You are on page 1of 28

Critical ReviewsTM in Immunology, 39(4):239 – 265 (2019)

Pathophysiology, Etiology, Epidemiology of Type


1 Diabetes and Computational Approaches for
Immune Targets and Therapy
Begum Dariya,a Gayathri Chalikonda,b Gowru Srivani,a Afroz Alam,a &
Ganji Purnachandra Nagarajub,*
a
Department of Bioscience and Biotechnology, Banasthali University, Vanasthali, Rajasthan, 304022, India;
b
Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322,
USA
*Address all correspondence to: Ganji Purnachandra Nagaraju, PhD, DSc, FAACC, Department of Hematology and Medical Oncology, Winship
Cancer Institute C#3025, Emory University, Atlanta, GA-30322, USA; Tel.: +1-470-585-8202, E-mail:pganji@emory.edu

ABSTRACT: Autoimmune diseases occur when the body’s natural defense system fails to differentiate its own cells
from the foreign cells and mistakenly attacks the healthy cells. Among the autoimmune diseases, the most common
serious disease is the type 1 diabetes (T1D). Biomarkers like c-peptide, autoantibodies, and glycated molecules are
now widely used for the early diagnosis of diabetes. However, the diverse nature of biomarkers and the available au-
toantibodies as biomarkers are not enough to differentiate the heterogeneity inherent in T1D. Novel biomarkers have
allowed the introduction of bioinformatics for assimilating the new data into clinical tools. Computer-aided drug design
contributes to the discovery of novel autoantibodies, and molecular docking promises to enhance it. Moreover, the study
of the pathophysiology of diabetes via molecular simulation has been proposed. In this review article, we focus on the
characterization of the etiology, epidemiological factors, and mechanisms of hyperglycemia that induce cellular damage
due to oxidative stress and proinflammatory responses. We also decribe novel biomarkers used for the detection of β-cell
destruction and diagnosis at early stages. Bioinformatics tools including molecular docking, sequence alignment, and
homology modeling are also presented. This report supports researchers in drug design, in disease detection at an early
phase, and in therapy development for T1D-associated complications.

KEY WORDS: type 1 diabetes, etiology, epidemiology, biomarkers, β-cells, insulin, hyperglycemia, complications,
bioinformatics, docking

ABBREVIATIONS: 2D DIGE, two-dimensional difference gel electrophoresis; AGE, advanced glycation end products; APC,
antigen-presenting cells; AR, aldose reductase; bdnf, brain-derived neutrophic factor; CD4, cluster of differentiation; DAG, diacyl
glycerol; DAN, diabetic autonomic neuropathy; DPN, diabetic peripheral neuropathy; ECM, extracellular matrix; ESI, electro-
spray ionization; esRAGE, endogenous secretion RAGE; GAD, glutamic acid decarboxylase; GFAT, glutamine:fructose-6-phos-
phate amidotranferase; GLUT, glucose transporter; GPPAD, global platform for the prevention of autoimmune diabetes; HbA1c,
glycated hemoglobin; HLA, human leukocyte antigen; IA-2, insulinoma 2–associated autoantibody; IAA, insulin autoantibod-
ies; ICA, islet cell cytoplasmic autoantibody; IFN, interferon; iNOS, inducible nitric oxide synthase; JM, juxta-transmembrane;
LC-MS, liquid chromatography-mass spectrometry; MA, microalbuminuria; MALDI, matrix-associated laser desorption ion-
ization; MHC, major histocompatibility complex; MNP, mononuclear phagocytic cells; NF-κB, nuclear factor kappa B; NIH,
National Institutes of Health; PBPP, platelet-based protein precursor; PDGFR, platelet-derived growth factor; PKC, protein ki-
nase C; PLN, pancreatic lymph node; PTMs, posttranslational modifications; PTP, protein tyrosine phosphatase; SDH, sorbitol
dehydrogenase; Shc, Src2 homology domain containing; SUR, sulfonylurea receptor; T1D, type 1 diabetes; TGF-β1, transcription
growth factor beta 1; TOF, time of flight; TNF, tumor necrosis factor; URP-GlcNAc, uridine diphosphate N-acetylglucosamine;
VEGF, vascular endothelial growth factor; ZNT8A, zinc transporter 8 autoantibody

I. INTRODUCTION distinguish its own cells from invaders, it attacks it-


self with autoantibodies. Thus, any individual with
The healthy immune system functions to shield the an autoimmune condition has an elevated risk of de-
body from any harmful foreign substances, includ- veloping excessive inflammation and tissue damage,
ing parasites, bacteria, and cancer cells. However, if which are hallmarks of autoimmune disorders. To
the immune system behaves abnormally and fails to date, researchers have identified > 80 autoimmune

1040-8401/19/$35.00 © 2019 by Begell House, Inc. www.begellhouse.com239


240 Dariya et al.

disorders, and the National Institutes of Health the disease can be treated with the adoption of in-
(NIH) estimates that ~ 23.5 million individuals in the tensive insulin therapy.11 Furthermore, pancreatic
United States are affected by such diseases. Autoim- transplantation or β-cell regeneration can restore
mune disorders can be categorized into two types, organ function. On the other hand, managing these
namely a systemic autoimmune disease that attacks complications is the crucial goal of the clinical and
many organs and a localized one that attacks a single research fields, that is, to save a patient’s life. A clear
organ. The diagnosis of an autoimmune disease is dif- knowledge of the structure of the proteins, their in-
ficult and misleading because these diseases change teractions, and detection of biomarkers for early di-
in severity over time. Common symptoms include agnosis are essential.
fatigue, rash, dizziness, and joint pain. Autoimmune Bioinformatics serves as an interdisciplinary
disease has no cure but needs therapy only to ease model for molecular biology and computational
the symptoms. The most common autoimmune dis- sciences. It includes interconnected databanks of
orders are type 1 diabetes (T1D), multiple sclerosis, clinical data. Additionally, computational tools and
rheumatoid arthritis, celiac disease and Alzheimer’s algorithms are used to determine similarity, homol-
disease. In this report, we have focused on T1D. ogy sequencing, and envisioning the 3D structures
T1D, insulin-dependent diabetes mellitus, is a of a protein along with the amino acid sequencing.
disorder in which the body cannot produce enough Thus, determining the interaction of a drug and a
insulin. This chronic disease is mediated by type IV protein becomes much easier with bioinformatics.
hypersensitivity, a cell-mediated immune response In this review article, we have focused on the epi-
driven by aberrant T lymphocytes that recognize pan- demiology, signaling pathways, and complications
creatic islets of Langerhans, damage insulin-making caused by T1D with an in silico approach.
β cells, and cause disease progression.1,2 This ge-
netic aberration causes the loss of self-recognition in II. EPIDEMIOLOGY AND ETIOLOGY
the T cells and targets β-cell antigens. Furthermore,
these T cells recruit other CD4+ and CD8+ T cells to A. Epidemiology
target β cells. The common symptoms experienced
by patients are polyphagia, glycosuria, polyuria, and The symptomatic and presymptomatic natures of
polydipsia. Hyperglycemia and deficiency in insu- the disease can be estimated for the analysis of ep-
lin are the prime reason for 5%–10% of diagnosed idemiology. The International Diabetes Federation
diabetes cases. T1D is the most prevalent disease, estimates that 8.8% of adults worldwide have dia-
with the incidence mostly in the population under 18 betes. Interestingly, T1D is much less prevalent than
years of age worldwide, with higher rates in Finland T2D; T1D is commonly diagnosed in children aged
and Sardinia and lower rates in China and Venezu- < 15 years, with > 500,000 children affected cur-
ela.3 Additionally, the international research DIA- rently and 900,000 new diagnoses every year.12 Oc-
MOND and EURODIAB reported the highest graph casionally, this disease is diagnosed after 50 years
peak for T1D occurs in the youngest population.3 of age,13 as evidenced by a genetic risk score from
Moreover, an increase in the incidence rate among the UK Biobank which states that the adult onset is
children was reported globally to be ~ 3%–5% every 42% for those aged 31–60 years.14 It is mostly fre-
year since 1960, mostly in developing countries.4–8 quently diagnosed in developed countries like Eu-
The exact cause for the disease is not understood; ropean countries, Australia, and North America and
environmental conditions, autoimmunity, and ge- less so in countries like Japan, China, and South Ko-
netic factors are all9 believed to play critical roles rea. These epidemiological variations may be due to
in the disease onset. T1D is characterized by the the genetic susceptibility associated with the class
dysregulation of glucose, which if untreated can II HLA (human leukocyte antigens) haplotypes (but
be fatal.10 Although there is no way to completely non-HLA loci are also reported to be susceptible),
prevent T1D, 1993 publications from the landmark environmental, and/or lifestyle factors. The geno-
diabetes control and complication trial reported that type HLA-DR-DQ always varies among countries,

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy241

and European people with this genotype are at B. Etiology


higher risk than those with an Asian background.15
Additionally, the risk is also high in children of im- The etiology factors for the autoimmunity targeted
migration parents.16 According to the studies from against islet cells include autoantibodies, genetic,
the UK Prospective Diabetes, younger adults diag- and environmental factors. The autoantibodies are
nosed with T2D are also consistent phenotypically first screened in infants with a T1D father or mother,
with T1D because they have β cells that target auto- allowing the targeting of the genetic and environ-
antibodies.17 When the incidence rates between gen- mental factors.29
ders were compared, girls had a higher peak than
boys.18,19 However, the incidence rate increases with
age; the incidence gradually decreases in women 1. Autoantibodies
but continues to be high for men.3 Environment and As reported, β-cell destruction by the antibodies
lifestyle are interlinked and are accompanied by ad- starts at an early stage, and ~ 90% of the cells are
vances in the economy and industry, and they play destroyed before the symptomatic stage. These au-
a key role in the pathogenesis of childhood T1D.20 toantibodies attack insulin and GAD65 starting at
Additionally, the nutritional factors and microbial > 1 year of age. Moreover, the incidence for the
infections are also risk factors for developing an au- insulin antibodies is highest in patients with the
toimmune disease.21 HLA-DR4-DQ8 haplotype and HLA-DR3-DQ2
In another aspect, presymptomatic T1D is for GAD65.29 In addition, other antibodies are lo-
considered a hallmark for epidemiology, and its cated in the secretory vesicles that attack tyrosine
autoantibodies act as biomarkers. The identifi- phosphatase molecules: IA-2, IA-2β, and ZNT8.
cation of these markers is an obstacle due to the Although the functions of IA-2 and IA-2β are not
heterogeneity in T1D in children. This heterogene- known, ZNT8 reportedly passes Zn ions from the
ity includes autoantibodies against glutamic acid cytoplasm to the secretory vesicles.29 The risk of
decarboxylase (GAD) and insulin autoantibodies these antibodies is illustrated in Table 1.
(IAA), which promote the disease by initiating an
autoimmune process at different ages.22–25 Stage 2
symptomatic progression of T1D is always diag- 2. Genetics
nosed after the detection of multiple autoantibod-
ies, whereas the initial antibodies can be diagnosed T1D is a polygenically inherited disease. Although
only at the seroconversion age of the patient. For the HLA genes are important for the immune re-
instance, for stage 3 symptomatic T1D, according sponse, other groups of genes, located on chromo-
to TEDDY’s study,26 those under 5 years of age de- some 6 that code for the major histocompatibility
tected with 1, 2, and 3 autoantibodies showed in- complex (MHC),33–35 help recognize foreign cells
cidence rates of 11%, 36%, and 47%, respectively. and maintain self-tolerance. The loci that play a key
According to the DAISY, BABYDIAB, DIPP, and role in developing autoimmunity are HLA-DR and
BABYDIET analyses, the multiple autoantibod- HLA-DQ, with their alleles HLA-DQB1, HLA-
ies targeting islets in 585 children of ages 5, 10, DRB1, and HLA-DQA1. Moreover, the grouping
and 15 years with higher risk for incidence were ~ of haplotypes DR3-DQ2 and DR4-DQ8 are cor-
44%, 70%, and 84%, respectively.26 In adults, the related with a higher risk of the disease.36 Addition-
prevalence of the detection of IAA decreased with ally, the non-loci HLA cells also contribute to >
increasing age; prevalences of 0.8% and 1.1% for 50% of the risk for T1D according to genome-wide
GAD65 in adults were detected in two studies.27,28 association studies.37 Nevertheless, other genes are
The progression of the autoantibodies was deter- involved in the growth and progression of autoan-
mined only within 2–4 years of age.26,28,29 Thus, this tibodies in stages 1 and 2, respectively. Such genes
could be characterized for developing preventive include INS, IL2RA, IFIH1, and CTSH, among oth-
measures for the therapy. ers (Table 2).

Volume 39, Issue 4, 2019


242 Dariya et al.

TABLE 1: Etiology: autoantibodies


Autoantibodies Age Genetic haplotype Ref.
GAD65 >1 HLA-DR3-DQ2 30
Insulin 1-2 HLA-DR4-DQ8 30
Protein phosphatase molecules
Autoantibodies Risk
IA-2 and IA-2β Reaching stage 3 31
ZNT8 (3 variants having Trp, Arg, Gln at Stage 1 and stage 2 32
position 325)

TABLE 2: Genes involved in progression of autoantibodies


Protein Gene Dysregulated function of the gene Phase
Proinsulin-insulin INS Low expression of thymus and Stage I and II
tolerant against proinsulin
Erb-b2 receptor tyrosine kinase 3 ERBB3 Regulates β-cell apoptosis Stage I and II
Protein tyrosine phosphatase, nonreceptor PTPN22 Expressed due to the activation of T Stage I and II
type 22 and B cell antigen receptor
Protein tyrosine phosphatase, nonreceptor PTPN2 Sensitive to the induction of β-cell Stage II
type 2 apoptosis
IL-2 receptor subunit α IL2RA Respond to IL-2 present in T cells

and T memory cells

3. Environment methylation, and histone modification. DNA meth-


ylation is the most commonly detected risk factor
Environmental factors include diet as well as re- associated with the secretion of insulin in the body.
gional toxicity differences. Diet takes into account Various studies have indicated that variation in
breast feeding in infancy, diversity among diets, methylation is seen at the 4 CpG site of the tran-
including gluten, cow milk, rye, egg, as well as vi- scription site of the insulin gene in T1D patients. The
tamins in foods. Other factors include gestational methylation of DNA has been observed at CpG-34,
infections, viral infections, and the gut microbiota. CpG-135, CpG-19, and CpG-180. These genetic al-
The chemical toxins include nitrites, nitrates, N-ni- terations near the pre-proinsulin coding sequences
troso compounds, and polychlorinated biphenyls, increase the risk of T1D development.38 Similarly,
which damage β cells through immunological path- for histone modifications, acetylation is increased in
ways. Due to such environmental factors, the epi- the lys 9 of the H3 histone protein (H3K9Ac) in the
genetic modifications promote autoimmunity. susceptible HLA-DRB1 and HLA-DQB1 in T1D in-
dividuals.39 Also, epigenetic modifications in miRNA
4. Epigenetics result in alterations in the immune response, cell cycle,
and apoptosis. As reported in studies about miRNA
Epigenetics is a term used to define what is beyond expression in the regulatory T cells of T1D patients,
genetics, and it acts as a mediator for environmen- a few miRNAs (including miRNA 31, miRNA-100,
tal risk factors. As such, these changes are stable miRNA-125b, miRNA-365, miRNA-335, miRNA-
and alter heredity due to changes in the chromatin 20b, miRNA-99a and miRNA-151) are underex-
without DNA sequence changes. Common epigen- pressed and miRNA-146a is overexpressed. These
etic modifications include miRNA regulation, DNA gene expressions indicate that miRNAs are also

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy243

associated with the regulation of immunity.40 Thus, a mostly screened in patients with a first-degree rel-
clear understanding of the association of epigenetics ative having an HLA genotype for T1D diagnosis.
and environmental risk factors for the occurrence of In addition, the appearance, epitope, number, and
T1D is essential for the moderation of the disease. affinity of autoantibodies are even correlated with
the risk of T1D.48 For instance, GADA and IAA are
III. BIOMARKERS most frequently screened in children as the first au-
toantibodies, yet they disappear with age at clinical
Abnormality in the pathology of any disease is onset.26,49–52 Furthermore, the seroconversion-linked
predicted for the onset of the disease by indicators susceptibility loci (including PTPN22, INS, SH2B3,
called biomarkers, which play a vital role in the di- and ERBB3) are reported as the higher-risk geno-
agnosis and prognosis in the clinical field. Various types, whereas the ERBB3 and SH2B3 are specifi-
biomarkers have been developed for the detection, cally connected with the autoantibody appearance.53
prevention, and therapy of a disease, such as the Post-translation modifications and splicing of β-cell
DAISY, DIPP, DIPIS and TEDDY screenings, with proteins are caused by the endoplasmic reticulum
biological samples based on age, sex, geography, and β-cell oxidative stress triggered by inflamma-
and risk factors. For instance, the most common tion. The post-translational modifications reported
biomarkers are serum diagnostic markers that de- in T1D are citrullination of endoplasmic reticulum
tect hyperglycemia, C-peptide, and autoantibodies. chaperon’s glucose-regulated protein 78,54 type II
These biomarkers involve monitoring glycated he- ROS modified collagen,55 phosphorylation of pe-
moglobin (HbA1c) for the recent history of blood ripherin,56 and oxidative insulin.57 Nucleic acids are
sugar levels. Furthermore, hyperglycemia condition also employed as biomarkers because they carry ge-
causes far-reaching oxidative and inflammatory ef- netic and epigenetic changes associated with disease
fects. Recently, a serum profile of 19 prediabetic development and progression. Among them, DNA
patients from the birth to the onset of disease was methylation is a biomarker for the diagnosis of dis-
prepared by Moulder et al.41 to narrow down markers ease. The presence or circulation of amylin DNA
for predicting the progression of the disease. Previ- and unmethylated insulin showing no methylation at
ously, a change in the levels of chemokines and cy- the CpG sites of the insulin gene in β cells have been
tokines expressions was detected in children at high detected in the bloodstream and have been explored
risk for T1D.42,43 Moreover, the increased expres- as markers for the destruction of β cells, which is an
sion of IL-16, IL-18, IL-6, and TNFα has been ob- early sign of disease onset.58 MiRNAs also play a
served in T1D children.44 Purohit et al.45 later noted key role in regulating the pathogenesis of T1D and
the downregulation of 4 cytokines (IL-8, MCP-1, β-cell function; thus, they represent potential bio-
MIP-Iβ and IL-Ira) in serum samples of T1D pa- markers. Approximately 200 miRNAs have been
tients. The C-peptide and insulin are released in observed in murine and human samples with T1D.59
equimolar concentrations from the vesicles packed Previous studies have reported upregulation in the
as proinsulin.46 C-peptide levels, which are sensitive expression of miRNAs detected in the serum samples
to β-cells, play a crucial role in differentiating T1D of T1D: miRNA-24,60–62 miRNA-21,60,61,63 miRNA-
from T2D.47 Additionally, predictive biomarkers are 210-5p,61,63 miRNA-181a-5p,61,64 miRNA-148a.60,61
essential for detecting the silent asymptomatic de- An increase in the circulation of miRNA-375 has
struction period of β cells. Autoantibodies are the been correlated with the destruction of β cells in
primary markers for diagnosis and prediction of T1D patients.60 Other miRNAs have been found
T1D. The autoantibodies produced in response to in the serum as well, including miRNA-21 and
autoantigens include insulin autoantibody (IAA), miRNA-210.63 Although miRNA-21a, miRNA-93,
glutamic acid decarboxylase (GADA), islet-cell and miR-146 are typically downregulated, miRNA-
cytoplasmic autoantibody (ICA), zinc transporter 326 expression is increased in T1D.65–67 Also, me-
8 autoantibody (ZNT8A), and insulinoma-2 associ- tabolomics biomarkers have been studied for their
ated autoantibody (IA-2). These autoantibodies are use in diagnosis; lipids, metabolites, and small

Volume 39, Issue 4, 2019


244 Dariya et al.

molecules have been evaluated. Amino acids (me- is cotranslationally inserted into the endoplasmic
thionine)68 and lipid metabolites (sphingolipids,69 reticulum lumen. The small peptide fragment pres-
phospholipids70) have also been explored for their ent at the N-terminal signal sequence region is re-
biomarker utility. Further advances that contribute moved by the signal peptidase to form proinsulin.
for the prediction and progression of the disease are The proinsulin then transits from the endoplasmic
needed to improve T1D detection and monitoring. reticulum to the Golgi apparatus (cisternae), where
it forms disulphide bonds between the A and B
IV. PHYSIOLOGY OF β-CELLS PRODUCING chains and is packaged into clatherin-coated ves-
INSULIN icles containing insulin and C-peptide (i.e., con-
necting peptide). Later, inside the vesicle, insulin
The pancreas is a heterocrine organ of the digestive and the C-peptide are separated and then exocy-
and endocrine systems that performs both exocrine tosed into the cytoplasm from the β cells in equi-
and endocrine functions. The endocrine functions molar concentrations. Thus, the C-peptide acts as
depend on the microscopic cluster of cells called is- a marker for insulin secretion during T1D diag-
lets of Langerhans, which constitute 1%–3% of the nosis.74 The active insulin initiates its pathway by
complete pancreatic mass. The islets in the pancreas binding to the membrane receptor proteins at the
are in turn divided into four cell types: glucagon-se- α subunit on the outside the cell and activates it.
creting α cells, insulin producing β-cells, somatosta- Later, the β subunit attached to the α subunit to-
tin producing δ-cells, and polypeptide-producing ward the inner side becomes autophosphorylated,
cells.71 Among these, the major source of insulin which further activates a local tyrosine kinase and
production is pancreatic β cells.71–73 The β cells eventually phosphorylates varied number of multi-
sense the glucose circulation in the blood, which ple intracellular enzymes including insulin recep-
stimulates insulin and concomitantly releases it.71 tor substrates. This process stimulates the glucose
The glucose enters the pancreatic β cells through transport channels, GLUT, on the cell surface, which
GLUT (GLUT1 in humans and GLUT2 in rodents) allows cells to take in glucose and store it for future
membrane-bound glucose transporters. Once in the use.
β cells, the glucose undergoes phosphorylation via
glucokinase and hexokinase, which further pro- VI. PATHOPHYSIOLOGY: THE IMMUNE CELL
duces ATP by glycolysis. Moreover, the ratio of TRAFFICKING AND β-CELL APOPTOSIS
glucokinase and hexokinase also determines the
glucose sensed by β cells. The gradual increase of In T1D, the insulin-producing pancreatic β cells are
ATPs in the β cells causes the KATP channel closure, reduced by 70%–80% due to insulitis and necrosis
membrane depolarization, and opening of the volt- of the tissue. This process eventually causes loss of
age-gated calcium channels. The rapid influx of Ca2+ function over years. This pathology is due to the au-
and subsequent increased intracellular Ca2+ lead to toimmune destruction mediated by the autoantibod-
the development of vesicles filled with insulin that ies released against the islet cell antigens.75,76 The
attach to the membrane and release via exocytosis. T cells are present inside the islets of Langerhans,
causing destruction of the insulin-producing β cells.
V. SYNTHESIS OF INSULIN Along with the T cells, macrophages, and mediators
(including oxygen free radicals, cytokines, and nitric
Insulin is an anabolic hormone that maintains glu- oxides) are all involved in insulitis. Earlier, Hostens
cose homeostasis. It functions to carry and transport et al.77 determined that the β cells, when exposed to
glucose to various tissues for energy metabolism. IL-1β and IFN-γ, show similar functional changes
Skeletal muscles and adipocytes comprise most as in prediabetic patients. Later, Ohara et al.78 also
glucose consumption. Its synthesis is similar to that determined that IL-1β also mediates decrease in the
of other peptide hormones and begins with the for- docking of insulin vesicles to the β cell membrane,
mation of linear polypeptide pre-proinsulin, which thereby resulting in the prevention of the first phase

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy245

of insulin secretion. Further introduction of β cells promoter of insulin, also impairs islet cells. Later,
to TNF-α, IL-1β, and IFN-γ eventually causes β-cell the autoimmune response includes islet cell autoan-
death.79 tibodies (ICA) and anti-GAD65 antibodies, which
Immune cell trafficking during T1D progression further contribute to autoimmune diseases. The
initially starts with T cells that escape the negative Cocksackie B virus is also involved in the progres-
selection in the thymus and make their way toward sion of T1D through molecular mimicry. The fol-
the pancreatic lymph node (PLN) to traffic into the lowing subsections describe the factors that promote
islets. On the other side, the antigen-presenting cells T1D.
(APCs) produce β-cell antigen to target PLN in a
pathological manner.80–82 The activated T cells in the A. Autoantibodies
PLN later infiltrate the islets via extravasation af-
ter recirculating within the thoracic, lymphatic duct, The islet autoantibodies are fundamentally rec-
and bloodstream. These further recruit additional ognized as the markers for T1D, and their respec-
lymphocytes into the islets.83 Additionally, the T tive antigens present on the secretory granules in
cells are stimulated repeatedly with their interaction the β cells, including the zinc transporter ZnT8-
with the APCs present on the CD11c+, resulting in (ZnT8A),90 insulin-(IAA),91 GAD65-(GADA)92 and
the production of cytokines, like IFN-γ. The specific the protein tyrosine phosphatase-like protein IA-2-
pancreatic T-cell response is detected during the on- (IA-2A).93 Autoantibodies also recognize the ho-
set of T1D due to MHC II rather than MHC I through mologous isoforms IA-2β94 and GAD67.95
the islet antigens produced by dendritic cells. Thus,
an inflammation environment with an increased 1. GAD65-GADA
number of B cells and mononuclear phagocytic cells
(MNP) is created due the IFN-γ production, which GAD65 is an enzyme that exists in the multiprotein
further upregulates the production of chemokines complex of the cytoplasmic side membrane of the
and vascular adhesion molecules in the endothelia synaptic like microvesicle. The structure includes 3
of the islets.81,84,85 The CD11c+ cells behave as gate- domains, a terminal NH2 domain, a middle domain
keeper cells that recruit both T and B cells to the with a proximal 5’-phosphate (PLP) cofactor bind-
infiltrated islet cells.86 In addition, the CD11c+ also ing site, and a terminal COOH catalytic domain.96,97
produce ~ 20 different chemokines with the ability It plays a central role in catalyzing the biosynthesis
to bind to the receptors on islet T cells.86 Further- of GABA. GABA is a gamma aminobutyric acid
more, the initial islet-targeting autoantibodies pro- that is the inhibitory of the neurotransmitter in the
duced reflect the autoantigens from the dendritic central nervous system (CNS). Additionally, they
cells, CD4+ and CD8+ T cells, in the islet. also exist in adult human pancreatic islets, more so
These processes result in the insufficient release in β cells than in α cells.98 The GAD67, an isoform
of insulin, eventually causing glucose overproduc- of GAD65 shows almost 76% homology with it.
tion and, thus, the descreased intake of glucose by Moreover, the GAD67 was detected to be expressed
the cells, resulting in hyperglycemia. Additionally, in the epithelial cells of thymic medulla that act as
the loss of insulin also causes the breakdown of fats self-antigens and are capable of escaping from the
and the oxidation of fatty acids, resulting in ketone auto reactive lymphocytes.99 GADA is characterized
overproduction and diabetic ketoacidosis.87–89 as such, it recognizes the epitopes present on the
The pathophysiology results from autoimmune antigen in T1D. GAD65 can be divided into three
destruction and nonautoimmune destruction of the different categories: GADA engaged against NH2
islet cells. As discussed earlier, the genetic com- terminal domain epitope, GADA engaged toward
ponents that play a crucial role are HLADR3 and the middle PLP binding domain site, and GADA to
HLADR4. Moreover, the parents with heterozygos- the COOH terminal domain epitope.100,101 The fre-
ity of these genes are at advanced risk for the oc- quency of positivity of the autoantibodies during the
currence of the disease. Additionally, PTPNN2, the onset of disease is > 80%.102

Volume 39, Issue 4, 2019


246 Dariya et al.

2. Insulin IA-2 -IA-2A for the ZnT8A are present at the NH2 and COOH ter-
minal domains90; moreover, ZnT8-positive patients
The insulinoma associated antigen is like an intrinsic are detected with 10% of the NH2 domain epitope
protein present at the membrane of secretory granules and many with the COOH domain epitope during
and is a receptor-like protein tyrosine phosphatase diagnosis. The frequency for the positive existence
(PTP). In the pancreas, it is present in the α, β, and of ZnT8A is > 65%.102
γ cells of the islets.103 Structurally, the ectodomain
promotes the localization into the lumen of secretory
4. Insulin Antibodies/IAA
granules, the cytoplasmic single juxta-transmem-
brane (JM), and a PTP-like cytoplasmic domain. The association between the autoreactive lympho-
During the mechanism of Ca++ promoting insulin se- cytes and the polymorphic variants of the insulin
cretion in the pancreas islets, the cytoplasmic domain gene are at the higher genetic risk for T1D. IAA
of IA-2 is cleaved to promote the secretory granule binds with the conformational epitopes; however,
mobilization toward the plasma membrane.104 Addi- they do not interact with the reduced antigen. More-
tionally, the cleaved IA-2 further migrates toward the over, the binding of IAA occurs with high affinity
nucleus, which promotes the expression of genes re- with the A chain of insulin at the 8–13 amino acid
sponsible for the secretory granules functions, includ- residues and with a low affinity with the B chain
ing insulin secretion and expansion of β cells.105 Thus, at the 28–30 amino acid residues. The frequency
IA-2 and heterodimers with IA-2β interact with the of the positive occurrence of this autoantibody
other diverse proteins for functioning of the β-cells at the onset of T1D is > 50% compared to other
in the islets of the pancreas for insulin secretion. The autoantibodies.102
expression of IA-2 at the mRNA and protein levels
has been detected in the splenocytes and thymus
5. HLA
and it escapes the negative selection in the thymus
of self-reactive immune cells.106 The IA-2A/antibody The human leukocyte antigen, HLA, weighs ~ 4,000
acts in two ways during the onset of disease: it is di- kb and is positioned on the 6p21.3 human chromo-
rected toward the epitope of cytoplasmic JM domain some. It functions via immune response to the envi-
or it is directed toward the epitope in the PTP-like ronmental factors in various autoimmune diseases.113
domain.107,108 The frequency of the presence of IA-2A HLA antigens comprise 2 classes: HLA class I that
at the onset of T1D is > 70%.102 encodes the genes (A, B, C) and HLA class II mole-
cules that encode for the genes (DP, DQ, DR). Most
3. ZnT8-ZnT8A are associated with autoantibodies. However, inter-
action with autoantibodies is strong only with the
The zinc transporter ZnT8 protein is a transmem- HLA class II loci in T1D, which contributes a risk of
brane multi mass protein that has 6 transmembrane ~ 40%–50%.114,115 GADA has been detected to bind
domains and a loop rich in histidine inserted between with more affinity with the HLA DR3-DQ2 haplo-
the IV and V domains.109 It forms a homodimer, in- type, whereas IA-2A and IAA are more strongly cor-
serted into the secretory granule membrane, where related with HLA DR3-DQ8.116 HLA class I and class
its histidine rich loop inserts towards the lumen and II antigens present on cells that are recognized by
the NH2/COOH terminal domains into the cyto- the CD8+ and CD4+ T lymphocytic cells, respectively.
plasm. The expression of ZnT8 at its mRNA level However, a stronger risk is detected with the HLA
is confined only for the islets of the pancreas110 with class II for the onset of T1D.117 The non-HLA–as-
its increased expression detected in β-cells than in sociated antigens that include susceptible genes for
α-cells.111,112 Functionally, it maintains the Zn con- T1D are INS, PTPN2, CTLA4, PTPN22, IFIHI, and
tent in the cells and is critical to get collected into the CLEC161. Approximately 50 non-HLA gene loci
secretary granules. Additionally, it also maintains have been determined as genetic risks for the onset of
the insulin tightly packed as hexamers. The epitopes T1D according to a genome-wide association study.

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy247

The allele scores and nucleotide polymorphisms of show increased glucose intolerance due to β-cell
non-HLA indicate the stratified risks for the develop- dysfunction and loss. The 5-year risk for this symp-
ment of islet autoantibodies that form the basis of the tomatic stage is ~ 75% and 100% for lifetime risk.124
progression to symptomatic T1D.118 Thus, both HLA The rate of progression in this stage is higher due
and non-HLA indicate a genetic risk ~ 10- to 100-fold to the influence of T1DM susceptible genes, but the
higher in individuals who have relatives with T1D. biomarkers for diagnosis at this stage are still lack-
Additionally, monozygotic twins have also been re- ing. The non-HLAs are extensively encouraged as
ported to have a 65%–70% higher risk of developing biomarkers for detection during stage 1; however,
T1D at an early age.10 Moreover, siblings that share the gene for non-HLAs that contribute to pathogen-
the HLA haplotypes DR3-DQ2/DR4-DQ8 are at 12- esis in the stages 2 and 3 have yet to be investigated.
fold higher risk for developing asymptomatic disease Additionally, the ZNT8 autoantibody variants in-
within 15 years if one sibling was already diagnosed clude tryptophan, glutamine, and arginine at the 325
with disease before 10 years of age.119 amino acid position.32 Moreover, stage 2 dysglyce-
mia exists for more than a year before the onset of
VII. STAGES OF T1D symptoms in stage 3 T1D.

The rate of progression starting from the stage of on- C. Stage 3


set of β-cell autoimmunity until the onset of glucose
intolerance and then to the symptomatic T1D lasts At this stage, the signs of diabetes and clinical symp-
from few months to decades in children or adults. It toms appear, including weight loss, fatigue, poly-
can be differentiated into stages basing on the clin- uria, ketoacidosis, and polydipsia. With the onset of
ical symptoms including the metabolic imbalance stage 3, the autoimmunity targeted against β cells
and hyperglycemia. Currently, T1D is classified ac- is increased and prolonged. The occurrence of the
cording to its prognostic significance. IA-2 autoantibody increases the risk for reaching the
stage 3 from stage 2.31 The data from the fine-needle
A. Stage 1 biopsy shows the occurrence of CD4+, CD8+, den-
dritic cells, macrophages, T cells, and B cells in the
The individuals in stage 1 are screened with at pancreatic islets of Langerhans.125,126 According to
least 2 islet autoantibodies including IA-2, ZnT8, estimates from the DAISY, BABYDIAD, DIPP, and
GAD65 and insulin; however, they have normogly- BABYDIET, children showed higher risks of 44%,
cemia. The children, if screened for genetic risk at 70%, and 84% at 5, 10, and 15 years, respectively.26
birth and if they reach stage 1, are at risk for disease In addition, the TEDDY studies determined that the
at age 5 or 10 years, with a frequency of 44% and seroconversions at 5 years were 11%, 36%, and 47%
70%, respectively, and they have a 100% lifetime for 1, 2, and 3 autoantibodies, respectively.127
risk.26 PTPN22 and INS expressions can be detected
in the development of stage 1 T1DM.120 Moreover, VIII. CLINICAL SYMPTOMS OF T1D
the immune stimuli encountered in the childhood
influence the development of stage 1 T1D. The de- The primary clinical symptoms of T1D include
velopment from stage 1 to stage 2 is detected via polydipsia, polyuria, and weight loss, and ~ 90%
the oral glucose tolerance test (OGTT)121,122 for the of the T1D patients present with these common
presence of dysglycemia as well as an increase in symptoms. Polyuria, weight loss, excess thirst, and
the levels of HbA1c.123 hunger are hyperglycemia-associated symptoms.
When the blood glucose level is > 180, it surpasses
B. Stage 2 the renal threshold, leading to osmotic diuresis and
polyuria. Polyuria is excess urination due to the high
Individuals with stage 2 are screened for more than levels of sugar present in the blood. The urine in the
two autoantibodies, like stage 1; however, they kidneys is reabsorbed due to the glucose present and

Volume 39, Issue 4, 2019


248 Dariya et al.

is further directed into the bloodstream. However, in Neuropathy is a neuropathic abnormality that
T1D, the kidneys lose the function of reabsorbing includes diverse clinical complications and a het-
glucose and it ends up in the urine. Similarly, poly- erogeneous group of disorders like diabetic periph-
dipsia is excessive thirst accompanied by prolonged eral neuropathy (DPN) and autonomic neuropathy
and temporary dryness of the mouth and prolonged (DAN).130 The risk factors for DPN include hyper-
dehydration causing headache, nausea, dizziness, glycemia, duration of diabetes, lipid levels, and
and fainting. If not diagnosed, this pathology further blood pressure; for DAN, the duration of diabetes,
leads to ketoacidosis, which further causes organ age, and uncontrolled glycemic levels are critical
failure, coma, and death. Ketoacidosis is a result of factors.134 DPN is asymptomatic but contributes to a
lipid breakdown to ketone byproducts to substitute higher risk of foot infection and amputation.134
for glucose88; it also causes fruity breath. The other
symptoms include hyperphagia, visual disturbances B. Macrovascular Disease
due to retinal defect, perianal candidiasis, and yeast
infection. Macrovascular complications are less well studied
than the microvascular. However, coronary artery
IX. COMPLICATIONS OF T1D disease is the most common risk factor that varies by
sex.135 Additionally, severity in retinopathy, higher
The complications of T1D are differentiated as body mass index, and blood pressure, proteinuria,
microvascular and macrovascular. Microvascular and depression are risk factors for cardiovascular
complications include neuropathy, nephropathy, and disease.136 Atherosclerosis is abundantly caused by
retinopathy, whereas macrovascular complications cardiovascular disease due to plaque development
includes peripheral vascular, cardiovascular, and in the arterial walls that impairs blood flow. Further-
cerebrovascular diseases. more, a deeper understanding of the mechanism in-
volved that potentiates the clinical interventions for
A. Microvascular Disease the prevention of diabetic complications is essential.

The serious disabling complication for T1D is reti- X. SIGNALING PATHWAYS INVOLVED IN PRE
nopathy, which affects adults aged 20–74 years.128 It AND POST T1D
develops in 4 stages: Microaneurysms develop due
to balloon-like swelling in minute blood vessels of The pathogenesis in T1D is generally related to the
the retina, causing blockage of the blood vessels and aberrant behavior of various signaling pathways in-
depriving them from the supply of blood. To com- cluding the insulin, AMPK, and PPAR pathways.
pensate the blood vessels, the retina starts develop- The signaling pathways play crucial roles in novel
ing new blood vessels, but this can lead to blindness drug targeting and therapies for diabetes.
because the retina has thin fragile walls and blood
may leak.128,129 In addition, cataracts and glaucoma A. Insulin Signaling Pathway
are also frequently diagnosed in diabetic patients.128
Nephropathy is another complication of T1D As discussed, earlier insulin maintains glucose ho-
that develops secondary to metabolic abnormali- meostasis and is facilitated by the binding of insulin
ties.130,131 According to clinical analysis, it is diag- to its receptors called insulin receptors. The receptor
nosed with increased albumin secretion in the urine is a tetrameric glycoprotein with 2 extracellular α
(30–299 mg/24 h). Thus, the renal end-stage results and 2 intracellular β subunits.137 The insulin binding
with the progression from microalbuminuria to to the α subunit leads to autophosphorylation and
macroalbuminuria. Additionally, it is also associated activation of the β subunit, further phosphorylating
with increased risk for cardiovascular disease and various intracellular substrates including IRS1-4
death. Thus, this may be a useful marker for cardio- (insulin receptor substrate) and Shc (Src 2 homol-
vascular risk.132,133 ogy domain containing). Further, IRS activates

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy249

PI3K serving as a docking site for the subunit p85 of cells in rodents.79 The expression of various inflam-
PI3-K. The activated PI3K further phosphorylates matory mediators including cytokines targets a com-
PIP2 to PIP3. Also, the PTEN pathway regulates the mon cascade of apoptosis, later loss of β-cell mass,
function of IRS and PIP3 by inhibiting its expression and finally diabetes.141,142 Thus, apoptosis of the β
whenever required. The increased concentration of cells is activated by various phosphorylation cas-
PIP3 recruits PDK (phosphoinositide-dependent cades, activated extracellular signals, and irregular
protein kinase), protein kinase C, and Akt (protein expression of anti- and proapoptotic genes and intra-
kinase B) toward the plasma membrane. The acti- cellular ATP levels.79 An extensive microarray analy-
vated PDK phosphorylates Akt at threonine 308. sis of the expression of genes during the exposure of
Because Akt requires serine for phosphorylation, β-cells to cytokines IFN-γ and IL-1β revealed ~ 700
the mTORC2 complex phosphorylates Akt at ser- genes that are downregulated or upregulated in their
ine 473. Thus, the activated Akt promotes glycogen, expressions.143–146 For instance, IL-1β in mice or in
protein, and lipid syntheses.138,139 Moreover, the ves- human islet cells was upregulated, whereas NF-κB
icles in the cells embedded with GLUT4 are trans- expression was downregulated. Exposure of β cells
located to the plasma membrane; they play a crucial to IL-1β led to loss of insulin secretion as well as a
role in glucose uptake. Therefore, the AS160 protein decline in the migration and docking of insulin gran-
activated by Akt translocates GLUT4 to embed it ules to the membrane of β-cells. Moreover, exposure
into the plasma membrane. Akt also follows another for longer periods to IFN-γ, IL-1β, and TNF, results
cascade in the activation of GSK3β that promotes in β-cell apoptosis.79 In general, the cytokines act by
glycogen synthesis and axon growth. Alternatively, promoting stress-response genes that either protect
the autophosphorylated β subunit also phosphory- or damage the survival of β cells. The transcription
lates Shc, which attracts Grb-2 and recruits the GTP factor NF-кB is activated by IL-1β and is inhibited
exchange factors SOS and GTP protein Ras. The by the super-repressor IкB, which protects β cells
activated Ras further follows a series of steps for against cytokine-mediated apoptosis. Additionally,
MAPK activation, thus promoting the expression of NF-кB regulates the expression of iNOS and other
genes, mitogenesis, and cell progression.138,140 How- transcription factors, including PDX-1 and Isl-1,
ever, in T1D patients, the lack of insulin disturbs its that are involved in the progression of β cells via
downstream signaling via dysregulation of the PI3K NO production. Thus, the endoplasmic reticulum
pathway. The IRS protein plays a crucial role in the Ca2+ homeostasis, progression, and apoptosis of β
insulin-signaling pathway. Although it is normally cells are directly or indirectly regulated by IL-1β–
phosphorylated at the serine to inhibit the insulin dependent NF-кB activation. Moreover, IFN-γ in
signaling pathway, in T1D it acts abnormally and support of IL-1β was determined to be involved in
remains overphosphorylated at the serine residue to the apoptosis of β-cells, wherein IFN-γ activates the
promote degradation via the ubiquitin-proteasome– JAK tyrosine kinase that further phosphorylates the
mediated pathway. This process results in the im- STAT-1 transcription factor, which translocates into
paired signaling cascade of insulin. the nucleus and binds at diverse γ-active genes.79
Thus, STAT potentiates IFN-γ on iNOS-mediated
B. Mechanism of β-Cell Apoptosis IL-1β expression.147 In support of this finding,
Cnopet al.148 also determined that mice deficient in
Autoimmune attacks result in an inflammatory reac- STAT-1 showed no damage in β cells because they
tion called insulitis in T1D. In this process pancreatic are protected against IL-1β and IFN-γ. Thus, it is
β cells target various immune cells including T cells evident that IL-1β, along other inflammatory modu-
and macrophages, causing invasion of mononuclear lators like IFN-γ or TNF-α, showed increased JNK
cells in the islets of the pancreas. The attack results activity and aberrant STAT-1 expression. In addi-
in the loss of β-cell mass in the advanced stages of tion, p38 MAPK targets ER homeostasis, and death
T1D. β-cell apoptosis is the most crucial part of the signals from the mitochondria also induced β-cell
T1D; it has been observed to cause depletion of β apoptosis.149–151 P53, the tumor suppressor protein

Volume 39, Issue 4, 2019


250 Dariya et al.

that lies downstream to NO-dependent MAPK acti- These AGEs are formed with an oxidation reaction
vation, is also upregulated.152 The dysregulated en- between carbonyl groups of sugar and amine groups
doplasmic reticulum Ca2+ concentration disturbs ER of proteins. The intermediates thereby include non-
homeostasis and promotes ER stress. The prolonged stable Schiff bases (lysine/arginine) and covalently
stress results in the upregulation of ER chaperons bound amadori products (ketoamine). Moreover,
JNK, MAPK and caspase-12, which that further reports indicate that AGE products cause diabetic
promotes apoptosis.153 The mitochondria not only complications.157 In addition to the interaction of
regulate the function and survival of β cells but also intra- and extracellular protein, AGEs also interact
promote apoptosis.154,155 The apoptotic functionality with their receptors, the central cell-surface recep-
of the mitochondria is mediated by the Bcl-2 fam- tors RAGE.158 The binding of the ligand with the re-
ily, which regulates the release of cytochrome c into ceptor activates proinflammatory cytokines, which
the cytosol and subsequently activates caspase 3 and promotes inflammation and oxidative stress. Thus,
caspase 9 to promote cell death.156 Thus, the fate of β under a hyperglycemic condition, RAGE expression
cells depends on the duration and severity of expo- is upregulated due to the increased availability of
sure to cytokines and the network of genes activated AGE products and the ablation of the RAGE gene,
by them, which finally cause β-cell apoptosis and Ager, which reduces insulin resistance. Thus, target-
eventually T1D. ing RAGE could be a therapeutic strategy for T1D
therapy and treatment of related complications. The
C. The Mechanism of Signaling Pathways interaction of AGE and RAGE also induces various
in T1D Associated with Hyperglycemia signaling pathways downstream, including MAPK/
ERK, PKC, JAK/PI3K, and Src. Additionally, this
Untreated or undetected T1D results in higher con- interaction activates NF-кB, which transcribes
centrations of intracellular glucose levels, clinically genes engaged in inflammation, and similarly Egr1,
called as hyperglycemia. This condition is reported which transcribes genes for adhesion and cell mo-
with excessive oxidative stress, production of ROS, tility. Thus, intracellular AGE production aberrantly
and tissue damage. These effects cause breaks in activates various signaling pathways and transcrip-
DNA strands, activating PARP, which results in the tion factors, which that ultimately results in diabetic
release of ADP ribose polymers that alter GAPDH complications. Moreover, as discussed earlier along
activation through polymers docking with them. with the development of insulin resistance, AGEs
This process eventually narrows glycolysis and di- also cause β-cell dysfunction and increased ROS
rects the glycolytic intermediates toward pathogenic production, which impairs mitochondrial function.
signaling pathways including the formation of ad- The T1D complications include kidney hypertrophy
vanced glycation end products (AGEs) in intracel- as well as increase in the glomerular filtration rate
lular, higher flux of polyol pathway, hyperactivation due to the lower secretion of endogenous secretory
of hexoamine pathway and PKC pathway. This pro- RAGE (esRAGE) and secretory RAGE (sRAGE).159
cess causes cellular stress, leading to micro- and Thus, an inverse relationship occurs between the
macrovascular complications. Thus, hyperglycemia concentration of endogenous secretion RAGE and
weakens glycolysis and causes glycolytic intermedi- kidney volume function; increased levels impair the
ates to accumulate, which deactivates the enzymes vascular complications of diabetes.160 Similarly, dia-
prostacyclin and NO synthase. betic retinopathy subsequently causes blindness due
to blood vessel formation on the retina. McVicar et
D. Advanced Glycosylation End Products al.161 determined that diabetic Ager (AGER gene)-
(AGE)-RAGE (Receptor) Pathway null mice showed lower retinal capillary degrada-
tion, adherent leukocytes in the retina, and decreases
A high blood glucose concentration results in the in F4/80+ microglia. Additionally, an increase in
formation of irreversible nonenzymatic glycation glyoxalase-1, which regulates the activity of meth-
products: advanced glycation end products (AGEs). ylglyoxal, is a precursor for the engagement of

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy251

RAGE and AGE compared with wild-type diabetic the genetic removal of the Ar gene in diabetic mice
mice. In another study, Han et al.162 determined that inhibited PKC and TGF-β1 activities, which play a
brain-derived neutrophic factor (BDNF) plays a major role in accruing extracellular matrix and glo-
crucial role in amending neuroplasticity and that its merulosclerosis. The use of inhibitor for AR also at-
overexpression in the hippocampus impairs hyper- tenuates ERK and Akt activity in renal cells, which
glycemic-induced neuroinflammation by blocking may be a novel strategy for diabetic nephropathy
NF-кB and RAGE pathways. Thus, blocking the ex- prevention.165 Moreover, Marko et al.166 determined
pression of RAGE and availability of AGE products that the early occurrence of cataracts in the pediatric
could reduce diabetes-related complications. patients of T1D in their first stage was due to oxi-
dative stress, the polyol pathway, and the osmotic
E. Polyol Pathway pathways. Recently, Akamine et al.167 described the
complications associated with T1D and T2D on the
The polyol pathway is aldose-reductase (AR) de- basis of circadian properties. In both cases, diabetic
pendent and plays a crucial role in complications neuropathic pain is due to the accumulation of sor-
including oxidative stress, nephropathy, cardio- bital in the peripheral nervous system, which results
myopathy, and platelet hyperaggregation.163 It is a in dysregulation or inhibition of Na+/K+-ATPase
dual-step process that includes the conversion of activity, which results in diabetic neuropathic pain.
hexose sugar (glucose) into polyol, a sugar alco- However, this pain is circadian dependent in T2D
hol. First, glucose is converted into sorbitol in the and not in T1D. Thus, proper knowledge of circa-
presence of the AR enzyme. The rate-limiting en- dian properties of the symptoms and their under-
zyme and NADPH cofactor are further oxidized into lying mechanisms will support the development of
fructose in the presence of sorbitol dehydrogenase more effective therapies.
(SDH) with NAD+ as a cofactor. The fructose is then
converted into advanced glycosylation end products F. Hexosamine Pathway
(AGE) that include fructose-3-phosphate and 3-de-
oxyglucosone.163 In the general process, glucose Excess intracellular glucose (~ 2%–5%) is also
metabolization is carried out via the hexokinase pushed into another pathogenic pathway called the
pathway; however, due to the hyperglycemic condi- hexosamine pathway, which ultimately contributes
tion, glucose levels saturate the hexokinase pathway to diabetes-related macrovascular disease. The path-
and become metabolized through the polyol path- way includes fructose-6-phosphate as a substrate
way. Thus, 30% of glucose is metabolized through obtained from glycolysis and glutamine, which are
this pathway and only 3% of aldohexose is altered further converted into glucosamine-6-phosphate
into sorbital under normoglycemic conditions. Ad- and glutamate, respectively, in the presence of glu-
ditionally, the polyol pathway also induces oxida- tamine: fructose-6-phosphate amidotransferase
tive stress by decreasing the availability of NADPH (GFAT) enzyme. Glucosamine-6-phosphate is then
for glutathione reductase to maintain a glutathione metabolized into uridine diphosphate N-acetylglu-
pool because glutathione is used vigorously by AR cosamine (UDP-GlcNAc).168,169 UDP-GlcNAc acts
as a cofactor. Moreover, SDH activity also pro- as a substrate for the O-GlcNAc transferase in gly-
motes the production of superoxide anion because cosylation, and it is involved in the addition of a sin-
it increases the concentration of NADH, a substrate gle sugar to proteins, which causes posttranslational
for NADH-dependent oxidase and AGE produc- protein modifications at serine or threonine residues.
tion, which subsequently enhances ROS production. This phosphorylation of the protein regulates the
Thus, the development of the oxidation stress via function of various enzymes, transcription factors,
the polyol pathway is the entire source for glucose and proteins that alter various cellular processes.
flux, which causes various complications including The hexosamine signaling pathway enhances O-Glc-
neuronal degeneration, retinopathy, and neovascu- NAcylation, which stimulates the expression of var-
larisation. Furthermore, Liu et al.164 determined that ious transcription factors including transforming

Volume 39, Issue 4, 2019


252 Dariya et al.

growth factor (TGF-β1) and plasminogen activator which further contributes to oxidative stress and
inhibitor (PAI-1). This process causes pathogenic lowers NO bioavailability.168,178 Furthermore, hy-
tissue damage in diabetic patients.170 Additionally, it perglycemic conditions also induce reproductive
also targets VEGF-A in retinal cells, which causes dysfunctions in males with T1D; the involved meta-
vascular lesions in diabetic retinopathy.171 However, bolic pathways are the PKC, polyol, and AGE recep-
the inhibition of GFAT downregulates the expres- tor pathways. Activated PKC and polyol pathways
sion of TGF-β1172 and PAI-1169 and increases O-Glc- have been detected within both in the testis and the
NAcylation, which causes diabetic complications.173 epididymis. Moreover, AGE activates CDC42 and
Thus, this process contributes to pathophysiological Akt1 to cause dysfunction in the reproductive sys-
abnormalities in T1D, and an in-depth exploration tem.179 All of these discoveries provide opportuni-
of these mechanisms may yield novel therapeutic ties for the development of therapies for diabetic
strategies for the therapy of T1D. complications.

G. Protein Kinase C (PKC) Pathway XI. UNDERSTANDING THE BIOLOGY AND


INTERACTIONS OF PROTEINS IN T1D
Intercellular hyperglycemia results from the activa- THROUGH BIOINFORMATICS
tion of the PKC pathway, and it is associated with
diabetic complications. PKC belongs to the phos- Bioinformatics is a field of interdisciplinary research
pholipid-dependent serine/threonine kinase family, of computer sciences and information technology
which plays a crucial role in cell progression, sur- applied together in the fields of research and med-
vival, glucose, and lipid metabolism. PKC has three icine. It deals with biological databases that play a
isoform groups: α, βI/II, and γ. The hyperglycemic central role in understanding the biological phenom-
condition enhances the expression of diacylglycerol ena and the interactions among various macromole-
(DAG), which activates the PKC pathway in the cules. Additionally, bioinformatics analyses unravel
heart (PKC-βII), kidneys, and glomeruli (PKC-βI), the genetic basis of various metabolic diseases, like
leading to various vascular complications. DAG is diabetes mellitus, to target after the completion of
the cofactor obtained from the products of glycol- the human genome project. Proteomics techniques
ysis, including glyceraldehyde-3-phosphate and di- are now being widely explored for a clearer under-
hydroxy-acetone phosphate. In addition, activated standing of T1D progression. Biomarkers are being
PKC under a hyperglycemic condition induces di- characterized for therapeutic purposes because they
abetic pathogenesis, contributing to the progression are specific, sensitive, and readily available in small
of extracellular matrix, angiogenesis, and alteration samples. Proteomic researchers are now focusing
in vascular permeability.174 Among the isoforms, on mapping the proteomes associated with the pan-
PKC-β is the major cause of pathogenesis in dia- creas and β cells,180 including the identification of a
betes, and it also promotes ECM (type IV collagen whole protein or the peptide obtained after protein
and fibronectin) proliferation and the expression of digestion. This process is initiated by determining
TGF-β protein.175 However, ECM accretion and the the mass to charge ratios and then searching for the
expression of TGDF-β have been reported to be de- protein in database. Ionization is also performed
layed when exposed to PKC inhibitor. Furthermore, prior to the mass analysis using electrospray ion-
NADPH oxidation and impaired ROS production ization (ESI) and matrix-associated laser desorp-
are absent in PKC-β null mice; thus, PKC-β also tion ionization (MALDI). These techniques use a
induces oxidative stress through the oxidation of time of flight (TOF) instrument. Merchant et al.181
NADPH.176 Similarly, PKC-γ, along with p38α and performed LC-MALDI-TOF, a tandem mass spec-
MAPK, promote Src homology-2, which causes trometry shotgun proteomics analysis, to determine
apoptosis independent of NF-кB activation and de- high-molecular-weight kininogens. Isolated samples
phosphorylating PDGFR β. In addition, PKC also from the Joslin study of the natural history of micro-
induces endothelial cell permeability in diabetes,177 albuminuria (MA) in the plasma peptidome of T1D

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy253

patients showed matching cystatin C eGFR and MA and t-tests to generate data and compare them be-
with patients with early progressive renal function tween patients with T1D and autoantibody-positive
decline, and with subsequent follow-up for 8–12 individuals. Then, univariate statistical analysis is
years. They reported three fragments of high-molec- performed between these populations. Transcript
ular-weight kininogen and a peptide des-Arg9-BK mapping is another technique that includes the use
(1-8) that promotes the expression of Erk1/2 phos- of an algorithm to determine the Hamilton path to
phorylation. Thus, this fragment was identified as a connect genes with the proteins they encode.191 A
protein correlated with early progressive renal func- 6-Mb map of chromosome 20, which is linked with
tion decline in T1D patients with MA. Although MA diabetes, has been designed for use in developing
is recorded as a diagnostic tool for the detection of novel targeted therapies.192
T1D,182 the association of MA with the future risk The most important database available for T1D
of renal function is always a question because most is the TIDbase (http://T1DBase.org).193 It comprises
MA patients reverted back to urine free of albu- data from various resources, such as BLAT and Beta
min,183 and only 20% of the patients showed protein- Cell Gene Bank, and includes various software tools
uria,184 and TID patients showed early progressive such as T1Dmart, Gbrowse, and Cytoscape. The
renal dysfunction.185 Thus, this technique has po- Jackson Laboratory Type 1 Diabetes Mouse Re-
tential to determine the protein for the cause of dis- pository (http://www.jax.org/resources/index.html)
ease. Crevecoeur et al.186 combined microarray with is another important resource. This nonprofit orga-
2D-DIGE for identifying the islet in 3-week-old di- nization contributes to medical research and their
abetic NOD mice. They suggested that the islet pro- database includes data about genetics, biology, and
tein showed posttranslational modifications (PTMs) genomics.
in the presence of peptidyl arginine deiminase 2. The major techniques used to extract the pro-
Moreover, the 2D-DIGE proteome also determined teomic data include microarray and mass spectrome-
that NIT-1 β cells are much prone to ER and ox- try; however, because these techniques are manually
idation stress than αTC-1 cells.187 Later, Zhang et performed, they are time-consuming. Thus, re-
al.188 compared the proteomic profile of the acinar searchers have collaborated with mathematics and
tissues with that of human islets using nano LC-MS. computer sciences to improve the analysis of protein
They profiled 706 and 1,104 proteins, respectively. data. Molecular docking is the bioinformatics tool
Later, they compared LCM islets and islets isolated used to predict the interaction between a drug and a
in the presence of enzymes and showed overlapping ligand before in vitro and in vivo studies. Thus, this
proteomic profiles. A detailed study of T1D-associ- field plays a major role in drug design. Presently, a
ated biomarkers is a significant tool for the clinical large amount of research on ligand–protein docking
management of T1D. As discussed earlier, the de- is being conducted to design drugs to treat diabe-
tection of islet antigen cells, including GAD, IA-2 tes mellitus. For instance, foot ulcers are a major
and S1c30A8 (Zn protein), could be a biomarker for complication for both T1D and T2D. As an initial
the initiation of autoimmunity in β cells.90,189 The en- step, a clinico-bioinformatics analysis is performed
vironmental determinants of diabetes in young con- for both male and female diabetic patients with foot
sortium and DASP remain undefined because not all ulcers.194 Male diabetic patients are highly prone to
of the antibodies involved in T1D progression and morality because a foot infected with MDR-GNB
pathogenesis have been determined.190 shows a more irregular glycemic secretion rate in
In another way, the prototyping of diseases like men than in women. Additionally, the interaction
diabetes is being used to validate bioinformatics. The sites for the ligand and protein are Asn132, Pro167,
researchers have used the discovery of biomarkers Lys234, Glu166, Val172, and Thr235 of enzyme
as the basis to detect disease. Initially, they found CTX-M-15 and the drug cefotaxime, which has an
genes that are highly associated with the occurrence enzyme drug complex.195 Homology modeling is the
of diabetes. Later, they identified other factors re- prediction of the structure of any protein due to its
lated to the causes of the disease using microarrays similarity with more than one protein sequence of

Volume 39, Issue 4, 2019


254 Dariya et al.

known or unknown structures.196,197 The commonly technologies to detect novel biomarkers involved
used homology modeling techniques are the SWISS- in the pathogenesis. They used global liquid chro-
MODEL (http://swissmodel.expasy.org/) and MOD- matography-mass spectrometry-based proteomics
ELLER (http://salilab.org/modeller/).198,199 Wiltgen analysis for almost 25 serum proteins. In addition
et al.200 performed homology modeling to predict the to the insulin autoantibodies, they identified cer-
structure of 65-kD GAD65. Initially they performed tain peptide markers from peptide assays. These
BLAST to compare the queried sequence of GAD65 peptide markers are independent of autoantibody
via pairwise comparison, resulting in a list of align- markers. Their results were negative for expression
ments showing varied homologous sequences. The but positive for the peptide NIQSLEVIGK, which
best alignment with the lowest expectation value matches the diabetic group. Moreover, they have
(E-value) is selected. Simultaneously, the crystal taken 2 polypeptides from the platelet-based protein
structure of DOPA decarboxylase of pig kidney with precursor (PPBP), which is upregulated in T1D pa-
and without inhibitor carbidopa is downloaded. The tients. These peptides include EESLDSDLYAELR
active site of GAD 65 is determined via sequence and NIQSLEVIGK of PPBP. These researchers
alignment. Moreover, a clear picture of the antigenic further performed sequence alignment with these
determinants is obtained through the visualization of peptides and proteins CTAPIII, β-TG, NAP-2, TC-
the epitopes on the GAD65 molecular surface. Thus, 1, and TC-2. They identified a similarity between
this process could facilitate our understanding of the NIQSLEVIGK and all proteins, although EESLDS-
immune response that is disrupted by the passive ad- DLYAELR shows similarity only with CTAP III and
ministration of molecules like DOPA. Assmann et TC-2 and β-TG. Meanwhile, TC-1, an antibacterial
al.59 performed a bioinformatics analysis and found protein released at the response of innate immunity
that almost 11 circulating miRNAs are dysregulated due to the active platelet α-granules, and NAP-2
in T1D patients: miR-1275, miR-21-5p, miR-375, are the activators of neutrophils released from the
miR-24-3p, miR-342-3p, miR-100-5p, miR210-5p, proteolytic cleavage of CTAP III and PPBP. Addi-
miR-181a-5p, miR-150-5p, miR-146a-5p, and miR- tionally, they also determined SERPING, a plasma
148a-3p. They regulate immune pathways such as protease C1 inhibitor as a sensitive marker for the
MAPK, Jak-STAT, PI3K-Akt, NF-κB, TNF, TCR, detection of T1D. Thus, they classified the samples
insulin, and TLR signaling pathways. However, fur- obtained from the control group and T1D patients
ther evaluation is necessary to determine the specific with 100% sensitivity and specificity of both the
role of each miRNA in pancreatic islets and β-cell PPBP and C1 inhibitors. In addition, their findings
functioning. suggest that T1D occurrence was also due to the
Sequence alignment is used to identify the simi- dysregulation of the innate immune response.
larities among primary sequences related to function The simulation method is another technique
and structure in DNA, RNA, and proteins. Pairwise used in the field of bioinformatics for tracking the
and multiple-sequence alignment are widely used pathophysiological process involved in various dis-
techniques by the researchers. Rao et al.201 constructed eases. The glucose-insulin simulation study could
a phylogenetic tree to determine the relationship be- be an essential process for diabetes research. Recent
tween diabetic complications of cardiomyopathy and simulation methods used for T1D include the mini-
pathogenically altered calcium homeostasis. Simi- mal model of Bergman under intensive care, which
larly, Rao et al.202 constructed a phylogram tree based is based on intravenous glucose tolerance test.204
on the protein data available from NCBI for deter- This method includes sampling glucose and insulin
mining the genes involved in the evolution of obesi- concentrations in plasma after a glucose intravenous
ty-related diabetes. They used the Clusta1W tool to injection. In addition, a new meal simulation method
perform multiple sequence alignments. is being used to compare the physiological processes
Various new studies are focusing on identify- in healthy individuals after a given meal.205
ing novel biomarkers using sequence alignments. Certain techniques called new-generation se-
For instance, Zhang et al.203 potentiated proteomics quencing (NGS) include sequencing millions of

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy255

DNA fragments either for an entire genome or for like diabetes and cancer. A novel derivative of cur-
a definite site using computational techniques. In cumin C66 was found to decrease hyperglycemia
T1D, the presence of DRB3*02:02 was identified promoted the inflammatory response and apopto-
on the DRB1*03:01 haplotype using NGS tech- sis by inhibiting JNK activity. JNK plays a crucial
nology, which showed a risk for T1D.206 Various role in regulating apoptotic effects of active TNF-α
NGS methods include single-molecule real-time se- cells in cardiac cells. The dysregulation causes dia-
quencer (SMRTTM), VisiGen Biotechnologies, and betes-associated cardiomyopathy. Furthermore, Pan
Nanopore DNA sequencer. However, they require et al.210 identified an interaction of the C66 and JNK
advanced bioinformatics techniques for analysis and proteins validated with SP600125 and using 2 JNK
determining the cause of disease.207 Mathieu et al.208 proteins (JNK1 and JNK2). The C66 was found in
explained ongoing projects in Europe, including the hydrophobic region in the JNK1 receptor bind-
Global Platform for the Prevention of Autoimmune ing pocket; the residues involved are Leu-168, Ile-
Diabetes (GPPAD; www.gppad.org/de) and INNO- 32, Val-40, and Ile-52. With JNK2, the binding
DIA, which have enabled biomarker discoveries occurred with 3 hydrogen bonds with Arg involved
based on patient samples such as urine and stool. in the pocket. Thus, C66 showed higher binding af-
INNODIA is a recent process that aims to facilitate finity with JNK2 (IC50-2.72) than JNK1 (IC50-81.9),
innovative ideologies for understanding and pre- which exhibited increased inhibition and anti-in-
vention. It collects samples from newly diagnosed flammatory effects. This research furthers the basis
T1D patients and first-degree relatives to elucidate for the drug designing and drug discovery related to
the pathogenesis and novel biomarkers. The clini- diabetic therapy.
cal data collected from omics and sequencing are
secured in the INNODIA database, which forms a XII. ADVANCED TECHNOLOGIES FOR THE
platform to support in silico modeling of T1D. MANAGEMENT OF T1D
Bioinformatics is also widely used for drug
designing and drug delivery. Kir6.2 is an integral Several novel technologies are now available for
membrane protein and a subunit of the ATP-sensi- the management of T1D that mainly adopt a stan-
tive K+ channel that allows the K+ flow from outside dard of care as the intensive insulin therapies as per
of the β-cell to the inside controlled by G-proteins the publication of landmark 1993 Diabetes Control
and sulfonylurea receptor (SUR). However, muta- and Complications Trial.211 Glycemic conditions are
tion in both Kir6.2 and SUR causes the permanent properly maintained via advanced glucose monitor-
opening of KATP channel and closing of calcium ing and insulin delivery. The most widely used tech-
channel. Jagadeb et al.209 reported the interaction of niques for the management include blood glucose
various phytochemicals including curcumin, pteros- meters, continuous glucose monitors, insulin pumps,
tilbene, piperine, and genistein with Kir6.2. The and a combination of monitors and pump that are
mutated amino acid residues found include H46Y, additionally automated driven by the algorithms.
Y330S, Q52R, R201H, G53D, L164P, K170T, Insulin pump therapy is administered via injection
R50Q, C166T, and V59M. However, due to the or continuous infusion, whereas intensive insulin
nonavailability of the crystal structure of Kir6.2, therapy is administered via subcutaneous injection
they first designed a secondary structure using an in of long-acting basal insulin given once or twice per
silico approach. All the phytochemicals that exhib- day. Additionally, a fast-acting insulin is given at
ited an inhibitory effect on the expression of Kir6.2 meal time. At present in this electronic world, the
were docked at an ATP-docking site that included advancement for insulin injections is replaced by
residues Ala-178, Ala-300, Phe-18, and Leu-181. the insulin pens and are potentiated because they
Hydrogen bonds formed between Arg-301 and are retrofitted with the smartphone applications that
Phe-183 in the binding pocket of mutated Kir6.2. offer data about a patient’s blood glucose, physical
Curcumin is a phytochemical that plays a crucial activity, and food intake. Blood glucose meters212
role in inhibiting pathogenesis of various diseases are user friendly and can be used at home, where

Volume 39, Issue 4, 2019


256 Dariya et al.

patients can self-manage their insulin regimens. biomarkers, the appropriate prediction of T1D re-
Moreover, the insulin delivery can also be auto- mains challenging. ‘Omics’ technology is now used
mated using an insulin delivery system, artificial for detecting and validating various biomarkers.
pancreas, and closed loop. These techniques have Circulating methylated DNA is a promising facet of
a built-in complete glucose monitor with an insu- the detection of dead β cells to predict T1D progres-
lin pump and algorithm for the delivery of insulin sion. The detection of biomarkers at every stage of
according to past insulin delivery data. Thus, more the disease along with stratified highest risk factors
advanced systems are developed and expected to potentiates the novel therapeutic procedure. Addi-
control both hyper- and hypoglycemic levels.213–216 tionally, the PTMs like C- peptide have attracted
However, exogenous therapy is for shorter periods research to assess the dysregulation of β cells. Ad-
and for recurrent episodes of hypoglycemia. There- vanced proteomic techniques are currently being
fore, replacing the functionality of β cells to create used to identify and validate β-cell biomarkers.
a normal glycemia level endogenously is essential. Circulating exosomes, including GAD65, proinsu-
Transplantation of pancreas from a healthy individ- lin, and IA-2, impair phenotyping and can also be
ual can bring glucose to normal levels. Transplan- used as predictive biomarkers. Thus, classifications
tation resulted in reducing autoimmune responses of biomarkers performed at novel stages provide a
and diabetes-related complications like micro- and superficial taxonomy of T1D and frame the clini-
macroangiopathy, with improvement in the survival cal trial designing. In addition, aberrantly activated
rate of the patient.217 However, these procedures signaling pathways, mitochondrial dysfunction, and
present high risk for surgical complications. Novel impaired metabolism of glucose and cholesterol are
approaches include extrahepatic site of implementa- the primary mechanisms for diabetic complications
tion, immunotolerance, and using substitutes for is- that are under investigation for T1D therapy. There-
lets. However, these procedures still require clinical fore, a profound study on the role of proteins and
testing in advanced phases. their molecular mechanisms would dramatically re-
duce T1D-related complications. The pathogenesis
XIII. CONCLUSIONS of T1D is highly confounded, and developing drugs
do treat this disease is challenging. The molecular
Type 1 diabetes (T1D) is a serious chronic autoim- description of signaling pathways involved in the
mune disease. The T1D phenotype remains a target progression of disease is essential for the develop-
and treatments are remain imperfect. Therefore, ment of personalized drug development and novel
perfect characterization of the etiology of diabetes drug discoveries. Many more advances in the field
to support further therapeutic developments is es- of genetics, epigenetics, and detection of autoanti-
sential. Genetic and autoimmune factors are always gens are needed to drive novel methods of diagnosis
risk predictors, although vitamin D deficiency and and therapy. T1D also enhances the risk for the pro-
drinking cow milk at an early age are still being in- gression of various other autoimmune diseases for
vestigated. Additionally, viral factors may also be patients who have T1D from adolescence. T1D with
causative triggers. Researchers have started work- other autoimmune complications prejudice the glu-
ing on vaccines on the premises that inequity in cose metabolism and hinder insulin therapy. Contin-
the gut microflora may cause certain pathogeneses uous monitoring of individuals with the autoimmune
(like T1D) and, thus, that microbial supplements diseases is essential to preventing damage to other
and probiotics released from the gut microbiota organs. Together, analyzing samples that are inter-
would reduce the risk of T1D to certain extent. linked with the clinical data, developing technolo-
Moreover, the efficient classification of biomark- gies with integrated methods, and pioneering data
ers based on the heterogeneity of the patient pop- analysis would provide an enormous breakthrough
ulation for diagnosing, monitoring, and preventing in the research of T1D. Recent advances in bioin-
disease is essential. Although dynamic genotyping formatics are effectively used for managing various
has been performed for multiple autoantibodies as diseases like cancer and diabetes. The development

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy257

of biological databases could form the foundation effect of intensive treatment of diabetes on the devel-
for drug discovery and bioinformatics. Comput- opment and progression of long-term complications in
insulin-dependent diabetes mellitus. New Engl J Med.
er-aided programs including modeling, homology
1993;329(14):977.
simulations, and docking have been widely used for 12. Aas Z, Babaei E, Feizi MAH, Dehghan G. Anti-prolif-
successful drug discovery and designing antidia- erative and apoptotic effects of dendrosomal farnesif-
betic drugs. Moreover, current research is providing erol C on gastric cancer cells. Asian Pac J Cancer Prev.
novel insights for the prediction and prevention of 2015;16(13):5325–29.
the disease pathogenesis, and clinical trials to cure 13. Thunander M, Petersson C, Jonzon K, Fornander J, Os-
siansson B, Torn C, Edvardsson S, Landin-Olsson M.
T1D are underway. Incidence of type 1 and type 2 diabetes in adults and
children in Kronoberg, Sweden. Diabet Res Clin Pract.
REFERENCES 2008;82(2):247–55.
14. Thomas NJ, Jones SE, Weedon MN, Shields BM, Oram
1. Bach J-F. The effect of infections on susceptibility to RA, Hattersley AT. Frequency and phenotype of type 1
autoimmune and allergic diseases. New Engl J Med. diabetes in the first six decades of life: a cross-sectional,
2002;347(12):911–20. genetically stratified survival analysis from UK Biobank.
2. Durinovic-Bello I, Schlosser M, Riedl M, Maisel N, Lancet Diabet Endocrinol. 2018;6(2):122–29.
Rosinger S, Kalbacher H, Deeg M, Ziegler M, Elliott 15. Baranda FS, Coto E, Déaz JM, Martinez-Naves E,
J, Roep B. Pro-and anti-inflammatory cytokine pro- Marténez VA, López-Larrea C. HLA class II and suscep-
duction by autoimmune T cells against preproinsulin in tibility and resistance to insulin-dependent diabetes mel-
HLA-DRB1* 04, DQ8 type 1 diabetes. Diabetologia. litus in a population from the northwest of Spain. Int J
2004;47(3):439–50. Immunogen. 1994;21(4):219–29.
3. Soltesz G, Patterson C, Dahlquist G, Group ES. World- 16. Delli AJ, Lindblad B, Carlsson A, Forsander G, Ivarsson
wide childhood type 1 diabetes incidence–what can we SA, Ludvigsson J, Marcus C, Lernmark Å, Group BDDS.
learn from epidemiology? Pediatr Diabet. 2007;8:6–14. Type 1 diabetes patients born to immigrants to Sweden
4. Jarosz-Chobot P, Polanska J, Szadkowska A, Kretowski increase their native diabetes risk and differ from Swedish
A, Bandurska-Stankiewicz E, Ciechanowska M, Deja G, patients in HLA types and islet autoantibodies. Pediatr Di-
Mysliwiec M, Peczynska J, Rutkowska J. Rapid increase abet. 2010;11(8):513–20.
in the incidence of type 1 diabetes in Polish children from 17. Turner R, Stratton I, Horton V, Manley S, Zimmet
1989 to 2004, and predictions for 2010 to 2025. Diabeto- P, Mackay IR, Shattock M, Bottazzo GF, Holman R,
logia. 2011;54(3):508–15. Group UPDS. UKPDS 25: autoantibodies to islet-cell
5. Group EAS. Variation and trends in incidence of child- cytoplasm and glutamic acid decarboxylase for predic-
hood diabetes in Europe. Lancet. 2000;355(9207):873–76. tion of insulin requirement in type 2 diabetes. Lancet.
6. Group DP. Incidence and trends of childhood type 1 diabetes 1997;350(9087):1288–93.
worldwide 1990–1999. Diabet Med. 2006;23(8):857–66. 18. Svensson J, Carstensen B, Mortensen HB, Borch-Johnsen
7. Patterson CC, Dahlquist GG, Gyürüs E, Green A, Soltész K, Diabetes DSGoC. Early childhood risk factors associ-
G, Group ES. Incidence trends for childhood type 1 diabe- ated with type 1 diabetes–is gender important? Eur J Epi-
tes in Europe during 1989–2003 and predicted new cases demiol. 2005;20(5):429–34.
2005–20: a multicentre prospective registration study. 19. Patterson C, Gyürüs E, Rosenbauer J, Cinek O, Neu A,
Lancet. 2009;373(9680):2027–33. Schober E, Parslow R, Joner G, Svensson J, Castell C.
8. Green A, Patterson CC, Group ETS. Trends in the inci- Trends in childhood type 1 diabetes incidence in Europe
dence of childhood-onset diabetes in Europe 1989–1998. during 1989–2008: evidence of non-uniformity over time
Diabetologia. 2001;44(3):B3–B8. in rates of increase. Diabetologia. 2012;55(8):2142–47.
9. Rönningen KS, Keiding N, Green A. Correlations between 20. Onkamo P, Väänänen S, Karvonen M, Tuomilehto J.
the incidence of childhood-onset type I diabetes in Europe Worldwide increase in incidence of type I diabetes—the
and HLA genotypes. Diabetologia. 2001;44(3):B51–B59. analysis of the data on published incidence trends. Diabe-
10. Insel RA, Dunne JL, Atkinson MA, Chiang JL, Dabelea tologia. 1999;42(12):1395–403.
D, Gottlieb PA, Greenbaum CJ, Herold KC, Krischer JP, 21. Rewers M, Ludvigsson J. Environmental risk factors for
Lernmark Å. Staging presymptomatic type 1 diabetes: type 1 diabetes. Lancet. 2016;387(10035):2340–48.
a scientific statement of JDRF, the Endocrine Society, 22. Ilonen J, Hammais A, Laine A-P, Lempainen J, Vaarala O,
and the American Diabetes Association. Diabet Care. Veijola R, Simell O, Knip M. Patterns of β-cell autoanti-
2015;38(10):1964–74. body appearance and genetic associations during the first
11. Nathan D, Genuth S, Lachin J, Cleary P, Crofford O, Da- years of life. Diabetes. 2013;62(10):3636–40.
vis M, Rand L, Siebert C, Control D, Group CTR. The 23. Lempainen J, Laine A-P, Hammais A, Toppari J, Simell O,

Volume 39, Issue 4, 2019


258 Dariya et al.

Veijola R, Knip M, Ilonen J. Non-HLA gene effects on the A. HL-A antigens and diabetes mellitus. Lancet. 1974;
disease process of type 1 diabetes: from HLA susceptibil- 304(7885):864–66.
ity to overt disease. J Autoimmun. 2015;61:45–53. 34. Singal D, Blajchman M. Histocompatibility (HL-A) an-
24. Giannopoulou EZ, Winkler C, Chmiel R, Matzke C, tigens, lymphocytotoxic antibodies and tissue antibodies
Scholz M, Beyerlein A, Achenbach P, Bonifacio E, Ziegler in patients with diabetes mellitus. Diabetes. 1973;22(6):
A-G. Islet autoantibody phenotypes and incidence in chil- 429–32.
dren at increased risk for type 1 diabetes. Diabetologia. 35. Cudworth A, Woodrow J. Evidence for HL-A-linked genes
2015;58(10):2317–23. in “juvenile” diabetes mellitus. Br Med J. 1975;3(5976):
25. Krischer JP, Lynch KF, Lernmark Å, Hagopian WA, Rew- 133–35.
ers MJ, She J-X, Toppari J, Ziegler A-G, Akolkar B, Group 36. Thomson G, Valdes A, Noble J, Kockum I, Grote M, Na-
TS. Genetic and environmental interactions modify the jman J, Erlich H, Cucca F, Pugliese A, Steenkiste A. Rela-
risk of diabetes-related autoimmunity by 6 years of age: tive predispositional effects of HLA class II DRB1-DQB1
the TEDDY study. Diabet Care. 2017;40(9):1194–202. haplotypes and genotypes on type 1 diabetes: a meta-anal-
26. Ziegler AG, Rewers M, Simell O, Simell T, Lem- ysis. Tiss Antigens. 2007;70(2):110–27.
painen J, Steck A, Winkler C, Ilonen J, Veijola R, Knip 37. Pociot F, Lernmark Å. Genetic risk factors for type 1 dia-
M. Seroconversion to multiple islet autoantibodies betes. Lancet. 2016;387(10035):2331–39.
and risk of progression to diabetes in children. JAMA. 38. Fradin D, Le Fur S, Mille C, Naoui N, Groves C, Zele-
2013;309(23):2473–79. nika D, McCarthy MI, Lathrop M, Bougneres P. Asso-
27. Gorus F, Vandewalle C, Dorchy H, Van Crombrugge P, ciation of the CpG methylation pattern of the proximal
Schuit F, Pipeleers D. Influence of age on the associa- insulin gene promoter with type 1 diabetes. PLoS One.
tions among insulin autoantibodies, islet cell antibodies, 2012;7(5):e36278.
and HLA DAQ1* 0301-DQB1* 0302 in siblings of pa- 39. Miao F, Chen Z, Zhang L, Liu Z, Wu X, Yuan Y-C, Na-
tients with type 1 (insulin-dependent) diabetes mellitus. tarajan R. Profiles of epigenetic histone post-translational
Belgian diabetes registry. J Clin Endocrinol Metabol. modifications at type 1 diabetes susceptible genes. J Bio
1994;78(5):1172–78. Chem. 2012;287(20):16335–45.
28. Rolandsson O, Hägg E, Hampe C, Sullivan Jr E, Nilsson 40. Hezova R, Slaby O, Faltejskova P, Mikulkova Z, Bu-
M, Jansson G, Hallmans G, Lernmark Å. Glutamate de- resova I, Raja KM, Hodek J, Ovesna J, Michalek J. mi-
carboxylase (GAD65) and tyrosine phosphatase-like pro- croRNA-342, microRNA-191 and microRNA-510 are
tein (IA-2) autoantibodies index in a regional population differentially expressed in T regulatory cells of type 1 dia-
is related to glucose intolerance and body mass index. Di- betic patients. Cell Immunol. 2010;260(2):70–74.
abetologia. 1999;42(5):555–59. 41. Moulder R, Bhosale SD, Erkkilä T, Laajala E, Salmi J,
29. Rewers M, Norris JM, Eisenbarth GS, Erlich HA, Beaty Nguyen EV, Kallionpää H, Mykkänen J, Vähä-Mäkilä M,
B, Klingensmith G, Hoffman M, Yu L, Bugawan TL, Blair Hyöty H. Serum proteomes distinguish children develop-
A. Beta-cell autoantibodies in infants and toddlers with- ing type 1 diabetes in a cohort with HLA-conferred sus-
out IDDM relatives: diabetes autoimmunity study in the ceptibility. Diabetes. 2015;64(6):2265–78.
young (DAISY). J Autoimmun. 1996;9(3):405–10. 42. Hanifi-Moghaddam P, Schloot NC, Kappler S, Seiβler
30. Ziegler A-G, Hummel M, Schenker M, Bonifacio E. Au- J, Kolb H. An association of autoantibody status and
toantibody appearance and risk for development of child- serum cytokine levels in type 1 diabetes. Diabetes.
hood diabetes in offspring of parents with type 1 diabetes: 2003;52(5):1137–42.
the 2-year analysis of the German BABYDIAB Study. 43. Hanifi-Moghaddam P, Kappler S, Seissler J, Müller-
Diabetes. 1999;48(3):460–68. Scholze S, Martin S, Roep B, Strassburger K, Kolb H,
31. Savola K, Bonifacio E, Sabbah E, Kulmala P, Vähäsalo Schloot N. Altered chemokine levels in individuals at
P, Karjalainen J, Tuomilehto-Wolf E, Meriläinen J, Åker- risk of type 1 diabetes mellitus. Diabet Med. 2006;23(2):
blom H, Knip M. IA-2 antibodies–a sensitive marker of 156–63.
IDDM with clinical onset in childhood and adolescence. 44. Zak K, Popova V, Mel’nichenko S, Tron’ko E,
Diabetologia. 1998;41(4):424–29. Man’kovskiĭ B. The level of circulating cytokines and
32. Lampasona V, Petrone A, Tiberti C, Capizzi M, Spoletini chemokines in the preclinical and early clinical stages of
M, Di Pietro S, Songini M, Bonicchio S, Giorgino F, Bon- type IA diabetes mellitus development. Terapevticheskii
ifacio E. Zinc transporter 8 antibodies complement GAD Arkhiv. 2010;82(10):10–15.
and IA-2 antibodies in the identification and characteriza- 45. Purohit S, Sharma A, Hopkins D, Steed L, Bode B, Ander-
tion of adult-onset autoimmune diabetes: Non Insulin Re- son SW, Reed JC, Steed RD, Yang T, She J-X. Large-scale
quiring Autoimmune Diabetes (NIRAD) 4. Diabet Care. discovery and validation studies demonstrate significant
2010;33(1):104–8. reductions in circulating levels of IL8, IL-1Ra, MCP-1,
33. Nerup J, Platz P, Andersen OO, Christy M, Lyngsoe J, and MIP-1β in patients with type 1 diabetes. J Clin Endo-
Poulsen J, Ryder L, Thomsen M, Nielsen LS, Svejgaard crinol Metabol. 2015;100(9):E1179–E87.

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy259

46. Schölin A, Törn C, Nyström L, Berne C, Arnqvist H, Crispim D. MicroRNA expression profiles and type 1 dia-
Blohme G, Bolinder J, Eriksson JW, Kockum I, Landin-Ols- betes mellitus: systematic review and bioinformatic anal-
son M. Normal weight promotes remission and low num- ysis. Endocr Connect. 2017;6(8):773–90.
ber of islet antibodies prolong the duration of remission in 60. Seyhan AA, Lopez YON, Xie H, Yi F, Mathews C, Pas-
type 1 diabetes. Diabet Med. 2004;21(5):447–55. arica M, Pratley RE. Pancreas-enriched miRNAs are al-
47. Leighton E, Sainsbury CA, Jones GC. A practical re- tered in the circulation of subjects with diabetes: a pilot
view of C-peptide testing in diabetes. Diabet Ther. cross-sectional study. Sci Rep. 2016;6:31479.
2017;8(3):475–87. 61. Nielsen LB, Wang C, Sørensen K, Bang-Berthelsen CH,
48. Bonifacio E. Predicting type 1 diabetes using biomarkers. Hansen L, Andersen M-LM, Hougaard P, Juul A, Zhang
Diabet Care. 2015;38(6):989–96. C-Y, Pociot F. Circulating levels of microRNA from chil-
49. Ilonen J, Lempainen J, Hammais A, Laine AP, Härkönen dren with newly diagnosed type 1 diabetes and healthy
T, Toppari J, Veijola R, Knip M, Register FPD. Primary is- controls: evidence that miR-25 associates to residual be-
let autoantibody at initial seroconversion and autoantibod- ta-cell function and glycaemic control during disease pro-
ies at diagnosis of type 1 diabetes as markers of disease gression. Exper Diabet Res. 2012;2012.
heterogeneity. Pediatr Diabet. 2018;19(2):284–92. 62. Erener S, Mojibian M, Fox JK, Denroche HC, Kieffer TJ.
50. Endesfelder D, Hagen M, Winkler C, Haupt F, Zillmer S, Circulating miR-375 as a biomarker of β-cell death and
Knopff A, Bonifacio E, Ziegler A-G, Zu Castell W, Achen- diabetes in mice. Endocrinology. 2013;154(2):603–8.
bach P. A novel approach for the analysis of longitudinal 63. Osipova J, Fischer D-C, Dangwal S, Volkmann I, Wid-
profiles reveals delayed progression to type 1 diabetes in a era C, Schwarz K, Lorenzen JM, Schreiver C, Jacoby U,
subgroup of multiple-islet-autoantibody-positive children. Heimhalt M. Diabetes-associated microRNAs in pediatric
Diabetologia. 2016;59(10):2172–80. patients with type 1 diabetes mellitus: a cross-sectional
51. Vehik K, Lynch KF, Schatz DA, Akolkar B, Hagopian W, cohort study. J Clin Endocrinol Metabol. 2014;99(9):
Rewers M, She J-X, Simell O, Toppari J, Ziegler A-G. Re- E1661–E1665.
version of β-cell autoimmunity changes risk of type 1 dia- 64. Nabih ES, Andrawes NG. The association between cir-
betes: TEDDY study. Diabet Care. 2016;39(9):1535–42. culating levels of miRNA-181a and pancreatic beta cells
52. Calderon B, Sacks DB. Islet autoantibodies and type 1 di- dysfunction via SMAD7 in type 1 diabetic children and
abetes: does the evidence support screening? Clin Chem. adolescents. J Clin Lab Analy. 2016;30(5):727–31.
2014;60(3):438–40. 65. Sebastiani G, Grieco FA, Spagnuolo I, Galleri L, Cataldo
53. Törn C, Hadley D, Lee H-S, Hagopian W, Lernmark Å, D, Dotta F. Increased expression of microRNA miR-326
Simell O, Rewers M, Ziegler A, Schatz D, Akolkar B. in type 1 diabetic patients with ongoing islet autoimmu-
Role of type 1 diabetes–associated SNPs on risk of au- nity. Diabet/Metabol Res Rev. 2011;27(8):862–66.
toantibody positivity in the TEDDY study. Diabetes. 66. Salas-Pérez F, Codner E, Valencia E, Pizarro C, Carrasco
2015;64(5):1818–29. E, Pérez-Bravo F. MicroRNAs miR-21a and miR-93 are
54. Yu W-L, Sun Y. Comment on Rondas et al. citrullinated down regulated in peripheral blood mononuclear cells
glucose-regulated protein 78 is an autoantigen in type 1 (PBMCs) from patients with type 1 diabetes. Immunobi-
diabetes. Diabetes 2015;64(3):573–586. ology. 2013;218(5):733–37.
55. Strollo R, Rizzo P, Spoletini M, Landy R, Hughes C, 67. Yang M, Ye L, Wang B, Gao J, Liu R, Hong J, Wang W,
Ponchel F, Napoli N, Palermo A, Buzzetti R, Pozzilli P. Gu W, Ning G. Decreased mi R-146 expression in periph-
HLA-dependent autoantibodies against post-translation- eral blood mononuclear cells is correlated with ongoing
ally modified collagen type II in type 1 diabetes mellitus. islet autoimmunity in type 1 diabetes patients 1 (downreg-
Diabetologia. 2013;56(3):563–72. ulation of miR-146 expression in peripheral blood mono-
56. Doran TM, Morimoto J, Simanski S, Koesema EJ, Clark nuclear cells in patients with type 2 diabetes is associated
LF, Pels K, Stoops SL, Pugliese A, Skyler JS, Kodadek with persistent immune imbalance in pancreatic islets). J
T. Discovery of phosphorylated peripherin as a major hu- Diabet. 2015;7(2):158–65.
moral autoantigen in type 1 diabetes mellitus. Cell Chem 68. Pflueger M, Seppänen-Laakso T, Suortti T, Hyötyläinen T,
Biol. 2016;23(5):618–28. Achenbach P, Bonifacio E, Orešič M, Ziegler A-G. Age-
57. Strollo R, Vinci C, Napoli N, Pozzilli P, Ludvigsson J, and islet autoimmunity–associated differences in amino
Nissim A. Antibodies to post-translationally modified in- acid and lipid metabolites in children at risk for type 1
sulin as a novel biomarker for prediction of type 1 diabetes diabetes. Diabetes. 2011;60(11):2740–47.
in children. Diabetologia. 2017;60(8):1467–74. 69. Fox TE, Bewley MC, Unrath KA, Pedersen MM, Ander-
58. Akirav EM, Lebastchi J, Galvan EM, Henegariu O, Akirav son RE, Jung DY, Jefferson LS, Kim JK, Bronson SK, Fla-
M, Ablamunits V, Lizardi PM, Herold KC. Detection of β nagan JM. Circulating sphingolipid biomarkers in models
cell death in diabetes using differentially methylated circu- of type 1 diabetes. J Lipid Res. 2011;52(3):509–17.
lating DNA. Proc Natl Acad Sci. 2011;108(47):19018–23. 70. La Torre D, Seppänen-Laakso T, Larsson HE, Hyötyläinen
59. Assmann TS, Recamonde-Mendoza M, De Souza BM, T, Ivarsson SA, Lernmark Å, Orešič M, Group DS.

Volume 39, Issue 4, 2019


260 Dariya et al.

Decreased cord-blood phospholipids in young age–at–on- is not required for diabetogenic CD4+ T cell migration.
set type 1 diabetes. Diabetes. 2013;62(11):3951–56. Front Immunol. 2018;9:2800.
71. Pickup JC, Hussain F, Evans ND, Sachedina N. In vivo 86. Sandor AM, Lindsay RS, Dyjack N, Whitesell JC, Rios
glucose monitoring: the clinical reality and the promise. C, Bradley BJ, Haskins K, Serreze DV, Geurts AM, Chen
Biosensors Bioelectronics. 2005;20(10):1897–902. Y-G. CD11c+ cells are gatekeepers for lymphocyte traf-
72. McClenaghan NH, Flatt P. Engineering cultured insu- ficking to infiltrated islets during type 1 diabetes. Frontiers
lin-secreting pancreatic B-cell lines. J Mol Med. 1999; Immunol. 2019;10:99.
77(1):235–43. 87. Gepts W. Pathologic anatomy of the pancreas in juvenile
73. Flatt P, Lenzen S. Frontiers of insulin secretion and pan- diabetes mellitus. Diabetes. 1965;14(10):619–33.
creatic B-cell research. London: Smith-Gordon; 1994. 88. Bottazzo GF, Dean BM, McNally JM, MacKay EH,
74. Markert LG. Prolonged exercise and its effects on type 1 Swift PG, Gamble DR. In situ characterization of auto-
diabetes mellitus [dissertation]. Laramie, WY: University immune phenomena and expression of HLA molecules
of Wyoming; 2017. in the pancreas in diabetic insulitis. New Engl J Med.
75. Cooper JD, Smyth DJ, Walker NM, Stevens H, Burren 1985;313(6):353–60.
OS, Wallace C, Greissl C, Ramos-Lopez E, Hyppönen E, 89. Lernmark Å, Stenger D, Baskin D, Palmer J, Li L,
Dunger DB. Inherited variation in vitamin D genes is as- Klöppel G, Vathanaprida C, Fält K, Landin-Olsson M,
sociated with predisposition to autoimmune disease type 1 Gown A. Heterogeneity of islet pathology in two infants
diabetes. Diabetes. 2011;60(5):1624–31. with recent onset diabetes mellitus. Virchows Archiv.
76. Atkinson MA, Eisenbarth GS, Michels AW. Type 1 diabe- 1995;425(6):631–40.
tes. Lancet. 2014;383(9911):69–82. 90. Wenzlau JM, Juhl K, Yu L, Moua O, Sarkar SA, Gottlieb
77. Hostens K, Pavlovic D, Zambre Y, Ling Z, Van Schrav- P, Rewers M, Eisenbarth GS, Jensen J, Davidson HW. The
endijk C, Eizirik DL, Pipeleers DG. Exposure of human cation efflux transporter ZnT8 (Slc30A8) is a major au-
islets to cytokines can result in disproportionately ele- toantigen in human type 1 diabetes. Proc Natl Acad Sci.
vated proinsulin release. J Clin Investig. 1999;104(1): 2007;104(43):17040–45.
67–72. 91. Palmer JP, Asplin CM, Clemons P, Lyen K, Tatpati O,
78. Ohara-Imaizumi M, Cardozo AK, Kikuta T, Eizirik DL, Raghu PK, Paquette TL. Insulin antibodies in insulin-de-
Nagamatsu S. The cytokine interleukin-1β reduces the pendent diabetics before insulin treatment. Science.
docking and fusion of insulin granules in pancreatic 1983;222(4630):1337–39.
β-cells, preferentially decreasing the first phase of exocy- 92. Baekkeskov S, Aanstoot H-J, Christgai S, Reetz A, Soli-
tosis. J Bio Chem. 2004;279(40):41271–74. mena M, Cascalho M, Folli F, Richter-Olesen H, Camilli
79. Eizirik DL, Mandrup-Poulsen T. A choice of death–the P-D. Identification of the 64K autoantigen in insulin-de-
signal-transduction of immune-mediated beta-cell apop- pendent diabetes as the GABA-synthesizing enzyme glu-
tosis. Diabetologia. 2001;44(12):2115–33. tamic acid decarboxylase. Nature. 1990;347(6289):151.
80. Wållberg M, Cooke A. Immune mechanisms in type 1 di- 93. Rabin DU, Pleasic SM, Palmer-Crocker R, Shapiro JA.
abetes. Trends Immunol. 2013;34(12):583–91. Cloning and expression of IDDM-specific human autoan-
81. Carrero JA, Calderon B, Towfic F, Artyomov MN, Un- tigens. Diabetes. 1992;41(2):183–86.
anue ER. Correction: defining the transcriptional and 94. Wasmeier C, Hutton JC. Molecular cloning of phog-
cellular landscape of type 1 diabetes in the NOD mouse. rin, a protein-tyrosine phosphatase homologue localized
PLoS One. 2014;9(1). doi: 10.1371/annotation/f277b29e- to insulin secretory granule membranes. J Bio Chem.
361b-4e56-b55b-612ebaca0432. 1996;271(30):18161–70.
82. Turley S, Poirot L, Hattori M, Benoist C, Mathis D. Phys- 95. Kaufman D, Erlander M, Clare-Salzler M, Atkinson M,
iological β cell death triggers priming of self-reactive T Maclaren N, Tobin A. Autoimmunity to two forms of glu-
cells by dendritic cells in a type-1 diabetes model. J Exper tamate decarboxylase in insulin-dependent diabetes melli-
Med. 2003;198(10):1527–37. tus. J Clin Investig. 1992;89(1):283–92.
83. Magnuson AM, Thurber GM, Kohler RH, Weissleder R, 96. Fenalti G, Law RH, Buckle AM, Langendorf C, Tuck K,
Mathis D, Benoist C. Population dynamics of islet-infil- Rosado CJ, Faux NG, Mahmood K, Hampe CS, Banga JP.
trating cells in autoimmune diabetes. Proc Natl Acad Sci. GABA production by glutamic acid decarboxylase is reg-
2015;112(5):1511–16. ulated by a dynamic catalytic loop. Nat Struct Mol Biol.
84. Calderon B, Carrero JA, Miller MJ, Unanue ER. Cellu- 2007;14(4):280.
lar and molecular events in the localization of diabeto- 97. Kass I, Hoke DE, Costa MG, Reboul CF, Porebski BT,
genic T cells to islets of Langerhans. Proc Natl Acad Sci. Cowieson NP, Leh H, Pennacchietti E, McCoey J, Kleifeld
2011;108(4):1561–66. O. Cofactor-dependent conformational heterogeneity of
85. Scott NA, Zhao Y, Krishnamurthy B, Mannering SI, Kay GAD65 and its role in autoimmunity and neurotransmitter
TW, Thomas HE. IFNγ-induced MHC class II expression homeostasis. Proc Natl Acad Sci. 2014;111(25):E2524–29.
on islet endothelial cells is an early marker of insulitis but 98. Mally MI, Cirulli V, Otonkoski T, Soto G, Hayek A.

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy261

Ontogeny and tissue distribution of human GAD expres- 111. Chimienti F, Devergnas S, Pattou F, Schuit F, Garcia-Cuenca
sion. Diabetes. 1996;45(4):496–501. R, Vandewalle B, Kerr-Conte J, Van Lommel L, Grunwald
99. Gotter J, Brors B, Hergenhahn M, Kyewski B. Medul- D, Favier A. In vivo expression and functional characteriza-
lary epithelial cells of the human thymus express a highly tion of the zinc transporter ZnT8 in glucose-induced insulin
diverse selection of tissue-specific genes colocalized in secretion. J Cell Sci. 2006;119(20):4199–206.
chromosomal clusters. J Exper Med. 2004;199(2):155–66. 112. Mohanasundaram D, Drogemuller C, Brealey J, Jessup
100. Piquer S, Belloni C, Lampasona V, Bazzigaluppi E, Vi- CF, Milner C, Murgia C, Lang CJ, Milton A, Zalewski PD,
anello M, Giometto B, Bosi E, Bottazzo GF, Bonifacio Russ GR. Ultrastructural analysis, zinc transporters, glu-
E. Humoral autoimmune responses to glutamic acid de- cose transporters and hormones expression in new world
carboxylase have similar target epitopes and subclass that primate (Callithrix jacchus) and human pancreatic islets.
show titer-dependent disease association. Clin Immunol. Gen Comp Endocrinol. 2011;174(2):71–79.
2005;117(1):31–35. 113. Horton R, Wilming L, Rand V, Lovering RC, Bruford EA,
101. Bonifacio E, Lampasona V, Bernasconi L, Ziegler A-G. Khodiyar VK, Lush MJ, Povey S, Talbot Jr CC, Wright
Maturation of the humoral autoimmune response to epi- MW. Gene map of the extended human MHC. Nat Rev
topes of GAD in preclinical childhood type 1 diabetes. Gen. 2004;5(12):889.
Diabetes. 2000;49(2):202–8. 114. Ounissi-Benkalha H, Polychronakos C. The molecular ge-
102. Kawasaki E, Yu L, Rewers MJ, Hutton JC, Eisenbarth netics of type 1 diabetes: new genes and emerging mecha-
GS. Definition of multiple ICA512/phogrin autoantibody nisms. Trends Mol Med. 2008;14(6):268–75.
epitopes and detection of intramolecular epitope spread- 115. Van Belle TL, Coppieters KT, Von Herrath MG. Type 1 di-
ing in relatives of patients with type 1 diabetes. Diabetes. abetes: etiology, immunology, and therapeutic strategies.
1998;47(5):733–42. Physiol Rev. 2011;91(1):79–118.
103. Solimena M, Dirkx R, Hermel J, Pleasic-Williams S, Sha- 116. Yu L, Zhao Z, Steck AK. T1D Autoantibodies: room for
piro J, Caron L, Rabin D. ICA 512, an autoantigen of type improvement? Curr Opin Endocrinol Diabet Obesity.
I diabetes, is an intrinsic membrane protein of neurosecre- 2017;24(4):285.
tory granules. EMBO J. 1996;15(9):2102–14. 117. Redondo MJ, Jeffrey J, Fain PR, Eisenbarth GS, Orban T.
104. Trajkovski M, Mziaut H, Schubert S, Kalaidzidis Y, Al- Concordance for islet autoimmunity among monozygotic
tkrüger A, Solimena M. Regulation of insulin granule twins. New Engl J Med. 2008;359(26):2849–50.
turnover in pancreatic β-cells by cleaved ICA512. J Bio 118. Maziarz M, Hagopian W, Palmer J, Sanjeevi C, Kockum I,
Chem. 2008;283(48):33719–29. Breslow N, Lernmark Å, Register TSCD, Graham J, Mac-
105. Trajkovski M, Mziaut H, Altkrüger A, Ouwendijk J, Neney B. Non-HLA type 1 diabetes genes modulate dis-
Knoch K-P, Müller S, Solimena M. Nuclear transloca- ease risk together with HLA-DQ and islet autoantibodies.
tion of an ICA512 cytosolic fragment couples granule Gene Immun. 2015;16(8):541.
exocytosis and insulin expression in β-cells. J Cell Biol. 119. Risch N. Assessing the role of HLA-linked and unlinked
2004;167(6):1063–74. determinants of disease. Am J Human Gen. 1987;40(1):1.
106. Diez J, Park Y, Zeller M, Brown D, Garza D, Ricordi C, 120. Concannon P, Rich SS, Nepom GT. Genetics of type 1A
Hutton J, Eisenbarth GS, Pugliese A. Differential splic- diabetes. New Engl J Med. 2009;360(16):1646–54.
ing of the IA-2 mRNA in pancreas and lymphoid organs 121. Sosenko JM, Skyler JS, DiMeglio LA, Beam CA, Krischer
as a permissive genetic mechanism for autoimmunity JP, Greenbaum CJ, Boulware D, Rafkin LE, Matheson D,
against the IA-2 type 1 diabetes autoantigen. Diabetes. Herold KC. A new approach for diagnosing type 1 diabe-
2001;50(4):895–900. tes in autoantibody-positive individuals based on predic-
107. Bearzatto M, Lampasona V, Belloni C, Bonifacio E. Fine tion and natural history. Diabet Care. 2015;38(2):271–76.
mapping of diabetes-associated IA-2 specific autoantibod- 122. Helminen O, Aspholm S, Pokka T, Ilonen J, Simell O,
ies. J Autoimmun. 2003;21(4):377–82. Veijola R, Knip M. OGTT and random plasma glucose
108. Dromey JA, Weenink SM, Peters GH, Endl J, Tighe PJ, in the prediction of type 1 diabetes and time to diagnosis.
Todd I, Christie MR. Mapping of epitopes for autoantibod- Diabetologia. 2015;58(8):1787–96.
ies to the type 1 diabetes autoantigen IA-2 by peptide phage 123. Helminen O, Aspholm S, Pokka T, Hautakangas M-R,
display and molecular modeling: overlap of antibody and Haatanen N, Lempainen J, Ilonen J, Simell O, Knip M,
T cell determinants. J Immunol. 2004;172(7):4084–90. Veijola R. HbA1c predicts time to diagnosis of type 1 dia-
109. Chimienti F, Devergnas S, Favier A, Seve M. Identifi- betes in children at risk. Diabetes. 2015;64(5):1719–27.
cation and cloning of a β-cell–specific zinc transporter, 124. Krischer JP, Group TDTS. The use of intermediate end-
ZnT-8, localized into insulin secretory granules. Diabetes. points in the design of type 1 diabetes prevention trials.
2004;53(9):2330–7. Diabetologia. 2013;56(9):1919–24.
110. Lemaire K, Chimienti F, Schuit F. Zinc transporters 125. Krogvold L, Wiberg A, Edwin B, Buanes T, Jahnsen
and their role in the pancreatic β-cell. J Diabet Investig. FL, Hanssen KF, Larsson E, Korsgren O, Skog O,
2012;3(3):202–11. Dahl-Jørgensen K. Insulitis and characterisation of

Volume 39, Issue 4, 2019


262 Dariya et al.

infiltrating T cells in surgical pancreatic tail resections Silva GM, Lopes CAP, Almeida AP, Donato MA, Peixoto
from patients at onset of type 1 diabetes. Diabetologia. C, Moura AA. The effects of insulin and follicle-simulat-
2016;59(3):492–501. ing hormone (FSH) during in vitro development of ovarian
126. Imagawa A, Hanafusa T, Tamura S, Moriwaki M, Itoh N, goat preantral follicles and the relative mRNA expression
Yamamoto K, Iwahashi H, Yamagata K, Waguri M, Nanmo for insulin and FSH receptors and cytochrome P450 aro-
T. Pancreatic biopsy as a procedure for detecting in situ au- matase in cultured follicles. Biol Reprod. 2012;87(3):69,
toimmune phenomena in type 1 diabetes: close correlation 1–11.
between serological markers and histological evidence of 140. de Oliveira Baraldi C, Moisés EC, Carvalho TMdJP, de
cellular autoimmunity. Diabetes. 2001;50(6):1269–73. Jesus Antunes N, Lanchote VL, Duarte G, Cavalli RC. Ef-
127. Krischer JP, Lynch KF, Schatz DA, Ilonen J, Lernmark Å, fect of type 2 diabetes mellitus on the pharmacokinetics of
Hagopian WA, Rewers MJ, She J-X, Simell OG, Toppari metformin in obese pregnant women. Clin Pharmacoki-
J. The 6 year incidence of diabetes-associated autoanti- net. 2012;51(11):743–49.
bodies in genetically at-risk children: the TEDDY study. 141. Donath M, Halban PA. Decreased beta-cell mass in dia-
Diabetologia. 2015;58(5):980–87. betes: significance, mechanisms and therapeutic implica-
128. Association AD. Standards of medical care in diabe- tions. Diabetologia. 2004;47(3):581–89.
tes—2010. Diabetes Care. 2010;33(Suppl 1):S11–S61. 142. Donath MY, Størling J, Maedler K, Mandrup-Poulsen
129. Frank RN. Potential new medical therapies for diabetic T. Inflammatory mediators and islet β-cell failure: a
retinopathy: protein kinase C inhibitors. New York: Else- link between type 1 and type 2 diabetes. J Mol Med.
vier; 2002. 2003;81(8):455–70.
130. Mahmud M. American diabetes association standard of 143. Rasschaert J, Liu D, Kutlu B, Cardozo A, Kruhøffer M,
medical care in diabetes. Diabet Care. 2009;32:13–61. Ørntoft TF, Eizirik D. Global profiling of double stranded
131. Gross JL, De Azevedo MJ, Silveiro SP, Canani LH, Car- RNA-and IFN-γ-induced genes in rat pancreatic beta
amori ML, Zelmanovitz T. Diabetic nephropathy: diagno- cells. Diabetologia. 2003;46(12):1641–57.
sis, prevention, and treatment. Diabet Care. 2005;28(1): 144. Kutlu B, Cardozo AK, Darville MI, Kruhøffer M, Mag-
164–76. nusson N, Ørntoft T, Eizirik DL. Discovery of gene
132. Garg JP, Bakris GL. Microalbuminuria: marker of vas- networks regulating cytokine-induced dysfunction and
cular dysfunction, risk factor for cardiovascular disease. apoptosis in insulin-producing INS-1 cells. Diabetes.
Vasc Med. 2002;7(1):35–43. 2003;52(11):2701–19.
133. Klausen K, Borch-Johnsen K, Feldt-Rasmussen B, Jen- 145. Cardozo AK, Heimberg H, Heremans Y, Leeman R, Kutlu
sen G, Clausen P, Scharling H, Appleyard M, Jensen JS. B, Kruhøffer M, Ørntoft T, Eizirik DL. A comprehensive
Very low levels of microalbuminuria are associated with analysis of cytokine-induced and nuclear factor-κB-de-
increased risk of coronary heart disease and death inde- pendent genes in primary rat pancreatic β-cells. J Bio
pendently of renal function, hypertension, and diabetes. Chem. 2001;276(52):48879–86.
Circulation. 2004;110(1):32–35. 146. Cardozo AK, Kruhøffer M, Leeman R, Ørntoft T, Eizirik
134. Boulton AJ, Vinik AI, Arezzo JC, Bril V, Feldman EL, DL. Identification of novel cytokine-induced genes in
Freeman R, Malik RA, Maser RE, Sosenko JM, Ziegler pancreatic β-cells by high-density oligonucleotide arrays.
D. Diabetic neuropathies: a statement by the American di- Diabetes. 2001;50(5):909–20.
abetes association. Diabet Care. 2005;28(4):956–62. 147. Darville M, Eizirik DL. Regulation by cytokines of the
135. Orchard TJ, Olson JC, Erbey JR, Williams K, Forrest KY- inducible nitric oxide synthase promoter in insulin-pro-
Z, Kinder LS, Ellis D, Becker DJ. Insulin resistance–re- ducing cells. Diabetologia. 1998;41(9):1101–8.
lated factors, but not glycemia, predict coronary artery 148. Cnop M, Welsh N, Jonas J-C, Jörns A, Lenzen S, Eizirik
disease in type 1 diabetes: 10-year follow-up data from the DL. Mechanisms of pancreatic β-cell death in type 1 and
pittsburgh epidemiology of diabetes complications study. type 2 diabetes: many differences, few similarities. Diabe-
Diabet Care. 2003;26(5):1374–79. tes. 2005;54(Suppl 2):S97–S107.
136. Roy M, Peng B, Roy A. Risk factors for coronary disease 149. Larsen CM, Wadt KA, Juhl LF, Andersen HU, Karlsen
and stroke in previously hospitalized African Americans AE, Su MS-S, Seedorf K, Shapiro L, Dinarello CA, Man-
with type 1 diabetes: a 6-year follow-up. Diabet Med. drup-Poulsen T. Interleukin-1β-induced rat pancreatic
2007;24(12):1361–68. islet nitric oxide synthesis requires both the p38 and ex-
137. Kim B, Feldman EL. Insulin resistance in the nervous sys- tracellular signal-regulated kinase 1/2 mitogen-activated
tem. Trend Endocrinol Metabol. 2012;23(3):133–41. protein kinases. J Bio Chem. 1998;273(24):15294–300.
138. Duarte FHG, Jallad RS, Amaro ACS, Drager LF, Loren- 150. Saldeen J, Lee JC, Welsh N. Role of p38 mitogen-ac-
zi-Filho G, Bronstein MD. The impact of sleep apnea tivated protein kinase (p38 MAPK) in cytokine-in-
treatment on carbohydrate metabolism in patients with duced rat islet cell apoptosis. Biochem Pharmacol.
acromegaly. Pituitary. 2013;16(3):341–50. 2001;61(12):1561–69.
139. Chaves RN, Duarte ABG, Rodrigues GQ, Celestino JJ, 151. Pavlovic D, Andersen NA, Mandrup-Poulsen T, Zizirik

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy263

D. Activation of extracellular signal-regulated kinase 166. Šimunović M, Paradžik M, Škrabić R, Unić I, Bućan K,
(ERK) 1/2 contributes to cytokine-induced apoptosis Škrabić V. Cataract as early ocular complication in chil-
in purified rat pancreatic b-cells. Eur Cytokine Netw. dren and adolescents with type 1 diabetes mellitus. Int J
2000;11(2):267–74. Endocrinol. 2018;2018:6763586.
152. Saldeen J, Tillmar L, Karlsson E, Welsh N. Nicotin- 167. Akamine T, Kusunose N, Matsunaga N, Koyanagi S, Ohdo
amide-and caspase-mediated inhibition of poly (ADP-ri- S. Accumulation of sorbitol in the sciatic nerve modulates
bose) polymerase are associated with p53-independent circadian properties of diabetes-induced neuropathic pain
cell cycle (G2) arrest and apoptosis. Mol Cell Biochem. hypersensitivity in a diabetic mouse model. Biochem Bio-
2003;243(1-2):113–22. phys Res Commun. 2018;503(1):181–87.
153. Schröder M, Kaufman RJ. ER stress and the unfolded 168. Brownlee M. Biochemistry and molecular cell biology of
protein response. Mutat Res/Fund Mol Mechan Mutagen. diabetic complications. Nature. 2001;414(6865):813.
2005;569(1-2):29–63. 169. Kolm-Litty V, Sauer U, Nerlich A, Lehmann R, Schle-
154. Maechler P, Wollheim CB. Mitochondrial function in nor- icher E. High glucose-induced transforming growth factor
mal and diabetic β-cells. Nature. 2001;414(6865):807. beta1 production is mediated by the hexosamine pathway
155. Newmeyer DD, Ferguson-Miller S. Mitochondria: re- in porcine glomerular mesangial cells. J Clin Investig.
leasing power for life and unleashing the machineries of 1998;101(1):160–69.
death. Cell. 2003;112(4):481–90. 170. Gurel Z, Sieg KM, Shallow KD, Sorenson CM, Sheibani
156. Friedlander RM. Apoptosis and caspases in neurodegener- N. Retinal O-linked N-acetylglucosamine protein modifi-
ative diseases. New Engl J Med. 2003;348(14):1365–75. cations: implications for postnatal retinal vascularization
157. Alberti KGMM, Zimmet PZ. Definition, diagnosis and and the pathogenesis of diabetic retinopathy. Mol Vision.
classification of diabetes mellitus and its complications. 2013;19:1047.
Part 1: diagnosis and classification of diabetes mellitus. 171. Vidro EK, Gee S, Unda R, Ma J-X, Tsin A. Glucose and
Provisional report of a WHO consultation. Diabet Med. TGFβ2 modulate the viability of cultured human ret-
1998;15(7):539–53. inal pericytes and their VEGF release. Curr Eye Res.
158. Goldin A, Beckman JA, Schmidt AM, Creager MA. Ad- 2008;33(11-12):984–93.
vanced glycation end products: sparking the development 172. Burt D, Gruden G, Thomas S, Tutt P, Dell’Anna C, Viberti
of diabetic vascular injury. Circulation. 2006;114(6): G, Gnudi L. P38 mitogen-activated protein kinase medi-
597–605. ates hexosamine-induced TGFβ1 mRNA expression in
159. Tam XH, Shiu SW, Leng L, Bucala R, Betteridge DJ, Tan human mesangial cells. Diabetologia. 2003;46(4):531–37.
KC. Enhanced expression of receptor for advanced glyca- 173. Buse MG. Hexosamines, insulin resistance, and the com-
tion end-products is associated with low circulating solu- plications of diabetes: current status. Am J Physiol-Endo-
ble isoforms of the receptor in type 2 diabetes. Clin Sci. crinol Metabol. 2006;290(1):E1–E8.
2010;120(2):81–89. 174. Geraldes P, King GL. Activation of protein kinase C iso-
160. Giannini C, D’Adamo E, De Giorgis T, Chiavaroli V, Ver- forms and its impact on diabetic complications. Circ Res.
rotti A, Chiarelli F, Mohn A. The possible role of esRAGE 2010;106(8):1319–31.
and sRAGE in the natural history of diabetic nephropathy 175. Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H,
in childhood. Pediatr Nephrol. 2012;27(2):269–75. Maeda S, Sugimoto T, Yasuda H, Kashiwagi A, Ways DK.
161. McVicar CM, Ward M, Colhoun LM, Guduric-Fuchs Amelioration of accelerated diabetic mesangial expan-
J, Bierhaus A, Fleming T, Schlotterer A, Kolibabka M, sion by treatment with a PKC β inhibitor in diabetic db/
Hammes H-P, Chen M. Role of the receptor for advanced db mice, a rodent model for type 2 diabetes. FASEB J.
glycation endproducts (RAGE) in retinal vasodegener- 2000;14(3):439–47.
ative pathology during diabetes in mice. Diabetologia. 176. Ohshiro Y, Ma RC, Yasuda Y, Hiraoka-Yamamoto J, Cl-
2015;58(5):1129–37. ermont AC, Isshiki K, Yagi K, Arikawa E, Kern TS, King
162. Han R, Liu Z, Sun N, Liu S, Li L, Shen Y, Xiu J, Xu Q. GL. Reduction of diabetes-induced oxidative stress, fi-
BDNF Alleviates neuroinflammation in the hippocampus brotic cytokine expression, and renal dysfunction in pro-
of type 1 diabetic mice via blocking the aberrant HMGB1/ tein kinase Cβ–null mice. Diabetes. 2006;55(11):3112–20.
RAGE/NF-κB pathway. Aging Dis. 2019;10(3):611. 177. Yuan SY, Ustinova EE, Wu MH, Tinsley JH, Xu W, Koro-
163. Tang W, Martin KA, Hwa J. Aldose reductase, oxidative mpai FL, Taulman AC. Protein kinase C activation con-
stress, and diabetic mellitus. Front Pharmacol. 2012;3:87. tributes to microvascular barrier dysfunction in the heart
164. Liu H, Luo Y, Zhang T, Zhang Y, Wu Q, Yuan L, Chung at early stages of diabetes. Circ Res. 2000;87(5):412–17.
S, Oates P, Yang J. Genetic deficiency of aldose reductase 178. Beckman JA, Creager MA, Libby P. Diabetes and athero-
counteracts the development of diabetic nephropathy in sclerosis: epidemiology, pathophysiology, and manage-
C57BL/6 mice. Diabetologia. 2011;54(5):1242–51. ment. JAMA. 2002;287(19):2570–81.
165. Oates PJ. Aldose reductase inhibitors and diabetic kidney 179. Maresch CC, Stute DC, Fleming T, Lin J, Hammes H-P,
disease. Curr Opin Investig Drugs. 2010;11(4):402–17. Linn T. Hyperglycemia induces spermatogenic disruption

Volume 39, Issue 4, 2019


264 Dariya et al.

via major pathways of diabetes pathogenesis. Sci Rep. type 2 diabetes susceptibility region on human chromo-
2019;9(1):1–12. some 20. Genomics. 2001;76(1-3):45–57.
180. Crèvecoeur I, Rondas D, Mathieu C, Overbergh L. The be- 193. Hulbert EM, Smink LJ, Adlem EC, Allen JE, Burdick
ta-cell in type 1 diabetes: what have we learned from pro- DB, Burren OS, Cavnor CC, Dolman GE, Flamez D, Fri-
teomic studies? Proteom–Clin Appl. 2015;9(7-8):755–66. ery KF. T1DBase: integration and presentation of com-
181. Merchant ML, Niewczas MA, Ficociello LH, Lukenbill plex data for type 1 diabetes research. Nucl Acid Res.
JA, Wilkey DW, Li M, Khundmiri SJ, Warram JH, Kro- 2006;35(Suppl 1):D742–46.
lewski AS, Klein JB. Plasma kininogen and kininogen 194. Lee HJ, Lee SH, Ha K-S, Jang HC, Chung W-Y, Kim JY,
fragments are biomarkers of progressive renal decline in Chang Y-S, Yoo DH. Ubiquitous healthcare service using
type 1 diabetes. Kidney Int. 2013;83(6):1177–84. Zigbee and mobile phone for elderly patients. Int J Med
182. Mogensen CE, Keane WF, Bennett PH, Jerums G, Parv- Inform. 2009;78(3):193–98.
ing H-H, Passa P, Steffes MW, Striker GE, Viberti GC. 195. Shakil S, Khan AU. Infected foot ulcers in male and fe-
Prevention of diabetic renal disease with special reference male diabetic patients: a clinico-bioinformative study.
to microalbuminuria. In: The Kidney and Hypertension in Ann Clin Microbiol Antimicrob. 2010;9(1):2.
Diabetes Mellitus. New York: Springer; 1996. p. 539–49. 196. Xu D, Xu Y, Uberbacher C. Computational tools for pro-
183. Giorgino F, Laviola L, Perin PC, Solnica B, Fuller J, tein modeling. Curr Protein Peptide Sci. 2000;1(1):1–21.
Chaturvedi N. Factors associated with progression to mac- 197. Schwede T, Diemand A, Guex N, Peitsch MC. Protein
roalbuminuria in microalbuminuric type 1 diabetic pa- structure computing in the genomic era. Res Microbiol.
tients: the EURODIAB prospective complications study. 2000;151(2):107–12.
Diabetologia. 2004;47(6):1020–28. 198. Schwede T, Kopp J, Guex N, Peitsch MC. SWISS-MODEL:
184. Perkins BA, Krolewski AS. Early nephropathy in type 1 an automated protein homology-modeling server. Nucleic
diabetes: a new perspective on who will and who will not Acids Res. 2003;31(13):3381–85.
progress. Curr Diabet Rep. 2005;5(6):455–63. 199. Sánchez R, Šali A. Comparative protein structure model-
185. Perkins BA, Ficociello LH, Ostrander BE, Silva KH, ing: introduction and practical examples with modeller.
Weinberg J, Warram JH, Krolewski AS. Microalbuminuria In: Protein Structure Prediction. New York: Springer;
and the risk for early progressive renal function decline in 2000. p. 97–129.
type 1 diabetes. J Am Soc Nephrol. 2007;18(4):1353–61. 200. Wiltgen M, Tilz GP. Homology modelling: a review about
186. Crèvecoeur I, Vig S, Mathieu C, Overbergh L. Under- the method on hand of the diabetic antigen GAD 65 struc-
standing type 1 diabetes through proteomics. Expert Rev ture prediction. Wiener Medizinische Wochenschrift.
Proteom. 2017;14(7):571–80. 2009;159(5-6):112–25.
187. Crèvecoeur I, Gudmundsdottir V, Vig S, Sodré FMC, 201. Rao AA, Thota H, Adapala R, Changalasetty SB,
D’Hertog W, Fierro AC, Van Lommel L, Gysemans C, Gumpeny RS, Akula A, Thota LS, Challa SR, Rao MN,
Marchal K, Waelkens E. Early differences in islets from Das UN. Proteomic analysis in diabetic cardiomyopathy
prediabetic NOD mice: combined microarray and pro- using bioinformatics approach. Bioinform Biol Insights.
teomic analysis. Diabetologia. 2017;60(3):475–89. 2008;2:BBI. S313.
188. Zhang L, Lanzoni G, Battarra M, Inverardi L, Zhang Q. 202. Rao AA, Tayaru NM, Thota H, Changalasetty SB, Thota
Proteomic profiling of human islets collected from frozen LS, Gedela S. Bioinformatic analysis of functional pro-
pancreata using laser capture microdissection. J Proteom. teins involved in obesity associated with diabetes. Int J
2017;150:149–59. Biomed Sci. 2008;4(1):70.
189. Knip M, Veijola R, Virtanen SM, Hyöty H, Vaarala O, 203. Zhang Q, Fillmore TL, Schepmoes AA, Clauss TR, Grit-
Åkerblom HK. Environmental triggers and determinants senko MA, Mueller PW, Rewers M, Atkinson MA, Smith
of type 1 diabetes. Diabetes. 2005;54(Suppl 2):S125–S36. RD, Metz TO. Serum proteomics reveals systemic dys-
190. Howson JM, Stevens H, Smyth DJ, Walker NM, Chan- regulation of innate immunity in type 1 diabetes. J Exper
dler KA, Bingley PJ, Todd JA. Evidence that HLA class Med. 2013;210(1):191–203.
I and II associations with type 1 diabetes, autoantibodies 204. Bergman R, Ider YZ, Bowden CR, Cobelli C. Quantitative
to GAD and autoantibodies to IA-2, are distinct. Diabetes. estimation of insulin sensitivity. Am J Physiol Endocrinol
2011;60(10):2635–44. Metab Gastrointest Physiol. 1979;236:E667–E77.
191. Collins C, Purohit S, Podolsky R, Zhao H, Schatz D, 205. Dalla Man C, Rizza RA, Cobelli C. Meal simulation
Eckenrode S, Yang P, Hopkins D, Muir A, Hoffman M. model of the glucose-insulin system. IEEE Trans Biomed
The application of genomic and proteomic technologies Eng. 2007;54(10):1740–49.
in predictive, preventive and personalized medicine. Vasc 206. Erlich HA, Valdes AM, McDevitt SL, Simen BB, Blake
Pharmacol. 2006;45(5):258–67. LA, McGowan KR, Todd JA, Rich SS, Noble JA, Consor-
192. Fossey SC, Mychaleckyj JC, Pendleton JK, Snyder JR, tium TDG. Next generation sequencing reveals the asso-
Bensen JT, Hirakawa S, Rich SS, Freedman BI, Bowden ciation of DRB3* 02: 02 with type 1 diabetes. Diabetes.
DW. A high-resolution 6.0-megabase transcript map of the 2013;62(7):2618–22.

Critical ReviewsTM in Immunology


Computational Approaches for Immune Targets and Therapy265

207. Pareek CS, Smoczynski R, Tretyn A. Sequencing technol- 212. Internationl Organization of Standardization (ISO). In
ogies and genome sequencing. J Appl Genet. 2011;52(4): vitro diagnostic test systems: requirements for blood-glu-
413–35. cose monitoring systems for self-testing in managing dia-
208. Mathieu C, Lahesmaa R, Bonifacio E, Achenbach P, Tree betes mellitus: ISO; 2003.
T. Immunological biomarkers for the development and 213. Nathan DM, Bayless M, Cleary P, Genuth S, Gubitosi-Klug
progression of type 1 diabetes. Diabetologia. 2018;61 R, Lachin JM, Lorenzi G, Zinman B, Group DER. Diabetes
(11):2252–58. control and complications trial/epidemiology of diabetes
209. Jagadeb M, Konkimalla VB, Rath SN, Das RP. Elucida- interventions and complications study at 30 years: advances
tion of the inhibitory effect of phytochemicals with Kir6. and contributions. Diabetes. 2013;62(12):3976–86.
2 wild-type and mutant models associated in type-1 diabe- 214. Skrivarhaug T, Bangstad H-J, Stene L, Sandvik L, Hans-
tes through molecular docking approach. Genom Inform. sen K, Joner G. Long-term mortality in a nationwide
2014;12(4):283. cohort of childhood-onset type 1 diabetic patients in Nor-
210. Pan Y, Wang Y, Zhao Y, Peng K, Li W, Wang Y, Zhang J, way. Diabetologia. 2006;49(2):298–305.
Zhou S, Liu Q, Li X. Inhibition of JNK phosphorylation 215. Foster NC, Beck RW, Miller KM, Clements MA, Rickels
by a novel curcumin analog prevents high glucose–in- MR, DiMeglio LA, Maahs DM, Tamborlane WV, Ber-
duced inflammation and apoptosis in cardiomyocytes and genstal R, Smith E. State of type 1 diabetes management
the development of diabetic cardiomyopathy. Diabetes. and outcomes from the T1D exchange in 2016–2018. Di-
2014;63(10):3497–511. abetes Technol Therap. 2019;21(2):66–72.
211. Cleary PA, Orchard TJ, Genuth S, Wong ND, Detrano R, 216. McCoy RG, Van Houten HK, Ziegenfuss JY, Shah ND,
Backlund J-YC, Zinman B, Jacobson A, Sun W, Lachin Wermers RA, Smith SA. Increased mortality of patients
JM. The effect of intensive glycemic treatment on coro- with diabetes reporting severe hypoglycemia. Diabetes
nary artery calcification in type 1 diabetic participants of Care. 2012;35(9):1897–901.
the diabetes control and complications trial/epidemiol- 217. Gruessner AC, Gruessner RW. Long-term outcome after
ogy of diabetes interventions and complications (DCCT/ pancreas transplantation: a registry analysis. Curr Op Or-
EDIC) study. Diabetes. 2006;55(12):3556–65. gan Transplant. 2016;21(4):377–85.

Volume 39, Issue 4, 2019

You might also like