Islet cells in human type 1 diabetes: from recent advances to novel therapies – a symposium-based roadmap for future research

in Journal of Endocrinology
Authors:
J Cantley School of Medicine, University of Dundee, Dundee, United Kingdom of Great Britain and Northern Ireland

Search for other papers by J Cantley in
Current site
Google Scholar
PubMed
Close
https://orcid.org/0000-0003-2509-1271
,
D L Eizirik ULB Center for Diabetes Research, Université Libre de Bruxelles Faculté de Médecine, Bruxelles, Belgium

Search for other papers by D L Eizirik in
Current site
Google Scholar
PubMed
Close
,
E Latres JDRF International, New York, NY, USA

Search for other papers by E Latres in
Current site
Google Scholar
PubMed
Close
,
C M Dayan Cardiff University School of Medicine, Cardiff, United Kingdom of Great Britain and Northern Ireland

Search for other papers by C M Dayan in
Current site
Google Scholar
PubMed
Close
, and
the JDRF-DiabetesUK-INNODIA-nPOD Stockholm Symposium 2022
Search for other papers by the JDRF-DiabetesUK-INNODIA-nPOD Stockholm Symposium 2022 in
Current site
Google Scholar
PubMed
Close
the JDRF-DiabetesUK-INNODIA-nPOD Stockholm Symposium 2022

Correspondence should be addressed to J Cantley: JCantley001@dundee.ac.uk
Open access

Sign up for journal news

There is a growing understanding that the early phases of type 1 diabetes (T1D) are characterised by a deleterious dialogue between the pancreatic beta cells and the immune system. This, combined with the urgent need to better translate this growing knowledge into novel therapies, provided the background for the JDRF–DiabetesUK–INNODIA–nPOD symposium entitled ‘Islet cells in human T1D: from recent advances to novel therapies’, which took place in Stockholm, Sweden, in September 2022. We provide in this article an overview of the main themes addressed in the symposium, pointing to both promising conclusions and key unmet needs that remain to be addressed in order to achieve better approaches to prevent or reverse T1D.

Abstract

There is a growing understanding that the early phases of type 1 diabetes (T1D) are characterised by a deleterious dialogue between the pancreatic beta cells and the immune system. This, combined with the urgent need to better translate this growing knowledge into novel therapies, provided the background for the JDRF–DiabetesUK–INNODIA–nPOD symposium entitled ‘Islet cells in human T1D: from recent advances to novel therapies’, which took place in Stockholm, Sweden, in September 2022. We provide in this article an overview of the main themes addressed in the symposium, pointing to both promising conclusions and key unmet needs that remain to be addressed in order to achieve better approaches to prevent or reverse T1D.

Introduction

Type 1 diabetes (T1D) is a chronic autoimmune disease with a strong inflammatory component that results from the interaction between predisposing genes and environmental factors (e.g. viral infections and diet). This interaction triggers an autoimmune assault against pancreatic beta cells that provokes local inflammation (insulitis) and progressive beta cell functional impairment as well as the loss due to apoptosis. The prevalence of T1D has doubled in 25 years in children, and people living with the disease lose on average ≥11 years of life expectancy (Livingstone et al. 2015, Gregory et al. 2022). Although interventions targeting the immune system have shown efficacy in delaying disease onset (Herold et al. 2019), there are presently no therapies that prevent or cure the disease. The limited success of drugs aiming to arrest/revert autoimmunity and to protect beta cells in T1D is mainly due to the poor understanding of the molecular pathogenesis driving the autoimmune destruction of human beta cells, as well as the heterogeneity of pathogenesis resulting in different patient ‘endotypes’ (Bluestone et al. 2021, Eizirik et al. 2021). Immune-mediated inflammation (‘initiation phase’) contributes both to the primary induction and secondary amplification of the immune assault, and inflammatory mediators trigger functional suppression and apoptosis of beta cells. This takes place in the context of a ‘dialogue’ between invading immune cells and the target beta cells, which is modulated by T1D candidate genes acting both on the immune system and on the beta cells (Polychronakos & Li 2011, Bergholdt et al. 2012, Eizirik et al. 2012, Eizirik et al. 2020). This dialogue is mediated by cytokines/chemokines released by beta cells, immune cells and by putative immunogenic signals delivered by dying or ‘altered’ beta cells. Immune cells – particularly CD8+ T cells that recognise antigens presented in the context of conventional human leukocyte antigen class I (HLA-I) – mediate beta cell apoptosis. Beta cells contribute to this process by upregulating the expression of the HLA-I molecules. The upregulated HLA-I may present neoantigens generated as a consequence of beta cell endoplasmic reticulum (ER) stress, posttranslational modifications, changes in alternative splicing (AS) and/or other mechanisms (Gonzalez-Duque et al. 2018, Azoury et al. 2020, Piganelli et al. 2020, Roep et al. 2021). The pathogenesis of T1D has been proposed to consist of stages 1–3, preceded by a putative stage 0 (prior to seroconversion), as depicted in Fig. 1. It is important to state that not all individuals at stage 1 (autoantibody positive) will progress to stage 3 (hyperglycaemia and clinical diagnosis), and there is heterogeneity in disease progression and presentation.

Figure 1
Figure 1

Progression to type 1 diabetes (T1D). Risk factors for T1D include genetic predisposition and environmental factors, such as viral infections and diet. Although the clinical diagnosis of T1D usually occurs at stage 3 when beta cell mass is depleted and hyperglycaemia is established, several stages precede this. There is increasing recognition of stage 0, in which beta cell dysfunction and impaired glycaemic control may occur in the absence of autoantibodies. Stage 1 represents seroconversion with the engagement of the adaptive immune system and the appearance of autoantibodies. During stage 2 beta cell dysfunction and apoptosis are increased impairing glycaemic control, leading to clinical diagnosis at stage 3. An important concept discussed is the reciprocal interaction between the beta cell and immune system throughout disease progression, along with interperson heterogeneity influenced by factors such as age of onset. Figure created with BioRender.com.

Citation: Journal of Endocrinology 259, 1; 10.1530/JOE-23-0082

The rationale for the JDRF–DiabetesUK–INNODIA–nPOD symposium entitled ‘Islet cells in human T1D: from recent advances to novel therapies’, which took place in Stockholm, Sweden, between September 23 and 25, 2022, was provided by the following observations: (i) the key role played by the deleterious dialogue between the immune system and the pancreatic beta cells in triggering the autoimmune process in T1D (Fig. 1); (ii) the urgent need to better translate this growing knowledge into novel (and probably combined) therapies for the disease, including better alignment of preclinical research with these goals. The main points discussed in the symposium are outlined later, culminating in a series of conclusions that should hopefully serve as a roadmap for future research in the field, with the ultimate goal to prevent the progressive beta cell dysfunction and loss characteristic of the disease.

The progressive beta cell immune system dialogue in T1D and functional follow-up in clinical trials

The meeting started with an overview lecture provided by Chantal Mathieu, emphasising the importance of the dialogue between beta cells and the immune system and the role of chemokines in this process. The understanding that beta cells are not simply inert victims of the immune system, but contribute to the natural history of the disease (Martens et al. 2021), is promoting the search for combined therapies to prevent or revert T1D, aiming to both ‘re-educate’ the immune system and boost the beta cell’s resistance to the immune assault. This provides the basis for novel and planned clinical trials implemented by the European consortium INNODIA (https://www.innodia.eu/).

Human islets in health and disease: the phenotype of human pancreatic beta cells in T1D

Mark Atkinson presented a lecture titled, ‘The Network for Pancreatic Organ Donors with Diabetes (nPOD) experience: histology of beta cells in T1D’ where he discussed the impact that improvements to existing technologies (e.g. confocal microscopy and light sheet fluorescent microscopy), new image analysis methods and the development of novel technologies (e.g. Co-Detection by Imaging (CODEX), Cytometry by Time of Flight (CyTOF)) have made on our mechanistic understanding of T1D pathogenesis. These technologies, combined with the availability of extremely rare and valuable tissues from individuals with or at increased risk of T1D, have led to many discoveries that might be considered ‘transformational’ in terms of their thought. Indeed, these recent advancements have uncovered multiple previously undescribed physiological contributors to T1D, many related to aspects of not only the endocrine but extending to the exocrine pancreas. To be clear, evidence for the autoimmune component in T1D remains strong, including the presentation of a moving 3D image of an nPOD donor (20-year-old T1D subject with 7 months disease duration) stained for glucagon and CD3, clearly demonstrating the presence of immune cells within the islet, interspersed amongst endocrine cells (Fig. 2).

Figure 2
Figure 2

Visualising insulitis in three dimensions. Image of a 3D reconstruction of an nPOD donor (20-year-old T1D subject with 7 months disease duration) stained for glucagon (blue) and CD3 (yellow), revealing the presence of infiltrating immune cells within the islet of Langerhans, interspersed with endocrine cells. 3D reconstruction was performed using Arivis Vision4D.

Citation: Journal of Endocrinology 259, 1; 10.1530/JOE-23-0082

Klaus Kaestner discussed the ‘Transcriptomics of beta cells from T1D pancreata’ and the role played by the Human Pancreas Analysis Programme (HPAP), a consortium undertaking deep phenotyping of the human endocrine pancreas and its interaction with the immune system. HPAP generates and distributes high-value data sets for the diabetes research community (Kaestner et al. 2019). Pancreatic and immune tissues undergo histology, multi-omics and cellular studies before data are integrated into PancDB (https://hpap.pmacs.upenn.edu/). The programme has made a rapid impact in the field (Wang et al. 2019, Fasolino et al. 2020, Japp et al. 2021, Bosi et al. 2022, Doliba et al. 2022, Fasolino et al. 2022). HPAP single-cell multi-omics is enabling new insights into pancreatic subpopulations in the T1D pancreas. For example, these data enabled the recent identification of major histocompatibility complex (MHC)-class II transcripts in a novel subset of pancreatic ductal epithelial cells during T1D, with a tolerogenic dendritic cell gene expression profile suggesting attempted immune regulation (Fasolino et al. 2022). Dr Kaestner went on to discuss alpha cell hypersecretion of glucagon from islets in non-diabetic glutamic acid decarboxylase (GAD) autoantibody-positive individuals: single-cell RNAseq (scRNAseq) has identified upregulation of alpha cell glycolytic genes, which may underpin the augmented glucagon release in the GADA+ pancreas (Doliba et al. 2022), consistent with other mechanistic studies by the group (Bahl et al. 2021). The lecture concluded with a recent transcriptional analysis of residual beta cells in the T1D pancreas (Wang et al. 2019).

Marcela Brissova considered the role of ‘islet cell physiology in T1D’, a disease of dysregulated glucose homeostasis. Currently, all forms of diabetes are diagnosed by elevated blood glucose. The failure of clinical diagnosis and treatment to incorporate molecular or genetic features of diabetes is a limiting factor in building efforts towards precision medicine and disease prevention. To bridge this gap, several resources in the USA are generating multimodal data from the human pancreas or islets supported by robust organ donation mechanisms. These resources include HPAP (Kaestner et al. 2019) (https://hpap.pmacs.upenn.edu/), nPOD (https://www.jdrfnpod.org), Human Islet Phenotyping Program of IIDP (Brissova et al. 2019) (https://iidp.coh.org), Human BioMolecular Atlas Program (2019) (https://hubmapconsortium.org) and the Human Atlas of the Neonatal and Early Life Pancreas (https://pancreatlas.org/datasets/531/overview). Dr Brissova and her colleagues at Vanderbilt University participate on various levels in all five programs, four of which are currently connected via the data-agnostic Pancreatlas platform (Saunders et al. 2020) (https://pancreatlas.org/), a curated resource to share imaging datasets developed by the Vanderbilt team. To integrate functional, genomic and genetic data with key donor characteristics across age, sex, ancestry and disease state, a robust islet phenotyping pipeline was developed (Brissova et al. 2018, Hart et al. 2018, Haliyur et al. 2019, Haliyur et al. 2021, Doliba et al. 2022) that allowed analyses of human islet preparations from >600 donors thus far including adult and paediatric individuals with and without T1D (Brissova et al. 2018, Haliyur et al. 2021). These approaches revealed that endocrine cell composition varies considerably in adult islets and is highly associated with hormone secretion, possibly influencing alpha–beta–delta cell paracrine interactions. In addition, the team found that beta cell composition and insulin secretion are influenced by sex and self-reported ancestry. During postnatal development, islet endocrine cell composition changes considerably and both beta and alpha cells undergo functional maturation, thus further raising questions about gene(s) and environmental factors influencing endocrine cell allocation, beta cell mass and diabetes predisposition. Studies of isolated islets from individuals diagnosed with T1D (stage 3) showed that residual beta cells have relatively normal function, whereas alpha cells have an intrinsic defect resulting in dysregulated glucagon secretion. Brissova postulated that several mechanisms could be responsible for alpha cell dysfunction in T1D such as extreme beta cell loss, chronic hyperglycaemia or repeated hypoglycaemia leading to changes in alpha cell state mediated by alterations in the expression of key islet-enriched transcription factors (TFs) (Brissova et al. 2018, Wang et al. 2019, Camunas-Soler et al. 2020). Recent work using islets from non-diabetic adult single GAD autoantibody-positive individuals led to a surprising discovery that alpha cells appear to have dysregulated secretory output prior to symptomatic T1D (Doliba et al. 2022). Collectively, these insights demonstrate that T1D is a complex disease requiring integrated studies by collaborative interdisciplinary teams to bring molecular insight into heterogeneity across multiple levels to achieve precision medicine in diagnosis and management. Integrating cellular function with molecular signatures and genetics, whilst fostering interactions of basic scientists and clinicians could inform future clinical trials aiming at T1D prevention.

‘What do the islets look like in T1D?’ To address this question, Sarah Richardson reflected on the value of pancreas biobanks in which T1D islets can be examined in their native environment. Globally there are <700 T1D donor pancreata, with the majority accessible today residing within the nPOD and the Exeter Archival Diabetes Biobanks (EADB; https://pancreatlas.org/datasets/960/explore). Advanced technologies such as scRNAseq and CyToF are providing exciting and informative insights into the pathology of T1D (Damond et al. 2019, Wang et al. 2019) but are frequently restricted to a small subset of individuals diagnosed between 10 and 30 years of age, with a duration of diabetes of >2 years. Donors with young-onset (age <10 years) and short duration (<2 years); those with long duration (>35 years); and those with late-onset (age >30 years) receive less attention due to limited access. The need to study ‘all’ groups is highlighted by the variation in hallmark T1D-related pathologies, which differ depending on ‘who you look in’ and ‘when you look’.

Collating data from EADB and nPOD to examine the presence of residual beta cells reveals proportionally fewer insulin-containing islets (ICIs) in donors diagnosed at age <7 years at the onset, with residual beta cells declining rapidly beyond this. In contrast, those diagnosed older (age ≥13 years) tend to have more residual ICIs at onset which can be preserved for many years post-diagnosis (Leete et al. 2016, Carr et al. 2022). Encouragingly, these data mirror circulating C-peptide levels observed in living individuals with comparable age-at-onset and disease duration (Carr et al. 2022), suggesting that C-peptide will be a valuable window into the pancreas in future intervention trials. The presence of insulitis (accumulation of infiltrating immune cells at the islet) is relatively rare in T1D pancreata (particularly with disease duration >1 year), being described as an ‘elusive lesion’ by Peter In’t Veld (In't Veld 2011). In those with a disease duration of <1 year, insulitis is more commonly found in those diagnosed young, highlighting again the importance of studying young-onset, short-duration donors to improve our understanding. Studies of short-duration donors reveal two forms of the disease, or endotypes: T1D Endotype 1(T1DE1) and 2(T1DE2) (Leete et al. 2016) (Fig. 3). These endotypes differ according to insulitis severity, infiltrating immune-cell profiles (particularly B-lymphocytes), the proportion of the residual beta cells and proinsulin localisation within the beta cells. T1DE1 is associated with young age-at-onset (age <7 years) and a rapid loss of circulating insulin, whereas individuals diagnosed ≥13 years of age (T1DE2) often retain detectable C-peptide long after diagnosis and retain normal proinsulin localisation in most islets. Individuals diagnosed later tend to maintain better blood glucose control and develop fewer complications, which aligns with these pancreatic studies where more beta cells are preserved. Interestingly, in these T1DE2 donors, there is evidence that beta cells may be defending themselves against the immune attack, through the upregulation of non-classical class I HLA molecules, such as HLA-E, -F and -G and the checkpoint inhibitor PD-L1 (Richardson et al. 2016, Wyatt et al. 2019, Colli et al. 2020a ). These can all negatively regulate, or inhibit, immune cells and may protect beta cells from bystander effects of infiltrating cells and reduce auto-reactive T-cell killing. This could be informative for beta cell replacement strategies in the future.

Figure 3
Figure 3

Summary of pancreas Type 1 diabetes endotypes (T1DE). Images of pancreatic sections from a T1DE1 (left panel) or T1DE2 (right panel) donor. Low magnification images of pancreas sections (left inset panels, 1 cm scale bars) with each islet annotated as being an uninflamed Insulin-containing islet (ICI) – red; inflamed, defined as >15 CD45+ immune cells/ islet, ICI – green; inflamed insulin-deficient islet (IDI) – blue; or uninflamed IDI – pink. T1DE1 donors have evidence of more inflammation (green and blue islet annotations) and fewer residual insulin-containing islets (red/green islet annotations) than individuals with T1DE2. The higher magnification images (right inset panels, 500 µm and 100 µm scale bars) demonstrate the presence of peri-insulitis and invasive insulitis in ICIs of a T1DE1 donor, and an example of low level peri-insulitis observed in a T1DE2 donor. Further immune and beta cell characteristics of the endotypes are described in the boxes.

Citation: Journal of Endocrinology 259, 1; 10.1530/JOE-23-0082

Carmella Evans-Molina opened her lecture on ‘in vivo beta cell function in pre-clinical T1D diagnostics’ by discussing the analysis of individuals with autoantibody positivity from cohorts such as the TrialNet Pathway to Prevention study and Diabetes Prevention Trial of T1D (DPT-1) (Ferrannini et al. 2010, Evans-Molina et al. 2018), which suggests there are several predictable metabolic checkpoints that are observed during the progression of T1D. Initially, there is a loss of early C-peptide responses, as well as reduced C-peptide area under the curve responses during oral glucose tolerance tests (OGTT) that can be detected up to 6 years prior to the onset of Stage 3 T1D (Evans-Molina et al. 2018). An important caveat is that individuals from these cohorts are identified as antibody positive in cross-sectional screening, so their exact timing of seroconversion is unknown. Nonetheless, during a second phase that continues until approximately 2 years before diagnosis, there is relative stability in OGTT C-peptide measures (Evans-Molina et al. 2018). Beginning about 2 years before diabetes onset, there is evidence of rising glycaemia and declining C-peptide secretion. Metabolic deterioration accelerates 1 year to 6 months prior to Stage 3 T1D onset, with marked declines in beta cell glucose sensitivity, rate sensitivity and potentiation coupled with decreased insulin sensitivity, and markedly rising blood glucose levels (Ferrannini et al. 2010, Evans-Molina et al. 2018). Furthermore, data published following the symposium revealed elevations in blood glucose prior to the appearance of islet autoantibodies (prior to stages 1–2) in children with a high genetic risk score for T1D (Warncke et al. 2022).

Dr Evans-Molina went on to consider age as an important modifier of T1D risk, as it is well-accepted that age is inversely associated with the risk of developing T1D in those with autoantibodies (Anand et al. 2021). Whether age influences patterns of C-peptide decline before and after the onset of Stage 3 disease is a critical question, especially as it relates to the selection of individuals for disease-modifying interventions and the timing of these interventions during periods of active, accelerated disease. In a recent analysis of DPT-1 data, Evans-Molina showed that younger and older progressors to T1D had a similar rate of decline of both beta cell glucose sensitivity and insulin sensitivity prior to the onset of Stage 3 T1D. This stands in contrast to the patterns observed after the onset of Stage 3 T1D where children manifest a more rapid loss of C-peptide following clinical diagnosis (Greenbaum et al. 2012, Hao et al. 2016). Consistent with these findings, measurable serum C-peptide and histologic evidence of residual insulin-containing islets in long-duration disease are less likely to be observed in individuals with younger age of onset (Davis et al. 2015, Lam et al. 2017, McKeigue et al. 2019, Oram et al. 2019).

The role of T1D candidate genes on beta cells – from genetics to new therapies

John Todd, in his lecture entitled Genes to clinic in type 1 diabetes’, showed that the candidate gene GLIS3 is clearly a beta cell fragility gene acting particularly in early life, predisposing to T1D in individuals diagnosed under the age of 7. Of note, the gut microbiome encodes functional insulin cross-reactive mimotopes of insulin that may promote tolerance to insulin and tolerance to similar bacterial proteins, for example, transketolase. Particularly in DR3/4-DQ8trans dimer-positive individuals, anti-insulin T cells escape from the thymus in the first months of life, especially during weaning (6 months onwards, 3 months before insulin autoantibodies appear). Translation of genetic information to the clinics should consider the following points: (i) Interval dosing of low-dose interleukin-2 (IL)-2 in children is safe and provides a steady-state increase in Tregs. If the Oxford IL-2 trial (ongoing) shows positive results, efforts to bring IL-2 into T1D prevention trials, and production of a non-invasive form of IL-2 should be commenced, including using IL-2 as maintenance therapy; (ii) healthy weaning should be promoted (especially in families with T1D and families in which anti-islet autoantibody screening has identified an autoantibody-positive child); (iii) healthy living during pregnancy could reduce the incidence of T1D; (iv) reduction of childhood obesity could reduce T1D incidence by ~30%; (v) promotion of gut microbiota and gut epithelial health could prevent T1D.

Lorenzo Pasquali addressed the theme ‘Inflammation and regulatory landscape of beta cells provides new insights into the genetics of type 1 diabetes’. He explained that the exposure of human islets to a pro-inflammatory environment causes profound changes in the beta cell gene expression and regulatory landscape. This process results in the activation of distal cis-regulatory elements bound by immune-responsive and tissue-specific TFs. Activation of these noncoding regulatory elements is coupled with 3D chromatin conformation changes enabling contact with their target gene promoters. Beta cell cytokine responsive enhancers harbour T1D-risk variants, suggesting a new mechanism whereby the variants act at a beta cell level but are functional only upon islet cell perturbation, for instance, as mediated by pro-inflammatory cytokines (Ramos-Rodríguez et al. 2019, Eizirik et al. 2020). Exploring these relationships could open an avenue to the identification of new T1D candidate genes acting at the beta cell level.

Single-cell approaches allow the detection of a heterogeneous beta cell response to the proinflammatory environment which could in turn inform on a specific, targetable, beta cell sub-population. Moreover, single-cell analyses may uncover a cell-type-specific noncoding regulatory response to cytokines; indeed, cellular responses may be islet beta cell-type specific, which might capture cues of the molecular basis of intrinsic beta cell fragility, as compared to other islet cell populations.

Closing the session, Decio L Eizirik discussed ‘From candidate genes to new therapies for T1D – the TYK2 story’, a project developed in close collaboration with Carmella Evans-Molina. He described that a challenge for studies on risk polymorphisms that may contribute to T1D is how to translate this knowledge into new therapies for the disease. Eizirik has previously described that 80% of the candidate genes for T1D are expressed in human islets (Eizirik et al. 2012), a finding also observed in the target tissues of other autoimmune diseases (Szymczak et al. 2021). He next compared the gene signatures induced by the cytokine interferon alpha (IFNα) (an ‘early cytokine’ that may contribute to three key features of the histology of human islets in T1D, namely upregulation of HLA class I, induction of ER stress and apoptosis) against the signatures observed in FACS-purified human beta cells obtained from patients affected by T1D and then ‘mined’ the common genes detected against gene signatures induced by a panoply of drugs in other cell systems (Colli et al. 2020b). This indicated that inhibition of TYK2, a kinase that transduces the early signals of IFNα and is a candidate gene for T1D (polymorphisms that decrease TYK2 signalling decrease disease risk), could revert the IFNα-induced signature. Additional testing of TYK2 inhibitors, done in collaboration with the Evans-Molina group, including agents already in an advanced state of evaluation for the treatment of other autoimmune diseases, indicates that TYK2 inhibition protects human beta cells in vitro against IFNα alone or in combination with IL-1β (Coomans de Brachène et al. 2020) and has clear beneficial effects in two models of autoimmune diabetes in mice (unpublished data). Importantly, TYK2 deletion in stem-cell-derived beta cells protects these cells against both IFN and CD8+ T-cell-induced damage (Chandra et al. 2022). As a whole, these data suggest that TYK2 inhibition is potentially an interesting approach to protect pancreatic beta cells in the early stages of T1D.

Beta cell stress in T1D

Heiko Lickert discussed ‘New insights into beta cell failure and recovery’. He described that beta cells are heterogeneous in health and disease and understanding the underlying mechanisms may allow to protect or even regenerate a functional beta cell mass (Bader et al. 2016, Roscioni et al. 2016, Yoshihara et al. 2020). Using an scRNA-seq-based evolutionary comparison of healthy mouse, pig and human islets revealed that alpha- and beta cell heterogeneity is conserved among species (Tritschler et al. 2022). Interestingly, isolated human islets across several healthy donors showed mature, immature, ER stressed and MHC classes I and II/autoantigen high-expressing clusters of beta cells, suggesting that the isolation procedure induces stress and unleashes beta cell plasticity. Streptozotocin (STZ)-induced diabetes and sustained hyperglycaemia for 100 days combined with scRNA-seq of mouse islets identified markers and pathways of beta cell dedifferentiation (Sachs et al. 2020). Single and combinatorial pharmacology revealed that exogenous insulin directly acts on beta cells, reducing glucotoxicity and contributing to restore beta cell identity and function. Interestingly, glucagon-like peptide 1 receptor (GLP-1R) can be targeted to deliver a GLP-1-oestrogen conjugate to beta cells to induce a nuclear response and protect beta cells from ER stress-mediated cell death, avoiding the side effects of oestrogen on female tissues (Sachs et al. 2020). It is well known that insulin/IGF signalling is essential for beta cell survival, proliferation and function and that beta cell insulin/IGF resistance causes overt diabetes (Goldfine & Kulkarni 2012, Jain et al. 2022). The discovery of the insulin inhibitory receptor in beta cells may allow specific sensitisation of insulin/IGF signalling in islet beta cells to trigger proliferation and survival (Ansarullah et al. 2021). In conclusion, understanding paths and mechanisms of beta cell stress, loss of identity and dedifferentiation and identifying and validating druggable molecular targets may allow to protect and/or regenerate beta cells in type 1 (and 2) diabetes.

Raghu Mirmira addressed the search for ‘Circulating biomarkers of beta cell stress and death’. In early T1D, beta cells are subjected to stresses such as inflammation, viral infections, hypoxia/ischemia and impaired nutrient handling as a result of insulin deficiency (Eizirik et al. 2009, Wilcox et al. 2016, Chen et al. 2017). Under these conditions, beta cells trigger emergency responses, including the ‘integrated stress response (ISR)’, which is highlighted by the activation of one or more of four kinases (PERK, HRI, PKR, GCN2) that lead to the phosphorylation of the translation factor eIF2α (Taniuchi et al. 2016). Phosphorylated eIF2α blocks the translational initiation of many mRNA transcripts in an attempt to divert energy expenditure toward cellular recovery processes (Muaddi et al. 2010). If the block is not alleviated, then the cell is bound for an apoptotic fate. Thus, like many emergency responses in the cell, the ISR balances the cellular fate. During the ISR, translationally inhibited mRNAs are compartmentalised into discrete processing bodies (P-bodies) and non-membranous inclusions known as stress granules (SGs) (Kedersha et al. 2013, Wheeler et al. 2016, Namkoong et al. 2018). PBs function in normal RNA decay, though this idea remains somewhat controversial (Luo et al. 2018). SGs, by contrast, are induced by cellular stressors (e.g. inflammation, viral infections) (Decker & Parker 2012) and function as cellular signalling ‘nodes’, whereby their predominant fate (recycling of their contents to translating ribosomes, diversion to lysosomes for degradation or fusion into endosomal cycling cascades) may ultimately determine the cell survival. Pancreatic tissue sections from autoantibody-positive non-diabetic individuals showed an increase in general SG formation in beta cells, whereas in longstanding T1D, SGs were significantly reduced. A perspective has been emerging that SGs may intercept other intracellular pathways (Buchan & Parker 2009, Buchan et al. 2013, Kedersha et al. 2013), including endosomes, which are eventually released as extracellular vesicles (EVs) (Baixauli et al. 2014, Jimenez et al. 2019). All EVs carry cell/stress-specific cargo that includes proteins, nucleic acids and lipids. The inclusion of SGs into forming endosomes and EVs (Jimenez et al. 2019) may augment a mechanism by which a cell’s ‘state of emergency’ can be communicated extracellularly. Dr Mirmira’s data demonstrate that the triggering of the ISR results in the inclusion of insulin mRNA within SGs. In humans with new onset T1D, insulin mRNA is detectable in circulation, whereas in longstanding T1D (where beta cells are few or dysfunctional), insulin mRNA is undetectable. These findings hold the promise that interrogation of EVs from individuals at risk for T1D might uncover mRNA species that reflect ongoing stress within beta cells. Such knowledge has the potential to identify high-risk individuals for whom intervention might prevent subsequent T1D.

Anath Shalev discussed the approaches her group is developing regarding ‘Reducing beta cell stress – clinical potential’. Recent clinical trial data (Ovalle et al. 2018, Xu et al. 2022) demonstrate that continuous oral use of the approved blood pressure medication, verapamil, in subjects with T1D delays loss of beta cell function and lowers insulin requirements for at least 2 years post-diagnosis. Verapamil downregulates the detrimental protein thioredoxin-interacting protein (TXNIP) and promotes an anti-oxidative, anti-apoptotic and immunomodulatory gene expression profile in human islets. Oral verapamil treatment reverses T1D-associated increases in serum chromogranin A (a T1D autoantigen) and in proinflammatory factors (IL-21 levels and T-follicular helper cells). This is the first indication that verapamil may also affect the autoimmunity of T1D in addition to its effects on beta cell survival. As such this could also help explain why verapamil is successful even without additional bona fide immunomodulation. Importantly, since verapamil is very well tolerated and its effects are not dependent on the presence of insulitis, testing it for the prevention of T1D in at-risk subjects seems warranted and would provide an attractive risk–benefit ratio. In addition, global longitudinal serum proteomics analysis revealed that chromogranin A, which can also be detected by a simple clinical blood lab test, provides a promising novel marker to help monitor successful therapy or disease progression in T1D. These studies provide an example of how findings in human islets and diabetes mouse models other than the non-obese diabetic (NOD) model of T1D can be successfully translated into clinical trials.

In the last lecture of the session, Alberto Pugliese discussed Exploring circulating miRNAs as biomarkers of disease progression, beta cell loss and stress’. miRNAs are critical regulators of gene expression and modulate pathways relevant to T1D. During the past decade, studies have explored associations of circulating miRNAs with T1D and to improve prediction of T1D risk and progression, including the decline of insulin secretion. Studies differed in design, cohorts, assays employed to measure miRNA levels, number of miRNAs measured, sample types tested (serum, plasma, rarely vesicles) and data analysis approaches. Despite the above mentioned differences, a review of published literature shows that among miRNAs whose circulating levels were associated with T1D/islet autoimmunity or disease progression, 35 were reported by more than 2 independent studies (range 2–8 studies reporting associations), including 12 miRNAs reported by at least 3 independent studies. These observations suggest that circulating miRNAs can be exploited as disease biomarkers with further advances and validation. This will require access to replication cohorts, and ideally, future studies should examine longitudinal samples. Ongoing studies are now relying on RNAseq-based platforms to examine the levels of higher numbers of miRNAs (extending to the 2000 range), and the sample volume requirement is down to microlitres, with minimal impact on existing repositories. It is possible that several of the earlier associations, based on RT-PCR assays that assessed a more limited number of miRNAs, may not be replicated due to differences in assay format and yet there is potential for the discovery of more and stronger associations. Reconciling RT-PCR and RNAseq data may be challenging but should be attempted and standardised assays should be developed for the clinical setting. Studies of miRNA responses in the context of disease-relevant metabolic/immunological stimulations would be helpful to better understand the biological impact of miRNAs, to unveil dysregulation of miRNAs that may not be evident in the steady state and to improve our understanding of the cellular origin of miRNAs in the circulation. To this end, initial studies showed the feasibility of these approaches, for example, by measuring changes in miRNA levels in patients, in vivo, during a mixed-meal tolerance test and in parallel during glucose-stimulated insulin secretion from isolated pancreatic tissue.

Beta cell replacement and protecting beta cells from the immune system

Timo Otonkoski addressed the question ‘Beta cells from ES- and iPS-cells: how close are we to endogenous beta cells?’ by presenting a comprehensive characterisation of human pluripotent stem cell-derived islets (SC islets) and primary human islets, focusing on their structure, function (both in vitro and in vivo following murine implantation), gene expression and metabolism as described in recent reports (Balboa et al. 2022, Barsby et al. 2022). Using a modified seven-stage differentiation protocol, he showed that when SC islets are given sufficient time to mature in vitro, they develop into islet-like aggregates that consist of nearly 100% endocrine cells (40–50% of both alpha and beta cells, 5% delta cells and 5–10% enterochromaffin-like cells, which may in fact represent endocrine progenitors) (Fig. 4). The SC islets present dynamic glucose-stimulated insulin secretion that closely matches adult islet controls, except for a somewhat lower extended second phase. When transplanted into immunodeficient mice, the SC islets become highly vascularised and innervated. However, as compared with implanted adult islets, there is a longer lag time before SC islets fully control the glycaemia of diabetic mice. The SC islets demonstrate signs of metabolic immaturity, for example, strong responsiveness to exogenous pyruvate and lower coupling of mitochondrial respiration to glucose-induced insulin secretion. However, these metabolic differences appear to quickly decrease in vivo, after implantation in mice, in agreement with transcriptomic data of maturation-associated genes and pathways. Overall, SC islets represent a valuable source of human pancreatic endocrine cells that can be effectively used both for the modelling of pathogenic mechanisms behind various types of diabetes (Balboa et al. 2021) and for the development of cell replacement therapies for insulin deficiency.

Figure 4
Figure 4

Human pluripotent stem cell-derived islets (SC islets). Double immunostaining for insulin and glucagon in the immature SC islets (stage 7, week 0) shows many double-positive cells. In the mature stage, the hormone-positive cells are clearly defined as separate populations in an architecture that resembles the human islet. Dynamic insulin secretion was studied with perifusion. The immature SC islets do not respond to high glucose, although they are able to respond to exendin-4 and membrane depolarisation induced by potassium chloride (KCl). In contrast, the mature-stage SC islets show a high response to glucose. (Data from Balboa et al. Nature Biotech 2022. Figure prepared by Väinö Lithovius.)

Citation: Journal of Endocrinology 259, 1; 10.1530/JOE-23-0082

In the lecture entitled ‘Stem cell to beta cell: state of the art and therapeutic potential’ Louise Winkel provided an industrial perspective on stem cell to beta cell technology, including a discussion of regulatory, manufacturing and logistical challenges that remain to bring a stem-cell-based therapy to the clinic. Novo Nordisk has been active in stem cell research for over 20 years. In late 2018, Cell Therapy R&D was established as an independent unit and has since grown to cover the full pharmaceutical value chain, from research to commercial, including a good manufacturing practice (GMP) manufacturing site in Fremont. In addition to several exploratory projects, three projects are moving towards the clinic: Parkinson’s disease, chronic heart failure and T1D, all but the latter in close collaboration with external partners.

The protocol for generating functional islet-like clusters from pluripotent stem cells has been developed internally over the past 14 years and is now matured to generate thoroughly characterised, functional cells that reproducibly reverse diabetes in pre-clinical models. This has been a major achievement not only accomplished by Novo Nordisk but also by several other groups in this field. However, it is ‘only’ the beginning of even bigger challenges to solve from an industrial perspective. Setting aside the major and complex challenges of the route of administration and protection from the immune system in people with T1D, upscaling and development of processes and assays that support large-scale GMP production, quality control and distribution beyond investigational new drug (IND)-enabling studies and Phase 1/2 trials is pivotal to the success of any cell-based therapy.

The development, delivery and commercialisation of stem cell-based therapies differ significantly from traditional drug development. Unlike protein- and peptide-based therapies, few standardised procedures are currently established across the industry or at the regulatory level. Each company must develop and mature both capabilities and processes as projects evolve, which requires very close collaboration across the research and development value chain. In addition, each cell type has different requirements and reliable delivery of cells into the body is extremely complex.

In summary, the field needs to focus on quality, scalability and cell delivery to accomplish our shared goal of helping people living with serious chronic diseases with safe and efficacious cell-based therapies.

Shareen Forbes presented work on ‘islet transplantation and mesenchymal stem cells’. Islet transplantation is of proven efficacy in T1D, reducing hypoglycaemia, stabilising blood glucose levels and improving the quality of life. However, there is a limited supply of donor pancreata and each recipient requires islets from two to three donors to substantially impact on the glycaemic control. Rates of insulin independence are only 17% in the UK, there is attrition in graft function over time and the scarcity of donor pancreases for transplantation further compounds the problem. Islets are avascular when transplanted into the liver, rendering them vulnerable to hypoxia and immune-mediated apoptosis and reliant on angiogenesis and vasculogenesis to engraft. As such, >60% of the transplanted islets die in the first 48 h post-transplant. Treatments to ameliorate this loss are urgently required.

Mesenchymal stromal cells (MSCs) are found in a number of organs and support cells within tissues. They are defined by the presence and absence of specific cell surface markers, are adherent to plastic and are able to differentiate into fat, bone and cartilage. MSCs from different sources are heterogeneous but can home to sites of inflammation and secrete immunomodulatory and anti-inflammatory cytokines as well as growth factors that are pro-angiogenic. MSCs are currently in Phase 3 clinical trials and have shown efficacy in a number of settings including acute graft vs host disease when given at a high dose intravenously (>1 × 106 MSCs). They are therefore potential candidates to improve islet engraftment and long-term graft function. Dr Forbes described their recent efforts to manufacture umbilical cord-derived human MSCs to GMP grade, with gene expression studies demonstrating anti-inflammatory, immunomodulatory and pro-regenerative properties (Forbes et al. 2020)). In a diabetic immunodeficient mouse model followed for 16 weeks, the Forbes team has shown superior glycaemic control, greater stimulated C-peptide concentrations, increased graft vascularity and delayed rejection with umbilical cord-derived MSCs plus islets vs islets alone (Forbes et al. 2020).

A current barrier to the translation of this therapy into humans is the demonstration of a homogeneous cryopreserved-banked MSC product, which is efficacious and safe at this proposed dose in islet transplantation. This would enable this allogeneic product to be thawed on demand and infused with islets into humans. Such a strategy would allow islet grafts to function better for longer periods and allow pancreases to be distributed to more people.

Kevan Herold provided an up-to-date review of ‘Immunotherapy and interventions in pre-diabetes and diabetes’. A number of immune interventions have been successfully trialled for the treatment of T1D in those with recent onset disease (Herold et al. 2002, 2005, 2013). Most recently, T1D clinical onset has been delayed in some individuals at risk for the diagnosis but who are free of symptoms for extended periods of time (Herold et al. 2019). However, even with the successes, the end of the disease has not yet been achieved, and further work is needed. The following observations and conclusions, based on the experience from clinical and preclinical studies, may help guide this work: first, there appears to be a window for successful interventions with the agents that have been able to modulate the natural disease course. This appears to be in the peri-diagnosis period (pre- and immediately post-diagnosis), which is thought to be characterised by active immune killing of beta cells. The reasons why this period is opportune have not been defined but it may relate to the susceptibility of immune cells to modulation. Second, the effects of immune therapies have not been permanent – in most cases, tolerance has not been achieved. With B cell depletion with anti-CD20 mAb (Pescovitz et al. 2009), which can modulate the decline in beta cell function when it is given to patients with new-onset disease, the recovery of B cells in the peripheral blood includes the recovery of autoreactive B cells (Chamberlain et al. 2016). Likewise with anti-CD3 mAb, autoreactive T cells are not permanently depleted. Recent preclinical and translational studies have highlighted the stem-cell-like features of autoreactive CD8+ T cells in T1D suggesting that these cells are resistant to elimination and may persist even after immune therapy. It suggests that rather than depleting the autoreactive repertoire, achieving tolerance will require an environment or ligands that prevent these autoreactive cells from reactivation. Finally, recent studies have described the contribution of beta cells themselves to their own demise (Rui et al. 2021, Mallone et al. 2022, Piñeros et al. 2022). They are an important source of inflammatory mediators that may recruit and activate autoreactive cells. Blocking these mechanisms in beta cells may solve both the problem of immune cell activation and beta cell killing.

With the progress of immune therapies, a new frontier is opened in which the goals for the treatment are directed towards the maintenance of the efficacy seen with single agents, timing of therapies, combinations of agents that may target immune and beta cells and the selection of patients who are most likely to respond to interventions. Much like the first descriptions of success with cancer immune therapy, progress in the field is likely to expand rapidly.

Coda: Since the symposium the US Food and Drug Administration have approved the use of Teplizumab (anti-CD3) injection to delay the onset of stage 3 T1D in patients >8 years of age with stage 2 T1D.

Measuring success in replacement/regeneration in pre-clinical and clinical models

Peter Senior opened this session by discussing ‘Clinical Assessment of Islet Cell Function’ from the standpoint of human islet transplantation. The University of Alberta has performed 729 islet transplants on 312 individuals since 1999 with a goal of improving both ‘metabolic health’ and ‘mental health’. Factors such as reducing glucose variability as well as reducing or abolishing hypoglycaemia and the fear of hypoglycaemia are important in improving ‘mental health’ beyond average glucose or HbA1c levels. Frequent assessments of islet cell function are required following islet transplantation to identify the need for additional islet loading (second or third islet transplants) and/or evidence of rejection. More invasive/extensive assessments of islet function provide more information, but a balance has to be struck with the patient burden of repeated assessments. In addition, stimulated C-peptide levels are very dependent on the stimulus such that glucose-potentiated arginine stimulation (with glucose = 360 mg/dL) could release three to four times as much C-peptide in islet transplants recipients than arginine alone (Rickels et al. 2005). The formulae to assess insulin sensitivity (e.g. HOMA or homeostatic model assessment) and to relate C-peptide to glucose (e.g. Suito Index) improve clinical correlation but are generally developed in the non-insulin-treated healthy population and may have limitations in the clinic population. More complex composite scores such as the BETA2 score which includes insulin dose and HbA1c as well as glucose and fasting C-peptide (Ryan et al. 2005, Forbes et al. 2016) are more convenient to use in clinical practice and correlate well with clinical outcomes (insulin independence) and invasive measures (r2 = 0.69) (Senior et al. 2020b, Lam et al. 2022). Continuous glucose monitoring (CGM) is required to capture variability and can show improvement following islet transplantation compared to hybrid closed-loop insulin therapy (Senior et al. 2020a). Further studies are required to indicate if this correlates well with ‘mental health’ in addition to ‘metabolic health’.

Michael Brehm discussed advancements in ‘Humanized models to understand the crosstalk between beta cells and the immune system’. Generating such models is a challenging task with several components. First, it is necessary to delete elements of the mouse immune system to ensure that any response to islet grafts is ‘human’ in origin. Deletion of the common gamma chain in NOD scid gamma (NSG) and NOD-Rag1null (NRG) mice impairs signalling through multiple key cytokines (IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21) resulting in the lack of T, B and natural killer (NK) cells (Ishikawa et al. 2005, Shultz et al. 2005, Pearson et al. 2008). Secondly, the deletion of murine insulin secretion allows the detection of the function of any transplanted islets via monitoring glucose levels. Such ‘hyperglycaemic’ mice can be generated either using the beta cell toxin streptozotocin or introducing the Akita insulin 2 gene mutation, which has a dominant negative effect on insulin production by impairing proinsulin folding in all forms of insulin (Pagliuca et al. 2014). Inducible beta cell depletion can be introduced using the rat insulin promotor-diphtheria toxin responsive element (RIP-DTR). Thirdly, the human immune system needs to be engrafted. This has been done by transferring either human peripheral blood mononuclear cells (PBMC) or haematopoietic stem cells (HSC). Unfortunately, such mice do not have a long lifespan due to Xeno-graft vs host disease, but this can be reduced by the deletion of mouse MHC class I and II without irradiation. NSG RIP-DTR MHC-DKO mice reconstituted with 50 million human PBMCs given intraperitoneally that receive diphtheria toxin reject a human islet allograft as shown by rising glucose levels (Brehm et al. 2019). Islet organoids derived from human pluripotent stem cells can be used in place of islet tissue and these systems have been used to demonstrate that overexpression of programmed death-ligand 1 (PD-L1) protected the organoids from rejection (Yoshihara et al. 2020). This platform has also been used recently by Doug Melton’s group to show that interferon-gamma signalling plays a major role in the rejection of stem cell-derived islets and this can be ameliorated to some degree by the deletion of CXCL10 in the stem cells (Sintov et al. 2022). Currently, work is underway to enhance the development of the engrafted human innate immune cells by expression of cytokines such as human IL-15 or human fms related receptor tyrosine kinase 3 (FLT3)-ligand. In addition, expression of mouse thymic stromal lymphopoietin has been shown to restore lymph node development in these composite mice, which is otherwise lacking.

James Cantley discussed ‘Preclinical models and assessment of islet function’, beginning by reviewing rodent models for T1D, including STZ-induced insulin-deficient diabetes, dosed to induce acute (Cantley et al. 2013) or progressive (Cantley et al. 2011) beta cell decline, along with the NOD/ShiLtJ mouse model of T1D. NOD mice are widely used in preclinical T1D studies with examples of successful translation to human trials including anti-CD3 immunotherapies (Herold et al. 1992, Chatenoud et al. 1994, Herold et al. 2019) and stem cell-derived pancreatic cell grafts (Agulnick et al. 2015, Ramzy et al. 2021, Shapiro et al. 2021), and some failures to translate. Despite steady improvements in pre-clinical study design, adopting many of the principles of clinical trials (Atkinson 2011, Grant et al. 2013, Gill et al. 2016), current pre-clinical endpoints are typically static-fed blood glucose measurements with a diabetes diagnostic threshold of >250 mg/dL. CGM has revolutionised the clinical management of diabetes, whilst providing new insights into glycaemic variability and its relationship with beta cell function (Gibb et al. 2020, Rickels et al. 2020, Carr et al. 2021, Yeşiltepe-Mutlu et al. 2021), risk of disease progression (Steck et al. 2022) and diabetes complications (Wilmot et al. 2021). Recent advances in telemetry have enabled remote CGM in rodents via surgically implanted carotid glucose sensors, providing new insights into glycaemic variability (Lo Martire et al. 2018), revealing marked daily glucose fluctuations during the prediabetic phase in NOD mice (Korstanje et al. 2017) and following minimal-mass islet transplantation in STZ-induced diabetic rats (King et al. 2017). This has important implications for binary pre-clinical study endpoints utilising the 250 mg/dL threshold, with a risk of false negatives and positives in prevention or reversal studies. Strategies for quantifying glycaemic variability during the pre-diabetic and diabetic phases were considered, such as rodent-adapted time-in-range, % coefficient of variation (%CV) or interquartile range, providing a graded assessment of disease progression and pre-clinical efficacy, whilst mirroring human glycaemic metrics. Barriers to wider implementation of preclinical CGM include expensive and invasive telemetry implants, with further innovation in rodent CGM, application of human patch systems to larger model species or better use of static blood glucose measurements needed to circumvent this.

In the second part of the lecture, Dr Cantley discussed the challenges of assessing beta cell mass and 3D structure of the rodent pancreas using conventional histology. A new Insulin1-tdTomato knock-in reporter mouse generated by the lab was presented which, combined with tissue clearing approaches and Mesolens imaging (Battistella et al. 2022), has enabled high-resolution imaging of total beta cell mass in the intact pancreas, whilst providing a proportional readout of insulin1 promoter activity.

What is missing for translation?

Steven Parks is a member of the Diabetes UK administrative team and was diagnosed with T1D aged 3 years. He shared key reflections on the translational gaps that need to be filled to address the needs of patients and their families. In particular, he emphasised that the needs differ at different ages – for example, between young children, teenagers and older adults. The sudden diagnosis of T1D after a very short (weeks) prodrome was a huge shock to his family and better markers able to monitor the decline in beta cell function in pre-diabetes, along with immune and genetic markers to predict who will develop T1D, are important to better prepare and therefore reduce the impact of diagnosis for families. At all stages, the idea of being immunosuppressed long-term as part of therapy raises concerns and risks need to be minimised, for example, by targeting treatments to islet cells. Following diagnosis, Steve confirmed that variability and unpredictability in glucose control is a major stress for people living with T1D. Reducing the complexity and burden of dealing with the different effects of food, exercise and psychological stress on glucose levels is essential. Although the technology of insulin replacement has advanced considerably, many people do not find it a sufficiently discrete, reliable or low burden to engage with it in its current form, especially among teenagers and young adults, emphasising the continued need for interventions such as prevention and stem cell replacement. At all stages of research, from planning to late stages, Steven emphasised the importance of including people living with diabetes and their families to ensure that what appears a good idea to research teams does really meet their needs and comprises an acceptable balance of benefits and risks.

In his lecture entitled ‘Clinical endpoints in beta cell preservation – Competing or complementary therapies?’, Colin Dayan built on Peter Senior’s comments, emphasising the need for improved C-peptide and clinical endpoint assessments in studies of islet cell function. Measurement of the area under the curve (AUC) of insulin/C-peptide over 2 or 4 h following a mixed-meal tolerance test has been the standard measure of beta cell function for 20 years. However, it is well known that beta cells secrete insulin in two phases – a rapid first phase and a slower second phase. The first phase is critical to regulate post-prandial peaks of glucose and reduce variability but occurs within 10–15 min, so it is not well captured by integration of the C-peptide and glucose response over 2–4 h. Consistent with this, there is only a modest correlation between AUC C-peptide or the detected peak C-peptide level (typically at around 90 min) and measures of glycaemic control, for example, HbA1c or CGM parameters (r values typically around 0.4) (Buckingham et al. 2015). Analysis of pooled data from 18 intervention trials of immunotherapy in new-onset T1D, 6 of which met their primary end point, conducted within the Trial Outcome Markers Initiative (TOMI) hosted by the C-Path Consortium has shown that immunotherapy (vs placebo) has a significant impact on AUC C-peptide at 6 months but not after 3 months. The same is true for insulin doses. By contrast, differences in HbA1c levels are seen at 3 months, suggesting that immunotherapy has an important early impact on beta cell function that is not detected by the AUC metric. Several alternative measures take into account the relationship between glucose and C-peptide levels, as Peter Senior indicated in his presentation, for example, Insulin dose-adjusted A1C (IDAA1c), the Suito Index, the B2 score, and glucose and C-peptide response curves (Sims et al. 2021); however, these do not reflect biphasic secretion. The oral minimal model (OMM) developed by Cobelli and colleagues (Cobelli et al. 2014, Galderisi et al. 2021) can estimate biphasic insulin secretion from the OGTT. The utility of hypoglycaemia as an outcome in clinical trials of beta cell preservation in new-onset T1D is also being explored within TOMI. Dr Dayan went on to describe the use of adaptive platform trials to study multiple combinations of agents (and agents at different doses) for beta cell preservation in a more rapid and efficient manner than with standard randomised controlled trial designs, as illustrated by the T1D Plus platform being developed in the INNODIA network. Work on the most sensitive and rapidly responsive measure of beta preservation will further accelerate these efforts and progress in drug development in this area.

‘The closed loop story – advantages and limitations’was the theme addressed by Charlotte Boughton, including recent advances in this field and the recently published Closed Loop from Onset of T1D (CLOuD) study applying closed-loop therapy from the diagnosis of T1D in children (Boughton et al. 2022). Closed-loop insulin pump therapy, in which insulin delivery is linked to CGM via an algorithm, aims to mimic islet function as closely as possible. She emphasised the challenge of glucose variability and unpredictability in insulin requirements that Steven Parks had highlighted, especially in very young children. Data from observation of over 2000 days in 114 different individuals showed that children aged 1–6 have a %CV in daily insulin requirements as high as 46%, as compared to values of around 36% in teenagers and young adults (Dovc et al. 2019). Closed-loop therapy reduces rather than increases the burden for patients, with users spending on average 36 min/day on diabetes management. Hyperglycaemia is potentially detrimental to islet cells (‘glucotoxicity’) including impairing beta cell function. It has therefore been hypothesised that improved glycaemic control immediately after diagnosis may improve islet cell function and survival. However, reports to date have produced conflicting results (Shah et al. 1989, Buckingham et al. 2013). The CLOuD study aimed to address this issue in young people aged 10–17 by comparing the use of hybrid closed-loop therapy with standard management (43% using pump therapy and 68% using CGM by 24 months) starting within 21 days of diagnosis (Ware et al. 2022). HbA1c levels were 0.4 percentage points lower at 12 months (6.9% vs 7.3%) and 1 percentage point lower at 24 months (6.9% vs 8.0%) in the hybrid closed-loop group, but no difference was seen in AUC C-peptide levels in the mixed-meal tolerance testing. Participants in the intensive arm used the closed-loop system for 76% of the time, which suggests that it had good levels of acceptability. However, five cases of severe hypoglycaemia and one case of diabetic ketoacidosis still occurred in the closed-loop group emphasising the remaining unmet need for replacement/preservation of islet function.

Conclusions and roadmap for future research

A suggested roadmap for future T1D research is summarised in Fig. 5. New imaging technologies applied to pancreatic biobank tissues and islets are transforming our view of the natural history of T1D. They demonstrate that beta and alpha cell changes occur early during pathogenesis, promote new hypotheses on the role of exocrine cells and suggest that a stage 0 T1D may exist (beta cell dysfunction and impaired glycaemic control, in the absence of autoimmunity). Further investigation of the role played by alpha cells in the pathophysiology of T1D, and why beta but not alpha cells are killed in long-term T1D, is clearly warranted.

Figure 5
Figure 5

A suggested roadmap for future type 1 diabetes (T1D) research. Summary of priorities for T1D research as described in detail in the main text. Pathophysiology (upper section). Risk factors for T1D involve a combination of genetic and environmental factors, including gut microbiota and viral infection. There is an urgent need for more research into the early stages of disease onset, including environmental triggers and the interaction between beta cells and the immune system. Progress in recent years has revealed marked heterogeneity in the age of diagnosis, disease progression and residual beta cell function in people with T1D. Further research into the factors underlying this heterogeneity is needed to generate deeper insights into disease aetiology, identify prognostic markers and enable personalised treatment strategies. With increased global obesity, including in childhood, more research into the role of obesity, sedentary lifestyle and insulin sensitivity on T1D progression is warranted. A recurring theme in the meeting was the dysregulation of glucagon secretion from pancreatic alpha cells (in spite of the fact that beta cells but not alpha cells are killed in long-term diabetes) and the imbalance of glucagon and insulin in the T1D pancreas. Finally, alterations in the exocrine and ductal system of the pancreas have been observed in T1D. Lower section, interventions. Summary of established and emerging approaches to treat T1D is provided, with estimated timescales to clinical impact (colour coded: pink, near-term 0–5 years; orange, mid-term 0–8 years; blue, long-term 5–10+ years). Improved pre-diabetes screening will support targeted immunotherapy, beta cell preservation and ultimately T1D prevention. Optimised combination therapies, reduced insulin requirements and smarter insulin delivery have the potential to improve clinical care in the near term. Stem-cell-derived cell transplant therapies are in clinical trials with positive early reports and have the potential to improve regulated insulin release in T1D in the mid-term. Likewise, strategies to improve the success of islet transplant therapy have the potential to reduce insulin dependence in the short term. Strategies to regenerate endogenous beta cells have long-term potential to restore regulated insulin secretion in the pancreas which, if coupled to beta cell protective therapies, may reverse T1D. Improvements in preclinical models and protocols can accelerate many of these therapeutic avenues. Figure created with BioRender.com.

Citation: Journal of Endocrinology 259, 1; 10.1530/JOE-23-0082

Histopathological differences between putative T1D endotypes continue to be explored. Invasive insulitis observed in childhood T1DE1 could be associated with alterations in islet basement membrane components and integrity. In contrast, the islet basement membrane appears structurally more intact in older onset T1DE2 with mild insulitis. Alterations in its integrity could account for different rates of disease progression and possibly for the sudden acceleration close to diagnosis after many years of ‘status quo preservation’. In addition, the lack of direct immune-beta cell contact in situations where the islet basement membrane is intact supports soluble factors mediating the dialogue between the two cell types and warrants further investigation. Insulin sensitivity may also play a role in shaping age-related endotypes. Research into the role of gut microbiota in T1D susceptibility, especially during weaning, may provide new insights and potential therapeutic strategies.

The hypothesis of the existence of T1D heterogeneity is starting to move from a phenomenological concept to one supported by novel mechanistic hypotheses. On one side, this invites enhanced efforts in the study of long-term non-progressors to identify disease-protective factors that may point to novel therapeutic targets. On the other, the often advocated immunomodulatory and beta cell-protective combination therapies may also be achieved by single agents targeting both immune and beta cells, for example, JAK or TYK2 inhibitors and verapamil. In addition, studying young organ donors with multiple autoantibodies but without overt hyperglycaemia (stages 1–2) will provide insights into disease pathogenesis and heterogeneity, interactions between the beta cell and immune system and potentially reveal new therapeutic targets.

It is likely that combination therapies with investigational products (reducing insulin dependence) and insulin delivery via closed-loop systems (refining insulin administration) will become standard therapeutic approaches in the near term. The ability of anti-CD3 administration to delay the onset of T1D provides a great source of optimism for the future of immunotherapies to preserve beta cell function. Improved screening and biomarkers to determine T1D risk will support-targeted implementation of these therapies. Although closed-loop systems did not improve the preservation of residual C-peptide in the recent CLOuD trial, it may act as a useful adjuvant for other immunomodulatory and beta cell-protective therapies. New strategies to improve the success of islet transplant therapies, such as encapsulation or co-transplantation with mesenchymal stromal cells, also have the potential to reduce insulin dependence in the near term. Recent progress in the advancement of stem cell to beta cell replacement therapy has clinical potential in the medium term, although regulatory, manufacturing and cost challenges remain. Finally, the development of strategies to drive regeneration of endogenous beta cells remains a long-term research goal which, in combination with recent advances in immunotherapy, could provide stable restoration of a functional pancreatic beta cell mass. Successful and timely development of these therapeutic approaches will require continued improvements in the assessment of glycaemic control/variability and beta cell function metrics as robust clinical/preclinical markers and endpoints. Improved understanding of T1D risk, heterogeneity and identification of prognostic markers will facilitate stratified treatment of the disease.

In summary, this symposium has captured a cross-section of current T1D research, including the aetiology and heterogeneity of T1D along with the current state-of-the-art and clinical potential of treatment strategies, both established, emerging and in development. This research landscape inspires optimism for the future management, treatment and even prevention of T1D.

Declaration of interest

JC and DE have no competing interests to declare. EL is a former employee of Regeneron Pharmaceuticals and owns company stock. CD has lectured for or been involved as an advisor to the following companies: Novonordisk, Sanofi-genzyme, Janssen, Servier, Lilly, Astrazeneca, Provention Bio, UCB, MSD, Vielo Bio, Avotres, Worg, Novartis. CD holds a patent jointly with Midatech plc.

Funding

We would like to thank the JDRF and DiabetesUK for sponsoring this symposium. JC is funded by a Steve Morgan Foundation Type 1 Diabetes Grand Challenge Senior Research Fellowship (22/0006505), the Medical Research Council (MR/W019590/1) and Tenovus Scotland (T20-69). CD holds research funding from JDRF, DiabetesUK and NIHR.

Acknowledgements

We would like to thank Frank Martin, Jodie Chan, Kamini Shah, Steven Parks, Jeannette Soderberg and Evan Levitt for their important roles in organising the symposium. JDRF-DiabetesUK-INNODIA-nPOD Symposium: Full list of chairs and speakers: Emily K Sims, Indiana University, IN, USA (chair). Chantal Mathieu, Katholieke Universiteit Leuven, Belgium (speaker). Decio L. Eizirik, Universite Libre de Bruxelles, Belgium (co-organiser, chair and speaker). James Cantley, University of Dundee, UK (co-organiser, chair and speaker). Colin M Dayan, Cardiff University, UK (co-organiser, chair and speaker). Esther Latres, JDRF Research, NY, USA (co-organiser and chair). Mark A Atkinson, University of Florida, FL, USA (chair and speaker). Sarah J Richardson, University of Exeter, UK (chair and speaker). Klaus H Kaestner, University of Pennsylvania, PA, USA (speaker). Marcela Brissova, Vanderbilt University, TN, USA (speaker). Carmella Evans-Molina, Indiana University, IN, USA (chair and speaker). Teresa Rodriguez-Calvo, Helmholtz Center Munich, Germany (chair). Francesco Dotta, University of Siena, Italy (chair). John A Todd, University of Oxford, UK (speaker). Lorenzo Pasquali, Universitat Pompeu Fabra Barcelona, Spain (speaker). Roberto Mallone, Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France (chair). Anath Shalev, The University of Alabama at Birmingham, AL, USA (chair and speaker). Heiko Lickert, Helmholtz Center Munich, Germany (speaker). Raghavendra G Mirmira, University of Chicago, IL, USA (speaker). Alberto Pugliese, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope, CA, USA (speaker). Shareen Forbes, University of Edinburgh, UK (chair and speaker). Timo Otonkoski, University of Helsinki, Finland (speaker). Louise Winkel, Novo Nordisk, Denmark (speaker). Kevan C Herold, Yale University, CT, USA (speaker). Peter Senior, University of Alberta, Canada (speaker). Michael A Brehm, University of Massachusetts, MA, USA (speaker). Richard A Oram, University of Exeter, UK (chair). Steven Parks, DiabetesUK, UK (speaker). Charlotte K Boughton, University of Cambridge (speaker).

References

  • Agulnick AD, Ambruzs DM, Moorman MA, Bhoumik A, Cesario RM, Payne JK, Kelly JR, Haakmeester C, Srijemac R, Wilson AZ, et al.2015 Insulin-producing endocrine cells differentiated in vitro from human embryonic stem cells function in macroencapsulation devices in vivo. Stem Cells Translational Medicine 4 12141222. (https://doi.org/10.5966/sctm.2015-0079)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Anand V, Li Y, Liu B, Ghalwash M, Koski E, Ng K, Dunne JL, Jönsson J, Winkler C, Knip M, et al.2021 Islet autoimmunity and HLA markers of presymptomatic and clinical type 1 diabetes: joint analyses of prospective cohort studies in Finland, Germany, Sweden, and the U.S. Diabetes Care 44 22692276. (https://doi.org/10.2337/dc20-1836)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ansarullah JC, Far FF, Homberg S, Wißmiller K, von Hahn FG, Raducanu A, Schirge S, Sterr M, Bilekova S, Siehler J. 2021 Inceptor counteracts insulin signalling in β-cells to control glycaemia. Nature 590 326331. (https://doi.org/10.1038/s41586-021-03225-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Atkinson MA 2011 Evaluating preclinical efficacy. Science Translational Medicine 3 96cm22. (https://doi.org/10.1126/scitranslmed.3002757)

  • Azoury ME, Tarayrah M, Afonso G, Pais A, Colli ML, Maillard C, Lavaud C, Alexandre-Heymann L, Gonzalez-Duque S, Verdier Y, et al.2020 Peptides derived from Insulin Granule Proteins are Targeted by CD8+ T cells across MHC Class I Restrictions in Humans and NOD Mice. Diabetes db200013. (https://doi.org/10.2337/figshare.12899315.v1)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bader E, Migliorini A, Gegg M, Moruzzi N, Gerdes J, Roscioni SS, Bakhti M, Brandl E, Irmler M, Beckers J, et al.2016 Identification of proliferative and mature β-cells in the islets of Langerhans. Nature 535 430434. (https://doi.org/10.1038/nature18624)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bahl V, Lee May C, Perez A, Glaser B & & Kaestner KH 2021 Genetic activation of α-cell glucokinase in mice causes enhanced glucose-suppression of glucagon secretion during normal and diabetic states. Molecular Metabolism 49 101193. (https://doi.org/10.1016/j.molmet.2021.101193)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Baixauli F, López-Otín C & & Mittelbrunn M 2014 Exosomes and autophagy: coordinated mechanisms for the maintenance of cellular fitness. Frontiers in Immunology 5 403. (https://doi.org/10.3389/fimmu.2014.00403)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Balboa D, Iworima DG & & Kieffer TJ 2021 Human pluripotent stem cells to model islet defects in diabetes. Frontiers in Endocrinology (Lausanne) 12 642152. (https://doi.org/10.3389/fendo.2021.642152)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Balboa D, Barsby T, Lithovius V, Saarimäki-Vire J, Omar-Hmeadi M, Dyachok O, Montaser H, Lund PE, Yang M, Ibrahim H, et al.2022 Functional, metabolic and transcriptional maturation of human pancreatic islets derived from stem cells. Nature Biotechnology 40 10421055. (https://doi.org/10.1038/s41587-022-01219-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Barsby T, Ibrahim H, Lithovius V, Montaser H, Balboa D, Vähäkangas E, Chandra V, Saarimäki-Vire J & & Otonkoski T 2022 Differentiating functional human islet-like aggregates from pluripotent stem cells. Star Protocols 3 101711. (https://doi.org/10.1016/j.xpro.2022.101711)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Battistella E, Schniete J, Wesencraft K, Quintana JF & & Mcconnell G 2022 Light-sheet mesoscopy with the Mesolens provides fast sub-cellular resolution imaging throughout large tissue volumes. iScience 25 104797. (https://doi.org/10.1016/j.isci.2022.104797)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bergholdt R, Brorsson C, Palleja A, Berchtold LA, Fløyel T, Bang-Berthelsen CH, Frederiksen KS, Jensen LJ, Størling J & & Pociot F 2012 Identification of novel Type 1 diabetes candidate genes by integrating genome-wide association data, protein-protein interactions, and human pancreatic islet gene expression. Diabetes 61 954962. (https://doi.org/10.2337/db11-1263)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bluestone JA, Buckner JH & & Herold KC 2021 Immunotherapy: building a bridge to a cure for type 1 diabetes. Science 373 510516. (https://doi.org/10.1126/science.abh1654)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bosi E, Marchetti P, Rutter GA & & Eizirik DL 2022 Human alpha cell transcriptomic signatures of types 1 and 2 diabetes highlight disease-specific dysfunction pathways. iScience 25 105056. (https://doi.org/10.1016/j.isci.2022.105056)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Boughton CK, Allen JM, Ware J, Wilinska ME, Hartnell S, Thankamony A, Randell T, Ghatak A, Besser REJ, Elleri D. 2022 Closed-loop therapy and preservation of C-peptide secretion in type 1 diabetes. New England Journal of Medicine 387 882893. (https://doi.org/10.1056/NEJMoa2203496)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brehm MA, Kenney LL, Wiles MV, Low BE, Tisch RM, Burzenski L, Mueller C, Greiner DL & & Shultz LD 2019 Lack of acute xenogeneic graft- versus-host disease, but retention of T-cell function following engraftment of human peripheral blood mononuclear cells in NSG mice deficient in MHC class I and II expression. FASEB Journal 33 31373151. (https://doi.org/10.1096/fj.201800636R)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brissova M, Haliyur R, Saunders D, Shrestha S, Dai C, Blodgett DM, Bottino R, Campbell-Thompson M, Aramandla R, Poffenberger G, et al.2018 Alpha cell function and gene expression are compromised in type 1 diabetes. Cell Reports 22 26672676. (https://doi.org/10.1016/j.celrep.2018.02.032)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Brissova M, Niland JC, Cravens J, Olack B, Sowinski J & & Evans-Molina C 2019 The Integrated Islet Distribution Program answers the call for improved human islet phenotyping and reporting of human islet characteristics in research articles. Diabetologia 62 13121314. (https://doi.org/10.1007/s00125-019-4876-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buchan JR & & Parker R 2009 Eukaryotic stress granules: the ins and outs of translation. Molecular Cell 36 932941. (https://doi.org/10.1016/j.molcel.2009.11.020)

  • Buchan JR, Kolaitis RM, Taylor JP & & Parker R 2013 Eukaryotic stress granules are cleared by autophagy and Cdc48/VCP function. Cell 153 14611474. (https://doi.org/10.1016/j.cell.2013.05.037)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buckingham B, Beck RW, Ruedy KJ, Cheng P, Kollman C, Weinzimer SA, Dimeglio LA, Bremer AA, Slover R, Tamborlane WV, et al.2013 Effectiveness of early intensive therapy on β-cell preservation in type 1 diabetes. Diabetes Care 36 40304035. (https://doi.org/10.2337/dc13-1074)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Buckingham B, Cheng P, Beck RW, Kollman C, Ruedy KJ, Weinzimer SA, Slover R, Bremer AA, Fuqua J, Tamborlane W, et al.2015 CGM-measured glucose values have a strong correlation with C-peptide, HbA1c and IDAAC, but do poorly in predicting C-peptide levels in the two years following onset of diabetes. Diabetologia 58 11671174. (https://doi.org/10.1007/s00125-015-3559-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Camunas-Soler J, Dai XQ, Hang Y, Bautista A, Lyon J, Suzuki K, Kim SK, Quake SR & & Macdonald PE 2020 Patch-seq links single-cell transcriptomes to human islet dysfunction in diabetes. Cell Metabolism 31 10171031.e4. (https://doi.org/10.1016/j.cmet.2020.04.005)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cantley J, Boslem E, Laybutt DR, Cordery DV, Pearson G, Carpenter L, Leitges M & & Biden TJ 2011 Deletion of protein kinase C delta in mice modulates stability of inflammatory genes and protects against cytokine-stimulated beta cell death in vitro and in vivo. Diabetologia 54 380389. (https://doi.org/10.1007/s00125-010-1962-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cantley J, Walters SN, Jung MH, Weinberg A, Cowley MJ, Whitworth TP, Kaplan W, Hawthorne WJ, O'connell PJ, Weir G, et al.2013 A preexistent hypoxic gene signature predicts impaired islet graft function and glucose homeostasis. Cell Transplantation 22 21472159. (https://doi.org/10.3727/096368912X658728)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Carr ALJ, Oram RA, Marren SM, Mcdonald TJ, Narendran P & & Andrews RC 2021 Measurement of peak C-peptide at diagnosis informs glycemic control but not hypoglycemia in adults with Type 1 diabetes. Journal of the Endocrine Society 5 bvab127. (https://doi.org/10.1210/jendso/bvab127)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Carr ALJ, Inshaw JRJ, Flaxman CS, Leete P, Wyatt RC, Russell LA, Palmer M, Prasolov D, Worthington T, Hull B, et al.2022 Circulating C-peptide levels in living children and young people and pancreatic β-cell loss in pancreas donors across Type 1 diabetes disease duration. Diabetes 71 15911596. (https://doi.org/10.2337/db22-0097)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chamberlain N, Massad C, Oe T, Cantaert T, Herold KC & & Meffre E 2016 Rituximab does not reset defective early B cell tolerance checkpoints. Journal of Clinical Investigation 126 282287. (https://doi.org/10.1172/JCI83840)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chandra V, Ibrahim H, Halliez C, Prasad RB, Vecchio F, Dwivedi OP, Kvist J, Balboa D, Saarimäki-Vire J, Montaser H, et al.2022 The type 1 diabetes gene TYK2 regulates β-cell development and its responses to interferon-α. Nature Communications 13 6363. (https://doi.org/10.1038/s41467-022-34069-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chatenoud L, Thervet E, Primo J & & Bach JF 1994 Anti-CD3 antibody induces long-term remission of overt autoimmunity in nonobese diabetic mice. Proceedings of the National Academy of Sciences of the United States of America 91 123127. (https://doi.org/10.1073/pnas.91.1.123)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chen C, Cohrs CM, Stertmann J, Bozsak R & & Speier S 2017 Human beta cell mass and function in diabetes: recent advances in knowledge and technologies to understand disease pathogenesis. Molecular Metabolism 6 943957. (https://doi.org/10.1016/j.molmet.2017.06.019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Cobelli C, Dalla Man C, Toffolo G, Basu R, Vella A & & Rizza R 2014 The oral minimal model method. Diabetes 63 12031213. (https://doi.org/10.2337/db13-1198)

  • Colli ML, Ramos-Rodríguez M, Nakayasu ES, Alvelos MI, Lopes M, Hill JLE, Turatsinze JV, Coomans de Brachène A, Russell MA, Raurell-Vila H, et al.2020a An integrated multi-omics approach identifies the landscape of interferon-α-mediated responses of human pancreatic beta cells. Nature Communications 11 2584. (https://doi.org/10.1038/s41467-020-16327-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Colli ML, Ramos-Rodríguez M, Nakayasu ES, Alvelos MI, Lopes M, Hill JLE, Turatsinze JV, Coomans de Brachène A, Russell MA, Raurell-Vila H, et al.2020b An integrated multi-omics approach identifies the landscape of interferon-α-mediated responses of human pancreatic beta cells. Nature Communications 11 2584. (https://doi.org/10.1038/s41467-020-16327-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Coomans de Brachène A, Castela A, Op De Beeck A, Mirmira RG, Marselli L, Marchetti P, Masse C, Miao W, Leit S, Evans-Molina C, et al.2020 Preclinical evaluation of tyrosine kinase 2 inhibitors for human beta-cell protection in type 1 diabetes. Diabetes, Obesity and Metabolism 22 18271836. (https://doi.org/10.1111/dom.14104)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Damond N, Engler S, Zanotelli VRT, Schapiro D, Wasserfall CH, Kusmartseva I, Nick HS, Thorel F, Herrera PL, Atkinson MA, et al.2019 A map of human Type 1 diabetes progression by imaging mass cytometry. Cell Metabolism 29 755768.e5. (https://doi.org/10.1016/j.cmet.2018.11.014)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Davis AK, Dubose SN, Haller MJ, Miller KM, Dimeglio LA, Bethin KE, Goland RS, Greenberg EM, Liljenquist DR, Ahmann AJ, et al.2015 Prevalence of detectable C-Peptide according to age at diagnosis and duration of type 1 diabetes. Diabetes Care 38 476481. (https://doi.org/10.2337/dc14-1952)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Decker CJ & & Parker R 2012 P-bodies and stress granules: possible roles in the control of translation and mRNA degradation. Cold Spring Harbor Perspectives in Biology 4 a012286. (https://doi.org/10.1101/cshperspect.a012286)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Doliba NM, Rozo AV, Roman J, Qin W, Traum D, Gao L, Liu J, Manduchi E, Liu C, Golson ML, et al.2022 α Cell dysfunction in islets from nondiabetic, glutamic acid decarboxylase autoantibody-positive individuals. Journal of Clinical Investigation 132. (https://doi.org/10.1172/JCI156243)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Dovc K, Boughton C, Tauschmann M, Thabit H, Bally L, Allen JM, Acerini CL, Arnolds S, De Beaufort C, Bergenstal RM, et al.2019 Young children have higher variability of insulin requirements: observations during hybrid closed-loop insulin delivery. Diabetes Care 42 13441347. (https://doi.org/10.2337/dc18-2625)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eizirik DL, Colli ML & & Ortis F 2009 The role of inflammation in insulitis and beta-cell loss in type 1 diabetes. Nature Reviews. Endocrinology 5 219226. (https://doi.org/10.1038/nrendo.2009.21)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eizirik DL, Sammeth M, Bouckenooghe T, Bottu G, Sisino G, Igoillo-Esteve M, Ortis F, Santin I, Colli ML, Barthson J, et al.2012 The human pancreatic islet transcriptome: expression of candidate genes for type 1 diabetes and the impact of pro-inflammatory cytokines. PLOS Genetics 8 e1002552. (https://doi.org/10.1371/journal.pgen.1002552)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eizirik DL, Pasquali L & & Cnop M 2020 Pancreatic β-cells in type 1 and type 2 diabetes mellitus: different pathways to failure. Nature Reviews. Endocrinology 16 349362. (https://doi.org/10.1038/s41574-020-0355-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Eizirik DL, Szymczak F, Alvelos MI & & Martin F 2021 From pancreatic β-cell gene networks to novel therapies for Type 1 diabetes. Diabetes 70 19151925. (https://doi.org/10.2337/dbi20-0046)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Evans-Molina C, Sims EK, Dimeglio LA, Ismail HM, Steck AK, Palmer JP, Krischer JP, Geyer S, Xu P, Sosenko JM, et al.2018 β Cell dysfunction exists more than 5 years before type 1 diabetes diagnosis. JCI Insight 3. (https://doi.org/10.1172/jci.insight.120877)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fasolino M, Goldman N, Wang W, Cattau B, Zhou Y, Petrovic J, Link VM, Cote A, Chandra A, Silverman M, et al.2020 Genetic variation in Type 1 diabetes reconfigures the 3D chromatin organization of T cells and alters gene expression. Immunity 52 257274.e11.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fasolino M, Schwartz GW, Patil AR, Mongia A, Golson ML, Wang YJ, Morgan A, Liu C, Schug J, Liu J, et al.2022 Single-cell multi-omics analysis of human pancreatic islets reveals novel cellular states in type 1 diabetes. Nature Metabolism 4 284299. (https://doi.org/10.1038/s42255-022-00531-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ferrannini E, Mari A, Nofrate V, Sosenko JM, Skyler JS & DPT-1 Study Group 2010 Progression to diabetes in relatives of type 1 diabetic patients: mechanisms and mode of onset. Diabetes 59 679685. (https://doi.org/10.2337/db09-1378)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Forbes S, Oram RA, Smith A, Lam A, Olateju T, Imes S, Malcolm AJ, Shapiro AM & & Senior PA 2016 Validation of the BETA-2 score: an improved tool to estimate beta cell function after clinical islet transplantation using a single fasting blood sample. American Journal of Transplantation 16 27042713. (https://doi.org/10.1111/ajt.13807)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Forbes S, Bond AR, Thirlwell KL, Burgoyne P, Samuel K, Noble J, Borthwick G, Colligan D, Mcgowan NWA, Lewis PS, et al.2020 Human umbilical cord perivascular cells improve human pancreatic islet transplant function by increasing vascularization. Science Translational Medicine 12 eaan5907. (https://doi.org/10.1126/scitranslmed.aan5907)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Galderisi A, Moran A, Evans-Molina C, Martino M, Santoro N, Caprio S & & Cobelli C 2021 Early impairment of insulin sensitivity, β-cell responsiveness, and insulin clearance in youth with Stage 1 Type 1 diabetes. Journal of Clinical Endocrinology and Metabolism 106 26602669. (https://doi.org/10.1210/clinem/dgab344)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gibb FW, McKnight JA, Clarke C & & Strachan MWJ 2020 Preserved C-peptide secretion is associated with fewer low-glucose events and lower glucose variability on flash glucose monitoring in adults with type 1 diabetes. Diabetologia 63 906914. (https://doi.org/10.1007/s00125-020-05099-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gill RG, Pagni PP, Kupfer T, Wasserfall CH, Deng S, Posgai A, Manenkova Y, Bel Hani A, Straub L, Bernstein P, et al.2016 A preclinical consortium approach for assessing the efficacy of combined anti-CD3 plus IL-1 blockade in reversing new-onset autoimmune diabetes in NOD mice. Diabetes 65 13101316. (https://doi.org/10.2337/db15-0492)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Goldfine AB & & Kulkarni RN 2012 Modulation of β-cell function: a translational journey from the bench to the bedside. Diabetes, Obesity and Metabolism 14(Supplement 3) 152160. (https://doi.org/10.1111/j.1463-1326.2012.01647.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gonzalez-Duque S, Azoury ME, Colli ML, Afonso G, Turatsinze JV, Nigi L, Lalanne AI, Sebastiani G, Carré A, Pinto S, et al.2018 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 Metabolism 28 946960.e6. (https://doi.org/10.1016/j.cmet.2018.07.007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Grant CW, Moran-Paul CM, Duclos SK, Guberski DL, Arreaza-Rubín G & & Spain LM 2013 Testing agents for prevention or reversal of Type 1 diabetes in rodents. PLoS One 8 e72989. (https://doi.org/10.1371/journal.pone.0072989)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Greenbaum CJ, Beam CA, Boulware D, Gitelman SE, Gottlieb PA, Herold KC, Lachin JM, Mcgee P, Palmer JP, Pescovitz MD, et al.2012 Fall in C-peptide during first 2 years from diagnosis: evidence of at least two distinct phases from composite Type 1 Diabetes TrialNet data. Diabetes 61 20662073. (https://doi.org/10.2337/db11-1538)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gregory GA, Robinson TIG, Linklater SE, Wang F, Colagiuri S, De Beaufort C, Donaghue KC, International Diabetes Federation Diabetes Atlas Type 1 Diabetes in Adults Special Interest Group Magliano DJ, Maniam J, et al.2022 Global incidence, prevalence, and mortality of type 1 diabetes in 2021 with projection to 2040: a modelling study. Lancet. Diabetes and Endocrinology 10 741760. (https://doi.org/10.1016/S2213-8587(2200218-2)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Haliyur R, Tong X, Sanyoura M, Shrestha S, Lindner J, Saunders DC, Aramandla R, Poffenberger G, Redick SD, Bottino R, et al.2019 Human islets expressing HNF1A variant have defective β cell transcriptional regulatory networks. Journal of Clinical Investigation 129 246251. (https://doi.org/10.1172/jci121994)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Haliyur R, Walker JT, Sanyoura M, Reihsmann CV, Shrestha S, Aramandla R, Poffenberger G, Ramirez AH, Redick SD, Babon JAB, et al.2021 Integrated analysis of the pancreas and islets reveals unexpected findings in human male with type 1 diabetes. Journal of the Endocrine Society 5 bvab162.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hao W, Gitelman S, Dimeglio LA, Boulware D, Greenbaum CJ & Type 1 Diabetes TrialNet Study Group 2016 Fall in C-peptide during first 4 years from diagnosis of Type 1 diabetes: variable relation to age, HbA1c, and insulin dose. Diabetes Care 39 16641670. (https://doi.org/10.2337/dc16-0360)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Hart NJ, Aramandla R, Poffenberger G, Fayolle C, Thames AH, Bautista A, Spigelman AF, Babon JAB, Denicola ME, Dadi PK, et al.2018 Cystic fibrosis-related diabetes is caused by islet loss and inflammation. JCI Insight 3. (https://doi.org/10.1172/jci.insight.98240)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Herold KC, Bluestone JA, Montag AG, Parihar A, Wiegner A, Gress RE & & Hirsch R 1992 Prevention of autoimmune diabetes with nonactivating anti-CD3 monoclonal antibody. Diabetes 41 385391. (https://doi.org/10.2337/diab.41.3.385)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Herold KC, Hagopian W, Auger JA, Poumian-Ruiz E, Taylor L, Donaldson D, Gitelman SE, Harlan DM, Xu D, Zivin RA, et al.2002 Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. New England Journal of Medicine 346 16921698. (https://doi.org/10.1056/NEJMoa012864)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Herold KC, Gitelman SE, Masharani U, Hagopian W, Bisikirska B, Donaldson D, Rother K, Diamond B, Harlan DM & & Bluestone JA 2005 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 54 17631769. (https://doi.org/10.2337/diabetes.54.6.1763)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Herold KC, Gitelman SE, Ehlers MR, Gottlieb PA, Greenbaum CJ, Hagopian W, Boyle KD, Keyes-Elstein L, Aggarwal S, Phippard D, et al.2013 Teplizumab (anti-CD3 mAb) treatment preserves C-peptide responses in patients with new-onset type 1 diabetes in a randomized controlled trial: metabolic and immunologic features at baseline identify a subgroup of responders. Diabetes 62 37663774. (https://doi.org/10.2337/db13-0345)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Herold KC, Bundy BN, Long SA, Bluestone JA, Dimeglio LA, Dufort MJ, Gitelman SE, Gottlieb PA, Krischer JP, Linsley PS, et al.2019 An anti-CD3 antibody, teplizumab, in relatives at risk for Type 1 diabetes. New England Journal of Medicine 381 603613. (https://doi.org/10.1056/NEJMoa1902226)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • HuBMAP Consortium 2019 The human body at cellular resolution: the NIH Human Biomolecular Atlas Program. Nature 574 187192. (https://doi.org/10.1038/s41586-019-1629-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • In't Veld P 2011 Insulitis in human type 1 diabetes: the quest for an elusive lesion. Islets 3 131138. (https://doi.org/10.4161/isl.3.4.15728)

  • Ishikawa F, Yasukawa M, Lyons B, Yoshida S, Miyamoto T, Yoshimoto G, Watanabe T, Akashi K, Shultz LD & & Harada M 2005 Development of functional human blood and immune systems in NOD/SCID/IL2 receptor {gamma} chain(null) mice. Blood 106 15651573. (https://doi.org/10.1182/blood-2005-02-0516)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jain C, Bilekova S & & Lickert H 2022 Targeting pancreatic β cells for diabetes treatment. Nature Metabolism 4 10971108. (https://doi.org/10.1038/s42255-022-00618-5)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Japp AS, Meng W, Rosenfeld AM, Perry DJ, Thirawatananond P, Bacher RL, Liu C, Gardner JS, Atkinson MA, Kaestner KH, et al.2021 TCR+/BCR+ dual-expressing cells and their associated public BCR clonotype are not enriched in type 1 diabetes. Cell 184 827839.e14. (https://doi.org/10.1016/j.cell.2020.11.035)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jimenez L, Yu H, McKenzie AJ, Franklin JL, Patton JG, Liu Q & & Weaver AM 2019 Quantitative proteomic analysis of small and large extracellular vesicles (EVs) reveals enrichment of adhesion proteins in small EVs. Journal of Proteome Research 18 947959. (https://doi.org/10.1021/acs.jproteome.8b00647)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kaestner KH, Powers AC, Naji A & HPAP Consortium & Atkinson MA 2019 NIH initiative to improve understanding of the pancreas, islet, and autoimmunity in Type 1 diabetes: the Human Pancreas Analysis Program (HPAP). Diabetes 68 13941402. (https://doi.org/10.2337/db19-0058)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kedersha N, Ivanov P & & Anderson P 2013 Stress granules and cell signaling: more than just a passing phase? Trends in Biochemical Sciences 38 494506. (https://doi.org/10.1016/j.tibs.2013.07.004)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • King AJ, Austin AL, Nandi M & & Bowe JE 2017 Diabetes in rats is cured by islet transplantation…but only during daytime. Cell Transplantation 26 171172. (https://doi.org/10.3727/096368916X692258)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Korstanje R, Ryan JL, Savage HS, Lyons BL, Kane KG & & Sukoff Rizzo SJ 2017 Continuous glucose monitoring in female NOD mice reveals daily rhythms and a negative correlation with body temperature. Endocrinology 158 27072712. (https://doi.org/10.1210/en.2017-00266)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lam CJ, Jacobson DR, Rankin MM, Cox AR & & Kushner JA 2017 β Cells Persist in T1D pancreata without Evidence of ongoing β-Cell turnover or neogenesis. Journal of Clinical Endocrinology and Metabolism 102 26472659. (https://doi.org/10.1210/jc.2016-3806)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lam A, Oram RA, Forbes S, Olateju T, Malcolm AJ, Imes S, Shapiro AMJ & & Senior PA 2022 Estimation of early graft function using the BETA-2 score following clinical islet transplantation. Transplant International 35 10335. (https://doi.org/10.3389/ti.2022.10335)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Leete P, Willcox A, Krogvold L, Dahl-Jørgensen K, Foulis AK, Richardson SJ & & Morgan NG 2016 Differential Insulitic profiles determine the extent of β-cell destruction and the age at onset of Type 1 diabetes. Diabetes 65 13621369. (https://doi.org/10.2337/db15-1615)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Livingstone SJ, Levin D, Looker HC, Lindsay RS, Wild SH, Joss N, Leese G, Leslie P, Mccrimmon RJ, Metcalfe W, et al.2015 Estimated life expectancy in a Scottish cohort with type 1 diabetes, 2008–2010. JAMA 313 3744. (https://doi.org/10.1001/jama.2014.16425)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Lo Martire V, Valli A, Bingaman MJ, Zoccoli G, Silvani A & & Swoap SJ 2018 Changes in blood glucose as a function of body temperature in laboratory mice: implications for daily torpor. American Journal of Physiology. Endocrinology and Metabolism 315 E662E670. (https://doi.org/10.1152/ajpendo.00201.2018)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Luo Y, Na Z & & Slavoff SA 2018 P-bodies: composition, properties, and functions. Biochemistry 57 24242431. (https://doi.org/10.1021/acs.biochem.7b01162)

  • Mallone R, Halliez C, Rui J & & Herold KC 2022 The β-cell in Type 1 diabetes pathogenesis: a victim of circumstances or an instigator of tragic events? Diabetes 71 16031610. (https://doi.org/10.2337/dbi21-0036)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Martens PJ, Gysemans C & & Mathieu C 2021 100 years of insulin: arresting or curing type 1 diabetes: an elusive goal, but closing the gap. Journal of Endocrinology 249 T1T11. (https://doi.org/10.1530/JOE-20-0552)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • McKeigue PM, Spiliopoulou A, McGurnaghan S, Colombo M, Blackbourn L, Mcdonald TJ, Onengut-Gomuscu S, Rich SS, Palmer CNA, McKnight JA, et al.2019 Persistent C-peptide secretion in Type 1 diabetes and its relationship to the genetic architecture of diabetes. BMC Medicine 17 165.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Muaddi H, Majumder M, Peidis P, Papadakis AI, Holcik M, Scheuner D, Kaufman RJ, Hatzoglou M & & Koromilas AE 2010 Phosphorylation of eIF2α at serine 51 is an important determinant of cell survival and adaptation to glucose deficiency. Molecular Biology of the Cell 21 32203231. (https://doi.org/10.1091/mbc.E10-01-0023)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Namkoong S, Ho A, Woo YM, Kwak H & & Lee JH 2018 Systematic characterization of stress-induced RNA granulation. Molecular Cell 70 175187.e8. (https://doi.org/10.1016/j.molcel.2018.02.025)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Oram RA, Sims EK & & Evans-Molina C 2019 Beta cells in type 1 diabetes: mass and function; sleeping or dead? Diabetologia 62 567577. (https://doi.org/10.1007/s00125-019-4822-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ovalle F, Grimes T, Xu G, Patel AJ, Grayson TB, Thielen LA, Li P & & Shalev A 2018 Verapamil and beta cell function in adults with recent-onset type 1 diabetes. Nature Medicine 24 11081112. (https://doi.org/10.1038/s41591-018-0089-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pagliuca FW, Millman JR, Gürtler M, Segel M, Van Dervort A, Ryu JH, Peterson QP, Greiner D & & Melton DA 2014 Generation of functional human pancreatic β cells in vitro. Cell 159 428439. (https://doi.org/10.1016/j.cell.2014.09.040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pearson T, Shultz LD, Miller D, King M, Laning J, Fodor W, Cuthbert A, Burzenski L, Gott B, Lyons B, et al.2008 Non-obese diabetic–recombination activating gene-1 (NOD–Rag 1 null) interleukin (IL)-2 receptor common gamma chain (IL 2 rγnull) null mice: a radioresistant model for human lymphohaematopoietic engraftment. Clinical and Experimental Immunology 154 270284. (https://doi.org/10.1111/j.1365-2249.2008.03753.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Pescovitz MD, Greenbaum CJ, Krause-Steinrauf H, Becker DJ, Gitelman SE, Goland R, Gottlieb PA, Marks JB, Mcgee PF, Moran AM, et al.2009 Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. New England Journal of Medicine 361 21432152. (https://doi.org/10.1056/NEJMoa0904452)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Piganelli JD, Mamula MJ & & James EA 2020 The role of β cell stress and neo-epitopes in the immunopathology of Type 1 diabetes. Frontiers in Endocrinology (Lausanne) 11 624590. (https://doi.org/10.3389/fendo.2020.624590)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Piñeros AR, Kulkarni A, Gao H, Orr KS, Glenn L, Huang F, Liu Y, Gannon M, Syed F, Wu W, et al.2022 Proinflammatory signaling in islet β cells propagates invasion of pathogenic immune cells in autoimmune diabetes. Cell Reports 39 111011. (https://doi.org/10.1016/j.celrep.2022.111011)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Polychronakos C & & Li Q 2011 Understanding type 1 diabetes through genetics: advances and prospects. Nature Reviews. Genetics 12 781792. (https://doi.org/10.1038/nrg3069)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ramos-Rodríguez M, Raurell-Vila H, Colli ML, Alvelos MI, Subirana-Granés M, Juan-Mateu J, Norris R, Turatsinze JV, Nakayasu ES, Webb-Robertson BM, et al.2019 The impact of proinflammatory cytokines on the β-cell regulatory landscape provides insights into the genetics of type 1 diabetes. Nature Genetics 51 15881595. (https://doi.org/10.1038/s41588-019-0524-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ramzy A, Thompson DM, Ward-Hartstonge KA, Ivison S, Cook L, Garcia RV, Loyal J, Kim PTW, Warnock GL, Levings MK, et al.2021 Implanted pluripotent stem-cell-derived pancreatic endoderm cells secrete glucose-responsive C-peptide in patients with type 1 diabetes. Cell Stem Cell 28 20472061.e5. (https://doi.org/10.1016/j.stem.2021.10.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Richardson SJ, Rodriguez-Calvo T, Gerling IC, Mathews CE, Kaddis JS, Russell MA, Zeissler M, Leete P, Krogvold L, Dahl-Jørgensen K, et al.2016 Islet cell hyperexpression of HLA class I antigens: a defining feature in type 1 diabetes. Diabetologia 59 24482458. (https://doi.org/10.1007/s00125-016-4067-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rickels MR, Schutta MH, Markmann JF, Barker CF, Naji C & Teff KL 2005 β-Cell function following human islet transplantation for type 1 diabetes. Diabetes 54 100106. (https://doi.org/10.2337/diabetes.54.1.100)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rickels MR, Evans-Molina C, Bahnson HT, Ylescupidez A, Nadeau KJ, Hao W, Clements MA, Sherr JL, Pratley RE, Hannon TS, et al.2020 High residual C-peptide likely contributes to glycemic control in type 1 diabetes. Journal of Clinical Investigation 130 18501862. (https://doi.org/10.1172/JCI134057)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Roep BO, Thomaidou S, Van Tienhoven R & & Zaldumbide A 2021 Type 1 diabetes mellitus as a disease of the β-cell (do not blame the immune system?). Nature Reviews. Endocrinology 17 150161. (https://doi.org/10.1038/s41574-020-00443-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Roscioni SS, Migliorini A, Gegg M & & Lickert H 2016 Impact of islet architecture on β-cell heterogeneity, plasticity and function. Nature Reviews. Endocrinology 12 695709. (https://doi.org/10.1038/nrendo.2016.147)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Rui J, Deng S, Perdigoto AL, Ponath G, Kursawe R, Lawlor N, Sumida T, Levine-Ritterman M, Stitzel ML, Pitt D, et al.2021 Tet2 Controls the Responses of β cells to Inflammation in Autoimmune Diabetes. Nature Communications 12 5074. (https://doi.org/10.1038/s41467-021-25367-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ryan EA, Paty BW, Senior PA, Lakey JR, Bigam D & & Shapiro AM 2005 Beta-score: an assessment of beta-cell function after islet transplantation. Diabetes Care 28 343347. (https://doi.org/10.2337/diacare.28.2.343)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sachs S, Bastidas-Ponce A, Tritschler S, Bakhti M, Böttcher A, Sánchez-Garrido MA, Tarquis-Medina M, Kleinert M, Fischer K, Jall S, et al.2020 Targeted pharmacological therapy restores β-cell function for diabetes remission. Nature Metabolism 2 192209. (https://doi.org/10.1038/s42255-020-0171-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Saunders DC, Messmer J, Kusmartseva I, Beery ML, Yang M, Atkinson MA, Powers AC, Cartailler JP & & Brissova M 2020 Pancreatlas: applying an adaptable framework to map the human pancreas in health and disease. Patterns 1 100120. (https://doi.org/10.1016/j.patter.2020.100120)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Senior P, Lam A, Farnsworth K, Perkins B & & Rabasa-Lhoret R 2020a Assessment of risks and benefits of beta cell replacement versus automated insulin delivery systems for Type 1 diabetes. Current Diabetes Reports 20 52. (https://doi.org/10.1007/s11892-020-01339-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Senior PA, Rickels MR, Eggerman T, Bayman L, Qidwai J, Alejandro R, Markmann JF, Hunsicker LG & Clinical Islet Transplant Consortium 2020b 360-OR: BETA-2 score is highly correlated with acute insulin response to intravenous glucose: an analysis of the clinical islet transplantation consortium trials. Diabetes 69. (https://doi.org/10.2337/db20-360-or)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shah SC, Malone JI & & Simpson NE 1989 A randomized trial of intensive insulin therapy in newly diagnosed insulin-dependent diabetes mellitus. New England Journal of Medicine 320 550554. (https://doi.org/10.1056/NEJM198903023200902)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shapiro AMJ, Thompson D, Donner TW, Bellin MD, Hsueh W, Pettus J, Wilensky J, Daniels M, Wang RM, Brandon EP, et al.2021 Insulin expression and C-peptide in type 1 diabetes subjects implanted with stem cell-derived pancreatic endoderm cells in an encapsulation device. Cell Reports. Medicine 2 100466. (https://doi.org/10.1016/j.xcrm.2021.100466)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shultz LD, Lyons BL, Burzenski LM, Gott B, Chen X, Chaleff S, Kotb M, Gillies SD, King M, Mangada J, et al.2005 Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells. Journal of Immunology 174 64776489. (https://doi.org/10.4049/jimmunol.174.10.6477)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sims EK, Cuthbertson D, Herold KC & & Sosenko JM 2021 The deterrence of rapid metabolic decline within 3 months after teplizumab treatment in individuals at high risk for type 1 diabetes. Diabetes 70 29222931. (https://doi.org/10.2337/db21-0519)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sintov E, Nikolskiy I, Barrera V, Hyoje-Ryu Kenty J, Atkin AS, Gerace D, Ho Sui SJ, Boulanger K & & Melton DA 2022 Whole-genome CRISPR screening identifies genetic manipulations to reduce immune rejection of stem cell-derived islets. Stem Cell Reports 17 19761990. (https://doi.org/10.1016/j.stemcr.2022.08.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Steck AK, Dong F, Geno Rasmussen C, Bautista K, Sepulveda F, Baxter J, Yu L, Frohnert BI, Rewers MJ, ASK Study Group, et al.2022 CGM metrics predict imminent progression to Type 1 diabetes: autoimmunity screening for kids (ASK) study. Diabetes Care 45 365371. (https://doi.org/10.2337/dc21-0602)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Szymczak F, Colli ML, Mamula MJ, Evans-Molina C & & Eizirik DL 2021 Gene expression signatures of target tissues in type 1 diabetes, lupus erythematosus, multiple sclerosis, and rheumatoid arthritis. Science Advances 7. (https://doi.org/10.1126/sciadv.abd7600)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Taniuchi S, Miyake M, Tsugawa K, Oyadomari M & & Oyadomari S 2016 Integrated stress response of vertebrates is regulated by four eIF2α kinases. Scientific Reports 6 32886. (https://doi.org/10.1038/srep32886)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tritschler S, Thomas M, Böttcher A, Ludwig B, Schmid J, Schubert U, Kemter E, Wolf E, Lickert H & & Theis FJ 2022 A transcriptional cross species map of pancreatic islet cells. Molecular Metabolism 66 101595. (https://doi.org/10.1016/j.molmet.2022.101595)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wang YJ, Traum D, Schug J, Gao L, Liu C, Atkinson MA, Powers AC, Feldman MD, Naji A, Chang KM, et al.2019 Multiplexed in situ imaging mass cytometry analysis of the human endocrine pancreas and immune system in Type 1 diabetes. Cell Metabolism 29 769783.e4.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Ware J, Allen JM, Boughton CK, Wilinska ME, Hartnell S, Thankamony A, De Beaufort C, Schierloh U, Fröhlich-Reiterer E, Mader JK, et al.2022 Randomized trial of closed-loop control in very young children with Type 1 diabetes. New England Journal of Medicine 386 209219. (https://doi.org/10.1056/NEJMoa2111673)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Warncke K, Weiss A, Achenbach P, Von Dem Berge T, Berner R, Casteels K, Groele L, Hatzikotoulas K, Hommel A, Kordonouri O, et al.2022 Elevations in blood glucose before and after the appearance of islet autoantibodies in children. Journal of Clinical Investigation 132. (https://doi.org/10.1172/JCI162123)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wheeler JR, Matheny T, Jain S, Abrisch R & & Parker R 2016 Distinct stages in stress granule assembly and disassembly. eLife 5 e18413. (https://doi.org/10.7554/eLife.18413)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wilcox NS, Rui J, Hebrok M & & Herold KC 2016 Life and death of β cells in Type 1 diabetes: a comprehensive review. Journal of Autoimmunity 71 5158. (https://doi.org/10.1016/j.jaut.2016.02.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wilmot EG, Lumb A, Hammond P, Murphy HR, Scott E, Gibb FW, Platts J & & Choudhary P 2021 Time in range: a best practice guide for UK diabetes healthcare professionals in the context of the COVID-19 global pandemic. Diabetic Medicine 38 e14433. (https://doi.org/10.1111/dme.14433)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Wyatt RC, Lanzoni G, Russell MA, Gerling I & & Richardson SJ 2019 What the HLA-I!-classical and non-classical HLA class I and their potential roles in type 1 diabetes. Current Diabetes Reports 19 159. (https://doi.org/10.1007/s11892-019-1245-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Xu G, Grimes TD, Grayson TB, Chen J, Thielen LA, Tse HM, Li P, Kanke M, Lin TT, Schepmoes AA, et al.2022 Exploratory study reveals far reaching systemic and cellular effects of verapamil treatment in subjects with type 1 diabetes. Nature Communications 13 1159. (https://doi.org/10.1038/s41467-022-28826-3)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yeşiltepe-Mutlu G, Çapaci M, Can E, Gökçe T, Bayrakçi G, Muradoğlu S, Incir S, Çakir EP & & Hatun S 2021 A comparison of glycemic parameters and their relationship with C-peptide and Proinsulin levels during partial remission and non-remission periods in children with type 1 diabetes mellitus - a cross-sectional study. BMC Endocrine Disorders 21 18.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Yoshihara E, O'connor C, Gasser E, Wei Z, Oh TG, Tseng TW, Wang D, Cayabyab F, Dai Y, Yu RT, et al.2020 Immune-evasive human islet-like organoids ameliorate diabetes. Nature 586 606611. (https://doi.org/10.1038/s41586-020-2631-z)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Figure 1

    Progression to type 1 diabetes (T1D). Risk factors for T1D include genetic predisposition and environmental factors, such as viral infections and diet. Although the clinical diagnosis of T1D usually occurs at stage 3 when beta cell mass is depleted and hyperglycaemia is established, several stages precede this. There is increasing recognition of stage 0, in which beta cell dysfunction and impaired glycaemic control may occur in the absence of autoantibodies. Stage 1 represents seroconversion with the engagement of the adaptive immune system and the appearance of autoantibodies. During stage 2 beta cell dysfunction and apoptosis are increased impairing glycaemic control, leading to clinical diagnosis at stage 3. An important concept discussed is the reciprocal interaction between the beta cell and immune system throughout disease progression, along with interperson heterogeneity influenced by factors such as age of onset. Figure created with BioRender.com.

  • Figure 2

    Visualising insulitis in three dimensions. Image of a 3D reconstruction of an nPOD donor (20-year-old T1D subject with 7 months disease duration) stained for glucagon (blue) and CD3 (yellow), revealing the presence of infiltrating immune cells within the islet of Langerhans, interspersed with endocrine cells. 3D reconstruction was performed using Arivis Vision4D.

  • Figure 3

    Summary of pancreas Type 1 diabetes endotypes (T1DE). Images of pancreatic sections from a T1DE1 (left panel) or T1DE2 (right panel) donor. Low magnification images of pancreas sections (left inset panels, 1 cm scale bars) with each islet annotated as being an uninflamed Insulin-containing islet (ICI) – red; inflamed, defined as >15 CD45+ immune cells/ islet, ICI – green; inflamed insulin-deficient islet (IDI) – blue; or uninflamed IDI – pink. T1DE1 donors have evidence of more inflammation (green and blue islet annotations) and fewer residual insulin-containing islets (red/green islet annotations) than individuals with T1DE2. The higher magnification images (right inset panels, 500 µm and 100 µm scale bars) demonstrate the presence of peri-insulitis and invasive insulitis in ICIs of a T1DE1 donor, and an example of low level peri-insulitis observed in a T1DE2 donor. Further immune and beta cell characteristics of the endotypes are described in the boxes.

  • Figure 4

    Human pluripotent stem cell-derived islets (SC islets). Double immunostaining for insulin and glucagon in the immature SC islets (stage 7, week 0) shows many double-positive cells. In the mature stage, the hormone-positive cells are clearly defined as separate populations in an architecture that resembles the human islet. Dynamic insulin secretion was studied with perifusion. The immature SC islets do not respond to high glucose, although they are able to respond to exendin-4 and membrane depolarisation induced by potassium chloride (KCl). In contrast, the mature-stage SC islets show a high response to glucose. (Data from Balboa et al. Nature Biotech 2022. Figure prepared by Väinö Lithovius.)

  • Figure 5

    A suggested roadmap for future type 1 diabetes (T1D) research. Summary of priorities for T1D research as described in detail in the main text. Pathophysiology (upper section). Risk factors for T1D involve a combination of genetic and environmental factors, including gut microbiota and viral infection. There is an urgent need for more research into the early stages of disease onset, including environmental triggers and the interaction between beta cells and the immune system. Progress in recent years has revealed marked heterogeneity in the age of diagnosis, disease progression and residual beta cell function in people with T1D. Further research into the factors underlying this heterogeneity is needed to generate deeper insights into disease aetiology, identify prognostic markers and enable personalised treatment strategies. With increased global obesity, including in childhood, more research into the role of obesity, sedentary lifestyle and insulin sensitivity on T1D progression is warranted. A recurring theme in the meeting was the dysregulation of glucagon secretion from pancreatic alpha cells (in spite of the fact that beta cells but not alpha cells are killed in long-term diabetes) and the imbalance of glucagon and insulin in the T1D pancreas. Finally, alterations in the exocrine and ductal system of the pancreas have been observed in T1D. Lower section, interventions. Summary of established and emerging approaches to treat T1D is provided, with estimated timescales to clinical impact (colour coded: pink, near-term 0–5 years; orange, mid-term 0–8 years; blue, long-term 5–10+ years). Improved pre-diabetes screening will support targeted immunotherapy, beta cell preservation and ultimately T1D prevention. Optimised combination therapies, reduced insulin requirements and smarter insulin delivery have the potential to improve clinical care in the near term. Stem-cell-derived cell transplant therapies are in clinical trials with positive early reports and have the potential to improve regulated insulin release in T1D in the mid-term. Likewise, strategies to improve the success of islet transplant therapy have the potential to reduce insulin dependence in the short term. Strategies to regenerate endogenous beta cells have long-term potential to restore regulated insulin secretion in the pancreas which, if coupled to beta cell protective therapies, may reverse T1D. Improvements in preclinical models and protocols can accelerate many of these therapeutic avenues. Figure created with BioRender.com.

  • Agulnick AD, Ambruzs DM, Moorman MA, Bhoumik A, Cesario