reduce IDDM in the Genetically at Risk.

*Data not yet available.

**Pilot study.

#No adverse events.

Table 2

Reversal trials in type 1 diabetes

Study nameTreatment(s)C-peptide at 1 year (nmol/L)Preservation of C-peptideReference
Cyclosporin Treatment in Children with Recent-Onset Type 1 Diabetes° Cyclosporin A 0.3 Yes* 72  
Continuous Insulin Infusion Throughout the First Two Weeks Following Type 1 Diabetes Onset° Intensive insulin therapy 0.5 Yes 73  
IMDIAB IV Vitamin E 0.2 Yes 74  
IMDIAB VI Nicotinamide 0.2 Yes 75  
Diabète Insuline Orale Oral insulin 0.1 No 76  
DIA-AID2 DiaPep277 0.2 Yes 77  
AbATE hOKT3gamma1(Ala-Ala) 0.2 Yes 78  
Diazoxide Treatment in Children with New-Onset Type 1 Diabetes° Diazoxide 0.2 Yes 79  
The Use of Polyclonal Anti–T-Lymphocyte Globulin to Prevent Progression of Autoimmune β-Cell Destruction in Recent Type 1 Diabetes ATG 0.2 Yes 80  
IMDIAB IX Nicotinamide + vitamin E 0.2 Yes** 81  
IMDIAB (retrospective analysis) Nicotinamide + intensive insulin therapy 0.1 Yes** 82  
TTEDD ChAglyCD3 (otelixizumab) 0.5 Yes 83  
Phase II Trial of hOKT3gamma1(Ala-Ala) Teplizumab for Treatment of Patients With Recent Onset Type 1 Diabetes hOKT3gamma1(Ala-Ala) 0.2 Yes 84  
IMDIAB XI Calcitriol + nicotinamide 0.1 No 85  
DIA-AID2 DiaPep277 0.2 Yes§ 86  
Autologous Hematopoietic Stem Cell Transplantation in Type 1 Diabetes Mellitus AHSCT 0.3 Yes 87  
Phase II Trial of DiaPep277 in Children with New-Onset Type 1 Diabetes° DiaPep277 0.2 No 88  
DIA-AID2 DiaPep277 0.4 Yes (trend)# 89  
A Phase II, Randomised, Double-Blind, Placebo-Controlled, Multi-Centre Study to Investigate the Impact of Diamyd on the Progression of Diabetes in Patients Newly Diagnosed With Type 1 Diabetes Mellitus GAD-alum 0.1 No 90  
Phase II Multiple Dose Treatment of Type 1 Diabetes Mellitus With hOKT3gamma1(Ala-Ala) Teplizumab 0.8 Yes (trend) 91  
A Randomized, Double-Blind, Placebo-Controlled, Multicenter, Parallel, Dose-Ranging Study to Evaluate the Efficacy, Safety, Tolerability, and Pharmacodynamics of NBI-6024 In Adult and Adolescent Patients With New Onset Type 1 Diabetes Mellitus NBI-6024 0.1 No 92  
Autologous Hematopoietic Stem Cell Transplantation in Type 1 Diabetes Mellitus AHSCT AUC = 30 Yes 53  
Effects of Rituximab on the Progression of Type 1 Diabetes in New Onset Subjects Rituximab 0.1 Yes¥ 93  
ENBREL (Etanercept) Administration to Patients Newly Diagnosed With Type 1 Diabetes Mellitus: Feasibility-Safety Study Etanercept 0.4 Yes 94  
Extension of Phase II Therapeutic Trial With a Humanized Non-Mitogenic CD3 (ChAgly CD3) Monoclonal Antibody in Recently Diagnosed Type 1 Diabetic Patients ChAgly CD3 AUC = 0.9 Yes# 95  
Efficacy of 6 Months Treatment With Diazoxide at Bedtime in Preventing β-Cell Demise in Newly Diagnosed Type 1 Diabetes Diazoxide 0.1 No 96  
Immunointervention With 1,25-dihydroxy-vitamin D3 in New-Onset Type 1 Diabetes 1,25(OH)2D3 0.1 No 97  
Protégé study Teplizumab 0.5 Yes# 98  
TrialNet GAD GAD-alum 0.3 No 99  
TrialNet Abatacept Abatacept 0.3 No 100  
DIATOR Atorvastatin 0.2 No 101  
Efficacy of ATG + Autologous CD34+ Stem Cells + GCSF in New-Onset Type 1 Diabetes° Mobilized hematopoietic CD34+ stem cells 0.4 Yes 102  
A Phase III, 3-Arm, Randomized, Double-Blind, Placebo-Controlled, Multicenter Study to Investigate the Impact of Diamyd on the Progression of Diabetes in Patients Newly Diagnosed With Type 1 Diabetes Mellitus (EU) GAD-alum 0.3 No 103  
DIATOR Atorvastatin 0.2 Yes 104  
Prospective Study of Autologous Hematopoietic Stem Cell Transplantation to Treat New Onset Type 1 Diabetes AHSCT 0.6 Yes 105  
Safety and Efficacy Study of Autologous Nonmyeloablative Hematopoietic Stem Cell Transplantation for Early Onset Type 1 Diabetes—A Phase II Study AHSCT  Yes 106  
A Phase I Trial of Proleukin and Rapamune in Recent-Onset Type 1 Diabetes Mellitus (ITN018AI) Rapamycin/interleukin (IL)-2 AUC = 6.3 No 107  
Canakinumab Study in Individuals With Newly Diagnosed Type 1 Diabetes (anti–IL-1)/ Anti–Interleukin-1 in Diabetes Action Canakinumab/Anakinra 0.1 No 108  
Reversing Type 1 Diabetes After it is Established: A Pilot Safety and Feasibility Study of Anti-Thymocyte Globulin (Thymoglobulin) and Pegylated GCSF (Neulasta) in Established Type 1 Diabetes ATG + GCSF 0.74 Yes 54  
Study nameTreatment(s)C-peptide at 1 year (nmol/L)Preservation of C-peptideReference
Cyclosporin Treatment in Children with Recent-Onset Type 1 Diabetes° Cyclosporin A 0.3 Yes* 72  
Continuous Insulin Infusion Throughout the First Two Weeks Following Type 1 Diabetes Onset° Intensive insulin therapy 0.5 Yes 73  
IMDIAB IV Vitamin E 0.2 Yes 74  
IMDIAB VI Nicotinamide 0.2 Yes 75  
Diabète Insuline Orale Oral insulin 0.1 No 76  
DIA-AID2 DiaPep277 0.2 Yes 77  
AbATE hOKT3gamma1(Ala-Ala) 0.2 Yes 78  
Diazoxide Treatment in Children with New-Onset Type 1 Diabetes° Diazoxide 0.2 Yes 79  
The Use of Polyclonal Anti–T-Lymphocyte Globulin to Prevent Progression of Autoimmune β-Cell Destruction in Recent Type 1 Diabetes ATG 0.2 Yes 80  
IMDIAB IX Nicotinamide + vitamin E 0.2 Yes** 81  
IMDIAB (retrospective analysis) Nicotinamide + intensive insulin therapy 0.1 Yes** 82  
TTEDD ChAglyCD3 (otelixizumab) 0.5 Yes 83  
Phase II Trial of hOKT3gamma1(Ala-Ala) Teplizumab for Treatment of Patients With Recent Onset Type 1 Diabetes hOKT3gamma1(Ala-Ala) 0.2 Yes 84  
IMDIAB XI Calcitriol + nicotinamide 0.1 No 85  
DIA-AID2 DiaPep277 0.2 Yes§ 86  
Autologous Hematopoietic Stem Cell Transplantation in Type 1 Diabetes Mellitus AHSCT 0.3 Yes 87  
Phase II Trial of DiaPep277 in Children with New-Onset Type 1 Diabetes° DiaPep277 0.2 No 88  
DIA-AID2 DiaPep277 0.4 Yes (trend)# 89  
A Phase II, Randomised, Double-Blind, Placebo-Controlled, Multi-Centre Study to Investigate the Impact of Diamyd on the Progression of Diabetes in Patients Newly Diagnosed With Type 1 Diabetes Mellitus GAD-alum 0.1 No 90  
Phase II Multiple Dose Treatment of Type 1 Diabetes Mellitus With hOKT3gamma1(Ala-Ala) Teplizumab 0.8 Yes (trend) 91  
A Randomized, Double-Blind, Placebo-Controlled, Multicenter, Parallel, Dose-Ranging Study to Evaluate the Efficacy, Safety, Tolerability, and Pharmacodynamics of NBI-6024 In Adult and Adolescent Patients With New Onset Type 1 Diabetes Mellitus NBI-6024 0.1 No 92  
Autologous Hematopoietic Stem Cell Transplantation in Type 1 Diabetes Mellitus AHSCT AUC = 30 Yes 53  
Effects of Rituximab on the Progression of Type 1 Diabetes in New Onset Subjects Rituximab 0.1 Yes¥ 93  
ENBREL (Etanercept) Administration to Patients Newly Diagnosed With Type 1 Diabetes Mellitus: Feasibility-Safety Study Etanercept 0.4 Yes 94  
Extension of Phase II Therapeutic Trial With a Humanized Non-Mitogenic CD3 (ChAgly CD3) Monoclonal Antibody in Recently Diagnosed Type 1 Diabetic Patients ChAgly CD3 AUC = 0.9 Yes# 95  
Efficacy of 6 Months Treatment With Diazoxide at Bedtime in Preventing β-Cell Demise in Newly Diagnosed Type 1 Diabetes Diazoxide 0.1 No 96  
Immunointervention With 1,25-dihydroxy-vitamin D3 in New-Onset Type 1 Diabetes 1,25(OH)2D3 0.1 No 97  
Protégé study Teplizumab 0.5 Yes# 98  
TrialNet GAD GAD-alum 0.3 No 99  
TrialNet Abatacept Abatacept 0.3 No 100  
DIATOR Atorvastatin 0.2 No 101  
Efficacy of ATG + Autologous CD34+ Stem Cells + GCSF in New-Onset Type 1 Diabetes° Mobilized hematopoietic CD34+ stem cells 0.4 Yes 102  
A Phase III, 3-Arm, Randomized, Double-Blind, Placebo-Controlled, Multicenter Study to Investigate the Impact of Diamyd on the Progression of Diabetes in Patients Newly Diagnosed With Type 1 Diabetes Mellitus (EU) GAD-alum 0.3 No 103  
DIATOR Atorvastatin 0.2 Yes 104  
Prospective Study of Autologous Hematopoietic Stem Cell Transplantation to Treat New Onset Type 1 Diabetes AHSCT 0.6 Yes 105  
Safety and Efficacy Study of Autologous Nonmyeloablative Hematopoietic Stem Cell Transplantation for Early Onset Type 1 Diabetes—A Phase II Study AHSCT  Yes 106  
A Phase I Trial of Proleukin and Rapamune in Recent-Onset Type 1 Diabetes Mellitus (ITN018AI) Rapamycin/interleukin (IL)-2 AUC = 6.3 No 107  
Canakinumab Study in Individuals With Newly Diagnosed Type 1 Diabetes (anti–IL-1)/ Anti–Interleukin-1 in Diabetes Action Canakinumab/Anakinra 0.1 No 108  
Reversing Type 1 Diabetes After it is Established: A Pilot Safety and Feasibility Study of Anti-Thymocyte Globulin (Thymoglobulin) and Pegylated GCSF (Neulasta) in Established Type 1 Diabetes ATG + GCSF 0.74 Yes 54  

Adapted with permission from Ben Nasr et al. (10). AbATE, Autoimmunity-Blocking Antibody for Tolerance in Recently Diagnosed Type 1 Diabetes; AHSCT, autologous hematopoietic stem cell transplantation; ATG, anti–T-lymphocyte globulin; AUC, area under the curve; DIA-AID, Efficacy and Safety Study of DiaPep277 in Newly Diagnosed Type 1 Diabetes Adults; DIATOR, Diabetes Intervention With Atorvastatin; GCSF, granulocyte-colony stimulating factor; IMDIAB, IMmunotherapy of DIABetes; TTED, TRX4 Monoclonal Antibody in Type 1 Diabetes (T1 DM).

◆Defined as author-reported interpretation.

°Official study name could not be determined.

*Effect lost on withdrawal of treatment.

**No synergistic benefit with combination.

§Differential outcomes depending on dose.

#Differential effects depending on age-group.

¥Reported partial preservation of β-cell function.

†Differential effects depending on baseline C-reactive protein concentrations.

Table 3

Features influencing the pathogenesis and natural history of type 1 diabetes likely underappreciated in therapeutic trials seeking to prevent and or reverse the disease

ImmunePancreas/β-CellsEnvironment/Genetics
Innate immunity Small pancreas Microbiome (gut, oral) 
Influence of age on immune response Vascular abnormalities Diet 
Immune cell metabolism Pancreatitis (role) Antibiotic use 
Acute versus chronic β-cell destruction Exocrine infiltration Exercise 
Limited/focal nature of insulitis β-Cell replication as a function of age Epigenetic modifications Relationship with gut (including celiac disease) 
ImmunePancreas/β-CellsEnvironment/Genetics
Innate immunity Small pancreas Microbiome (gut, oral) 
Influence of age on immune response Vascular abnormalities Diet 
Immune cell metabolism Pancreatitis (role) Antibiotic use 
Acute versus chronic β-cell destruction Exocrine infiltration Exercise 
Limited/focal nature of insulitis β-Cell replication as a function of age Epigenetic modifications Relationship with gut (including celiac disease) 

Perhaps no segment of the historical model for type 1 diabetes pathogenesis has been as rigorously investigated as that of the immune response of persons with or at various levels of risk for the disease. While such studies have yielded success stories with practical outcomes (e.g., autoantibody staging for disease risk, biomarker development, identification of subjects for disease prevention trials), they all suffer from a variety of limitations. By their nature, nearly all studies of human immune responses involve analysis of peripheral blood rather than at the site of β-cell destruction. In addition, while the potential importance of the so-called effector and regulatory components in type 1 diabetes pathogenesis have been extensively studied, only recently have serious considerations been given to the effects of aging, diet, immune cell metabolism, microbial pathogens, microbiomes, and epigenetic changes on the immune response affording this disease (Table 3) (1418). These factors, individually and in combination, clearly influence immune responses in general and, thus, must be associated by default with the pathogenesis of type 1 diabetes.

As a number of type 1 diabetes reviews have recently been published highlighting features of the immune response in peripheral blood (4,19,20), this Perspective will focus on studies of human pancreas and other tissues obtained from organ donors with or at risk for type 1 diabetes that have largely, but not exclusively, been made possible through the efforts of the Belgian Beta Cell Bank and the JDRF Network for Pancreatic Organ Donors with Diabetes (nPOD) program (21,22).

Immunological Characteristics of the Pancreas in Type 1 Diabetes—Evidence for Disease Subtypes

Recent studies of human pancreata have added support to the growing concept that subtypes of type 1 diabetes truly exist. For example, when histological studies of type 1 diabetes pancreata are combined, patients with disease onset at age 0–14 years and within 1 year of diagnosis show more inflamed islets (68%) and fewer islets with residual β-cells (39%) than do patients with onset at 15–39 years of age (23). This suggests a more vigorous autoimmune response occurs when disease develops in young children.

Additional evidence in support this concept was the recent finding that younger age of onset is associated with higher levels of CD20+ B cells, CD45+ cells, and CD8+ T cells in insulitis lesions, with fewer insulin-positive islets (15,24). Conversely, infiltrates with fewer CD20+ cells were observed in patients with type 1 diabetes who were older at onset and were associated with lower levels of CD45+ cells and CD8+ T cells, as well as more insulin-positive islets. These same studies also noted that islet CD8+ T cells expressed T-cell receptors that bound MHC class I tetramers loaded with the β-cell autoantigen IGRP and other target peptides in patients with recent-onset type 1 diabetes (24). These studies are also in agreement with recent findings that the T cells invading pancreatic islets are, in fact, directed at β-cell antigens (25).

It is also noteworthy that pancreata from patients in each age-group greater than 1 year from diabetes onset have similarly low levels of insulitis (3–4%) and equivalent maintenance of insulin-positive islets (13%). Therefore, it appears that the process of insulitis formation in the two age-groups may have equalized and lessened over time, avoiding complete β-cell annihilation. As discussed below, recent studies indicate that some β-cells survive in type 1 diabetes for many decades (24,26). Thus, key questions become: What type of inflammation is present in these disease subtypes at various stages at what stages and can these be pushed toward the resolution phase that may be part and parcel of the inflammatory process?

Inflammation Is Also Present in Pancreatic Exocrine Tissue

Recent studies emanating from nPOD demonstrate chronic inflammation, including enhanced CD8+ T-cell infiltration (and, to a lesser degree, CD4+ and CD11c+ cells) in the exocrine pancreas in subjects with type 1 diabetes (27). Other studies report a similar propensity for neutrophil invasion of the pancreas (with decreased peripheral neutrophil counts) in type 1 diabetes (28). The propensity of this organ for inflammation/pancreatitis induced by multiple factors (e.g., hypertriglyceridemia, virus infection, drugs) could be a result of a susceptibility gene that affects tissue-based inflammation or other facets that have, in the past, been considered “leakiness” (29,30). Importantly, the inflammatory process in the pancreas appears to be subclinical, as most new-onset patients do not present with symptoms of pancreatic inflammation.

Clarifying the Immune: Viral Connections in Type 1 Diabetes

While numerous viruses have been posed as potential contributors to the pathogenesis of type 1 diabetes, the vast majority of investigation continues to focus on coxsackievirus (CVB). Indeed, recent efforts have provided evidence for the presence of viral capsid protein expression, VP1, as well as viral RNA and type 1 interferon (IFN) in islets (27,31).

Recently, we and others have noted expression of multiple type 1 IFN signature proteins, including MDA5, in association with CVB capsid protein, VP1, in type 1 diabetes pancreata (32,33). The islets in new-onset organ donors (Fig. 1B) and in autoantibody-positive donors (data not shown) demonstrate infiltrate; however, it is substantially less than that seen in the NOD mouse (Fig. 1A). In addition, immune cells tend to invade the NOD islet but manifest largely as peri-insulitis in humans (Fig. 1A and B). In both NOD (data not shown) and the islets from humans with new-onset disease, there is evidence for type 1 IFN activity as MDA5 and other IFN-signature proteins are expressed (Fig. 1C). However, in pancreata from patients with diabetes, for several years there are many insulin-positive cells, MDA5-positive islet cells, and VP1 capsid protein all expressed in the absence of CD45+ cell infiltration (Fig. 1C and M.C.-S., unpublished data). These data suggest a chronic, persistent, low-level viral infection may perpetuate a form of islet inflammation long-term that is tissue based. As to potential underlying mechanisms for such observations, CVB RNA may undergo 5′ terminal deletions in vivo, leading to a replication-deficient virus, affording a potential explanation for viral persistence and lack of immune activation resulting in viral clearance (34). Alternatively, genetic contributions to acute viral responses regulated by type 1 diabetes susceptibility genes (e.g., IFIH1, Tyk2, or PTPN22) or increased numbers of plasmacytoid dendritic cells producing high levels of type 1 IFN may modulate downstream antiviral responses (32,35). Persistent viral infections were recently shown to propagate chronic type 1 IFN responses that downgrade inflammation and immune responses, thereby contributing to continued viral infection (3638). If CVB or other viral infections cause chronic type 1 IFN–based inflammation, immune modulation of this response or the use of antiviral therapy may provide a means to clear the virus or reduce the inflammation that affects β-cell function and/or survival.

Figure 1

Emerging features regarding pancreatic islets in type 1 diabetes. Recent descriptions of insulitis (23,25,51,55) have placed an emphasis on the quantitative differences in this lesion when comparing human pancreatic samples to those observed in the NOD mouse model of disease. For example, the intensity and pattern of lymphocytic infiltration in NOD mice at or immediately prior to disease onset (A; 14-week-old new-onset case) is quite pronounced relative to that of human type 1 diabetes (B; 13-year-old with type 1 diabetes <1 year, nPOD 6228). C: Consistent with a notion ascribing a role for viral infections with type 1 diabetes, an nPOD organ donor from a patient with disease onset at 10.2 years of age and a 4-year duration was examined. An islet from this donor expressed abundant insulin (blue), CVB capsid protein VP1 (green), and MDA5 (red) in islet cells. β-Cells expressing both MDA5 and insulin are purple. This islet was also negative for CD45 staining, demonstrating a lack of insulitis (representative image = 40×).

Figure 1

Emerging features regarding pancreatic islets in type 1 diabetes. Recent descriptions of insulitis (23,25,51,55) have placed an emphasis on the quantitative differences in this lesion when comparing human pancreatic samples to those observed in the NOD mouse model of disease. For example, the intensity and pattern of lymphocytic infiltration in NOD mice at or immediately prior to disease onset (A; 14-week-old new-onset case) is quite pronounced relative to that of human type 1 diabetes (B; 13-year-old with type 1 diabetes <1 year, nPOD 6228). C: Consistent with a notion ascribing a role for viral infections with type 1 diabetes, an nPOD organ donor from a patient with disease onset at 10.2 years of age and a 4-year duration was examined. An islet from this donor expressed abundant insulin (blue), CVB capsid protein VP1 (green), and MDA5 (red) in islet cells. β-Cells expressing both MDA5 and insulin are purple. This islet was also negative for CD45 staining, demonstrating a lack of insulitis (representative image = 40×).

Close modal

Over the past decade, knowledge regarding human pancreatic islets and β-cells has increased dramatically. Amplified by studies on human pancreatic islets from the National Institutes of Health (NIH) Integrated Islet Distribution Program (IIDP) and nPOD, a greater understanding of human islet gene expression, function, proliferation, and regeneration has emerged. A consensus has materialized that human pancreatic islets have fundamental differences with the more widely studied rodent islet, including dissimilarities in islet cell composition, basal insulin secretion, susceptibility to toxins such as streptozotocin, amyloid formation, and cell proliferation (3942).

In response to the increased metabolic demands of insulin resistance and obesity, rodent β-cells increase insulin biosynthesis and cellular proliferation, leading to a marked increase in β-cell mass. In contrast, pancreatic samples from obese humans show only a minimal or modest expansion in β- or islet-cell mass (43). These differences between rodent and human islets do not invalidate rodent models of islet biology but require increased attention and integration of findings in rodents to human islets and the human pancreas.

β-Cell Mass Is Not Equal in All Individuals

Historical models of type 1 diabetes have assumed a normal β-cell mass at birth that declines once the autoimmune attack occurs. However, recent studies of cadaveric pancreata have shown that β-cell mass in normal humans without diabetes varies three- to fivefold, independent of adult age or BMI, with β-cell mass likely mostly determined in the first two decades of life (4345). This has important implications when one considers the starting point for declining β-cell mass during autoimmune β-cell destruction. Thus, an individual’s timeline to diabetes onset could be determined not by the severity of the autoimmune attack but the starting point for β-cell mass (Fig. 2). The reasons for this variation in β-cell mass are unknown but could include the in utero environment, events during the first decade of life, and yet unknown genetic or environmental determinants. Further emphasizing the need to understand the timeline and determinants of human β-cell mass is the recent observation of a smaller pancreatic mass in individuals with new-onset type 1 diabetes or with islet-cell autoantibodies (46,47). This observation raises the possibility that determinants of both pancreatic mass and β-cell mass might be impacted, as endocrine islet cells and exocrine cells share a common embryologic heritage.

Figure 2

Summary of recent changes in our understanding of the pathogenesis of type 1 diabetes. The y-axis shows β-cell mass in three individuals with a different baseline β-cell mass over time (x-axis). Likewise, the loss of β-cell mass may differ among individuals as shown by the dashed lines. Finally, β-cell mass is markedly inadequate, but does not become “zero” in many individuals with type 1 diabetes as shown by the horizontal dashed lines. While these lines are drawn as smooth, it is likely that the decline in β-cell mass or function is intermittent and possibly episodic. Recent discoveries also highlight the presence of viral particles in the type 1 pancreas, the variable insulitis, the role of B and T lymphocytes, and the presence of inflammatory cells in the exocrine pancreas. These processes are shown as occurring after an unknown “trigger event,” but this is speculative.

Figure 2

Summary of recent changes in our understanding of the pathogenesis of type 1 diabetes. The y-axis shows β-cell mass in three individuals with a different baseline β-cell mass over time (x-axis). Likewise, the loss of β-cell mass may differ among individuals as shown by the dashed lines. Finally, β-cell mass is markedly inadequate, but does not become “zero” in many individuals with type 1 diabetes as shown by the horizontal dashed lines. While these lines are drawn as smooth, it is likely that the decline in β-cell mass or function is intermittent and possibly episodic. Recent discoveries also highlight the presence of viral particles in the type 1 pancreas, the variable insulitis, the role of B and T lymphocytes, and the presence of inflammatory cells in the exocrine pancreas. These processes are shown as occurring after an unknown “trigger event,” but this is speculative.

Close modal

Are All β-Cells Equally Susceptible to Destruction in Type 1 Diabetes?

It has also been assumed that all human β-cells are equally susceptible to autoimmune attack and that differences in the timeline of type 1 diabetes pathogenesis relate to immune differences. In reality, variations in β-cell susceptibility to cytokines or immune cell attack could be an important determinant of when an individual develops clinical diabetes (Fig. 2). While certain immunomodulatory approaches appear to improve C-peptide production (6), improved β-cell function is not synonymous with prevention of β-cell loss or recovery of β-cell mass. An ongoing debate surrounding type 2 diabetes is whether loss of β-cell function or reduction in mass is the reason for inadequate insulin secretion, but most agree that both pathogenic processes are important. In addition, metabolic derangements clearly impact key islet-enriched transcription factors or may promote loss of β-cell identity (48,49). Therefore, potential parallels with β-cell dysfunction and/or loss in type 1 diabetes seem clear. A challenge is that there are no markers (other than insulin secretion) or noninvasive imaging modalities that reflect β-cell mass in humans. As insulin secretion (basal or stimulated as part of the intravenous glucose tolerance test, oral glucose tolerance test, or mixed-meal tolerance test) can be affected by chronic elevations in the blood glucose and secretory capacity has not been shown to truly correlate closely with β-cell mass over time, improvements in how to assess β-cell mass in humans are needed.

Subjects With Long-standing Type 1 Diabetes, in Fact, Have β-Cells

One of the longer-standing dogmas in type 1 diabetes is that eventually all β-cells are lost in long-standing disease, but the emerging reality is quite different. Many individuals with type 1 diabetes produce small amounts of C-peptide, and studies of the pancreata from individuals with type 1 diabetes show the presence of insulin-positive cells, sometimes within glucagon-rich islets or as single insulin-positive cells scattered throughout the pancreatic exocrine tissue (24,26,50). Interestingly, C-peptide levels were higher in patients >18 years of age at onset and with shorter duration of diabetes (26). These findings raise the question of why some β-cells escape the autoimmune attack or are somehow resistant to it. Are the surviving β-cells somehow “different”? Alternatively, new β-cells may be constantly being regenerated and subsequently destroyed by the ongoing autoimmune process. Now that the transcriptional profile and molecular signatures of normal human β-cells are being defined, it should be possible to determine whether these residual insulin-positive cells are “normal” β-cells.

Inducing Human β-Cells to Proliferate

Intense efforts to induce human β-cell proliferation are under way and our improved understanding of human β-cell biology is providing clues regarding signaling pathways and cell cycle determinants important for human β-cell proliferation (4244). As recently summarized, we currently lack an approach to induce sustained human β-cell proliferation with an acceptable safety profile (42). A clear challenge is the need to induce only β-cell proliferation as many of the current approaches and growth factors being tested target pathways present in many cell types. The ability to specifically target β-cells in vivo with either a proliferative signal or a protective intervention is needed. Moreover, successful strategies in prevention of type 1 diabetes and/or in preservation of β-cell function may require interventions targeting immune pathways in combination with approaches that promote β-cell proliferation (Fig. 3).

Figure 3

A conceptual model for improving efforts to prevent and or cure type 1 diabetes. Previous clinical trials using monotherapies (i.e., single agents) have shown various degrees of success, albeit even in the best of situations partial, in terms of achieving desired therapeutic outcomes. Here, we diagram the potential, albeit a theoretical, model for the principle that combination therapies would provide a substantial improvement over singular monotherapies, with respect to preservation of β-cell mass. Mono, monotherapy.

Figure 3

A conceptual model for improving efforts to prevent and or cure type 1 diabetes. Previous clinical trials using monotherapies (i.e., single agents) have shown various degrees of success, albeit even in the best of situations partial, in terms of achieving desired therapeutic outcomes. Here, we diagram the potential, albeit a theoretical, model for the principle that combination therapies would provide a substantial improvement over singular monotherapies, with respect to preservation of β-cell mass. Mono, monotherapy.

Close modal

When and How Fast Are β-Cells Lost in Type 1 Diabetes and Is It a True Loss That Occurs or a Mere Loss of Their Function?

At odds with studies of NOD mice (Fig. 1A), it has been difficult to document insulitis in many type 1 diabetes cases during the prediabetic phase (Fig. 1B), when individuals already have clear signs of autoimmunity (51). This observation implies that attacks on β-cells likely occur in a relapsing-remitting fashion (52). As currently available biomarkers fail to indicate precisely when periods of attack occur, such periods could be missed with short-term therapies. Therefore, future trials should consider longer-term treatment periods or utilization of agents whose effects would be lasting (e.g., tolerance inducting). However, agents used for long-term treatment must also avoid adverse side effects in order to gain widespread acceptance.

It has also recently become apparent that β-cell mass does not decrease in a linear fashion (T. Rodriguez-Calvo, K. Herold, M.A.A., and M.v.H., unpublished data). Indeed, substantial β-cell mass might still be present until just prior to the time when oral glucose tolerance testing becomes abnormal. This latter observation is potentially encouraging in that more β-cells may be present than once thought prior to diagnosis. As a result, efforts to preserve β-cell mass and metabolic capacity in settings of secondary disease prevention might have more potential for success than previously assumed.

Questions also abound regarding the degree of β-cell function (or lack thereof) following diagnosis of the disease. It was previously thought that approximately 90% of β-cell mass and function are irrevocably lost by the time type 1 diabetes is diagnosed. However, we now know that strong immune suppression can result in a rather rapid recovery of β-cell function (53). Factors that contribute to reduced β-cell function at the time of diagnosis include inflammatory stress, excessive demand for insulin, and endoplasmic reticulum stress—deleterious processes that are at least in part reversible. Furthermore, as noted previously, many adults with type 1 diabetes of extended duration still retain a degree of C-peptide production (54). This realization points toward the possibility that maintenance of remaining β-cell function in adults, even many years postdiagnosis, may provide clinical benefit.

Nature of the BeastWho Attacks and Destroys the β-Cells and How Do We Need to Deal With It?

The most prominent cell found in human islets in the setting of type 1 diabetes is the CD8+ cytotoxic T cell, which is also a likely candidate to aid in β-cell killing due to its ability to recognize targets via antigen in the context of MHC class I, which is elevated in many islets in those with the disease (55). Therapeutically, lymphocytes and memory lymphocytes of the adaptive immune response can be targeted by anti–T-cell drugs, such as anti-CD3, anti-CD2 (LFA3Ig), and certain costimulatory blockers (56,57). Indeed, partial success of such compounds in recently diagnosed type 1 diabetes, defined by preservation of β-cell function (i.e., C-peptide production) over several months to years, speaks toward an important role for such autoreactive lymphocytes in β-cell destruction, at least late during the pathogenesis of type 1 diabetes.

Are β-cell antigen-specific CD8+ T cells the only factor? Certainly not, as it has become clear that general low-grade inflammation can be observed in the exocrine pancreas and inflammatory cytokines known to harm islets are also thought to be elevated during type 1 diabetes pathogenesis. Thus, anti-inflammatory therapies targeting cytokines may hold promise, and a recent trial blocking tumor necrosis factor has shown initial promise in preserving β-cells (58). These observations provide further support for the concept of combination therapies. Examples of such combinations would include an induction component using drugs targeting inflammation and T-/B-cell memory, as well as a maintenance component that could involve antigens to induce tolerance to β-cells.

One additional important question involves how the β-cell appears on the radar screen of the immune system in the first place. Is autoimmunity the primary cause, or might it be that metabolic derailment exerts stress on β-cells and in this way makes them visible to the immune system? In reality, this might at least be a contributing factor to the pathogenesis of type 1 diabetes and type 2 diabetes, as metabolic markers can precede the diagnosis of the former by several years. Considering this, priority should be given to the addition of drugs to combination therapies that stabilize and maintain β-cells and β-cell function (Fig. 3).

Within a few years, those involved in the care of persons with type 1 diabetes as well as researchers seeking to make impactful discoveries for those living with the disease will celebrate the centennial anniversary of the discovery of therapeutic insulin. Thankfully, the era since that monumental event has seen a multitude of improvements in diabetes care (59). At the same time, significant research efforts have been directed at finding the underlying cause(s) of type 1 diabetes, in large part guided by the notion of developing a means to prevent as well as provide a true “cure” for the disease. While progress has clearly been made toward understanding the initiating and sustaining events in the pathogenesis of type 1 diabetes (Fig. 2), much more investigation and discovery are needed. We believe that future attempts to prevent and/or reverse type 1 diabetes are most likely to be successful if they incorporate the recent advances in our evolving understanding of pathogenesis of the disease.

See accompanying articles, pp. 968, 971, 989, 997, 1008, 1016, 1030, and 1036.

Acknowledgments. The authors would like to thank Dr. Amanda Posgai for her remarkable editorial efforts toward publication of the manuscript.

Funding. Concepts that underlie much of the communication were developed through research supported by JDRFhttp://dx.doi.org/10.13039/100000901 (25-2013-268, 17-2012-3, and 25-2012-516, to M.A.A.), NIH National Institute of Allergy and Infectious Diseases (AI42288, to M.A.A. and M.C.-S.), NIH National Institute of Diabetes and Digestive and Kidney Diseases (DK89572, DK72473, DK104211, to A.C.P.), The Leona M. and Harry B. Helmsley Charitable Trust, the Department of Veterans Affairs, and the Vanderbilt Diabetes Research and Training Center (DK20593).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

1.
Eisenbarth
GS
.
Type I diabetes mellitus. A chronic autoimmune disease
.
N Engl J Med
1986
;
314
:
1360
1368
[PubMed]
2.
Atkinson
MA
.
George S. Eisenbarth, 1947-2012
.
Diabetologia
2013
;
56
:
435
438
[PubMed]
3.
Bluestone
JA
,
Herold
K
,
Eisenbarth
G
.
Genetics, pathogenesis and clinical interventions in type 1 diabetes
.
Nature
2010
;
464
:
1293
1300
[PubMed]
4.
Peakman
M
.
Immunological pathways to β-cell damage in Type 1 diabetes
.
Diabet Med
2013
;
30
:
147
154
[PubMed]
5.
Atkinson
MA
,
Eisenbarth
GS
,
Michels
AW
.
Type 1 diabetes
.
Lancet
2014
;
383
:
69
82
[PubMed]
6.
Skyler
JS
,
Ricordi
C
.
Stopping type 1 diabetes: attempts to prevent or cure type 1 diabetes in man
.
Diabetes
2011
;
60
:
1
8
[PubMed]
7.
Greenbaum
C
,
Atkinson
MA
.
Persistence is the twin sister of excellence: an important lesson for attempts to prevent and reverse type 1 diabetes
.
Diabetes
2011
;
60
:
693
694
[PubMed]
8.
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
[PubMed]
9.
Skyler
JS
.
Primary and secondary prevention of type 1 diabetes
.
Diabet Med
2013
;
30
:
161
169
[PubMed]
10.
Ben Nasr
M
,
D’Addio
F
,
Usuelli
V
,
Tezza
S
,
Abdi
R
,
Fiorina
P
.
The rise, fall, and resurgence of immunotherapy in type 1 diabetes
.
Pharmacol Res.
6 August
2014
[Epub ahead of print]. DOI: 10.1016/j.phrs.2014.07.004
[PubMed]
11.
Shoda
LK
,
Young
DL
,
Ramanujan
S
, et al
.
A comprehensive review of interventions in the NOD mouse and implications for translation
.
Immunity
2005
;
23
:
115
126
[PubMed]
12.
Todd
JA
,
Knip
M
,
Mathieu
C
.
Strategies for the prevention of autoimmune type 1 diabetes
.
Diabet Med
2011
;
28
:
1141
1143
[PubMed]
13.
Lernmark
A
,
Larsson
HE
.
Immune therapy in type 1 diabetes mellitus
.
Nat Rev Endocrinol
2013
;
9
:
92
103
[PubMed]
14.
Frederiksen BN, Steck AK, Kroehl M, et al. Evidence of stage- and age-related heterogeneity of non-HLA SNPs and risk of islet autoimmunity and type 1 diabetes: the Diabetes Autoimmunity Study in the Young. Clin Dev Immunol 2013;2013:417657
15.
Stankov
K
,
Benc
D
,
Draskovic
D
.
Genetic and epigenetic factors in etiology of diabetes mellitus type 1
.
Pediatrics
2013
;
132
:
1112
1122
[PubMed]
16.
Crume
TL
,
Crandell
J
,
Norris
JM
, et al
.
Timing of complementary food introduction and age at diagnosis of type 1 diabetes: the SEARCH Nutrition Ancillary Study (SNAS)
.
Eur J Clin Nutr
2014
;
68
:
1258
1260
[PubMed]
17.
de Goffau
MC
,
Fuentes
S
,
van den Bogert
B
, et al
.
Aberrant gut microbiota composition at the onset of type 1 diabetes in young children
.
Diabetologia
2014
;
57
:
1569
1577
[PubMed]
18.
Richardson
SJ
,
Leete
P
,
Dhayal
S
, et al
.
Detection of enterovirus in the islet cells of patients with type 1 diabetes: what do we learn from immunohistochemistry? Reply to Hansson SF, Korsgren S, Pontén F et al. [letter]
.
Diabetologia
2014
;
57
:
647
649
[PubMed]
19.
Arif
S
,
Leete
P
,
Nguyen
V
, et al
.
Blood and islet phenotypes indicate immunological heterogeneity in type 1 diabetes
.
Diabetes
2014
;
63
:
3835
3845
[PubMed]
20.
Chen
YG
,
Cabrera
SM
,
Jia
S
, et al
.
Molecular signatures differentiate immune states in type 1 diabetic families
.
Diabetes
2014
;
63
:
3960
3973
[PubMed]
21.
In’t Veld
P
,
Lievens
D
,
De Grijse
J
, et al
.
Screening for insulitis in adult autoantibody-positive organ donors
.
Diabetes
2007
;
56
:
2400
2404
[PubMed]
22.
Pugliese
A
,
Yang
M
,
Kusmarteva
I
, et al
.
The Juvenile Diabetes Research Foundation Network for Pancreatic Organ Donors with Diabetes (nPOD) Program: goals, operational model and emerging findings
.
Pediatr Diabetes
2014
;
15
:
1
9
[PubMed]
23.
In’t Veld
P
.
Insulitis in human type 1 diabetes: a comparison between patients and animal models
.
Semin Immunopathol
2014
;
36
:
569
579
[PubMed]
24.
Skowera
A
,
Ladell
K
,
McLaren
JE
, et al
.
β-Cell–specific CD8 T cell phenotype in type 1 diabetes reflects chronic autoantigen exposure
.
Diabetes
2015
;
64
:
916
925
[PubMed]
25.
Coppieters
KT
,
Dotta
F
,
Amirian
N
, et al
.
Demonstration of islet-autoreactive CD8 T cells in insulitic lesions from recent onset and long-term type 1 diabetes patients
.
J Exp Med
2012
;
209
:
51
60
[PubMed]
26.
Davis AK, DuBose SN, Haller MJ, et al.; TID Exchange Clinic Network. Prevalence of detectable C-peptide according to age at diagnosis and duration of type 1 diabetes. Diabetes Care 2015;38:476–481
27.
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
[PubMed]
28.
Valle
A
,
Giamporcaro
GM
,
Scavini
M
, et al
.
Reduction of circulating neutrophils precedes and accompanies type 1 diabetes
.
Diabetes
2013
;
62
:
2072
2077
[PubMed]
29.
Vaarala
O
,
Atkinson
MA
,
Neu
J
.
The “perfect storm” for type 1 diabetes: the complex interplay between intestinal microbiota, gut permeability, and mucosal immunity
.
Diabetes
2008
;
57
:
2555
2562
[PubMed]
30.
Bogdani
M
,
Korpos
E
,
Simeonovic
CJ
,
Parish
CR
,
Sorokin
L
,
Wight
TN
.
Extracellular matrix components in the pathogenesis of type 1 diabetes
.
Curr Diab Rep
2014
;
14
:
552
[PubMed]
31.
Krogvold
L
,
Edwin
B
,
Buanes
T
, et al
.
Detection of a low-grade enteroviral infection in the islets of Langerhans of living patients newly diagnosed with type 1 diabetes
.
Diabetes
2015
;
64
:
1682
1687
[PubMed]
32.
Santin
I
,
Eizirik
DL
.
Candidate genes for type 1 diabetes modulate pancreatic islet inflammation and β-cell apoptosis
.
Diabetes Obes Metab
2013
;
15
(
Suppl. 3
):
71
81
[PubMed]
33.
Tanaka
S
,
Aida
K
,
Nishida
Y
,
Kobayashi
T
.
Pathophysiological mechanisms involving aggressive islet cell destruction in fulminant type 1 diabetes
.
Endocr J
2013
;
60
:
837
845
[PubMed]
34.
Tracy
S
,
Smithee
S
,
Alhazmi
A
,
Chapman
N
.
Coxsackievirus can persist in murine pancreas by deletion of 5′ terminal genomic sequences
.
J Med Virol
2015
;
87
:
240
247
[PubMed]
35.
Xia
CQ
,
Peng
R
,
Chernatynskaya
AV
, et al
.
Increased IFN-α-producing plasmacytoid dendritic cells (pDCs) in human Th1-mediated type 1 diabetes: pDCs augment Th1 responses through IFN-α production
.
J Immunol
2014
;
193
:
1024
1034
[PubMed]
36.
Wilson
EB
,
Yamada
DH
,
Elsaesser
H
, et al
.
Blockade of chronic type I interferon signaling to control persistent LCMV infection
.
Science
2013
;
340
:
202
207
[PubMed]
37.
Sullivan
BM
,
Teijaro
JR
,
de la Torre
JC
,
Oldstone
MB
.
Early virus-host interactions dictate the course of a persistent infection
.
PLoS Pathog
2015
;
11
:
e1004588
[PubMed]
38.
Teijaro
JR
,
Ng
C
,
Lee
AM
, et al
.
Persistent LCMV infection is controlled by blockade of type I interferon signaling
.
Science
2013
;
340
:
207
211
[PubMed]
39.
Brissova
M
,
Fowler
MJ
,
Nicholson
WE
, et al
.
Assessment of human pancreatic islet architecture and composition by laser scanning confocal microscopy
.
J Histochem Cytochem
2005
;
53
:
1087
1097
[PubMed]
40.
Cabrera
O
,
Berman
DM
,
Kenyon
NS
,
Ricordi
C
,
Berggren
PO
,
Caicedo
A
.
The unique cytoarchitecture of human pancreatic islets has implications for islet cell function
.
Proc Natl Acad Sci USA
2006
;
103
:
2334
2339
[PubMed]
41.
Dai
C
,
Brissova
M
,
Hang
Y
, et al
.
Islet-enriched gene expression and glucose-induced insulin secretion in human and mouse islets
.
Diabetologia
2012
;
55
:
707
718
[PubMed]
42.
Bernal-Mizrachi
E
,
Kulkarni
RN
,
Scott
DK
,
Mauvais-Jarvis
F
,
Stewart
AF
,
Garcia-Ocaña
A
.
Human β-cell proliferation and intracellular signaling part 2: still driving in the dark without a road map
.
Diabetes
2014
;
63
:
819
831
[PubMed]
43.
Saisho
Y
,
Butler
AE
,
Manesso
E
,
Elashoff
D
,
Rizza
RA
,
Butler
PC
.
β-Cell mass and turnover in humans: effects of obesity and aging
.
Diabetes Care
2013
;
36
:
111
117
[PubMed]
44.
Butler
PC
,
Meier
JJ
,
Butler
AE
,
Bhushan
A
.
The replication of beta cells in normal physiology, in disease and for therapy
.
Nat Clin Pract Endocrinol Metab
2007
;
3
:
758
768
[PubMed]
45.
Gregg
BE
,
Moore
PC
,
Demozay
D
, et al
.
Formation of a human β-cell population within pancreatic islets is set early in life
.
J Clin Endocrinol Metab
2012
;
97
:
3197
3206
[PubMed]
46.
Williams
AJ
,
Thrower
SL
,
Sequeiros
IM
, et al
.
Pancreatic volume is reduced in adult patients with recently diagnosed type 1 diabetes
.
J Clin Endocrinol Metab
2012
;
97
:
E2109
E2113
[PubMed]
47.
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
[PubMed]
48.
Talchai
C
,
Xuan
S
,
Lin
HV
,
Sussel
L
,
Accili
D
.
Pancreatic β cell dedifferentiation as a mechanism of diabetic β cell failure
.
Cell
2012
;
150
:
1223
1234
[PubMed]
49.
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
[PubMed]
50.
Keenan
HA
,
Sun
JK
,
Levine
J
, et al
.
Residual insulin production and pancreatic β-cell turnover after 50 years of diabetes: Joslin Medalist Study
.
Diabetes
2010
;
59
:
2846
2853
[PubMed]
51.
In’t Veld
P
.
Insulitis in human type 1 diabetes: the quest for an elusive lesion
.
Islets
2011
;
3
:
131
138
[PubMed]
52.
von Herrath
M
,
Sanda
S
,
Herold
K
.
Type 1 diabetes as a relapsing-remitting disease?
Nat Rev Immunol
2007
;
7
:
988
994
[PubMed]
53.
Couri
CE
,
Oliveira
MC
,
Stracieri
AB
, et al
.
C-peptide levels and insulin independence following autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus
.
JAMA
2009
;
301
:
1573
1579
[PubMed]
54.
Haller
MJ
,
Gitelman
SE
,
Gottlieb
PA
, et al
.
Anti-thymocyte globulin/G-CSF treatment preserves beta cell function in patients with established type 1 diabetes
.
J Clin Invest
2015
;
125
:
448
455
[PubMed]
55.
Richardson
SJ
,
Willcox
A
,
Bone
AJ
,
Morgan
NG
,
Foulis
AK
.
Immunopathology of the human pancreas in type-I diabetes
.
Semin Immunopathol
2011
;
33
:
9
21
[PubMed]
56.
Rigby
MR
,
DiMeglio
LA
,
Rendell
MS
, et al.;
T1DAL Study Team
.
Targeting of memory T cells with alefacept in new-onset type 1 diabetes (T1DAL study): 12 month results of a randomised, double-blind, placebo-controlled phase 2 trial
.
Lancet Diabetes Endocrinol
2013
;
1
:
284
294
[PubMed]
57.
Pope
JE
,
Rampakakis
E
,
Sampalis
J
.
The durability of abatacept as a first and subsequent biologic and improvement in HAQ from a large multi-site real-world study
.
Semin Arthritis Rheum.
28 September
2014
[Epub ahead of print]. DOI: 10.1016/j.semarthrit.2014.09.00
[PubMed]
58.
Nepom
GT
,
Ehlers
M
,
Mandrup-Poulsen
T
.
Anti-cytokine therapies in T1D: concepts and strategies
.
Clin Immunol
2013
;
149
:
279
285
[PubMed]
59.
Cefalu
WT
,
Boulton
AJ
,
Tamborlane
WV
, et al
.
Status of Diabetes Care: “It just doesn’t get any better . . . or does it?”
Diabetes Care
2014
;
37
:
1782
1785
[PubMed]
60.
Chase
HP
,
Lescheck
E
,
Rafkin-Mervis
L
, et al
.; Type 1 Diabetes TrialNet NIP Study Group.
Nutritional Intervention to Prevent (NIP) Type 1 Diabetes A Pilot Trial. ICAN
2009
;
1
:
98
107
61.
Knip
M
,
Virtanen
SM
,
Seppä
K
, et al.;
Finnish TRIGR Study Group
.
Dietary intervention in infancy and later signs of beta-cell autoimmunity
.
N Engl J Med
2010
;
363
:
1900
1908
[PubMed]
62.
Hummel
S
,
Pflüger
M
,
Hummel
M
,
Bonifacio
E
,
Ziegler
AG
.
Primary dietary intervention study to reduce the risk of islet autoimmunity in children at increased risk for type 1 diabetes: the BABYDIET study
.
Diabetes Care
2011
;
34
:
1301
1305
[PubMed]
63.
Knip
M
,
Åkerblom
HK
,
Becker
D
, et al.;
TRIGR Study Group
.
Hydrolyzed infant formula and early β-cell autoimmunity: a randomized clinical trial
.
JAMA
2014
;
311
:
2279
2287
[PubMed]
64.
Vaarala
O
,
Ilonen
J
,
Ruohtula
T
, et al
.
Removal of bovine insulin from cow’s milk formula and early initiation of beta-cell autoimmunity in the FINDIA Pilot Study
.
Arch Pediatr Adolesc Med
2012
;
166
:
608
614
[PubMed]
65.
Lampeter
EF
,
Klinghammer
A
,
Scherbaum
WA
, et al.;
DENIS Group
.
The Deutsche Nicotinamide Intervention Study: an attempt to prevent type 1 diabetes
.
Diabetes
1998
;
47
:
980
984
[PubMed]
66.
Carel JC, Landais P, Bougnères P.
Therapy to prevent type 1 diabetes mellitus [letter]
.
N Engl J Med
2002
;
347
:
1115
1116
67.
Harrison
LC
,
Honeyman
MC
,
Steele
CE
, et al
.
Pancreatic beta-cell function and immune responses to insulin after administration of intranasal insulin to humans at risk for type 1 diabetes
.
Diabetes Care
2004
;
27
:
2348
2355
[PubMed]
68.
Gale
EA
,
Bingley
PJ
,
Emmett
CL
,
Collier
T
;
European Nicotinamide Diabetes Intervention Trial (ENDIT) Group
.
European Nicotinamide Diabetes Intervention Trial (ENDIT): a randomised controlled trial of intervention before the onset of type 1 diabetes
.
Lancet
2004
;
363
:
925
931
[PubMed]
69.
Skyler
JS
,
Krischer
JP
,
Wolfsdorf
J
, et al
.
Effects of oral insulin in relatives of patients with type 1 diabetes: the Diabetes Prevention Trial—Type 1
.
Diabetes Care
2005
;
28
:
1068
1076
[PubMed]
70.
Näntö-Salonen
K
,
Kupila
A
,
Simell
S
, et al
.
Nasal insulin to prevent type 1 diabetes in children with HLA genotypes and autoantibodies conferring increased risk of disease: a double-blind, randomised controlled trial
.
Lancet
2008
;
372
:
1746
1755
[PubMed]
71.
Vandemeulebroucke
E
,
Gorus
FK
,
Decochez
K
, et al.;
Belgian Diabetes Registry
.
Insulin treatment in IA-2A-positive relatives of type 1 diabetic patients
.
Diabetes Metab
2009
;
35
:
319
327
[PubMed]
72.
Bougneres
PF
,
Carel
JC
,
Castano
L
, et al
.
Factors associated with early remission of type I diabetes in children treated with cyclosporine
.
N Engl J Med
1988
;
318
:
663
670
[PubMed]
73.
Shah
SC
,
Malone
JI
,
Simpson
NE
.
A randomized trial of intensive insulin therapy in newly diagnosed insulin-dependent diabetes mellitus
.
N Engl J Med
1989
;
320
:
550
554
[PubMed]
74.
Pozzilli
P
,
Visalli
N
,
Cavallo
MG
, et al
.
Vitamin E and nicotinamide have similar effects in maintaining residual beta cell function in recent onset insulin-dependent diabetes (the IMDIAB IV study)
.
Eur J Endocrinol
1997
;
137
:
234
239
[PubMed]
75.
Visalli
N
,
Cavallo
MG
,
Signore
A
, et al
.
A multi-centre randomized trial of two different doses of nicotinamide in patients with recent-onset type 1 diabetes (the IMDIAB VI)
.
Diabetes Metab Res Rev
1999
;
15
:
181
185
[PubMed]
76.
Chaillous
L
,
Lefèvre
H
,
Thivolet
C
, et al
.
Oral insulin administration and residual beta-cell function in recent-onset type 1 diabetes: a multicentre randomised controlled trial. Diabète Insuline Orale group
.
Lancet
2000
;
356
:
545
549
[PubMed]
77.
Raz
I
,
Elias
D
,
Avron
A
,
Tamir
M
,
Metzger
M
,
Cohen
IR
.
Beta-cell function in new-onset type 1 diabetes and immunomodulation with a heat-shock protein peptide (DiaPep277): a randomised, double-blind, phase II trial
.
Lancet
2001
;
358
:
1749
1753
[PubMed]
78.
Herold
KC
,
Hagopian
W
,
Auger
JA
, et al
.
Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus
.
N Engl J Med
2002
;
346
:
1692
1698
[PubMed]
79.
Ortqvist
E
,
Björk
E
,
Wallensteen
M
, et al
.
Temporary preservation of beta-cell function by diazoxide treatment in childhood type 1 diabetes
.
Diabetes Care
2004
;
27
:
2191
2197
[PubMed]
80.
Saudek
F
,
Havrdova
T
,
Boucek
P
,
Karasova
L
,
Novota
P
,
Skibova
J
.
Polyclonal anti-T-cell therapy for type 1 diabetes mellitus of recent onset
.
Rev Diabet Stud
2004
;
1
:
80
88
[PubMed]
81.
Crinò
A
,
Schiaffini
R
,
Manfrini
S
, et al.;
IMDIAB group
.
A randomized trial of nicotinamide and vitamin E in children with recent onset type 1 diabetes (IMDIAB IX)
.
Eur J Endocrinol
2004
;
150
:
719
724
[PubMed]
82.
Crinó
A
,
Schiaffini
R
,
Ciampalini
P
, et al.;
IMDIAB Group
.
A two year observational study of nicotinamide and intensive insulin therapy in patients with recent onset type 1 diabetes mellitus
.
J Pediatr Endocrinol Metab
2005
;
18
:
749
754
[PubMed]
83.
Keymeulen
B
,
Vandemeulebroucke
E
,
Ziegler
AG
, et al
.
Insulin needs after CD3-antibody therapy in new-onset type 1 diabetes
.
N Engl J Med
2005
;
352
:
2598
2608
[PubMed]
84.
Herold
KC
,
Gitelman
SE
,
Masharani
U
, et al
.
A single course of anti-CD3 monoclonal antibody hOKT3gamma1(Ala-Ala) results in improvement in C-peptide responses and clinical parameters for at least 2 years after onset of type 1 diabetes
.
Diabetes
2005
;
54
:
1763
1769
[PubMed]
85.
Pitocco
D
,
Crinò
A
,
Di Stasio
E
, et al.;
IMDIAB Group
.
The effects of calcitriol and nicotinamide on residual pancreatic beta-cell function in patients with recent-onset Type 1 diabetes (IMDIAB XI)
.
Diabet Med
2006
;
23
:
920
923
[PubMed]
86.
Huurman
VA
,
Decochez
K
,
Mathieu
C
,
Cohen
IR
,
Roep
BO
.
Therapy with the hsp60 peptide DiaPep277 in C-peptide positive type 1 diabetes patients
.
Diabetes Metab Res Rev
2007
;
23
:
269
275
[PubMed]
87.
Voltarelli
JC
,
Couri
CE
,
Stracieri
AB
, et al
.
Autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus
.
JAMA
2007
;
297
:
1568
1576
[PubMed]
88.
Lazar
L
,
Ofan
R
,
Weintrob
N
, et al
.
Heat-shock protein peptide DiaPep277 treatment in children with newly diagnosed type 1 diabetes: a randomised, double-blind phase II study
.
Diabetes Metab Res Rev
2007
;
23
:
286
291
[PubMed]
89.
Schloot
NC
,
Meierhoff
G
,
Lengyel
C
, et al
.
Effect of heat shock protein peptide DiaPep277 on beta-cell function in paediatric and adult patients with recent-onset diabetes mellitus type 1: two prospective, randomized, double-blind phase II trials
.
Diabetes Metab Res Rev
2007
;
23
:
276
285
[PubMed]
90.
Ludvigsson
J
,
Faresjö
M
,
Hjorth
M
, et al
.
GAD treatment and insulin secretion in recent-onset type 1 diabetes
.
N Engl J Med
2008
;
359
:
1909
1920
[PubMed]
91.
Herold
KC
,
Gitelman
S
,
Greenbaum
C
, et al.;
Immune Tolerance Network ITN007AI Study Group
.
Treatment of patients with new onset type 1 diabetes with a single course of anti-CD3 mAb teplizumab preserves insulin production for up to 5 years
.
Clin Immunol
2009
;
132
:
166
173
[PubMed]
92.
Walter
M
,
Philotheou
A
,
Bonnici
F
,
Ziegler
AG
,
Jimenez
R
;
NBI-6024 Study Group
.
No effect of the altered peptide ligand NBI-6024 on beta-cell residual function and insulin needs in new-onset type 1 diabetes
.
Diabetes Care
2009
;
32
:
2036
2040
[PubMed]
93.
Pescovitz
MD
,
Greenbaum
CJ
,
Krause-Steinrauf
H
, et al.;
Type 1 Diabetes TrialNet Anti-CD20 Study Group
.
Rituximab, B-lymphocyte depletion, and preservation of beta-cell function
.
N Engl J Med
2009
;
361
:
2143
2152
[PubMed]
94.
Mastrandrea
L
,
Yu
J
,
Behrens
T
, et al
.
Etanercept treatment in children with new-onset type 1 diabetes: pilot randomized, placebo-controlled, double-blind study
.
Diabetes Care
2009
;
32
:
1244
1249
[PubMed]
95.
Keymeulen
B
,
Walter
M
,
Mathieu
C
, et al
.
Four-year metabolic outcome of a randomised controlled CD3-antibody trial in recent-onset type 1 diabetic patients depends on their age and baseline residual beta cell mass
.
Diabetologia
2010
;
53
:
614
623
[PubMed]
96.
Radtke
MA
,
Nermoen
I
,
Kollind
M
, et al
.
Six months of diazoxide treatment at bedtime in newly diagnosed subjects with type 1 diabetes does not influence parameters of beta-cell function and autoimmunity but improves glycemic control
.
Diabetes Care
2010
;
33
:
589
594
[PubMed]
97.
Walter
M
,
Kaupper
T
,
Adler
K
,
Foersch
J
,
Bonifacio
E
,
Ziegler
AG
.
No effect of the 1alpha,25-dihydroxyvitamin D3 on beta-cell residual function and insulin requirement in adults with new-onset type 1 diabetes
.
Diabetes Care
2010
;
33
:
1443
1448
[PubMed]
98.
Sherry
N
,
Hagopian
W
,
Ludvigsson
J
, et al.;
Protégé Trial Investigators
.
Teplizumab for treatment of type 1 diabetes (Protégé study): 1-year results from a randomised, placebo-controlled trial
.
Lancet
2011
;
378
:
487
497
[PubMed]
99.
Wherrett
DK
,
Bundy
B
,
Becker
DJ
, et al.;
Type 1 Diabetes TrialNet GAD Study Group
.
Antigen-based therapy with glutamic acid decarboxylase (GAD) vaccine in patients with recent-onset type 1 diabetes: a randomised double-blind trial
.
Lancet
2011
;
378
:
319
327
[PubMed]
100.
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
[PubMed]
101.
Martin
S
,
Herder
C
,
Schloot
NC
, et al.;
DIATOR Study Group
.
Residual beta cell function in newly diagnosed type 1 diabetes after treatment with atorvastatin: the Randomized DIATOR Trial
.
PLoS One
2011
;
6
:
e17554
[PubMed]
102.
Snarski
E
,
Milczarczyk
A
,
Torosian
T
, et al
.
Independence of exogenous insulin following immunoablation and stem cell reconstitution in newly diagnosed diabetes type I
.
Bone Marrow Transplant
2011
;
46
:
562
566
[PubMed]
103.
Ludvigsson
J
,
Krisky
D
,
Casas
R
, et al
.
GAD65 antigen therapy in recently diagnosed type 1 diabetes mellitus
.
N Engl J Med
2012
;
366
:
433
442
[PubMed]
104.
Strom
A
,
Kolb
H
,
Martin
S
, et al.;
DIATOR Study Group
.
Improved preservation of residual beta cell function by atorvastatin in patients with recent onset type 1 diabetes and high CRP levels (DIATOR trial)
.
PLoS One
2012
;
7
:
e33108
[PubMed]
105.
Li
L
,
Shen
S
,
Ouyang
J
, et al
.
Autologous hematopoietic stem cell transplantation modulates immunocompetent cells and improves β-cell function in Chinese patients with new onset of type 1 diabetes
.
J Clin Endocrinol Metab
2012
;
97
:
1729
1736
[PubMed]
106.
Gu
W
,
Hu
J
,
Wang
W
, et al
.
Diabetic ketoacidosis at diagnosis influences complete remission after treatment with hematopoietic stem cell transplantation in adolescents with type 1 diabetes
.
Diabetes Care
2012
;
35
:
1413
1419
[PubMed]
107.
Long
SA
,
Rieck
M
,
Sanda
S
, et al.;
Diabetes TrialNet and the Immune Tolerance Network
.
Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs β-cell function
.
Diabetes
2012
;
61
:
2340
2348
[PubMed]
108.
Moran
A
,
Bundy
B
,
Becker
DJ
, et al.;
Type 1 Diabetes TrialNet Canakinumab Study Group
;
AIDA Study Group
.
Interleukin-1 antagonism in type 1 diabetes of recent onset: two multicentre, randomised, double-blind, placebo-controlled trials
.
Lancet
2013
;
381
:
1905
1915
[PubMed]