REVIEW doi: 10.1111/sji.12338 ..................................................................................................................................................................

New Insights and Biomarkers for Type 1 Diabetes: Review for Scandinavian Journal of Immunology M. T. Heinonena, R. Mouldera & R. Lahesmaa

Abstract Turku Centre for Biotechnology, University of Turku,  Abo Akademi University, Turku, Finland

Received 21 June 2015; Accepted in revised form 25 June 2015 Correspondence to: R. Lahesmaa, Turku Centre for Biotechnology, University of Turku and  Abo Akademi University, Tykist€okatu 6 FI-20520 Turku, Finland. E-mail: [email protected] a

These authors have contributed equally to this work.

The increasing incidence of type 1 diabetes observed in the past 60 years has spawned massive efforts in multiple research fields to elucidate the aetiology of this disease. While GWAS studies provide a good genetic basis for the current knowledge, it is clear that environmental triggers and their influence in disease prevalence and origin are highly important. The realization of disease heterogeneity has created a requirement for better biomarkers to complement the known autoantibody markers and to more successfully predict the severity and onset time of the disease. Such biomarkers would be needed both for prevention as well as for monitoring disease activity and response to preventive and therapeutic measures. Systematic holistic approaches concentrating on the triggering molecular mechanisms, pancreatic beta cells, immune response, as well as the influence of diet and environment, are necessary to understand the disease pathogenesis and find a cure. The current genomic knowledge is being broadened with accompanying studies in epigenetics and transcriptomic regulation, metabolomics, proteomics and lipidomics, covering the whole system from beta cells, the profile and cellular balance of the infiltrating lymphocytes, to gut microbiota and viral infections. Here we highlight interesting recent findings in type 1 diabetes research.

Introduction Type 1 diabetes (T1D) is an autoimmune disease that occurs when the body can no longer produce insulin due to the destruction of the insulin producing b cells of the Islet of Langerhans in the pancreas [1]. The disease is associated with genetic susceptibility and there is a large body of evidence supporting the influence of early childhood diet, infections, environmental components and other nongenetic factors in its aetiology. Alarmingly there has been a steady increase in the incidence of T1D during the past 60 years [2, 3], leading to increased efforts to understand the disease phenotype. A number of longitudinal sample collections from at risk subjects have thus been established, particularly in geographic regions where the risk is highest. The Diabetes Auto Immunity Study in the Young (DAISY) was started in July 1993 to study how genes and the environment interact to trigger the onset of T1D. Similarly, the Diabetes Prediction and Prevention study (DIPP) was established in Finland in 1994 [4], and has been since mirrored in other regions, e.g. Sweden (DIPIS [5]), Germany (BabyDiab, [6]) and the USA (The Environmental Determinants of Diabetes in the Young, TEDDY [7]). In these studies, newborns are screened for

244

T1D risk and at risk children are recruited to periodically visit the clinic where biological samples are collected (every 3 or 6 months). Notably, due to the level of participation and adherence to the study and sample collection, there is age, risk, gender and geographically matched samples for diseased and healthy children. These unique longitudinal sample series provide an exceptional opportunity to find markers that would reflect the disease process from the birth to the diagnosis, give insights into potential pathogenetic mechanisms as well as biomarkers that could be used to predict the progression to T1D and monitor its activity at different stages. Moreover, studying such follow-up samples from children who represent distinct clinical outcomes will enable us to identify markers that can be used for stratification of children according to different disease subsets. This will be particularly important for preventive and therapeutic strategies and trials to test them. Recent studies using these biobanks and exploiting a range of methods including transcriptomics, lipidomics, metabolomics, proteomics and metagenomics have provided compelling new insights into disease process and pathogenesis. In this review, we consider recent progress in the identification of biomarkers and understanding the influence of the diet and environment on the pathogenesis

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

M. T. Heinonen et al. New Insights and Biomarkers for Type 1 Diabetes 245 ..................................................................................................................................................................

of T1D, with particular focus on the use of data from these cohorts, in addition to recent data from pancreatic organ donors and autopsies. Figure 1 provides an illustration of the triggering factors and potential sources of biomarkers that have received recent attention in T1D research.

T1D associated autoantibodies To date the only truly established markers that have been detected in association with the autoimmune attack are a panel of serum borne autoantibodies that include islet cell autoantibodies (ICA), antibodies to insulin (IAA), glutamic acid decarboxylase (GAD), protein tyrosine phosphatase (IA2) and zinc transporter 8 (ZnT8). These have been evaluated in terms the underlying risks of their order of presentation and their different combinations. For instance, in the evaluation of data from the Finnish DIPP cohort, the influence of the identity of the first primary antibody and the age of seroconversion was assessed in 520 children who progressed to T1D. IAA was the most common (320 children), which appeared in the second year of life [8]. Recently, the data from three major long term studies in Finland, Germany and United States were combined to evaluate their collective observations. In keeping with earlier conclusions, the majority of children at risk of T1D

who displayed multiple autoantibody seroconversion progressed to diabetes within 15 years [9]. However, as in the order of 2–5% of patients clinically diagnosed with T1D are found negative for known antibodies, there may still be other autoantibodies to be found. Although efforts to detect additional autoantibodies have been made, the former five autoantibodies remain as the key markers [10, 11]. In addition, the effort to find SNP markers associated with autoantibody appearance continues. In the frame of the TEDDY study, recent results revealed new insights on this matter [12]. T€orn et al. reported that among the high-risk genotype carriers aged under 9 years, they could verify four T1D and seroconversion linked susceptibility loci from the eight previously reported ones; PTPN22, ERBB3, SH2B3 and INS. They showed that although all of these are associated with T1D, ERBB3, SH2B3 are especially associated with autoantibody appearance. They also suggested that some SNPs are rather predictive for autoantibody appearance, whereas others are for T1D, and this could be caused by the environmental factors associated with the disease.

From the genome to the transcriptome and beyond The risk for developing T1D can be assessed by genotyping, whereby polymorphisms of the human leucocyte

Figure 1 Summary of triggering factors and biomarkers in type 1 Diabetes. There is a multitude of epidemiological factors that may trigger the onset of type 1 diabetes. Suspected environmental factors include some that may begin their influence already in utero, as well as dietary elements, such as introduction of cow’s milk and wheat, and lack of exposure to sunlight/ low vitamin D levels. The pancreas, in which the beta cells are destroyed by autoimmune reaction, has a major role in the disease. The lymphocyte driven destruction is mostly mediated by the imbalanced immune homeostasis. The effect of viral infections has recently been supported by new data rising in the field. The gut microbiome has also been shown to have an impact. The quest to find more accurate biomarkers, causative genes and molecular mechanisms, besides the classical autoantibody markers and GWAS studies, has created new and noteworthy transcriptomic, epigenetic and proteomic data. Recent discoveries are paving the way to improved biomarkers to predict and prevent T1D and to stratify patients for therapeutic and prevention trials.

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

246 New Insights and Biomarkers for Type 1 Diabetes M. T. Heinonen et al. .................................................................................................................................................................. antigen genes (HLA-DQB1) have been found to be the most common linkage. Notably, however, only 10% of the subjects displaying these genetic traits develop disease. A wider panel of (less frequent) risk alleles has been identified and GWAS studies have been conducted in search of other genetic variants and combinations that may either increase or protect from the risk [13]. Altogether, the past era of GWAS has produced close to 60 T1D susceptibility loci in various chromosomal regions. Currently the focus is upon analysis and combination of the vast resource of data produced in the GWAS field, from which the aims are to pinpoint the causative genes and their function in disease aetiology, and to discover mechanistic pathways. How T1D susceptibility loci influence the function of the immune system, pancreatic beta cells and their interactions with environmental triggers is an active area of investigation. In this review, we will highlight selected recent findings. For a broader historical perspective of what has been discovered in the past from GWAS studies, the recent detailed reviews, such as the one by Bakay et al. [14], are recommended. As an example where researchers have undertaken the re-analysis of GWAS data and combined these with other data types, Guo et al. [15] recently used Bayesian colocalization analysis to study whether genetic associations to different immune-mediated diseases, including T1D, were consistent with a causal variant that regulates gene expression in B cells and monocytes. They integrated disease GWAS with eQTL data and highlighted candidate causal genes and cells. In the course of their study they also refined the earlier finding that regulation of the CTSH gene is contributory to T1D aetiology, and defined the putative associations of this marker in detail [16]. CTSH overexpression has been shown to protect the insulinsecreting cells against cytokine-induced apoptosis, making this a valuable finding with regard to potential treatment and prevention. In further work by the same group, Guo and Wallace [15] used a new bioinformatic approach for variant set enrichment analysis, VSEAMS, to identify novel transcription regulators contributing to T1D. They extended a non-parametric SNP set enrichment method and tested for enrichment of GWAS signals in functionally defined loci. Their results revealed enrichment of T1D GWAS associations near genes targeted by IKZF3, which localizes within a known T1D susceptibility region, BATF and ESRRA, which are associated with other autoimmune disorders. Another interesting approach has been to focus on the genes in GWAS studies rather than just for the SNP markers. New susceptibility genes were found by Qiu et al. [17] with a gene-based GWAS study. Finally, regarding gene regulation, the epigenetic factors together with emerging knowledge about non-coding RNAs as transcriptional regulators have also been integrated into T1D research [18–20]. A recently reported major study, fine-

mapped the T1D susceptibility loci and discovered new markers that localize in active thymic gene enhancer regions, in addition to active T and B cell enhancer regions [21]. They indicated that a potential follow-up from this would be to discover causal genes and regulators by analysing which promoter regions are interacting with the enhancer regions. In parallel with the development of new approaches for the analysis of susceptibility loci and causative genes, pathway analysis approaches have been used in order to elucidate the T1D disease mechanisms. In a recent study by Evangelou et al. [22], they used summary level GWAS data from the Wellcome Trust Case Control Consortium (WTCCC) project and succeeded in finding new associations and reported more refined pathways. In comparison to previous pathway analyses, e.g. Carbonetto and Stephens [23], differences in these results were notable in terms of the larger sample size and the removal of the MHC genes from the analysis. Nevertheless, the results were consistent with earlier findings. Additionally, in the context of future studies that might aim to define the mechanistic interactions between the causative genes or gene regulators and/or build a holistic view about the network causing the disease phenotype, it is worth considering genetic epistasis i.e. how gene interactions can affect the phenotype [24]. Moreover, there are a number of genes important for the proper function of the immune system that should be taken into consideration, even though they have not been highlighted by GWAS studies. This is especially applicable for studies of gene–gene interactions and disease promoting pathways. In our recent study, we investigated the role of T helper cell balance regulators, GIMAP genes, in human immune-mediated diseases [25]. We discovered an association of GIMAP5 in the Finnish South-Western subpopulation, but most importantly, we observed a GIMAP5-INS interaction in the data set. The GIMAP5 genotype provided a protective effect against an INS conferred risk, and this was further emphasized by the putative regulation of GIMAP5 by the insulin induced transcription factor FOXO1. In addition to immune system specific genes not indicated by GWAS, the genes involved in beta cell function are also an important piece of the puzzle. Sartori et al. [26] found GATA factors, which are known to be central in tissue development and survival, to have an important role in beta cell function and T1D pathogenesis. Their functional studies were conducted in mice, but they also showed that multiple polymorphisms in the GATA4 region are associated with human T1D. In their summary, Santin and Eizrik [27] indicated that the polymorphisms in T1D candidate genes might affect the function and inflammation of beta cells in response to environmental cues. Stroling et al. [28] reviewed the status of beta cell function in T1D, and using a published RNA sequencing data set made a systematic human islet expression analysis of all genes located in 50

Scandinavian Journal of Immunology, 2015, 82, 244–253

M. T. Heinonen et al. New Insights and Biomarkers for Type 1 Diabetes 247 ..................................................................................................................................................................

T1D-associated GWAS loci, reporting that that 336 out of the 857 genes they tested were expressed in human islets and that many of these interact in protein networks. Beyond sequencing the genome as a potential template for risk-associated aberrations, researchers have investigated how the risk and onset is reflected in the dynamics of the whole-blood transcriptome in the pre-diabetic phases [29– 32]. Following on from preliminary studies [29], Kallionpaa et al. [31] compared the transcriptome of healthy controls with children on route to T1D and seroconverted children. The activity of genes and pathways related to innate immunity functions was observed, and notably type 1 interferon (IFN) response genes and IFN response factors were identified as central mediators of the IFN-related transcriptional changes observed prior to seroconversion. In keeping with this, and other observations from seroconverted children [30], Ferreira et al. [32] similarly reported differential abundance of transcripts associated IFN response in the PBMCs of children before seroconversion, while linking this to be the likely influence of viral infection. Kallionpaa et al. [31] also considered the influence of disease associated SNPs, including Immunochip measurements of the subjects. Differences were detected between children progressing to diabetes and their matched healthy controls with the detection of variants of a number of genes that have been previously linked to several autoimmune diseases. In terms of the regulation of gene expression, Stefan et al. [20] compared the DNA methylation patterns of lymphocyte cell lines from monozygotic twin pairs who were discordant and concordant for T1D and (3 versus 6 pairs). With the integration of this and GWAS data, significant differences were observed in the DNA-methylation of several known T1D associated genes, i.e. HLA, INS, IL-2RB, CD226.

compared serum profiles from longitudinal sample series from birth to the onset of T1D from 19 prediabetic children with control children in the same genetic risk group, and detected new markers for prediciting and monitoring the disease progression [40]. The work indicated that the protein profiles could distinguish healthy controls from children en route to diabetes and included proteomic differences detected before seroconversion. The detected differences and profiles emphasized alteration in the relative abundance of proteins associated with inflammation and immune responses as the children progressed to T1D. In the future, it will be of great interest to find out which of the findings can be generalized through crossvalidation in independent cohorts and which are more specific to a given cohort. Inflammation of the beta cells is accompanied by elevated levels of pro-inflammatory cytokines, and altered levels of chemokines and cytokines have been observed in the serum of children with a high T1D risk [41, 42]. Increased levels of IL6, IL-16, TNFa and IL-8 were detected in children with T1D associated antibodies long before the diagnosis of T1D [43]. Moreover, in relation to viral triggers for T1D (see below), distinct cytokine profiles have been observed with islet autoimmunity and enterovirus infection [44]. Purohit et al. [45] recently presented an overview on the scope for multiplex cytokine assays in the study of environmental triggers of T1D, citing their advantages in terms of throughput and cost, although emphasizing the importance of thorough assay development. Another viable route to protein markers from serum may be from the analysis of extracellular vesicles, such as the micro-particulate fraction [46], exosomes or other microvesicles [47].

Serum proteomics

Inflammation plays an important role for beta cell function in different ways at various stages of T1D development (reviewed in Eizirik et al.) [48]. The efforts of the Network for Pancreatic Organ Donors with Diabetes (nPOD) project have provided fresh insights into beta cell biology and its role in T1D. On the basis of the results from the nPOD project, Reddy et al. [49] observed a population of putatively pathogenic leucocytes among insulin producing islet cells, suggesting that the leucocytes detected in the exocrine region possibly may be pathogenic to residual beta cells in long-standing diabetic individuals and participate in the disease pathogenesis. Similarly, immune cell infiltrates have been detected also in exocrine pancreas pointing out that the underlining aetiology of T1D may not be restricted to beta cells [50, 51]. In the Diabetes Virus Detection (DiViD) project, pancreatic tissue from patients recently diagnosed with diabetes and organ donors (diabetic and no diabetic) has been collected to study the aetiology of T1D. Importantly, for patients where glucose-induced insulin secretion could

The targeted measurement of serum protein markers is regularly used in the clinic as general evaluation of health status, where for instance glycated haemoglobin (HbA1c) is monitored to assess blood sugar levels and the diabetic condition [33]. Serum potentially carries thousands of proteins. It is a complex mixture with significant interindividual variation and dominated by several highly abundant proteins [34]. As with the case of transcripts, changes in the serum proteome could reflect or predict the onset of T1D. However, while a number of studies have compared the serum proteome of T1D patients [35–37], only a few reports have considered markers prior to diagnosis [38–40]. McGuire et al. [38] targeted analysis of the cord blood of T1D developing children, and although discriminating patterns were discerned between healthy controls and children who later developed T1D, the distinguishing peaks were not identified. Recently, we

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

Pancreatic beta cells

248 New Insights and Biomarkers for Type 1 Diabetes M. T. Heinonen et al. .................................................................................................................................................................. be still observed after the onset of T1D, a normal glucoseinduced insulin response could be observed after several days in a non-diabetogenic environment in vitro [52]. All in all, these publications, among others, have shown the importance of the pancreas and considerations of the roles of both endocrine and exocrine cells in the disease aetiology. Other important aspect rising from the beta cell research is the role of viral infections [53], which is discussed later in this review.

proposed that low phospholipid levels at birth could contribute to early induction of islet autoimmunity [61]. With similar measurements from the German BABYDIAB study, distinct metabolic profiles were associated with age and islet autoimmunity [58]. The observations from the cord blood measurements were taken as an indication of the development of a metabolomics phenotype in utero. Notably, data considering regional difference in developed and emerging societies have similarly supported the role of in utero effects on the offspring (see below) [62].

Proteomics of the pancreas, islets and beta cells The progress made using proteomics to study beta cells has been recently reviewed by Crevecoeur et al. [39]. In brief the developments in this area of research has followed a course consistent with the evolution of proteomics technologies of the past 10 years. From initial studies using 2D gel electrophoresis to create reference maps of the pancreas and islets, to studies using multidimensional chromatography together with mass spectrometry. Studies of the beta cell proteome have generally investigated the effect of stimulation with inflammatory cytokines (IL-1b and IFNϒ) [39], although as with the other work with samples of pancreatic origin, the use of human material has been the exception rather than the rule. Studies of the stimulation of human beta cells indicated changes in proteins involved in cytoskeleton remodelling and vesicle motility. Investigations have also considered T cell reactivity against beta cell peptides [54], and post-translationally modified beta cell autoantigens have been reported that demonstrate recognition by T cells of T1D patients [55, 56].

Metabolomics The interaction of an organism and its environment is primarily reflected by changes in its metabolic profile. In this manner, the detection of perturbations in the metabolome or distinctive patterns can provide early signatures of T1D risk. Studies of the serum metabolome of subjects developing T1D have included global analysis of polar metabolites and lipids in longitudinal samples from infancy (including cord blood) through to diagnosis [57–61]. Data from the cord blood studies have suggested early metabolite traits may predispose the subjects to a diseased outcome. Finnish children who progressed to T1D had decreased phosphatidylcholines (PCs) at birth (cord blood) and diminished ether phospholipids throughout the followup [57]. Increased levels of pro-inflammatory lysophosphatidylcholine, glutamate, and branched chain amino acids, and decreased levels of several TCA cycle metabolites were also observed in serum samples collected before the appearance of autoantibodies. In related metabolomics studies of umbilical cord blood from a Swedish cohort, it was similarly indicated that the profiles could be used to identify children at increased risk for T1D. The authors

Gut microbiome Beyond the makeup of the human genome, the body is host to a subpopulation of microbiota that outnumbers its own cells 10 times over. From the moment of birth there is a colonization of the gut microbiome, and thus our immediate environment and the manner of delivery can influence its diversity [63, 64]. Gut microbiota affect both the innate and adaptive immune system. Colonization of the gut can thus serve to educate the immune system, and while some gastrointestinal infections can be protective others may predispose to risk [65, 66]. Evaluation of how aberrations of this symbiosis can precipitate or prevent diabetes has included analysis of stool samples prospectively collected T1D risk cohorts (i.e. DIPP and DIABIMMUNE) [67–69]. Alterations in the gut microbiome of infants in the period between seroconversion and diagnosis were interpolated from these data. Decreased alpha diversity (how many types of species of microbiota there are in a sample) was observed in seroconverted children prior to diagnosis, although not in the seroconverted children who were not diagnosed with T1D during the time frame of the study [67]. Higher levels of human b-defensin 2 (hBD2) were also found in the early samples of children who developed T1D. In keeping with emerging evidence of the immuno-metabolic effects of butyrate and other short-chain fatty acids, de Goffau et al. [68] observed that lactate-producing and butyrate-producing bacteria were less abundant in autoantibody positive children and thus implicated in b cell autoimmunity. Similar observations have also been made in studies of microbiota and type 2 diabetes [70]. Davis-Richardson et al. studied the early development of the gut microbiomes from stool samples of 76 children at high genetic risk for T1D. A higher abundance of Bacteroides dorei was observed before seroconversion, indicating that changes in the composition of the microbiome could be useful in the prediction of T1D autoimmunity in genetically susceptible infants [69]. In summary, there is much evidence supporting the correlations of the diversity of microbiota in the gut with the balance of the immune responses. To complement the information obtained from stool samples, analysis of gut biopsies if available would be valuable. Moreover, in terms of proteomics measurements, powerful new technologies have recently been combined such that detailed character-

Scandinavian Journal of Immunology, 2015, 82, 244–253

M. T. Heinonen et al. New Insights and Biomarkers for Type 1 Diabetes 249 ..................................................................................................................................................................

ization of tissue from biopsy samples and their associated microbiota can be achieved in a time efficient manner with minimal sample requirements [71].

Viral triggers of T1D The role of viral infection has long been considered as a potential trigger for the development of T1D [72]. The candidate list of risk-associated viruses has ranged from mumps to Rubella and one hypothesis has been that the observed trend for increasing T1D could reflect the growth of widespread vaccination programs. Overall, enteroviruses have remained as a main candidate [72, 73]. Enterovirus RNA has been detected in the pancreatic islets from newborn babies who died from fulminant coxsackievirus infection [74], and enteroviral capsid protein has been detected in the beta cells of patients who had either been recently diagnosed with or had established T1D [75, 76]. In keeping with evidence that the effect of enterovirus infection on the risk of T1D development is strain specific [77], it was shown that Coxsackievirus B1 is associated with an increased risk of beta cell autoimmunity [78]. In the latter study, serum samples were screened for neutralizing antibodies against 41 different enterovirus types in longitudinal sample series from the DIPP study. The strongest risk was observed when the infection occurred a few months before autoantibodies appeared, while the presence of maternal antibodies against the virus reduced the risk, as did the coxsackieviruses B3 and B6. On the basis of evaluation of the effect of enterovirus infection in pregnancy it was concluded that this was not a major risk factor but might play a role in some susceptible subjects. Further consideration of the data from other European nations supported the notion of the diabetogenic nature of the CBV1 virus [79]. The detection of enteroviral RNA in stool samples from T1D children has been previously reported, and recent investigations have considered changes in the gut virome at the onset of autoimmunity. In this study (19 versus 19) no dramatic changes were observed [80].

Diet environment and prevalence The accumulation of several prospective longitudinal sample collections has provided a valuable resource to study the regional differences, changes and the influence of diet and various interventions. Nested in these studies has been the evaluation of variables such as breast feeding [81], food diversity during the first year of life, the intakes of cow milk and fruit and berry juices [82, 83], early introduction of gluten [84], different types of wheat, rye, cereals, fish, fish oil and egg [82]. These data have thus been considered in terms of T1D/beta cell immunity as well as for the risks of asthma and allergies in childhood. Clinical trials conducted in parallel have considered nasal

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

and oral insulin as well as hydrolyzed formula milk [85– 87]. To date these have not proved effective. Evaluation of the cumulative data from Finland has shown that the growth of T1D incidence in children below the age of 15 years has now plateaued [88]. Further data suggest that the observed trend could be associated with the more frequent use of vitamin D supplements [89]. Conflicting evidence has also been reported [90, 91]. Further evaluations of environmental influences upon T1D and allergy have extended to the comparison of cohorts from neighbouring geographic regions. The DIABIMMUNE study has compared and followed newborns from the towns of Espoo (Finland), Tartu (Estonia) and Petrovoska (Russia). The comparison represents the contrast between developing and developed society, and city and rural environment. The interpretations of this study follow many of the considerations of the hygiene hypothesis, i.e. that recent changes in living conditions, family sizes, exposure to animals and an increasing hygiene may influence the maturation of the immune system [92]. The data from the DIABIMMUNE study support the contrasting rates of T1D incidence between these regions, and implicates the plasticity in the developing immune system towards environmental factors that may begin already in utero [62]. Notably, in the cord blood samples from Petrovoska, several pivotal innate immune response mediators were upregulated and the gene expression profiles were clearly different than those of cord blood samples of babies born in Estonia or Finland. The observed profiles were compared with published data comparing immune responses in newborns versus young children, bacterially challenged cells from children, and children with sepsis [62]. This analysis demonstrated that the immune responses of newborns from Petrovoska resembled that of 1 year old children. Such an early maturation of immune response already in utero could thus be protective against the development of immune-mediated disease in later life.

Cellular profiles The human leucocyte antigen gene encodes for major histocompatibility complex (MHC) proteins, which are responsible for regulation of the immune system in humans. The genes of the MHC locus include MHC class I and MHC class II. MHC class I molecules present antigens binding to CD8+ cells, and MHC class II molecules are expressed on antigen presenting cells and present antigens to T helper cells (CD4+). Polymorphisms in these genes may thus affect the nature of the antigen presented. There has been a long established association of T cells with T1D, most notably with involvement of autoreactive CD4+ T helper cells and CD8+ T cells in destruction of the beta cells. Recent studies have isolated the cellular subtypes from at risk

250 New Insights and Biomarkers for Type 1 Diabetes M. T. Heinonen et al. .................................................................................................................................................................. subjects/diseased and studied for instance the diversity of the T cell receptor in patients [93]. With the recent recognition of other Th cells subtypes, Th17 cells have also come into focus as a player in the events leading to autoimmunity. The observation of increasing Th1/Th17 plasticity with progression from b cell autoantibody positivity to type 1 diabetes, was reported, suggesting a Th1/Th17 influence on tissue destruction in autoimmune conditions [94]. The emerging new area of Tfh cells and their role in autoimmunity has produced interesting new insights into the mechanisms of T1D [95]. Kenefeck et al. reported that in comparison to healthy controls, the memory CD4+ T cells (CD4+ CD45RA ) of T1D patients have increased levels of molecules characteristic for Tfh cells and have a higher percentage of the CXCR5 + ICOS+ effector cells. The role of Treg cells has also received much attention [96, 97]. Ryba-Stanislawowska et al. have shown that the balance between Treg cells and Th17 cells is disturbed in T1D patients [98]. A major line of investigation also includes the interplay of IL2 signalling and Treg cells. The genetic variation in IL2Ra is known to be causative for T1D, but it has also been shown that it correlates with defects in Treg function [99]. This has been taken into consideration in clinical trials, as low doses of IL2 stimulate Treg cell proliferation without an effect on effector T cells [100]. It has also been shown that Treg cells inhibit inflammation and contribute to the disease progression via IL10 and TGF-b secretion [101]. IL10 production by Treg cells was shown to be essential in the immune response at environmental interfaces, such as lung and intestine, and Th1 responses can be suppressed by the TGF-b produced by Treg cells [102, 103]. Beyond investigations of the causative nature of cellular types, research has also been made towards prevention by the use of expanded peripheral or cord blood T regulatory cells for immune cell therapy to stop immune-mediated destruction of the beta cells [104].

Conclusion While a universal phenotype or trigger has yet to be identified for T1D, its aetiology remains under investigation. The aspects of genetic risk together with autoimmunity remain as predictors of risk and onset, and yet the evidence for such triggering elements as limited exposure to sunlight/vitamin D or dietary factors, such as cow’s milk remain inconclusive. Re-occurring evidence has supported the influence of viral factors for several decades, thus presenting the question if indeed T1D is triggered by a virus, (at least in some cases/regions), would vaccination reduce the incidence? Similarly, if an imbalance in the gut microflora may precipitate the illness, could diet, probiotics, other microbial supplements, or specific factors or metabolites released from microbes reduce the risk of T1D.

Evidence suggesting in utero effects for protection and predisposition has also emerged, with further indication that these are dependent on the environment. However, any subsequent health/preventative directives related to these would be less subtle. Better biomarkers for classifying the heterogeneous patient population and for the prediction, prevention and monitoring the disease are urgently needed. Analysis of longitudinal samples linked to carefully collected clinical information with cutting-edge emerging technologies and advanced data analysis and integration methods provides an enormous opportunity for breakthroughs in T1D research. The near future is likely to provide further important insights into disease pathogenesis, new practical assays for predictive and preventive medicine and for clinical trials to find cure for T1D.

Acknowledgment This work was supported by the Academy of Finland Centre of Excellence in Molecular Systems Immunology and Physiology Research, 2012–2017, Decision No. 250114, JDRF, The Sigrid Juselius Foundation and the National Technology Agency of Finland.

References 1 Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature 2010;464:1293– 300. 2 Gale EAM. The rise of childhood type 1 diabetes in the 20th century. Diabetes 2002;51:3353–61. 3 Gale EAM. Epidemiology of type 1 diabetes. Diapedia. Aug 13; 21042821128 rev. no. 42.:http://dx.doi.org/ 10.14496/dia.21042821128.42, 2014 4 Kupila A, Muona P, Simell T et al.; Juvenile Diabetes Research Foundation Centre for the Prevention of Type I Diabetes in Finland. Feasibility of genetic and immunological prediction of type I diabetes in a population-based birth cohort. Diabetologia 2001;44:290–7. 5 Larsson K, Elding-Larsson H, Cederwall E et al. Genetic and perinatal factors as risk for childhood type 1 diabetes. Diabetes Metab Res Rev 2004;20:429–37. 6 Hummel S, Ziegler AG. Early determinants of type 1 diabetes: experience from the BABYDIAB and BABYDIET studies. Am J Clin Nutr 2011;94:1821S–3S. 7 TEDDY Study Group. The environmental determinants of diabetes in the young (TEDDY) study. Ann N Y Acad Sci 2008;1150:1–13. 8 Ilonen J, Hammais A, Laine AP et al. Patterns of beta-cell autoantibody appearance and genetic associations during the first years of life. Diabetes 2013;62:3636–40. 9 Ziegler AG, Rewers M, Simell O et al. Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. JAMA 2013;309:2473–9. 10 Koo BK, Chae S, Kim KM et al. Identification of novel autoantibodies in type 1 diabetic patients using a high-density protein microarray. Diabetes 2014;63:3022–32. 11 Miersch S, Bian X, Wallstrom G et al. Serological autoantibody profiling of type 1 diabetes by protein arrays. J Proteomics 2013;94:486–96.

Scandinavian Journal of Immunology, 2015, 82, 244–253

M. T. Heinonen et al. New Insights and Biomarkers for Type 1 Diabetes 251 .................................................................................................................................................................. 12 Torn C, Hadley D, Lee HS et al.; TEDDY Study Group. Role of type 1 diabetes-associated SNPs on risk of autoantibody positivity in the TEDDY study. Diabetes 2015;64:1818–29. 13 Polychronakos C, Li Q. Understanding type 1 diabetes through genetics: advances and prospects. Nat Rev Genet 2011;12:781–92. 14 Bakay M, Pandey R, Hakonarson H. Genes involved in type 1 diabetes: an update. Genes (Basel) 2013;4:499–521. 15 Guo H, Fortune MD, Burren OS, Schofield E, Todd JA, Wallace C. Integration of disease association and eQTL data using a bayesian colocalisation approach highlights six candidate causal genes in immune-mediated diseases. Hum Mol Genet 2015;24:3305–13. 16 Floyel T, Brorsson C, Nielsen LB et al. CTSH regulates beta-cell function and disease progression in newly diagnosed type 1 diabetes patients. Proc Natl Acad Sci U S A 2014;111:10305–10. 17 Qiu YH, Deng FY, Li MJ, Lei SF. Identification of novel risk genes associated with type 1 diabetes mellitus using a genome-wide genebased association analysis. J Diabetes Investig 2014;5:649–56. 18 Mirza AH, Kaur S, Brorsson CA, Pociot F. Effects of GWASassociated genetic variants on lncRNAs within IBD and T1D candidate loci. PLoS ONE 2014;9:e105723. 19 Ke X, Cortina-Borja M, Silva BC, Lowe R, Rakyan V, Balding D. Integrated analysis of genome-wide genetic and epigenetic association data for identification of disease mechanisms. Epigenetics 2013;8:1236–44. 20 Stefan M, Zhang W, Concepcion E, Yi Z, Tomer Y. DNA methylation profiles in type 1 diabetes twins point to strong epigenetic effects on etiology. J Autoimmun 2014;50:33–7. 21 Onengut-Gumuscu S, Chen WM, Burren O et al. Fine mapping of type 1 diabetes susceptibility loci and evidence for colocalization of causal variants with lymphoid gene enhancers. Nat Genet 2015;47:381–6. 22 Evangelou M, Smyth DJ, Fortune MD et al. A method for genebased pathway analysis using genomewide association study summary statistics reveals nine new type 1 diabetes associations. Genet Epidemiol 2014;38:661–70. 23 Carbonetto P, Stephens M. Integrated enrichment analysis of variants and pathways in genome-wide association studies indicates central role for IL-2 signaling genes in type 1 diabetes, and cytokine signaling genes in crohn’s disease. PLoS Genet 2013;9:e1003770. 24 Setsirichok D, Tienboon P, Jaroonruang N et al. An omnibus permutation test on ensembles of two-locus analyses can detect pure epistasis and genetic heterogeneity in genome-wide association studies. Springerplus 2013;2:230-1801-2-230, eCollection 2013. 25 Heinonen MT, Laine AP, Soderhall C et al.; Finnish Pediatric Diabetes Registry. GIMAP GTPase family genes: potential modifiers in autoimmune diabetes, asthma, and allergy. J Immunol 2015;194:5885–94. 26 Sartori DJ, Wilbur CJ, Long SY et al. GATA factors promote ER integrity and beta-cell survival and contribute to type 1 diabetes risk. Mol Endocrinol 2014;28:28–39. 27 Santin I, Eizirik DL. Candidate genes for type 1 diabetes modulate pancreatic islet inflammation and beta-cell apoptosis. Diabetes Obes Metab 2013;15 (Suppl 3):71–81. 28 Storling J, Brorsson CA. Candidate genes expressed in human islets and their role in the pathogenesis of type 1 diabetes. Curr Diab Rep 2013;13:633–41. 29 Elo LL, Mykkanen J, Nikula T et al. Early suppression of immune response pathways characterizes children with prediabetes in genome-wide gene expression profiling. J Autoimmun 2010;35:70–6. 30 Reynier F, Pachot A, Paye M et al. Specific gene expression signature associated with development of autoimmune type-I diabetes using whole-blood microarray analysis. Genes Immun 2010;11:269–78. 31 Kallionpaa H, Elo LL, Laajala E et al. Innate immune activity is detected prior to seroconversion in children with HLA-conferred type 1 diabetes susceptibility. Diabetes 2014;63:2402–14.

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

32 Ferreira RC, Guo H, Coulson RM et al. A type I interferon transcriptional signature precedes autoimmunity in children genetically at risk for type 1 diabetes. Diabetes 2014;63:2538–50. 33 Helminen O, Aspholm S, Pokka T et al. HbA1c predicts time to diagnosis of type 1 diabetes in children at risk. Diabetes 2014;64: 1719–27. 34 Nanjappa V, Thomas JK, Marimuthu A et al. Plasma proteome database as a resource for proteomics research 2014 update. Nucleic Acids Res 2014;42:D959–65. 35 Metz TO, Qian WJ, Jacobs JM et al. Application of proteomics in the discovery of candidate protein biomarkers in a diabetes autoantibody standardization program sample subset. J Proteome Res 2008;7:698–707. 36 Zhi W, Purohit S, Carey C, Wang M, She JX. Proteomic technologies for the discovery of type 1 diabetes biomarkers. J Diabetes Sci Technol 2010;4:993–1002. 37 Zhi W, Sharma A, Purohit S et al. Discovery and validation of serum protein changes in type 1 diabetes patients using high throughput two dimensional liquid chromatography-mass spectrometry and immunoassays. Mol Cell Proteomics 2011;10:M111012203. 38 McGuire JN, Eising S, Wagner AM, Pociot F. Screening newborns for candidate biomarkers of type 1 diabetes. Arch Physiol Biochem 2010;116:227–32. 39 Crevecoeur I, Rondas D, Mathieu C, Overbergh L. The beta-cell in type 1 diabetes: what have we learned from proteomic studies? Proteomics Clin Appl 2015; Epub ahead of print. 40 Moulder R, Bhosale SD, Erkkila T et al. Serum proteomes distinguish children developing type 1 diabetes in a cohort with HLA-conferred susceptibility. Diabetes 2015;64:2265–78. 41 Hanifi-Moghaddam P, Schloot NC, Kappler S, Seissler J, Kolb H. An association of autoantibody status and serum cytokine levels in type 1 diabetes. Diabetes 2003;52:1137–42. 42 Hanifi-Moghaddam P, Kappler S, Seissler J et al. Altered chemokine levels in individuals at risk of type 1 diabetes mellitus. Diabet Med 2006;23:156–63. 43 Zak KP, Popova VV, Mel’nichenko SV, Tron’ko EN, Man’kovskii BN. The level of circulating cytokines and chemokines in the preclinical and early clinical stages of type IA diabetes mellitus development. Ter Arkh 2010;82:10–5. 44 Yeung WC, Al-Shabeeb A, Pang CN et al. Children with islet autoimmunity and enterovirus infection demonstrate a distinct cytokine profile. Diabetes 2012;61:1500–8. 45 Purohit S, Sharma A, She JX. Luminex and other multiplex high throughput technologies for the identification of, and host response to, environmental triggers of type 1 diabetes. Biomed Res Int 2015;2015:326918. 46 Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol 2009;19:43–51. 47 El Andaloussi S, Mager I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov 2013;12:347–57. 48 Eizirik DL, Colli ML, Ortis F. The role of inflammation in insulitis and beta-cell loss in type 1 diabetes. Nat Rev Endocrinol 2009;5:219–26. 49 Reddy S, Zeng N, Al-Diery H et al. Analysis of peri-islet CD45positive leucocytic infiltrates in long-standing type 1 diabetic patients. Diabetologia 2015;58:1024–35. 50 Rodriguez-Calvo T, Ekwall O, Amirian N, Zapardiel-Gonzalo J, von Herrath MG. Increased immune cell infiltration of the exocrine pancreas: a possible contribution to the pathogenesis of type 1 diabetes. Diabetes 2014;63:3880–90. 51 Atkinson MA. Losing a grip on the notion of beta-cell specificity for immune responses in type 1 diabetes: can we handle the truth? Diabetes 2014;63:3572–4. 52 Krogvold L, Skog O, Sundstrom G et al. Function of isolated pancreatic islets from patients at onset of type 1 diabetes; insulin

252 New Insights and Biomarkers for Type 1 Diabetes M. T. Heinonen et al. ..................................................................................................................................................................

53

54

55

56

57

58

59

60

61

62

63

64 65 66

67

68

69

70 71

72 73 74

secretion can be restored after some days in a non-diabetogenic environment in vitro. results from the DiViD study. Diabetes 2015;64:2506–12. Krogvold L, Edwin B, Buanes T et al. Detection of a low-grade enteroviral infection in the islets of langerhans of living patients newly diagnosed with type 1 diabetes. Diabetes 2015;64:1682–7. Nadine LD, Giam K, Purcell AW. The Use of Proteomics to Dissect the Molecular Specificities of T Cells in Type 1 Diabetes. J Diabetes Metab 2013;S12:1–7. McGinty JW, Chow IT, Greenbaum C, Odegard J, Kwok WW, James EA. Recognition of posttranslationally modified GAD65 epitopes in subjects with type 1 diabetes. Diabetes 2014;63:3033–40. Rondas D, Crevecoeur I, D’Hertog W et al. Citrullinated glucoseregulated protein 78 is an autoantigen in type 1 diabetes. Diabetes 2015;64:573–86. Oresic M, Simell S, Sysi-Aho M et al. Dysregulation of lipid and amino acid metabolism precedes islet autoimmunity in children who later progress to type 1 diabetes. J Exp Med 2008;205:2975–84. Pflueger M, Seppanen-Laakso T, Suortti T et al. Age- and islet autoimmunity-associated differences in amino acid and lipid metabolites in children at risk for type 1 diabetes. Diabetes 2011;60:2740–7. Sysi-Aho M, Ermolov A, Gopalacharyulu PV et al. Metabolic regulation in progression to autoimmune diabetes. PLoS Comput Biol 2011;7:e1002257. Oresic M, Gopalacharyulu P, Mykkanen J et al. Cord serum lipidome in prediction of islet autoimmunity and type 1 diabetes. Diabetes 2013;62:3268–74. La Torre D, Seppanen-Laakso T, Larsson HE et al.; DiPiS Study Group. Decreased cord-blood phospholipids in young age-at-onset type 1 diabetes. Diabetes 2013;62:3951–6. Kallionpaa H, Laajala E, Oling V et al.; DIABIMMUNE Study Group. Standard of hygiene and immune adaptation in newborn infants. Clin Immunol 2014;155:136–47. Gronlund MM, Lehtonen OP, Eerola E, Kero P. Fecal microflora in healthy infants born by different methods of delivery: permanent changes in intestinal flora after cesarean delivery. J Pediatr Gastroenterol Nutr 1999;28:19–25. Dunne JL, Triplett EW, Gevers D et al. The intestinal microbiome in type 1 diabetes. Clin Exp Immunol 2014;177:30–7. Noverr MC, Huffnagle GB. Does the microbiota regulate immune responses outside the gut? Trends Microbiol 2004;12:562–8. Penders J, Gerhold K, Thijs C et al. New insights into the hygiene hypothesis in allergic diseases: mediation of sibling and birth mode effects by the gut microbiota. Gut Microbes 2014;5:239–44. Kostic AD, Gevers D, Siljander H et al. The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe 2015;17:260–73. de Goffau MC, Luopajarvi K, Knip M et al. Fecal microbiota composition differs between children with beta-cell autoimmunity and those without. Diabetes 2013;62:1238–44. Davis-Richardson AG, Ardissone AN, Dias R et al. Bacteroides dorei dominates gut microbiome prior to autoimmunity in finnish children at high risk for type 1 diabetes. Front Microbiol 2014;5:678. Tilg H, Moschen AR. Microbiota and diabetes: an evolving relationship. Gut 2014;63:1513–21. Guo T, Kouvonen P, Koh CC et al. Rapid mass spectrometric conversion of tissue biopsy samples into permanent quantitative digital proteome maps. Nat Med 2015;21:407–13. Kondrashova A, Hyoty H. Role of viruses and other microbes in the pathogenesis of type 1 diabetes. Int Rev Immunol 2014;33:284–95. Filippi CM, von Herrath MG. Viral trigger for type 1 diabetes: pros and cons. Diabetes 2008;57:2863–71. Ylipaasto P, Klingel K, Lindberg AM et al. Enterovirus infection in human pancreatic islet cells, islet tropism in vivo and receptor

75

76

77

78

79

80

81

82

83

84

85

86

87

88 89

90

91

involvement in cultured islet beta cells. Diabetologia 2004;47:225– 39. Richardson SJ, Leete P, Bone AJ, Foulis AK, Morgan NG. Expression of the enteroviral capsid protein VP1 in the islet cells of patients with type 1 diabetes is associated with induction of protein kinase R and downregulation of mcl-1. Diabetologia 2013;56:185–93. Richardson SJ, Willcox A, Bone AJ, Foulis AK, Morgan NG. The prevalence of enteroviral capsid protein vp1 immunostaining in pancreatic islets in human type 1 diabetes. Diabetologia 2009;52:1143–51. Hamalainen S, Nurminen N, Ahlfors H et al. Coxsackievirus B1 reveals strain specific differences in plasmacytoid dendritic cell mediated immunogenicity. J Med Virol 2014;86:1412–20. Laitinen OH, Honkanen H, Pakkanen O et al. Coxsackievirus B1 is associated with induction of beta-cell autoimmunity that portends type 1 diabetes. Diabetes 2014;63:446–55. Oikarinen S, Tauriainen S, Hober D et al.; VirDiab Study Group. Virus antibody survey in different european populations indicates risk association between coxsackievirus B1 and type 1 diabetes. Diabetes 2014;63:655–62. Kramna L, Kolarova K, Oikarinen S et al. Gut virome sequencing in children with early islet autoimmunity. Diabetes Care 2015;38:930– 3. Sorkio S, Cuthbertson D, Barlund S et al.; TRIGR Study Group. Breastfeeding patterns of mothers with type 1 diabetes: results from an infant feeding trial. Diabetes Metab Res Rev 2010;26:206–11. Virtanen SM, Nevalainen J, Kronberg-Kippil€a C et al. Food consumption and advanced ß cell autoimmunity in young children with HLA-conferred susceptibility to type 1 diabetes: a nested casecontrol design. Am J Clin Nutr 2012;95:471–8. Lamb MM, Miller M, Seifert JA et al. The effect of childhood cow’s milk intake and HLA-DR genotype on risk of islet autoimmunity and type 1 diabetes: the diabetes autoimmunity study in the young. Pediatr Diabetes 2015;16:31–8. Beyerlein A, Chmiel R, Hummel S, Winkler C, Bonifacio E, Ziegler AG. Timing of gluten introduction and islet autoimmunity in young children: updated results from the BABYDIET study. Diabetes Care 2014;37:e194–5. N€ant€o-Salonen K, Kupila A, Simell S et al. Nasal insulin to prevent type 1 diabetes in children with HLA genotypes and autoantibodies conferring increased risk of disease: A double-blind, randomised controlled trial. Lancet 2008;372:1746–55. Bonifacio E, Ziegler AG, Klingensmith G et al.; Pre-POINT Study Group. Effects of high-dose oral insulin on immune responses in children at high risk for type 1 diabetes: the pre-POINT randomized clinical trial. JAMA 2015;313:1541–9. Knip M, Akerblom HK, Becker D et al.; TRIGR Study Group. Hydrolyzed infant formula and early beta-cell autoimmunity: a randomized clinical trial. JAMA 2014;311:2279–87. Harjutsalo V, Sund R, Knip M, Groop PH. Incidence of type 1 diabetes in finland. JAMA 2013;310:427–8. Makinen M, Simell V, Mykkanen J et al. An increase in serum 25hydroxyvitamin D concentrations preceded a plateau in type 1 diabetes incidence in finnish children. J Clin Endocrinol Metab 2014;99:E2353–6. Simpson M, Brady H, Yin X et al. No association of vitamin D intake or 25-hydroxyvitamin D levels in childhood with risk of islet autoimmunity and type 1 diabetes: the diabetes autoimmunity study in the young (DAISY). Diabetologia 2011;54:2779–88. Reinert-Hartwall L, Honkanen J, Harkonen T et al.; DIABIMMUNE Study Group. No association between vitamin D and betacell autoimmunity in finnish and estonian children. Diabetes Metab Res Rev 2014;30:749–60.

Scandinavian Journal of Immunology, 2015, 82, 244–253

M. T. Heinonen et al. New Insights and Biomarkers for Type 1 Diabetes 253 .................................................................................................................................................................. 92 Strachan DP. Hay fever, hygiene, and household size. BMJ 1989;299:1259–60. 93 Eugster A, Lindner A, Catani M et al. High diversity in the TCR repertoire of GAD65 autoantigen-specific human CD4 + T cells. J Immunol 2015;194:2531–8. 94 Reinert-Hartwall L, Honkanen J, Salo HM et al.; DIABIMMUNE Study Group. Th1/Th17 plasticity is a marker of advanced beta cell autoimmunity and impaired glucose tolerance in humans. J Immunol 2015;194:68–75. 95 Kenefeck R, Wang CJ, Kapadi T et al. Follicular helper T cell signature in type 1 diabetes. J Clin Invest 2015;125:292–303. 96 Lehtimaki S, Lahesmaa R. Regulatory T cells control immune responses through their non-redundant tissue specific features. Front Immunol 2013;4:294. 97 Bluestone JA, Tang Q, Sedwick CE. T regulatory cells in autoimmune diabetes: past challenges, future prospects. J Clin Immunol 2008;28:677–84. 98 Ryba-Stanislawowska M, Skrzypkowska M, Mysliwiec M, Mysliwska J. Loss of the balance between CD4(+)Foxp3(+) regulatory T cells and CD4(+)IL17A(+) Th17 cells in patients with type 1 diabetes. Hum Immunol 2013;74:701–7.

Ó 2015 The Foundation for the Scandinavian Journal of Immunology

99 Garg G, Tyler JR, Yang JH et al. Type 1 diabetes-associated IL2RA variation lowers IL-2 signaling and contributes to diminished CD4 + CD25 + regulatory T cell function. J Immunol 2012;188:4644–53. 100 Waldron-Lynch F, Kareclas P, Irons K et al. Rationale and study design of the adaptive study of IL-2 dose on regulatory T cells in type 1 diabetes (DILT1D): a non-randomised, open label, adaptive dose finding trial. BMJ Open 2014;4:e005559-2014005559. 101 Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 2012;30:531–64. 102 Rubtsov YP, Rasmussen JP, Chi EY et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 2008;28:546–58. 103 Li MO, Wan YY, Flavell RA. T cell-produced transforming growth factor-beta1 controls T cell tolerance and regulates Th1- and Th17cell differentiation. Immunity 2007;26:579–91. 104 Theil A, Wilhelm C, Guhr E, Reinhardt J, Bonifacio E. The relative merits of cord blood as a cell source for autologous T regulatory cell therapy in type 1 diabetes. Horm Metab Res 2015;47:48–55.

New Insights and Biomarkers for Type 1 Diabetes: Review for Scandinavian Journal of Immunology.

The increasing incidence of type 1 diabetes observed in the past 60 years has spawned massive efforts in multiple research fields to elucidate the aet...
253KB Sizes 1 Downloads 6 Views