Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Received: 7 March 2020 Revised: 4 May 2020 Accepted: 4 May 2020

DOI: 10.1111/cge.13772

REVIEW

Genetic predisposition in type 2 diabetes: A promising


approach toward a personalized management of diabetes

Mahmoud M. Sirdah Ph.D.1,2 | N. Scott Reading Ph.D.3,4

1
Division of Hematology, Department of
Internal Medicine, University of Utah School Abstract
of Medicine, Salt Lake City, Utah Diabetes mellitus, also known simply as diabetes, has been described as a chronic
2
Biology Department, Al Azhar University-
and complex endocrine metabolic disorder that is a leading cause of death across the
Gaza, Gaza, Palestine
3
Institute for Clinical and Experimental globe. It is considered a key public health problem worldwide and one of four impor-
Pathology, ARUP Laboratories, Salt Lake tant non-communicable diseases prioritized for intervention through world health
City, Utah
4
campaigns by various international foundations. Among its four categories, Type
Department of Pathology, University of Utah
School of Medicine, Salt Lake City, Utah 2 diabetes (T2D) is the commonest form of diabetes accounting for over 90% of
worldwide cases. Unlike monogenic inherited disorders that are passed on in a simple
Correspondence
Dr. Mahmoud M. Sirdah, Biology Department, pattern, T2D is a multifactorial disease with a complex etiology, where a mixture of
Al Azhar University-Gaza, P O Box 1277, Gaza,
genetic and environmental factors are strong candidates for the development of the
Palestine.
Email: sirdah@alazhar.edu.ps, m.sirdah@ clinical condition and pathology. The genetic factors are believed to be key
utah.edu
predisposing determinants in individual susceptibility to T2D. Therefore, identifying
the predisposing genetic variants could be a crucial step in T2D management as it
may ameliorate the clinical condition and preclude complications. Through an under-
standing the unique genetic and environmental factors that influence the develop-
ment of this chronic disease individuals can benefit from personalized approaches to
treatment. We searched the literature published in three electronic databases:
PubMed, Scopus and ISI Web of Science for the current status of T2D and its associ-
ated genetic risk variants and discus promising approaches toward a personalized
management of this chronic, non-communicable disorder.

KEYWORDS

diabetes mellitus, genetic predisposition, genome-wide association studies, personalized


medicine, pharmacogenomics, T2DM

1 | I N T RO DU CT I O N in medicinal practice.5-8 The concept of personalized medicine has


been with humanity throughout generations and as time passes we
Medicine is science-based conceptual framework and practices deal- are increasing able to act upon our knowledge to affect the course of
ing with the maintenance of health through prevention, diagnosis, disease at the individual level.4,9,10 The most advanced aspect of per-
treatment, alleviation of symptoms, and cure of disease. Hence, medi- sonalized medicine has been in the field of pharmacogenomics: to
cal practices are based on the current understanding of a disease or ill- address the right drug at the right dosage for the right response.11,12
ness and gradually improve as new information and data fills in the Today advances in many aspects of medicine, biology, chemistry have
details.1-3 From the four humors of ancient Greece to today the prac- opened the doors to practical application of personalized medicine in
tice of medicine has increasingly become more exact in defining dis- the treatment of different disorders.6,13,14 Advancements such as the
4
ease and their treatments. Science has opened Pandora's genetic box sequencing of the human genome, genome-wide association studies,
to discover another level of disease interaction to explore and address genetic architecture and molecular technologies brought forth a

Clinical Genetics. 2020;1–23. wileyonlinelibrary.com/journal/cge © 2020 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 1
2 SIRDAH AND READING

genomic revolution to the medical field. This has the potential of intervention by various health foundations: The International Diabetes
improving health treatments by understanding genetic etiologies of Federation (IDF), Centers for Disease Control and Prevention (CDC)
diseases. and the World Health Organization (WHO).23-26 The estimates pro-
The incidence of Type 2 Diabetes (T2D) has risen rapidly in many vided by international specialized organizations like the IDF, WHO
nations in the 21st century and due to its complex etiology, devastat- and CDC for the prevalence of diabetes are frightening. The global
ing pathology and humanistic and economic burdens it has drawn estimates for worldwide prevalence of diabetes among adults over
attention of scientists from many diverse disciplines to better under- 18 years has increased from 4.7% (108 million) in 1980 to 8.8% (425
stand the underlying mechanisms behind its severe complica- million) in 2017 (95% CI 7.2%-11.3%), emphasizing a rapid rise in
tions.15-17 These studies (177 855 according to PubMed, accessed on middle- and low-income countries. Currently, diabetes is affecting
April 20, 2020) cover wide range of diabetes related topics including nearly half billions worldwide and its prevalence is anticipated to fur-
but limited to: clinical trials, complications, diagnosis, prevention, risk ther increase by 10.2% by 2030 and to a global prevalence of 10.9%
factors, and more recently the genetics issues. We believe it is impor- giving rise to more than 700 million people affected by diabetes in the
tant to address the heritable aspects of T2D as a key public health year 2045.23,27Figure 1 illustrates the IDF recent estimates of global
problem that severely impacting the national and global economies to diabetes prevalence (9.3%, 95%CD: 7.4-12.1) in adults (20-79 years
promote for a greater understanding of T2D pathophysiology and old), and the prevalence within seven geographical regions of the
advocate for novel managements supportive of a personalized medi- world. It is worth mentioning that North America-Caribbean region
cine approach. In this work we review the current status of genetic and the Middle East and North Africa (MENA) region are ranked first
risk factors associated with T2D and discuss promising approaches and second in the diabetic prevalence with 13.3% and 12.9%,
toward personalized management of this widely distributed disorder. respectively.
We searched the literature published in three electronic databases: According to the CDC 2017 report, 30.3 million (9.4% of the
PubMed, Scopus and ISI Web of Science for relevant journal articles U.S. population) people had diabetes, and 1 in 4 of them does not know
using “diabetes mellitus,” “Type 2 diabetes,” “personalized medicine,” they have it. The CDC report also showed that the number of adults
“personalized diabetes,” “precision diabetes,” “Type 2 diabetes diagnosed with diabetes has more than doubled in the last two decades.
Genetic risk score,” “Type 2 diabetes genome-wide association studies Recently, the CDC considered diabetes as the seventh leading cause of
or GWAS,” “Type 2 diabetes Genetic architecture,” “Type 2 diabetes death.25,28 According to IDF recent estimate report, someone is dying
candidate gene association studies,” “Type 2 diabetes linkage analy- from diabetes or its deleterious complications every 7 seconds contribut-
sis”, as the main keywords of the queries. The search was restricted to ing to more than 4.2 million deaths per year, with large proportion of
English language publications despite the ethnicity of the population. those deaths occurring in individuals under the age of 60 years.23,27

2 | D I A B E T E S M E LL I T U S 2.2 | History and classification of diabetes

Diabetes mellitus, more commonly referred to as “diabetes,” has been Diabetes occurs either due to insufficient insulin production or when
described as a complex, chronic, non-communicable, endocrine chal- the body cannot efficiently utilize the produced insulin.29 The com-
lenging, metabolic disorder characterized by persistent hyperglycemia mon clinical features and symptoms of diabetes rapid weight loss and
of such magnitude requiring continuous medical management. Other- frequent urination, were described by ancient civilizations: Egyptians
wise, over time it may cause serious pathologic damage and microvas- on a 3600-year-old papyrus (1552 B.C.), as well as other civilizations
cular complications especially in the nerves (neuropathy), kidney including the Greeks, Chinese, Arab and Indians.30-32 However, the
(nephropathy), eye (retinopathy), as well as, macrovascular complica- term diabetes (Greek word meaning to go through or siphon) has been
tions of blood vessels, leading to the development of cardiovascular, credited to Apollonius of Memphis around 250 to 230 B.C. to
cerebrovascular, and peripheral vascular diseases.18-20 describe a disease that drains patients of more liquid than they can
drink.33-35 In the 17th century, diabetes in Europe was known as the
“pissing evil,” and the term “mellitus” (meaning honeyed or sweet) was
2.1 | Epidemiology of diabetes coined by British anatomist and physician Thomas Willis due to the
sweetness of urine from diabetic patients.30,36,37
The prevalence of diabetes is dramatically increasing across the The classification and diagnosis of diabetes are multifaceted and
15,21
globe. Although it has been described as a disease of wealthy and over the last decades many discussions, deliberation and amendments
developed nations, diabetes is now growing rapidly in poor and low- were performed in order to agree on and establish the current widely
income, developing nations as well.22 Owing to its morbidity, mortal- accepted classifications that included four main types of diabetes,
ity, and economic burdens, diabetes is considered a key public health type 1 diabetes (T1D), T2D, gestational diabetes (GD) and specific
problem worldwide and one of four important non-communicable dis- types of diabetes due to other causes,29 the first three classes are also
eases for which public health campaigns have been initiated used by other international organizations including the IDF, CDC
(The World Diabetes Day, The World Health Day) and prioritized for and WHO.
SIRDAH AND READING 3

F I G U R E 1 Global and regional prevalence of diabetes in adults (20-79 years) according to IDF 2019 Atlas [Colour figure can be viewed at
wileyonlinelibrary.com]

2.2.1 | T1D T1D is believed to have a genetic predisposition component. Sup-


port for this hypothesis has come from numerous GWAS linkage and
T1D is an organ-specific autoimmune disease usually leading to rela- association studies, and candidate gene studies, which have identified
tive or absolute insulin deficiency. Loss of insulin production is primar- many T1D risk regions across the human genome. The earliest region
ily due to the infiltration of Langerhans islets by autoreactive identified included the human leukocyte antigen (HLA) class II genes
lymphocytes and the consequent antibodies directed against and and accounted for nearly half of T1D genetic risk. Other non-HLA
destroying the insulin-producing β-cells of the islets. The result is usu- genes (eg, INS, CTLA4, PTPN22, and IL2RA) have also been identified
ally an absolute dependence on exogenous insulin to regulate blood as contributing to the T1D genetic predisposition risk.52 Owing to
glucose. T1D can develop at any age, yet it most frequently develops rapid analytical and technological development about 50 susceptibility
before adulthood.29,38 T1D represents about 5% of the overall dia- regions have identified as contributing to T1D with some loci com-
betic cases, with an increasing incidence worldwide that varies sub- monly associated with immune-mediated disorders.53Onengut-
25
stantially by geographical regions. Although, the highest T1D rates Gumuscu et al., (2016) identified in their comparative analysis study
(incidence of 62.5/100000 per year in Finland) were reported in Scan- with 15 immune-mediated disorders three novel loci that associated
dinavia and north-westEurope,39-42 it is becoming more common in with T1D. Their discussion stressed that there was a genetic similarity
other ethnic groups such as (American Indians, Alaskan Natives, His- between T1D and other autoimmune conditions such as juvenile idio-
43-46
panics), with Western Pacific ethnicities having the lowest rates. pathic arthritis and ulcerative colitis. They also illustrated credible sets
In the United States, CDC estimated the incidence rate, from 2001 to for the T1D SNPs that mainly centralized to enhancer regions in thy-
2015, of T1D in youth increased by 1.9% annually and now exceeds mus, T and B cells, and CD34+ stem cells, opening the door for further
20 per 100 000 population.25,47,48 In the Middle East and North Africa researches to identify causative genes and sequences.53
(MENA) region, where Arabs constitute the ethnic majority, the IDF
recently estimated 149 400 children and adolescents (< 19 years old)
have T1D with an increase of 20 800 newly diagnosed cases annually. 2.2.2 | T2D
Among MENA nations, the incidence of T1D varies from 1 per
100 000 population in Bahrain to about 420 per 100 000 population T2D is the commonest form of diabetes and a risk factor for cardio-
in Algeria.49 Part of this variation is due to under-reporting among the vascular associated morbidity and mortality. Hyperglycemia in this
sub-Saharan nations where only 6.1% of the nations provided infor- form of diabetes is due to ineffective use of insulin, insulin resistance,
mation to the IDF. A substantial variation in the number of T1D cases or relative (rather than absolute) insulin deficiency that impairs glu-
23
was reported among Arab countries. It worthwhile mentioning that cose transport and glucose homeostasis. Because autoimmune
T1D incidence rate from African region (49 diverse sub-Saharan coun- destruction of pancreatic β-cells does not occur in T2D, most but not
tries and territories) should be taken with caution as incidence rates all T2D patients do not need insulin treatment to survive.23,25,29,54-56
are available only from three countries (6.1%). The reasons behind the The specific causes of T2D are not known. The current hypothesis is
global T1D incidence variations are still uncertain but a multifactorial, T2D is the result of multifactorial etiology where a mixture of genetic
complex interplay, including genetic and environmental factors, are and environmental factors are the underlying basis for the pathology
suspected..15,43,44,50,51 of the disease. A number of different genetic variants, but risk is small
4 SIRDAH AND READING

(small effect size with odds ratio of <1.5), have been identified as risk diet and physical activity. These figures do not reflect the state of pre-
factors for an individual's susceptibility for T2D development. How- diabetic patients whose blood glucose is elevated, but not high
ever, these risk factors, alone or in combination may have the poten- enough to be classified as diabetes.23 Unfortunately, many people
tial to substantially affect the clinical picture and cure of T2D with prediabetes have T2D within 10 years.81-83
56-63
patients. The high prevalence of T2D, especially in low- and middle-income
countries, together with the high risk of developing both microvascu-
lar complications (including retinopathy, nephropathy and neuropathy)
2.2.3 | Gestational diabetes and macrovascular complications (such as cardiovascular com-
orbidities) make T2D a seriously expanding global public health prob-
Any degree of glucose intolerance that first develops during preg- lem. A number of studies have shown T2D is formed by a complex or
nancy is called gestational diabetes (GD), where the blood glucose multifactorial etiology where a mixture of genetic and environmental
level is above normal range, yet not of the diagnostic value for diabe- (obesity, an unhealthy diet and physical inactivity) factors are strong
tes.29,64 In pregnant women with GD, some hormones made by the candidates for the impaired glucose homeostasis and subsequent
placenta obstructs the body from using insulin effectively (blocking development of T2D clinical pathology. Therefore, T2D should be
effect on insulin), so glucose builds up in the blood instead of being considered, treated and managed as a chronic heterogeneous disorder
utilized by the cells. This phenomenon usually occurs late in the sec- with multiple pathophysiological abnormalities, diverse susceptibility
ond trimester (20-24 weeks of pregnancy) and is called contra-insulin to complications, and varied response to medical cure and strategic
effect.65,66 Gestational diabetes is reported in almost 7% of all preg- interventions.84-86 Interventions may include key modifiable risk fac-
nancies, however the prevalence is ranging from 1% to 14% tors such as lifestyle, adiposity, physical inactivity, unhealthy diet
depending on the population and the diagnostic criteria.67-69 In the (quality of staple foods) as well as pharmacological issues.21,87-91
United States between 7% and >20% of pregnant women are diag-
nosed with GD.70 Women with GD have a higher risk of complica-
tions in pregnancy and delivery, developing T2D within 5-15 years of 3.1 | Etiology and pathogenesis of T2D
their diagnosis,67,71 and their children have an elevated risk of devel-
oping T2D.72-74 The American Diabetes Association (ADA) recom- T2D is a heterogeneous syndrome characterized by persistent hyper-
mends that women diagnosed with GD should receive lifestyle glycemia (increased hepatic glucose production and decreased periph-
interventions and have regular screenings for the development of eral glucose uptake in target tissues) due to insulin resistance and/or
diabetes.29 relative impairment in insulin secretion as the main pathophysiological
features contributing to the development of symptoms, while disease
progression is attributed to gradual decrease of the functional pancre-
2.2.4 | Specific types of diabetes due to other atic β cell mass and secretory capacity overtime.29,76,92,93
causes However, the causes of T2D are multifactorial and involve, to
varying extents, both genetic and environmental risk factors including,
There are other less frequent types of diabetes with various known but not limited to, those of carbohydrate and fat metabolism, obesity,
etiologies that constitutes 1% to 5% of the overall hyperglycemic dia- lack of exercise, stress, as well as aging.94-99 There are an increasing
betic patients. These less common types are either genetically deter- number of scientific studies that support the possibility of genetic fac-
mined or result from some specific disease, infection, medication, or tors being involved as key determinants in individual susceptibility to
chemical exposure.29,75-77 This class of diabetes include the subtypes T2D, potentially some people inherit a predisposition for the disease
associated with monogenic defects (neonatal diabetes and maturity- and become more likely to develop T2D than others.84-86 Therefore,
onset diabetes of the young [MODY]), pancreatitis, endocrinopathies, identifying T2D genetic risk variants is a crucial step in disease man-
drug-induced, and infection (congenital rubella, cytomegalovirus, ade- agement as it may ameliorate the clinical condition and preclude the
novirus and mumps).29,78-80 complications.56,100-103 Until recently T2D was seen only in adults but
it is nowadays also reporting increasingly frequently in chil-
dren.15,104,105 Remarkably, during the last two decades the number of
3 | OVERVIEW OF T2D T2D has tripled in the aged and overweight or obese populations
making it the fastest-increasing disease worldwide.15,106,107
Over the past few decades there have been significant advancements The worldwide explosion of obesity in the past decade has
in research toward a better understanding of T2D. According to resulted in a markedly ever-increasing prevalence of T2D. Therefore,
recent IDF estimates diabetes affects more than 463 million people understanding the pathogenesis of T2D is of great importance. This
worldwide. The number of people living with this lifelong chronic dis- effort is complicated by different risk factors that likely contribute to
ease continues to grow and is expected to reach 578 million in 2030 the development of T2D and the varied clinical features emerging
and 700 million in 2045. This epidemic is taking place on the back of from one or more synergistic combination of these risk fac-
profound and pervasive detrimental alterations in lifestyle, principally tors.17,106,108 An increasing number of studies suggest the possibility
SIRDAH AND READING 5

of many genes, mostly in carbohydrate and fat metabolism, as transcription factor-7-like2 (TCF7L2) were the firstly identified as T2D
predisposing for T2D which with various environmental factors and susceptibility loci through GWAS.125-128 The fundamental mecha-
lifestyle changes are considered responsible for the onset and the rap- nisms by which these genetic loci participate in T2D development or
idly increasing affected individuals with T2D. The genetic predisposi- even susceptibility is still lacking. So far, many susceptibility and pre-
tion of T2D is not only affecting the development of the clinical disposition genetic loci have been described and found to be
symptoms but also it has been linked to the disease serious pathologic influenced by other environmental factors such as obesity and life
84,109-115
microvascular and macrovascular complications. Eventually, style effects.84,103,129,130 However, there are an increasing molecular
the proper management and treatment of T2D necessitate the eluci- evidence that emphasized on the role of some variants in coding
dation of its molecular etiology by empirically defining the genes and regions as causative genes that associated with T2D.112,131,132
variants involved, as well as, effective interventions to combat the The sibling-relative risk for T2D is 3 compared with 15 for
obesity epidemic that is known to be a major risk factor for develop- T1D.84,133 Genetic studies revealed the concordance of T2D in mono-
ing T2D. zygotic twins is about 70% to 90%, while it is only 20% to 30% in
dizygotic twins.58,59,84,134 The lifetime risk of developing T2D is 40%
for individuals who have one T2D parent and higher (70%) if both par-
3.2 | Genetics and architecture of T2D ents are affected, however the risk of developing T2D is much higher
when the mother has the disease.130,135-137
We owe a debt of gratitude to the many researchers and patients As mentioned above, the polygenic, −multifactorial nature of T2D
who have contributed to our current understanding of the genetic is now well-established and the genetic architecture of T2D is
variants associated with the development and prognosis of T2D. suggesting allelic heterogeneity with multiple genetic risk variants
These advances over the past 20 years have opened new vistas and identified to be associated mainly, but not limited, with β-cell function
established the foundations toward elucidating the genetic architec- (insulin production and release), insulin action (resistance/low sensitiv-
ture risk and molecular basis underlying the pathophysiology of T2D. ity), and adiposity. Despite some debates about the consistency of the
Early linkage studies revealed a few genes (CAPN10, TCF7L2) and can- T2D genetic architectures and the possible disease models, GWAS
didate gene studies built upon those findings to add a handful more studies have provided great understanding and insights into the
genes (PPARG, IRS1, IRS-2, KCNJ11, WFS-1, HNF1A, HNF1B, HNF4A) genetic architecture of T2D, including the number, frequency and
that lead to T2D risk. However, none of these genes could provide a effect sizes of risk variants in different ethnicities and populations
substantial association to the observed heritability of T2D. Such worldwide. GWAS and genetic architecture studies should be contin-
approach in researches was expanded through the linkage studies, ued for T2D, as well as for other polygenetic multifactorial diseases, in
GWAS, and meta-analyses in diverse ethnic groups and multiple diverse populations and including gene-environment interaction
populations.58,84,109,110,112-115 Currently large-scale genetic studies, which would likely contribute crucially to widening the terrain of
have identified more than 400 distinct genomic regions (genetic sig- shared and unique population genetic effects for T2D.138,139 The
nals) influencing T2D, with few exclusions, most of them are common overarching goal of these studies should emphasize on the translation
and shared across different ethnicities and populations. Most of these and utilization of the gathered genetic information into daily clinical
variants have modest predisposition effects on T2D and with a few practice as well as clarifying the molecular mechanism for early dis-
exceptions, most signals mapped to regulatory sequence, non-coding ease prediction, minimizing the complications and therapeutic
sequences outside (intergenic) or within (intronic) the genes that are decision-making.139-143
probably to perturb gene expression rather than alter protein function
in disease-relevant tissues, like pancreatic islets. However, some have
been identified as human validated therapeutic targets through coding 3.2.1 | β-cell function and insulin secretion
variants that are causal for disease.60,116,117 These loci, usually
referred to by the name of the nearest genes, have substantially A foremost metabolic characteristic for diabetes in general is insulin
improved the estimation of T2D genetic predisposition. However, deficiency resulting from relative to absolute pancreatic β-cell dys-
many other genes have been found to predispose to T2D in smaller- function.144,145 Accounting for less than 10% of the diabetic preva-
84,109,112,115,118,119
scale studies in solitary specific populations. lence, the genetic basis of T1D with its autoimmune etiology has been
In 1998 peroxisome proliferator-activatedreceptor-gamma successfully determined that involved genetic variants associated
(PPARG) gene was the first candidate gene to be reproducibly associ- directly with β-cell functions as well as pancreatic development.146-149
120
ated with T2D. This nuclear receptor gene has been identified as a On the other hand, in T2D and due to increased peripheral insulin
key regulator of adipogenesis, glucose homeostasis, and lipid metabo- resistance, the β-pancreatic islets can no longer sustain higher com-
lism. A relatively common substitution (Pro12Ala) in the PPARG gene pensatory rates of insulin production get exhausted and dysfunc-
was found to be associated with lower body mass index and improved tional.93,150-152 In addition, increased β-cell apoptosis leading to
insulin sensitivity among middle-aged and elderly individuals. 120-124
deficit in β-cell mass to a variable extent is also indicated in T2D
The variation at the hematopoietically expressed Homeobox (HHEX), patients. This loss may be due a high secretory demand in overtly
the Solute Carrier Family 30 Member 8 (SLC30A8), and the hyperglycemic or obese individuals, high oxidative stress, misfolding in
6

TABLE 1 Genetic risk variants associated with T2D through β-Cell function and/or insulin secretion

Gene/nearest Genetic
gene Name of gene/nearest gene variant Context location RA OA RAF OR P value Population Study design Reference
NOTCH2 Notch receptor 2 rs10923931 Intron 1p13-p11 T G 0.11 1.13 4.1E−08 European GWAS meta-analysis 172
FAF1 Fas associated factor 1 rs17106184 Intron 1p32.3 G A 0.92 1.10 4.1E−09 Multi ethnic GWAS-meta-analysis 84,173,174
MACF1 Microtubule actin crosslinking factor 1 rs3768321 Intron 1p34.3 T G 0.19 1.13 2.4E−09 Multi ethnic GWAS-meta-analysis 175
PROX1 Prospero homeobox 1 rs340874 Intron 1q32.2-q32.3 C T 0.52 1.07 7.2E−10 European GWAS-meta-analysis 174,176
PROX1 Prospero homeobox 1 rs2075423 Intron 1q32.2-q32.3 G T 0.62 1.07 8.1E−09 European GWAS- meta-analysis 177
BCL11A B-cell lymphoma/leukemia 11A rs243021 Intergenic 2p16.1 A G 0.46 1.08 2.9E−15 European GWAS meta-analysis 178
THADA Thyroid adenoma associated rs7578597 Exon/missense 2p21 T C 0.90 1.15 1.1E−09 European GWAS meta-analysis 172
THADA Thyroid adenoma associated rs35720761 Exon/missense 2p21 C T 0.10 1.12 3.3E−10 Multi ethnic GWAS Meta-analysis 115
TMEM163 Transmembrane protein 163 rs6723108 Intergenic 2q21.3 T G 0.85 1.31 3.3E−09 Indian GWAS 179
COBLL1 Cordon-Bleu WH2 repeat protein like 1 rs7607980 Exon/missense 2q24.3 C T 0.12 1.15 8.3E−15 Multi ethnic GWAS meta-analysis 115
UBE2E2 Ubiquitin conjugating enzyme E2E2 rs7612463 Intron 3p24.3 C A 0.83 1.19 2.3E−09 Japanese GWAS 180
ADCY5 Adenylate cyclase 5 rs11708067 Intron 3q13.2-q21 A G 0.78 1.12 9.9E−21 European GWAS- meta-analysis 176
IGF2BP2 Insulin like growth factor 2 MRNA rs11927381 Intron 3q27.2 C T 0.33 1.14 3.0E−14 Multi ethnic GWAS- meta-analysis 175
binding protein 2
IGF2BP2 Insulin like growth factor 2 MRNA rs4402960 Intron 3q27.2 T G 0.30 1.17 8.9E−16 European GWAS 181,182
binding protein 2
ST6GAL1 ST6 beta-galactoside alpha-2,6-sialyltransferase rs16861329 Intron 3q27.3 G A 0.75 1.09 3.4E−08 South Asia GWAS 183
1
LPP LIM domain containing preferred translocation rs6808574 Intergenic 3q27.3-q28 C T 0.79 1.07 5.8E−09 Multi ethnic GWAS- meta-analysis 173,174
partner in lipoma
WFS1 Wolframin ER transmembrane glycoprotein rs6446482 Intron 4p16.1 G C 0.48 0.90 3.4E−07 UK populations Pooled analysis 184
WFS1 Wolframin ER transmembrane glycoprotein rs10010131 Intron 4p16.1 G A 0.59 1.12 0.001 Swedish and GWAS 185
Finnish
MAEA Macrophage erythroblast attacher rs6815464 Intron 4p16.3 C G 0.58 1.13 1.6E−20 East Asians GWAS meta-analysis 186
TMEM154 Transmembrane protein 154 rs6813195 Intergenic 4q31.3 C T 0.59 1.08 4.1E−14 Multi ethnic GWAS-meta-analysis 173,174
PAM Peptidylglycine alpha-amidating rs35658696 Exon/missense 5q21.1 G A 0.051 1.17 2.5E−17 European GWAS meta-analysis 187
monooxygenase. using
PPIP5K2 Diphosphoinositol pentakisphosphate kinase 2 rs36046591 Exon/missense 5q21.1 G A 0.05 1.19 3.3E−08 Multi ethnic GWAS meta-analysis 115
ZFAND3 Zinc finger AN1-type containing 3 rs9470794 Intron 6p21.2 C T 0.27 1.12 2.1E−10 East Asians GWAS meta-analysis 174,186
KCNK16 Potassium two pore domain channel subfamily rs1535500 Exon/missense 6p21.2 T G 0.42 1.08 2.3E−08 East Asians GWAS meta-analysis 174,186
K member 16
POU5F1/ POU class 5 homeobox 1./transcription factor rs3130501 Intron 6p21.33 G A 0.38 1.07 4.2E−09 Multi ethnic GWAS- meta-analysis 173,174
TCF19 19
CDKAL1 CDK5 regulatory subunit associated rs7766070 Intron 6p22.2 A C 0.27 1.12 1.9E−11 Multi ethnic GWAS- meta-analysis 175
protein 1 like 1
SIRDAH AND READING
TABLE 1 (Continued)

Gene/nearest Genetic
gene Name of gene/nearest gene variant Context location RA OA RAF OR P value Population Study design Reference
SSR1/ RREB1 Signal sequence receptor subunit 1/Ras rs9505118 Intron 6p24.3 A G 0.45 1.06 1.4E−09 Multi ethnic GWAS- meta-analysis 173,174
responsive element binding protein 1
SIRDAH AND READING

RREB1 Ras responsive element binding protein 1 rs9379084 Exon/missense 6p24.3 A G 0.11 1.13 4.0E−09 Multi ethnic GWAS meta-analysis 115
JAZF1 Juxtaposed with another zinc finger protein 1 rs849134 Intron 7p15.2-p15.1 A G 0.53 1.12 6.4E−13 Multi ethnic GWAS- meta-analysis 175
GCK Glucokinase (hexokinase 4) rs4607517 Intergenic 7p15.3-p15.1 A G 0.16 1.07 5.0E−08 European GWAS- meta-analysis 174,176
DGKB/ Diacylglycerol kinase, beta 90 kDa rs2191349 Intergenic 7p21.2 T G 0.52 1.06 1.0E−08 European GWAS- meta-analysis 174,176
TMEM195
GCC1/PAX4 GRIP and coiled-coil domain containing rs6467136 Intergenic 7q32.1 G A 0.79 1.11 5.0E−11 East Asians GWAS meta-analysis 174,186
1/paired box 4
PAX4 Paired box 4 rs2233580 Exon/missense 7q32.1 T C 0.1 1.79 9.3E−09 Multi ethnic GWAS meta-analysis 115
ANK1 Ankyrin-1 rs516946 Intron 8p11.21 C T 0.76 1.09 2.5E−10 European GWAS meta-analysis
TP53INP1 Tumor protein P53 inducible nuclear protein 1 rs896854 Exon/missense 8q22.1 T C 0.48 1.06 9.9E−10 European GWAS meta-analysis
SLC30A8 Solute carrier family 30 member 8 rs13266634 Exon/missense 8q24.11 C T 0.69 1.12 1.4E−11 Multi ethnic GWAS- meta-analysis 172,175
CDKN2A/ Cyclin dependent kinase inhibitor rs10811661 Intergenic 9p21 T C 0.82 1.14 2.3E−10 Multi ethnic GWAS- meta-analysis 175
CDKN2B 2A/cyclin dependent kinase inhibitor 2B
GLIS3 GLIS Family Zinc Finger 3 rs7041847 Intron 9p24.2 A G 0.41 1.10 2.0E−14 East Asians GWAS meta-analysis 186
CHCHD9/ Coiled-coil-helix-coiled-coil-helix domain rs13292136 Intergenic 9q21.31 C T 0.93 1.11 2.8E−08 European GWAS meta-analysis 178
TLE4 containing 2 pseudogene 9/TLE family
member 4, transcriptional corepressor
TLE1 TLE family member 1, transcriptional rs2796441 Intron 9q21.32 G A 0.57 1.07 5.4E−09 European GWAS meta-analysis 177
corepressor
GPSM1 G protein signaling modulator rs11787792 Intron 9q34.3 A G 0.87 1.15 1.7E−10 East Asia GWAS 188
GPSM1 G protein signaling modulator rs60980157 Exon/missense 9q34.3 T C 0.26 1.09 1.7E−09 Multi ethnic GWAS meta-analysis 115,174,177,
179-189
CDC123/ cell division cycle 123 homolog/calcium/ rs12779790 Intergenic 10p13 G A 0.18 1.11 1.2E−10 European GWAS meta-analysis 172
CAMK1D calmodulin dependent protein kinase type
1D
VPS26A VPS26, retromer complex component A rs1802295 30 UTR 10q22.1 A G 0.26 1.08 4.1E−08 South Asia GWAS 183
ZMIZ1 Zinc finger MIZ-type containing 1 rs12571751 Intron 10q22.3 A G 0.52 1.08 1.0E−10 European GWAS meta-analysis 177
HHEX/IDE Hematopoietically expressed homeobox/ rs1111875 Intergenic 10q23.33 C T 0.52 1.13 6.0E−10 European GWAS 181
insulin degrading enzyme
TCF7L2 transcriptional factor 7 like 2 rs7903146 Intron 10q25.2-q25.3 T C 0.18 1.37 1.0E−48 Finn GWAS 181
TCF7L2 transcriptional factor 7 like 2 rs7903146 Intron 10q25.2-q25.3 T C 0.30 1.65 1.5E−34 French GWAS 125
TCF7L2 transcriptional factor 7 like 2 rs34872471 Intron 10q25.2-q25.3 C T 0.28 1.31 6.4E−53 Multi ethnic GWAS- meta-analysis 175
ABCC8 ATP Binding Cassette Subfamily rs757110 Exon/missense 11p15.1 C A 0.40 1.07 1.7E−08 Multi ethnic GWAS meta-analysis 115
C Member 8
7

(Continues)
8

TABLE 1 (Continued)

Gene/nearest Genetic
gene Name of gene/nearest gene variant Context location RA OA RAF OR P value Population Study design Reference
KCNJ11 potassium channel, inwardly rectifying rs5215 Exon/missense 11p15.1 C T 0.27 1.14 5.0E−11 UK GWAS 190
subfamily J, member 11
KCNJ11 potassium channel, inwardly rectifying rs5219 Exon/missense 11p15.1 T C 0.46 1.14 6.7E−11 Finns GWAS 181
subfamily J, member 11
KCNQ1 Potassium channel voltage-gated channel, rs231362 Intron 11p15.5 G A 0.52 1.08 2.8E−13 European GWAS meta-analysis 178
KQT-like subfamily, member 1
ARAP1 ArfGAP with RhoGAP domain, ankyrin repeat rs1552224 50 UTR 11q13.4 A C 0.88 1.14 1.4E−22 European GWAS meta-analysis 178
and PH domain 1
MTNR1B Melatonin receptor 1B rs1387153 Intergenic 11q21-q22 T C 0.28 1.09 7.8E−15 European GWAS meta-analysis 178
TSPAN8/ Tetraspanin 8/leucine rich repeat containing G rs7961581 Intergenic 12q21.1 C T 0.27 1.09 1.1E−09 European GWAS meta-analysis 172
LGR5 protein-coupled receptor
CCDC63 Coiled-coil domain containing protein 63 rs11065756 Intron 12q24.11 G A 0.83 1.40 9.4E−09 Korean GWAS meta-analysis 191
SPRY2 Sprouty RTK signaling antagonist 2 rs1359790 Intergenic 13q31.1 G A 0.71 1.15 6.0E−09 Asian GWAS 192
RASGRP1 RAS guanyl releasing protein 1 rs7403531 Intron 15q14 T C 0.35 1.10 3.9E−09 Chinese Han GWAS 193
C2CD4A C2 calcium dependent domain containing rs7172432 Intergenic 15q22.2 A G 0.58 1.11 8.8E−14 Multi ethnic Meta-analysis 180
protein 4A
HMG20A High mobility group 20A rs7177055 Intron 15q24.3 A G 0.68 1.08 4.6E−09 European GWAS meta-analysis 177
HMG20A High mobility group 20A rs7178572 Intron 15q24.3 G A 0.52 1.09 7.1E−11 South Asia GWAS 183
ZFAND6 Zinc finger AN1-type containing 6 rs11634397 Intergenic 15q25.1 G A 0.60 1.06 2.4E−09 European GWAS meta-analysis 178
AP3S2 Adaptor related protein complex 3 subunit rs2028299 Non coding RNA 15q26.1 C A 0.31 1.10 1.9E−11 South Asia GWAS 183
sigma 2
PRC1 Protein regulator of cytokinesis 1 rs8042680 Intron 15q26.1 A C 0.22 1.07 2.4E−10 European GWAS meta-analysis 178
BCAR1 Breast cancer anti-estrogen resistance 1 rs7202877 Intergenic 16q23.1 T G 0.89 1.12 3.5E−08 European GWAS meta-analysis 177
SRR Serine racemase rs391300 Intron 17p13.3 G A 0.62 1.28 3.1E−09 Chinese Hans GWAS 194
HNF1B HNF1 Homeobox B rs4430796 Intron 17q12 G A 0.28 1.19 1.5E−11 Chinese Hans GWAS 193
TM6SF2 Gene transmembrane 6 superfamily member rs58542926 Exon/ missense 19p13.11 T C 0.08 1.14 3.2E−10 Multi ethnic GWAS meta-analysis 115
GIPR Gastric inhibitory polypeptide receptor rs1800437 Exon/missense 19q13.32 C G 0.21 1.09 4.6 × 10–9 European GWAS-meta-analysis 187
HNF4A Hepatocyte nuclear factor 4 alpha rs4812829 Intron 20q13.12 A G 0.29 1.09 2.6E−10 South Asia GWAS 183
MTMR3 Myotubularin related protein 3 rs41278853 Exon/missense 22q12.2 A G 0.08 1.14 5.6E−09 Multi ethnic GWAS meta-analysis 115
ASCC2 Activating signal cointegrator 1 complex rs11549795 Exon/missense 22q12.2 C T 0.08 1.13 1.0E−07 Multi ethnic GWAS meta-analysis 115
Subunit 2

Abbreviations: RA, risk allele; RAF, risk allele frequency; OA, other allele; OR, odds ratio.
SIRDAH AND READING
SIRDAH AND READING 9

the endoplasmic reticulum, or inflammatory signals of interleukin-1 β receptor activity, lower body mass index and improved insulin sensi-
produced within β-pancreatic islets cells. Unfortunately, β-cell dys- tivity suggesting it may have as protecting effect for an individual
function and the deficit β-cell mass progressively deteriorate insulin against T2D. The PPARG gene encodes a nuclear receptor protein that
secretion and aggravate the hyperglycemia, causing severe diabetic regulates adipocyte differentiation, and possibly lipid metabolism and
complications over time.153-156 expression of adipocyte-specific genes. The PPARG gene expressed in
Involving different populations, GWAS data are providing an adipocytes has an extra exon B and a common polymorphic substitu-
ever-increasing number of T2D susceptibility variants. Interestingly, tion of a Proline for alanine at position 12 (Pro12Ala) of this protein,
and despite the fact that insulin resistance is the main etiological fac- which is cited in about 15% of the European population. This
tor in T2D development,61,63,131,140,141,157-160 the majority variants Pro12Ala polymorphism was reproducibly shown to be linked with
appear to influence insulin secretion and β-cell function rather than increased transcriptional activity, increased insulin sensitivity and
insulin action. The TCF7L2 gene, a pleiotropic transcription factor hence protection against T2D. The strong association of the Pro12Ala
involved in glucose metabolism in many tissues, carried the strongest polymorphism of PPARG gene to T2D was supported by other studies
association with T2D in multiple genetic studies (refs). Several SNP including GWAS, large scale, and meta-analysis.120-123,177,219-224
(ie, rs12255372 and rs7903146) carried by TCF7L2 in several large Table 2 summarizes other genetic variants that were reproducibly
scale case–control studies, meta-analyses, and GWAS revealing the identified by candidate gene association studies, linkage analysis and
remarkable consistency and relatively large effect size OR = 1.37-1.65, GWAS to be associated to T2D probably through insulin action. In
among different ethnicities.161-171Table 1 provides a list of reproduc- addition to genetic risks, it is worthwhile mentioning that obesity and
ibly identified genetic risk variants linked to T2D possibly thought decreased physical activity are also significant indicators of risk
affecting β-cell dysfunction and reduced β-cell mass. toward developing an insulin-resistant condition. When these condi-
tions are added to the genetic predisposition for insulin resistance,
the development of T2D is accelerated.231-235
3.2.2 | Insulin action (insulin resistance/low insulin
sensitivity loci)
3.2.3 | Obesity and altered BMI
Insulin is a peptide hormone produced by the β-cells in the pancreatic
islets. It regulates the metabolism of carbohydrates by promoting the Obesity is a diabetogenic determinant that has an increasing world-
uptake of glucose from the blood by liver, fat and skeletal muscle cells. wide prevalence, especially in developing countries. While lifestyle
In T2D hyperglycemia is due to ineffective insulin response by the changes have driven obesity prevalence to epidemic proportions,
body's cells (known as insulin resistance) resulting in disrupted glucose genetic predisposition is a substantial contributor that together with
homeostasis. Therefore, and despite sufficient insulin in the blood- environmental influences significantly increase the risk of obesity,
stream, the target cells remain locked and do not let blood glucose insulin resistance and ultimately T2D.17,108,236-239 The genetic vari-
195-198
into the cells. Unfortunately, individuals with insulin resistance ants contributing to alternation in BMI, and hence obesity, have been
have built up a tolerance to insulin, making normal levels less effec- the subjects for several GWAS and provide a better understanding of
tive. As a result, more insulin is produced and released in greater the biological basis of obesity across different populations and ethnic-
quantities from the pancreas to persuade the target cells and tissues ities.131,240-246 The considerable correlation between obesity from
to take up and store glucose. Over time, the β-cells of the pancreatic one side and insulin resistance and β-cell dysfunction from the other
islets get exhausted (dysfunction) and can no longer sustain these side is fundamental to answering the obesity-diabetes association
higher rates of insulin production. Eventually, dysfunction of β-cells question and is expected to exacerbate the development of T2D and
result in higher blood glucose levels (prediabetes) and, ultimately, its severe complications much earlier than in non-obese individuals.
T2D.93,150-152,199-201 Evidently, the excess fat in the glucose target cells makes these cells
The importance of insulin resistance in the T2D etiology is well- less responsive to signals of insulin hormone, thus insulin resistance is
established and acknowledged as the most powerful early predictor more probably to occur in overweight and obese
93,202-206 153,155,200,247-250
and therapeutic target for T2D. There are several risk factors individuals.
that are assumed to play a role in developing insulin resistance includ- Table 2 lists many of the genetic variants that have been discov-
ing genetic predisposition, ethnicity, overweight/obesity, and ered through many genetic studies showing and association of BMI
aging.207-213 Some candidate genetic variants that make a person and T2D in different populations. A significant variant with a high
more or less probably to develop insulin resistance leading to T2D T2D association is rs9939609 located in the intronic region of the Fat
have been identified and confirmed in reproducible studies among dif- mass and obesity-associated (FTO) gene, imposing an allelic 0.39 kg/
ferent populations.214-218 m2 increase in BMI.251,252 Other polymorphisms in the FTO locus
In the pre-GWAS era the first genetic locus linked to T2D was were shown to be strongly associated with extreme childhood obe-
identified as a negative association with insulin resistance. Deeb et al, sity.181,243,251,253-256 Obesity and T2D association are largely
1998 identified through a candidate gene approach that a Pro12Ala described from epidemiological and pathophysiological perspectives.
polymorphism within the PPARG gene was associated with decreased Many genetic risk genes (FTO, MC4R, ADAMTS9, GRB14/COBLL1 and
10 SIRDAH AND READING

QPCTL/GIPR) identified in obesity and T2D association studies carry 4.1 | Personalized medicine in monogenic diabetes
only a limited “indirect” or modest risk.86,112,138,160,243,253,255,257
Monogenic forms of diabetes characterized by pancreatic β-cell dys-
function, such as MODY and neonatal diabetes (ND), are eminent
4 | PERSONALIZED MEDICINE IN examples of successful pharmacogenomics management that found a
DIABETES place in clinical practice and where personalized diabetic care has
already become a reality.269,270 There are 13 distinct types of MODY
Personalized medicine is a medical approach that stratify people or based on the dysfunctional gene causing the phenotype. The most
patients into different groups based on their specific characteristics, common type is MODY3 resulting from a mutation in HNF1A gene
clinical presentations, and/or genetic makeup, thus directing diagnos- that affects the transcription of multiple genes related to glucose
tic and/or therapeutic strategies toward most effective treatment out- metabolism, insulin secretion, and insulin production.269 MODY
comes. Although the term “personalized medicine,” interchangeably patients with HNF1A mutation have been found to be hypersensitive
precision medicine, individualized medicine or stratified medicine, has to sulfonylureas as antidiabetic drugs due to decreased expression of
become more practiced in the last few decades, the concept of tailor- HNF1A regulated genes in the liver, leading to decreased uptake of
ing of treatment to patients dates back to at least 2500 years ago, to sulfonylureas. Therefore, very low doses of short acting sulfonylureas
the time of Hippocrates, the Greek physician and the father of mod- has been recommended as initial treatment for MODY3 patients with
ern medicine. Hippocrates was practitioner and advocate of personal- poor glycemic control on an appropriate diet.270,271
ized medicine and provided written scripts about the individuality of On the other hand, ND was found to have a number of gene eti-
disease and the importance of giving different treatments to different ologies including, SNPs, chromosome duplication, and hyp-
patients.258 omethylation. The clinical symptoms are most commonly caused by
Personalized medicine has been considered a quantum leap from activating mutations in KCNJ11 or ABCC8 genes thus affecting the
the 20th century paradigm of Western or conventional medicine ATP-sensitive potassium channel in pancreatic β cells, resulting in
which was very useful for communicable diseases with a single etiol- diminished insulin secretion and absolute dependence on exogenous
ogy to medicine for the 21st century which not only maintains the insulin.269,272 High doses of sulfonylureas have been found to fix
progresses of the 20th century but also meritoriously handled these alternations in the ATP-sensitive potassium channels, thus regu-
etiology-complex disorders.2 Among the most crucial advances that late the release of insulin. Nowadays, ND patients with KCNJ11 or
favor the development of personalized medicine approach are GWAS ABCC8 mutations are being successfully managed with oral sulfonyl-
and massively parallel sequencing and other molecular technologies ureas instead injectable insulin. Sulfonylureas not only substitute for
which open the door for applying genetic-based risk assessment in the more expensive, more invasive insulin injections, but it also mini-
complex, non-monogenic disorders.143,259,260 Personalized medicine mizes the greater risk for hypoglycemic episodes in ND patients. Per-
approach not only provides superior care and improved welfare and sonalized monogenic diabetes is now stretching beyond therapeutic
health than other approaches, it has the potential to save money and concerns to provide future information about additional clinical fea-
alleviate economic burden of diseases on national health care sys- tures, complications, and probability of disease remission.273
tems.261-264 By practicing personalized medicine in the treatment of
diabetes, health professionals are individualizing health care and man-
agement plans based on a patient's susceptibility to disease and 4.2 | Personalized medicine in T2D
response to treatment, thus maximizing benefits and minimizing
harms.6,265 In the case of diabetes, where the estimated total annual The etiology, clinical presentation, and complications of T2D are
global healthcare expenditures was more than United States 720 bil- greatly varied among patients complicating prevention and manage-
lion dollars,23,27 the potential savings to individuals and economies are ment strategies. T2D remains effectively a diagnosis of exclusion. The
significant. hallmark feature of patients categorized as T2D, after excluding the
This DNA-centered view of personalized diabetes should not over- other more defined causative forms of diabetes, is chronic hyperglyce-
shadow other factors, such as environmental, lifestyle and behavioral mia.102 The roadmap in the T2D care protocol is based on remarkably
factors, that besides the genetic factors may affect the development of robust evidence and follows an algorithmic sequence that starts with
the symptoms.266,267 So far, personalized medicine has been success- lifestyle (diet and exercise) adjustments plus the generally well toler-
fully practiced in diabetes subtypes of simple etiology, monogenic dia- ated biguanide antihyperglycemic agent (metformin HCL) as the first
betes, and become part of the clinical tools used at advanced diabetes recommended pharmacological treatment. Later, the protocols may
institutions. Nevertheless, in diabetes subtypes with complex or multi- proceed to other drugs and/or insulin depending on the tolerability
factorial etiology interesting and promising leads are emerging despite and glycemic goals needed. Nevertheless, T2D patients have shown
some bridgeable challenges and gaps. With the ongoing technological variable responsiveness to drug treatment mainly due to genetic pre-
developments, smart utilization of big data, genomics and other disposition and makeup, with a noteworthy number of patients having
“-omics,” there is hope to make personalized medicine for complex eti- exhibited some degree of drug resistance that lessen intervention
ology diseases, for example, T2D, a reality in the near future.268 effectiveness.274-278 The benefits of lifestyle changes in diabetes
TABLE 2 Genetic risk variants associated with T2D through insulin action (resistance/low sensitivity) and/or adiposity

Gene/nearest
gene Name of Gene/nearest gene Genetic Variant Context location RA OA RAF OR P value Population Study design Reference
GCKR Glucokinase (hexokinase 4) regulator rs780094 Intron 2p23 C T 0.39 1.06 1.0E−09 European GWAS-meta-analysis 174,176
GCKR Glucokinase (hexokinase 4) regulator rs1260326 Exon/missense 2p23 C T 0.61 1.05 9.1E−10 European GWAS-meta-analysis 187
SIRDAH AND READING

RBMS1/ITGB6 RNA binding motif single stranded rs7593730 Intron 2q24.2 C T 0.82 1.11 4.0E−08 European GWAS 225
interacting protein 1/integrin subunit
beta 6
GRB14 Growth factor receptor bound protein 14 rs3923113 Intergenic 2q24.3 A C 0.74 1.09 1.0E−08 South Asian GWAS 183
IRS1 Insulin receptor substrate 1 rs7578326 Intron 2q36 A G 0.64 1.11 5.4E−20 European GWAS meta-analysis 178
CAPN10 Calcium-activated neutral proteinase 10 rs3792267 SNP-43 Intron 2q37.3 G A 0.88 1.38 0.03 African-Americans Meta-analysis 226
CAPN10 Calcium-activated neutral proteinase 10 rs3792267 SNP-43 Intron 2q37.3 G A 0.94 1.18 0.03 Asia Meta-analysis 227,228
CAPN10 Calcium-activated neutral proteinase 10 rs2975760 SNP-44 Intron 2q37.3 C T 0.16 1.1 0.0005 Multi ethnic Meta-analysis 229
ADAMTS9 ADAM metallopeptidase with rs4607103 Intron 3p14.1 C T 0.76 1.09 1.2E−08 European GWAS meta-analysis 172
thrombospondin type 1 motif, 9
PSMD6 Proteasome 26S subunit, non-ATPase 6 rs831571 Intergenic 3p14.1 C T 0.61 1.09 8.41E−11 East Asians GWAS meta-analysis 174,186
PPARG Peroxisome proliferator activated rs17036101 Intergenic 3p25.2 G A 0.93 1.15 2.0E−07 European GWAS meta-analysis 172
receptor gamma
PPARG Peroxisome proliferator activated rs1801282 Exon/missense 3p25.2 C G 0.88 1.16 6E−10 Multi ethnic GWAS-meta-analysis 173,174
receptor gamma
ANKRD55 Ankyrin repeat domain 55 rs459193 Intergenic 5q11.2 G A 0.70 1.08 6.0E−09 European GWAS meta-analysis 177
ANKRD55 Ankyrin repeat domain 55 rs9687833 Intron 5q11.2 A G 0.20 1.12 2.9E−09 Multi ethnic GWAS-meta-analysis 175
177
ARL15 ADP ribosylation factor like GTPase 15 rs702634 Intron 5q11.2 A G 0.76 1.06 6.9E−09 Multi ethnic GWAS-meta-analysis 84,173,174
ZBED3 Zinc finger BED-type containing 3 rs4457053 Intron 5q13.3 G A 0.26 1.08 2.8E−12 European GWAS meta-analysis 178
MIR129/LEP MicroRNA 129-1/Leptin rs791595 Intron 7q32.1 A G 0.08 1.17 2.6E−13 East Asian GWAS 188
KLF14 Krüppel-like factor 14 rs972283 Intergenic 7q32.2 G A 0.55 1.07 2.2E−10 European GWAS meta-analysis 178
PTPRD Protein tyrosine phosphatase receptor rs17584499 Intron 9p24.1-p23 T C 0.06 1.57 8.5E−10 Han Chinese GWAS 194
type D
GRK5 G protein-coupled receptor kinase 5 rs10886471 Intron 10q26.11 C T 0.78 1.12 7.0E−09 Han Chinese GWAS 193
HMGA2 High mobility group AT-hook 2 rs1531343 Intron 12q14.3 C G 0.10 1.1 3.6E−09 European GWAS meta-analysis 178
IGF1 Insulin like growth factor 1 rs35767 Near gene 5 12q23.2 G A 0.85 1.04 2.2E−09 European GWAS. meta-analysis 176
HNF1A HNF1 homeobox A rs7957197 Intron 12q24.31 T A 0.85 1.07 2.4E−08 European GWAS meta-analysis 178
FTO Fat mass and obesity-associated gene rs8050136 Intron 16q12.2 A C 0.38 1.17 1.3E−12 European GWAS 181,190
SLC16A11/A13 Solute carrier family 16 member 11/13 rs312457 Intron 17p13.1 G A 0.08 1.20 7.7E−13 East Asian GWAS 188
MC4R Melanocortin 4 receptor. rs12970134 Intergenic 18q21.32 A G 0.27 1.08 1.2E−08 European GWAS meta-analysis 177
BCL2 BCL2 apoptosis regulator rs12454712 Intron 18q21.33 T C 0.63 1.09 2.1E−08 multiethnic GWAS meta-analysis 230
CILP2 Cartilage intermediate layer protein 2 rs10401969 Intron 19p13.11 C T 0.08 1.13 7.0E−09 European GWAS meta-analysis 84,173,174
11

(Continues)
12 SIRDAH AND READING

prevention and effectiveness of several antihyperglycemic drugs are

84,173,174
84,173,174
84,173,174
Reference
counterbalanced by the globally continuing increasing prevalence of
T2D.102 This may justifies the necessity for designing concomitant
prevention and an appropriate pharmacological treatment strategies
including personalized approaches to enhanced tolerability and
effectiveness.100,279-283
GWAS meta-analysis

GWAS meta-analysis

Based on the success in applying personalized approaches to


Study design

treating monogenic diabetes, T2D with its complex polygenic multi-


factorial etiology, could be only a few steps away from successful
GWAS

implementations. Stratification of T2D patients into multiple subtypes


is a major changeling issue for personalized care (Figure 2). Because
patients suffering from pancreatic islet β-cell dysfunction are unlikely
to respond to therapeutics that enhance insulin secretion identifying
East Asians
Population

South Asia
European

the mechanistic pathway of the T2D pathology is essential for more


tailored treatment choices.
0.56 1.10 1.3E−08
0.29 1.09 2.6E−10
0.79 1.27 3.0E−10
P value

4.2.1 | Pharmacogenomics in T2D


RA OA RAF OR

Pharmacogenomics applied to the T2D management is making


advancements in understanding the interaction between genetics and
molecular mechanisms of T2D drugs. Several studies have described
G
G
G

the impact of polymorphisms in important genes for sulfonylurea


metabolism and the sulfonylurea receptor, β-cell function, and insulin
A
A
A

action on the effectiveness of sulphonylureas as an antidiabetic drugs.


19q13.11
20q13.12
location

These include: ABCC8, KCNJ11, TCF7L2, CYP2C9, IRS1, CDKAL1,


Xq28

CDKN2A, CDKN2B, KCNQ1, NOS1AP and CAPN10 genes273 Similarly,


the influences of other T2D associated polymorphism on the effi-
ciency of other antidiabetic drugs such as biguanides and
Intergenic
Context

thiazolidinediones, as well as lipid-lowering drugs have been men-


Intron
Intron

tioned in several studies to include common variants in genes like


Abbreviations: RA, risk allele; RAF, risk allele frequency; OA, other allele; OR, odds ratio.

SLC22A1 and PPARG.284-290


Genetic Variant
rs3786897
rs4812829
rs5945326

4.2.2 | Polygenic risk score

In this era of advanced GWAS and other “omics” big data researchers
have identified about 400 distinct genetic signals influencing T2D risk
with a growing number of risk-alleles with lower frequency and small
effect size (modest effect on risk). However, when considered in com-
Hepatocyte nuclear factor 4 alpha

bination, the impact of these risk alleles could be significant and offer
dual-specificityphosphatase-9
Name of Gene/nearest gene

the capability to capture information on individual patterns of disease


predisposition.116,291 Expressed as a polygenic risk score these aggre-
gated genetic risks can identify individuals with a high future risk of
diabetes. The polygenic risk score is probabilistic rather than an
Peptidase D

evidence-based model and most suitably used as a predictor to quan-


(Continued)

tify a discrete increment in overall risk for disease. Although polygenic


risk score approach had been used for three T2D associated genetic
variants (KCNJ11, PPARG and TCF7L2) previously,292 the utilization of
Gene/nearest

this approach becomes more reliable in the GWAS, “omics,” and big
TABLE 2

data era.
HNF4A
DUSP9
PEPD
gene

As a part of the personalized medicine approach, different large


scale studies from various populations and ethnicities (European, and
SIRDAH AND READING 13

F I G U R E 2 Conventional and
personalized treatment approaches
[Colour figure can be viewed at
wileyonlinelibrary.com]

non-European) have emphasized the success of polygenic risk scores Such sexual variations in T2D predisposition justifies the need for
to identify individuals or populations at high risk to develop sex-specific approaches in conventional and personalized diabetes
T2D.116,291,293,294 Polygenic risk scores have also been used in differ- management.299
ential diagnosis in those with diabetes, providing a growing optimism
for offering significant clinical advantages and reducing morbidity and
mortality associated with diabetes.116 In their recent study, Ahlqvist 4.2.4 | Phenome-wide association studies
and coworkers illustrated the superiority of genetic risk score over
classical diabetes classifications in clustering, stratification and differ- Very recently a new genotype-to-phenotype approach known as
entiation of adult-onset diabetic cases into five distinct clusters that phenome-wide association study (PheWAS) has been developed to
include T1D and T2D novel subgroups, and its importance in enhanc- statistically estimate the association between single-nucleotide poly-
ing and promoting precision medicine in diabetes. The researchers morphisms and many different phenotypes.300 This approach is not a
also showed that clustering based on genetic risk score central to the substitution of phenotype-to-genotype GWAS approach, but could
development of diabetes successfully identified patients at high risk work complement GWAS as a reliable tools to advance personalized
of diabetic complications at diagnosis and provided information about Medicine300-302 This PheWAS approach has re-validatedprior-known
the pathological mechanistic pathway, thereby guiding choice of genetic variants and diabetes associations,303 and appears to have
295
therapy. application in understanding the complexities of many polygenic dis-
As rare variants with small effect sizes are difficult to identify orders.300 Through this auxiliary PheWAS approach, further crucial
296
using GWAS, the prediction value of polygenic risk scores derived steps in personalized diabetes could be achieved.
from GWAS could be complementary augmented by data from
whole-genome or whole-exome sequencing.297
5 | CHALLENGES AND LIMITATIONS

4.2.3 | Sexual dimorphism in T2D 5.1 | Genome wide association studies

Adding to the complex etiology gender has been shown to play a role Despite the contribution of GWAS in developing genomics to its
in T2D. T2D has a higher occurrence in males than females, with dif- understanding of genetic risk for a growing number of complex poly-
fering manifestations in predisposition, glucose metabolism and pro- genic disorders including T2D, there are many challenges and limita-
gression of diabetic complications. Implementation of personalized tions associated with this technique and the generated data. These
management for diabetes could not be successful without considering include: small effect sizes of associations, difficulty in determining true
the sexual dimorphic issues of this metabolic disorder. Although the causal associations, conducting studies in highly admixed multi-ethnic
process of sexual dimorphism is not fully understood, gender specific- populations, heterogeneity of subjects in multicenter studies of very
ities in neonatal development, epigenetic modification, and the basic large sample size, insufficient sample size, and a lack of well-defined
disparities in sex hormones are possible inflecting factors. Recently, case and control groups (specifically for population stratifica-
the effect of T2D associated allele, Kruppel Like Factor 14 (KLF14) on tion).296,304,305 In addition, due to the steep infrastructural require-
pre-adipocyte proliferation, lipogenesis disruption, and body fat distri- ments this technique is not ubiquitously available in areas that are
298
bution were found to be female specific. In addition, human and dealing with T2D.143 We cannot ignore ethical concerns306 as well as
animal model experiments elucidated on the female-specific protec- the financial, technological requirements, which are extremely costly
tion of endogenous Estrogen from T2D during their childbearing age. and hardly available for many research institutions worldwide.296,307
14 SIRDAH AND READING

5.2 | Personalized medicine in T2D specially in low and middle income countries. Ultimately, local and
global ethical legislations should be laid down to protect the rights
Increasing application of personalized medicine based on individual and privacy of the individuals' genetic records from being abused
genetic and phenotypic profiles has provided promising medical worldwide.
advantages for patient care and disease management. However, fur-
ther steps are necessary to integrate our current understanding of AC KNOWLEDG EME NT S
complex disease architectures into a personalized medicine approach. Dr. Mahmoud Sirdah would like to thank Dr. Josef T. Prchal of the
In the context of T2D diagnosis and treatment there are some chal- Huntsman Cancer Institute, University of Utah, for hosting him as a
lenges and limitations of the personalized medicine approach should visiting scholar during his sabbatical leave. Also the authors would like
be taken into consideration. Many of which could impact the individ- to thank Dr. Saleh N. Mwafy for his fine touches on the sketches.
uals, healthcare systems, economy and society. These challenges and
limitations include: the heterogeneity of T2D, ethical concerns about CONFLIC T OF INT ER E ST
individual privacy and confidentiality, handling of genomic information The authors declare no conflict of interest related to this work.
by insurance companies, quality and standardizations of phenotypic
data, stratification and genetic discrimination based on ethnicity, and DATA AVAILABILITY STAT EMEN T
financial factors in areas with limited resources.101,273,308-310 Data sharing is not applicable to this article as no new data were cre-
Despites the remarkable success of personalized approach in the ated or analyzed in this study.
management of monogenic forms of diabetes the great heterogeneity
of the multifactorial T2D remains a challenge in defining etiological OR CID
Mahmoud M. Sirdah https://orcid.org/0000-0002-1146-5570
subgroups, refining the diagnostic and therapeutic pathways for opti-
N. Scott Reading https://orcid.org/0000-0003-0806-5661
mal management. There are a growing number of presumptive phar-
macogenomics associations, however, only robustly replicated
RE FE RE NCE S
associations from larger prospective studies in ethnically diverse
1. Fokunang CN, Ndikum V, Tabi OY, et al. Traditional medicine: past,
populations across a range of age group should be exploited. In this
present and future research and development prospects and integra-
manner will we bridge the gap in clinical trial information regarding
tion in the National Health System of Cameroon. Afr J Tradit Comple-
T2D management in youth, adolescents, elderly, and pregnant ment Altern Med. 2011;8(3):284-295. https://doi.org/10.4314/
women.273 Notwithstanding of these challenges and limitations, the ajtcam.v8i3.65276.
technological developments in diagnosis and analytical tools are 2. Whitcomb DC. What is personalized medicine and what should it
replace? Nat Rev Gastroenterol Hepatol. 2012;9(7):418-424. https://
expected to enhance our understanding of T2D heterogeneity and
doi.org/10.1038/nrgastro.2012.100.
broaden our capabilities to apply them to personalized medicine. 3. Croft P, Altman DG, Deeks JJ, et al. The science of clinical practice:
disease diagnosis or patient prognosis? Evidence about “what is
likely to happen” should shape clinical practice. BMC Med. 2015;13:
20. https://doi.org/10.1186/s12916-014-0265-4.
6 | C O N CL U S I O N 4. Steele FR. Personalized medicine: something old, something new.
Pers Med. 2009;6(1):1-5. https://doi.org/10.2217/17410541.6.1.1.
Personalized medicine shows promise in providing cost savings and 5. Korf BR. Genetics and medical practice: new approaches to “old” dis-
efficiencies in the treatment and management of T2D. Genetic variant orders. Curr Opin Pediatr. 2002;14(6):688-690. https://doi.org/10.
1097/00008480-200212000-00007.
identification on the individual level and understanding their influence
6. Vogenberg FR, Isaacson Barash C, Pursel M. Personalized medicine:
in the predisposition and pathogenesis of T2D would be a critical part 1: evolution and development into theranostics. P T. 2010;35
advance in disease management improving the clinical condition and (10):560-576. http://www.ncbi.nlm.nih.gov/pubmed/21037908.
precluding complications. In our review we have identified the current 7. Offit K. Personalized medicine: new genomics, old lessons. Hum Genet.
2011;130(1):3-14. https://doi.org/10.1007/s00439-011-1028-3.
state of genetic risk variants associated with T2D and shown the
8. Milunsky A. The “new genetics” in clinical practice: a brief primer.
importance pharmacogenomics studies have in associating actionable J Am Board Fam Med. 2017;30(3):377-379. https://doi.org/10.3122/
relationships between genetic and pharmacologic treatments. It is our jabfm.2017.03.160316.
desire to forward the advance of personalized approaches toward 9. Tutton R. Personalizing medicine: futures present and past. Soc Sci
Med. 2012;75(10):1721-1728. https://doi.org/10.1016/j.socscimed.
management of this growing diabetic epidemic.
2012.07.031.
We recommend that all involved stakeholders should start 10. Dirette DP. Personalized medicine: definitions, history, and implica-
national and international preparedness plans for establishing and tions for the OT profession. Open J Occup Ther. 2015;3(4):8. https://
strengthening the infrastructure necessary for promoting T2D person- doi.org/10.15453/2168-6408.1213.
alized medicine. The ongoing technological advances are the corner- 11. de Leon J. Pharmacogenomics: the promise of personalized medicine
for CNS disorders. Neuropsychopharmacology. 2009;34(1):159-172.
stones for developing the data to improve our understanding of the
https://doi.org/10.1038/npp.2008.147.
etiology and architecture of T2D and translate those discoveries into 12. Mini E, Nobili S. Pharmacogenetics: implementing personalized med-
more effective prevention and therapeutic strategies. The economic icine. Clin Cases Miner Bone Metab. 2009;6(1):17-24. http://www.
burden of implementing this approach should be considered very well ncbi.nlm.nih.gov/pubmed/22461093.
SIRDAH AND READING 15

13. Redekop WK, Mladsi D. The faces of personalized medicine: a 34. Lakhtakia R. The history of diabetes mellitus. Sultan Qaboos Univ
framework for understanding its meaning and scope. Value Health. Med J. 2013;13(3):368-370. https://doi.org/10.12816/
2013;16(6 Suppl):S4-S9. https://doi.org/10.1016/j.jval.2013. 0003257.
06.005. 35. Goldman R, Zajac J, Shrestha A, Patel P, Poretsky L. The Main events
14. Hayes DF, Markus HS, Leslie RD, Topol EJ. Personalized medicine: in the history of diabetes mellitus. In: Poretsky L, ed. Principles of
risk prediction, targeted therapies and mobile health technology. Diabetes Mellitus. 3rd ed. New York: Springer International Publish-
BMC Med. 2014;12:37. https://doi.org/10.1186/1741-7015-12-37. ing AG; 2017. https://doi.org/10.1007/978-3-319-18741-9.
15. Cataloguing WL. Global Report on Diabetes. Vol 978. Geneva, Swit- 36. Gregory R. Book review: the pissing evil, a comprehensive history of
zerland: World Health Organization; 2016. http://www.who.int/ diabetes. Br J Diabetes. 2018;18:1-2.
about/licensing/. 37. Bradwel S. The pissing evil: a comprehensive history of diabetes
16. Ginter E, Simko V. Type 2 diabetes mellitus, pandemic in 21st cen- mellitus. Soc Hist Med. 2019;32(3):3-654. https://doi.org/10.1093/
tury. Adv Exp Med Biol. 2012;771:42-50. https://doi.org/10.1007/ shm/hkz036.
978-1-4614-5441-0_6. 38. Organization WH. Classification of Diabetes Mellitus. Geneva: World
17. Zimmet PZ. Diabetes and its drivers: the largest epidemic in human Health Organization; 2019. https://apps.who.int/iris/rest/
history? Clin Diabetes Endocrinol. 2017;3:1. https://doi.org/10.1186/ bitstreams/1233344/retrieve.
s40842-016-0039-3. 39. Andersson T, Ahlbom A, Carlsson S. Diabetes prevalence in Sweden
18. Standards of medical care in diabetes- -2014. Diabetes Care. 2014; at present and projections for year 2050. PLoS ONE. 2015;10(11):
37(Suppl 1):S14-S80. https://doi.org/10.2337/dc14-S014. e0143084. https://doi.org/10.1371/journal.pone.0143084.
19. Beckman JA, Creager MA. Vascular complications of diabetes. Circ 40. Hussen HI, Yang D, Cnattingius S, Moradi T. Type I diabetes among
Res. 2016;118(11):1771-1785. https://doi.org/10.1161/ children and young adults: the role of country of birth, socioeco-
CIRCRESAHA.115.306884. nomic position and sex. Pediatr Diabetes. 2013;14(2):138-148.
20. Kosiborod M, Gomes MB, Nicolucci A, et al. Vascular complications https://doi.org/10.1111/j.1399-5448.2012.00904.x.
in patients with type 2 diabetes: prevalence and associated factors 41. Saraswathi S, Al-Khawaga S, Elkum N, Hussain K. A systematic
in 38 countries (the DISCOVER study program). Cardiovasc Diabetol. review of childhood diabetes research in the middle east region.
2018;17(1):150. https://doi.org/10.1186/s12933-018-0787-8. Front Endocrinol. 2019;10:805. https://doi.org/10.3389/fendo.2019.
21. Ogurtsova K, da Rocha Fernandes JD, Huang Y, et al. IDF diabetes 00805.
Atlas: global estimates for the prevalence of diabetes for 2015 and 42. Hakkarainen P, Sund R, Arffman M, et al. Working people with type
2040. Diabetes Res Clin Pr. 2017;128:40-50. https://doi.org/10. 1 diabetes in the Finnish population. BMC Public Health. 2017;17(1):
1016/j.diabres.2017.03.024. 805. https://doi.org/10.1186/s12889-017-4723-8.
22. Hu FB. Globalization of diabetes: the role of diet, lifestyle, and 43. Ji J, Hemminki K, Sundquist J, Sundquist K. Ethnic differences in
genes. Diabetes Care. 2011;34(6):1249-1257. https://doi.org/10. incidence of type 1 diabetes among second-generation immigrants
2337/dc11-0442. and adoptees from abroad. J Clin Endocrinol Metab. 2010;95(2):847-
23. IDF, Federation ID. IDF DIABETES ATLAS; Ninth Edition. 9th ed. 850. https://doi.org/10.1210/jc.2009-1818.
Brussels, Belgium: International Diabetes Federation; 2019. 44. Maahs DM, West NA, Lawrence JM, Mayer-Davis EJ. Epidemiology
24. (PAHO/WHO) PAHO-WRHO. Health Status Of The Population: of type 1 diabetes. Endocrinol Metab Clin N Am. 2010;39(3):481-497.
Noncommunicable Disease Prevention and Control. 2017. https://doi.org/10.1016/j.ecl.2010.05.011.
25. National Diabetes Statistics Report, 2017. Estimates of Diabetes and 45. Atkinson MA, Eisenbarth GS, Michels AW. Type 1 diabetes. Lancet.
its Burden in the United States. Atlanta, GA, USA: National Center for 2014;383(9911):69-82. https://doi.org/10.1016/S0140-6736(13)
Chronic Disease Prevention and Health Promotion: Division of Dia- 60591-7.
betes Translation; 2017. 46. Dzidzonu DK, Skrivarhaug T, Joner G, Moger TA. Ethnic differences
26. Organization WH, Organization WH. Global Status Report on Non- in the incidence of type 1 diabetes in Norway: a register-based study
communicable Diseases 2014. Geneva: World Health Organization; using data from the period 2002-2009. Pediatr Diabetes. 2016;17(5):
2014. 337-341. https://doi.org/10.1111/pedi.12294.
27. Saeedi P, Petersohn I, Salpea P, et al. Global and regional diabetes 47. Rogers MAM, Kim C, Banerjee T, Lee JM. Fluctuations in the inci-
prevalence estimates for 2019 and projections for 2030 and 2045: dence of type 1 diabetes in the United States from 2001 to 2015: a
results from the international diabetes federation diabetes Atlas, 9 longitudinal study. BMC Med. 2017;15(1):199. https://doi.org/10.
(th) edition. Diabetes Res Clin Pr. 2019;157:107843. https://doi.org/ 1186/s12916-017-0958-6.
10.1016/j.diabres.2019.107843. 48. Mayer-Davis EJ, Lawrence JM, Dabelea D, et al. Incidence trends of
28. Stokes A, Preston SH. The contribution of rising adiposity to the type 1 and type 2 diabetes among youths, 2002-2012. N Engl J Med.
increasing prevalence of diabetes in the United States. Prev Med. 2017;376(15):1419-1429. https://doi.org/10.1056/NEJMoa1610187.
2017;101:91-95. https://doi.org/10.1016/j.ypmed.2017.05.031. 49. Patterson CC, Karuranga S, Salpea P, et al. Worldwide estimates of
29. 2. Classification and diagnosis of diabetes: standards of medical Care incidence, prevalence and mortality of type 1 diabetes in children
in Diabetes-2019. Diabetes Care. 2019;42(Suppl 1):S13-S28. https:// and adolescents: results from the international diabetes federation
doi.org/10.2337/dc19-S002. diabetes Atlas, 9th edition. Diabetes Res Clin Pract. 2019;157:
30. Karamanou M, Protogerou A, Tsoucalas G, Androutsos G, Poulakou- 107842. https://doi.org/10.1016/j.diabres.2019.107842.
Rebelakou E. Milestones in the history of diabetes mellitus: the main 50. Spanakis EK, Golden SH. Race/ethnic difference in diabetes and dia-
contributors. World J Diabetes. 2016;7(1):1-7. https://doi.org/10. betic complications. Curr Diab Rep. 2013;13(6):814-823. https://doi.
4239/wjd.v7.i1.1. org/10.1007/s11892-013-0421-9.
31. Ahmed AM. History of diabetes mellitus. Saudi Med J. 2002;23(4): 51. Diaz-Valencia PA, Bougneres P, Valleron AJ. Global epidemiology of
373-378. http://www.ncbi.nlm.nih.gov/pubmed/11953758. type 1 diabetes in young adults and adults: a systematic review.
32. Loriaux DL. Diabetes and the Ebers papyrus 1552 B.C. Endocrinolo- BMC Public Health. 2015;15:255. https://doi.org/10.1186/s12889-
gist. 2006;16(2):1. 015-1591-y.
33. Das AK, Shah S. History of diabetes: from ants to analogs. J Assoc 52. Rich SS. Genetics and its potential to improve type 1 diabetes care.
Physicians India. 2011;59(Suppl 59):6-7. http://www.ncbi.nlm.nih. Curr Opin Endocrinol Diabetes Obes. 2017;24:279-284. https://doi.
gov/pubmed/21818991. org/10.1097/MED.0000000000000347.
16 SIRDAH AND READING

53. Onengut-Gumuscu S, Chen WM, Burren O, et al. Fine mapping of 72. Buchanan TA, Xiang AH, Page KA. Gestational diabetes mellitus:
type 1 diabetes susceptibility loci and evidence for colocalization of risks and management during and after pregnancy. Nat Rev
causal variants with lymphoid gene enhancers. Nat Genet. 2015;47: Endocrinol. 2012;8(11):639-649. https://doi.org/10.1038/nrendo.
381-386. https://doi.org/10.1038/ng.3245. 2012.96.
54. Chatterjee S, Khunti K, Davies MJ. Type 2 diabetes. Lancet. 2017; 73. Jarvela IY, Juutinen J, Koskela P, et al. Gestational diabetes iden-
389(10085):2239-2251. https://doi.org/10.1016/S0140-6736(17) tifies women at risk for permanent type 1 and type 2 diabetes in
30058-2. fertile age: predictive role of autoantibodies. Diabetes Care. 2006;
55. Martin-Timon I, Sevillano-Collantes C, Segura-Galindo A, Del 29(3):607-612. https://doi.org/10.2337/diacare.29.03.06.dc05-
Canizo-Gomez FJ. Type 2 diabetes and cardiovascular disease: have 1118.
all risk factors the same strength? World J Diabetes. 2014;5(4):444- 74. Al-Hakeem MM. Pregnancy outcome of gestational diabetic mothers:
470. https://doi.org/10.4239/wjd.v5.i4.444. experience in a tertiary center. J Fam Community Med. 2006;13(2):55-
56. Malandrino N, Smith RJ. Personalized medicine in diabetes. Clin 59. http://www.ncbi.nlm.nih.gov/pubmed/23012105.
Chem. 2011;57(2):231-240. https://doi.org/10.1373/clinchem. 75. Forlenza GP, Moran A, Nathan B. Other specific types of diabe-
2010.156901. tes. In: Cowie CC, Menke A, Cissell MA, et al., eds. Diabetes in
57. Nair AK, Baier LJ. Complex genetics of type 2 diabetes and effect America. 3rd ed.National Institutes of Health: Bethesda, MD;
size: what have we learned from isolated populations? Rev Diabet 2018.
Stud. 2015;12(3–4):299-319. https://doi.org/10.1900/RDS.2015. 76. Mayer-Davis EJ, Kahkoska AR, Jefferies C, et al. ISPAD clinical prac-
12.299. tice consensus guidelines 2018: definition, epidemiology, and classi-
58. Lyssenko V, Laakso M. Genetic screening for the risk of type 2 diabe- fication of diabetes in children and adolescents. Pediatr Diabetes.
tes: worthless or valuable? Diabetes Care. 2013;36(Suppl 2):S120- 2018;19(Suppl 2):7-19. https://doi.org/10.1111/pedi.12773.
S126. https://doi.org/10.2337/dcS13-2009. 77. Bullard KM, Cowie CC, Lessem SE, et al. Prevalence of diagnosed
59. Ali O. Genetics of type 2 diabetes. World J Diabetes. 2013;4(4):114- diabetes in adults by diabetes type - United States, 2016. MMWR
123. https://doi.org/10.4239/wjd.v4.i4.114. Morb Mortal Wkly Rep. 2018;67(12):359-361. https://doi.org/10.
60. Mahajan A, Taliun D, Thurner M, et al. Fine-mapping type 2 diabetes 15585/mmwr.mm6712a2.
loci to single-variant resolution using high-density imputation and 78. Feingold KR. Atypical forms of diabetes. In: Feingold KR, Anawalt B,
islet-specific epigenome maps. Nat Genet. 2018;50(11):1505-1513. Boyce A, et al., eds. Endotext. South Dartmouth, MA: MDText.com,
https://doi.org/10.1038/s41588-018-0241-6. Inc.; 2000. http://www.ncbi.nlm.nih.gov/pubmed/25905351.
61. Billings LK, Florez JC. The genetics of type 2 diabetes: what have we 79. Resmini E, Minuto F, Colao A, Ferone D. Secondary diabetes associ-
learned from GWAS? Ann N Y Acad Sci. 2010;1212:59-77. https:// ated with principal endocrinopathies: the impact of new treatment
doi.org/10.1111/j.1749-6632.2010.05838.x. modalities. Acta Diabetol. 2009;46(2):85-95. https://doi.org/10.
62. Marouli E, Kanoni S, Mamakou V, et al. Evaluating the glucose rais- 1007/s00592-009-0112-9.
ing effect of established loci via a genetic risk score. PLoS ONE. 80. Filippi CM, von Herrath MG. Viral trigger for type 1 diabetes: pros
2017;12(11):e0186669. https://doi.org/10.1371/journal.pone. and cons. Diabetes. 2008;57(11):2863-2871. https://doi.org/10.
0186669. 2337/db07-1023.
63. Xue A, Wu Y, Zhu Z, et al. Genome-wide association analyses iden- 81. Tabak AG, Herder C, Rathmann W, Brunner EJ, Kivimaki M. Predia-
tify 143 risk variants and putative regulatory mechanisms for type betes: a high-risk state for diabetes development. Lancet. 2012;379
2 diabetes. Nat Commun. 2018;9(1):2941. https://doi.org/10.1038/ (9833):2279-2290. https://doi.org/10.1016/S0140-6736(12)
s41467-018-04951-w. 60283-9.
64. Coustan DR, Carpenter MW. The diagnosis of gestational diabetes. 82. DeJesus RS, Breitkopf CR, Rutten LJ, Jacobson DJ, Wilson PM,
Diabetes Care. 1998;21(Suppl 2):B5-B8. http://www.ncbi.nlm.nih. Sauver JS. Incidence rate of prediabetes progression to diabetes:
gov/pubmed/9704220. modeling an optimum target group for intervention. Popul Health
65. Ruiz-Palacios M, Ruiz-Alcaraz AJ, Sanchez-Campillo M, Larque E. Manag. 2017;20(3):216-223. https://doi.org/10.1089/pop.2016.
Role of insulin in placental transport of nutrients in gestational dia- 0067.
betes mellitus. Ann Nutr Metab. 2017;70(1):16-25. https://doi.org/ 83. Yudkin JS. “Prediabetes”: are there problems with this label? Yes,
10.1159/000455904. the label creates further problems! Diabetes Care. 2016;39(8):1468-
66. Dean L, McEntyre J. The Genetic Landscape of Diabetes. Bethesda 1471. https://doi.org/10.2337/dc15-2113.
(MD): National Center for Biotechnology Information (US); 2004. 84. Prasad RB, Groop L. Genetics of type 2 diabetes-pitfalls and possibil-
67. Herath H, Herath R, Wickremasinghe R. Gestational diabetes ities. Genes (Basel). 2015;6(1):87-123. https://doi.org/10.3390/
mellitus and risk of type 2 diabetes 10 years after the index preg- genes6010087.
nancy in Sri Lankan women-a community based retrospective cohort 85. van Tilburg J, van Haeften TW, Pearson P, Wijmenga C. Defining
study. PLoS ONE. 2017;12(6):e0179647. https://doi.org/10.1371/ the genetic contribution of type 2 diabetes mellitus. J Med Genet.
journal.pone.0179647. 2001;38(9):569-578. https://doi.org/10.1136/jmg.38.9.569.
68. Madhu SV. Diabetes in pregnancy—a critical window of opportunity. 86. Ingelsson E, McCarthy MI. Human genetics of obesity and type 2 dia-
Int J Diabetes Dev Ctries. 2018;38(1):3. betes mellitus: past, present, and future. Circ Genom Precis Med.
69. Federico C, Pridjian G. An overview of gestational diabetes. In: 2018;11(6):e002090. https://doi.org/10.1161/CIRCGEN.118.
Nair DB, ed. Nutritional and Therapeutic Interventions for Diabetes 002090.
and Metabolic Syndrome. Cambridge, MA: Elsevier B.V; 2018. 87. Manne-Goehler J, Siedner MJ, Montana L, et al. Hypertension and
https://doi.org/10.1016/C2016-0-01904-6. diabetes control along the HIV care cascade in rural South Africa.
70. Sacks DB, Coustan DR, Cundy T, Donovan L, Hod M. Gestational J Int AIDS Soc. 2019;22(3):e25213. https://doi.org/10.1002/jia2.
diabetes mellitus: why the controversy? Clin Chem. 2018;64(3):431- 25213.
438. https://doi.org/10.1373/clinchem.2016.266577. 88. Dunachie S, Chamnan P. The double burden of diabetes and global
71. Baptiste-Roberts K, Barone BB, Gary TL, et al. Risk factors for type infection in low and middle-income countries. Trans R Soc Trop Med
2 diabetes among women with gestational diabetes: a systematic Hyg. 2019;113(2):56-64. https://doi.org/10.1093/trstmh/try124.
review. Am J Med. 2009;122(3):207-214 e4. https://doi.org/10. 89. Dagenais GR, Gerstein HC, Zhang X, et al. Variations in diabetes
1016/j.amjmed.2008.09.034. prevalence in low-, middle-, and high-income countries: results from
SIRDAH AND READING 17

the prospective urban and rural epidemiological study. Diabetes Care. 108. Thibault V, Belanger M, LeBlanc E, et al. Factors that could
2016;39(5):780-787. https://doi.org/10.2337/dc15-2338. explain the increasing prevalence of type 2 diabetes among adults
90. Narayan KM, Rhodes EC. Addressing noncommunicable diseases in in a Canadian province: a critical review and analysis. Diabetol
primary care: the case of type 2 diabetes. J R Coll Physicians Edinb. Metab Syndr. 2016;8:71. https://doi.org/10.1186/s13098-016-
2016;46(4):272-277. https://doi.org/10.4997/JRCPE.2016.414. 0186-9.
91. Shirinzadeh M, Afshin-Pour B, Angeles R, Gaber J, Agarwal G. The 109. Mohlke KL, Boehnke M. Recent advances in understanding the
effect of community-based programs on diabetes prevention in low- genetic architecture of type 2 diabetes. Hum Mol Genet. 2015;24
and middle-income countries: a systematic review and meta-analy- (R1):R85-R92. https://doi.org/10.1093/hmg/ddv264.
sis. Glob Health. 2019;15(1):10. https://doi.org/10.1186/s12992- 110. Grant RW, Moore AF, Florez JC. Genetic architecture of type 2 dia-
019-0451-4. betes: recent progress and clinical implications. Diabetes Care. 2009;
92. Lin Y, Sun Z. Current views on type 2 diabetes. J Endocrinol. 2010; 32(6):1107-1114. https://doi.org/10.2337/dc08-2171.
204(1):1-11. https://doi.org/10.1677/JOE-09-0260. 111. Jonsson A, Ladenvall C, Ahluwalia TS, et al. Effects of common
93. Cantley J, Ashcroft FM. Q&A: insulin secretion and type 2 diabetes: genetic variants associated with type 2 diabetes and glycemic
why do beta-cells fail? BMC Biol. 2015;13:33. https://doi.org/10. traits on alpha- and beta-cell function and insulin action in
1186/s12915-015-0140-6. humans. Diabetes. 2013;62(8):2978-2983. https://doi.org/10.
94. Colberg SR, Sigal RJ, Fernhall B, et al. Exercise and type 2 diabetes: 2337/db12-1627.
the American College of Sports Medicine and the American Diabe- 112. Sun X, Yu W, Hu C. Genetics of type 2 diabetes: insights into the
tes Association: joint position statement executive summary. Dia- pathogenesis and its clinical application. Biomed Res Int. 2014;2014:
betes Care. 2010;33(12):2692-2696. https://doi.org/10.2337/ 926713-926715. https://doi.org/10.1155/2014/926713.
dc10-1548. 113. Gan W, Bragg F, Walters RG, et al. Genetic predisposition to type
95. Kaku K, Daida H, Kashiwagi A, et al. Long-term effects of 2 diabetes and risk of subclinical atherosclerosis and cardiovascular
pioglitazone in Japanese patients with type 2 diabetes without a diseases among 160,000 Chinese adults. Diabetes. 2019;68(11):
recent history of macrovascular morbidity. Curr Med Res Opin. 2009; 2155-2164. https://doi.org/10.2337/db19-0224.
25(12):2925-2932. https://doi.org/10.1185/03007990903328124. 114. Hara K, Shojima N, Hosoe J, Kadowaki T. Genetic architecture of
96. Wu Y, Ding Y, Tanaka Y, Zhang W. Risk factors contributing to type type 2 diabetes. Biochem Biophys Res Commun. 2014;452(2):213-
2 diabetes and recent advances in the treatment and prevention. Int 220. https://doi.org/10.1016/j.bbrc.2014.08.012.
J Med Sci. 2014;11(11):1185-1200. https://doi.org/10.7150/ijms. 115. Fuchsberger C, Flannick J, Teslovich TM, et al. The genetic architec-
10001. ture of type 2 diabetes. Nature. 2016;536(7614):41-47. https://doi.
97. Sigal RJ, Kenny GP, Wasserman DH, Castaneda-Sceppa C. Physical org/10.1038/nature18642.
activity/exercise and type 2 diabetes. Diabetes Care. 2004;27(10): 116. Udler MS, McCarthy MI, Florez JC, Mahajan A. Genetic risk scores
2518-2539. https://doi.org/10.2337/diacare.27.10.2518. for diabetes diagnosis and precision medicine. Endocr Rev. 2019;40:
98. Chia CW, Egan JM, Ferrucci L. Age-related changes in glucose 1500-1520. https://doi.org/10.1210/er.2019-00088.
metabolism, hyperglycemia, and cardiovascular risk. Circ Res. 2018; 117. Wesolowska-Andersen A, Yu GZ, Nylander V, et al. Deep learning
123(7):886-904. https://doi.org/10.1161/CIRCRESAHA.118. models predict regulatory variants in pancreatic islets and refine
312806. type 2 diabetes association signals. elife. 2020;9:1-21. https://doi.
99. Boles A, Kandimalla R, Reddy PH. Dynamics of diabetes and obesity: org/10.7554/eLife.51503.
epidemiological perspective. Biochim Biophys Acta Mol basis Dis. 118. Gamboa-Melendez MA, Huerta-Chagoya A, Moreno-Macias H, et al.
2017;1863(5):1026-1036. https://doi.org/10.1016/j.bbadis.2017. Contribution of common genetic variation to the risk of type 2 dia-
01.016. betes in the Mexican mestizo population. Diabetes. 2012;61(12):
100. Liao WL, Tsai FJ. Personalized medicine in type 2 diabetes. Biomedi- 3314-3321. https://doi.org/10.2337/db11-0550.
cine. 2014;4:8. https://doi.org/10.7603/s40681-014-0008-z. 119. Talmud PJ, Cooper JA, Morris RW, et al. Sixty-five common genetic
101. Grant RW, Wexler DJ. Personalized medicine in type 2 diabetes: variants and prediction of type 2 diabetes. Diabetes. 2015;64(5):
what does the future hold? Diabetes Manag. 2012;2(3):199-204. 1830-1840. https://doi.org/10.2337/db14-1504.
https://doi.org/10.2217/dmt.12.15. 120. Deeb SS, Fajas L, Nemoto M, et al. A Pro12Ala substitution in
102. Fitipaldi H, McCarthy MI, Florez JC, Franks PW. A global overview PPARgamma2 associated with decreased receptor activity, lower
of precision medicine in type 2 diabetes. Diabetes. 2018;67(10): body mass index and improved insulin sensitivity. Nat Genet. 1998;
1911-1922. https://doi.org/10.2337/dbi17-0045. 20(3):284-287. https://doi.org/10.1038/3099.
103. Udler MS, Kim J, von Grotthuss M, et al. Type 2 diabetes genetic loci 121. Mansoori A, Sotoudeh G, Djalali M, et al. Effect of DHA-rich fish oil
informed by multi-trait associations point to disease mechanisms on PPARgamma target genes related to lipid metabolism in type
and subtypes: a soft clustering analysis. PLoS Med. 2018;15(9): 2 diabetes: a randomized, double-blind, placebo-controlled clinical
e1002654. https://doi.org/10.1371/journal.pmed.1002654. trial. J Clin Lipidol. 2015;9(6):770-777. https://doi.org/10.1016/j.
104. Alberti G, Zimmet P, Shaw J, Bloomgarden Z, Kaufman F, Silink M. jacl.2015.08.007.
Type 2 diabetes in the young: the evolving epidemic: the interna- 122. Dong C, Zhou H, Shen C, et al. Role of peroxisome proliferator-
tional diabetes federation consensus workshop. Diabetes Care. activated receptors gene polymorphisms in type 2 diabetes and met-
2004;27(7):1798-1811. https://doi.org/10.2337/diacare.27.7.1798. abolic syndrome. World J Diabetes. 2015;6(4):654-661. https://doi.
105. Reinehr T. Lifestyle intervention in childhood obesity: changes and org/10.4239/wjd.v6.i4.654.
challenges. Nat Rev Endocrinol. 2013;9(10):607-614. https://doi.org/ 123. Li S, Chen W, Srinivasan SR, Boerwinkle E, Berenson GS. The peroxi-
10.1038/nrendo.2013.149. some proliferator-activatedreceptor-gamma2 gene polymorphism
106. Bhupathiraju SN, Hu FB. One (small) step towards precision nutri- (Pro12Ala) beneficially influences insulin resistance and its tracking
tion by use of metabolomics. Lancet Diabetes Endocrinol. 2017;5(3): from childhood to adulthood: the Bogalusa heart study. Diabetes.
154-155. https://doi.org/10.1016/S2213-8587(17)30007-4. 2003;52(5):1265-1269. https://doi.org/10.2337/diabetes.52.5.
107. Arslanian S, Bacha F, Grey M, Marcus MD, White NH, Zeitler P. 1265.
Evaluation and management of youth-onset type 2 diabetes: a posi- 124. Ochoa M, Razquin C, Miguel M-G, Marti A. Role of PPAR-γ2
tion statement by the American Diabetes Association. Diabetes Care. polymorphisms inbodyweight regulation. Futur Lipidol. 2008;3
2018;41(12):2648-2668. https://doi.org/10.2337/dci18-0052. (1):11.
18 SIRDAH AND READING

125. Sladek R, Rocheleau G, Rung J, et al. A genome-wide association 143. Bush WS, Moore JH. Chapter 11: genome-wide association studies.
study identifies novel risk loci for type 2 diabetes. Nature. 2007;445 PLoS Comput Biol. 2012;8(12):e1002822. https://doi.org/10.1371/
(7130):881-885. https://doi.org/10.1038/nature05616. journal.pcbi.1002822.
126. Pascoe L, Tura A, Patel SK, et al. Common variants of the novel type 144. Remedi MS, Emfinger C. Pancreatic beta-cell identity in diabetes.
2 diabetes genes CDKAL1 and HHEX/IDE are associated with Diabetes Obes Metab. 2016;18(Suppl 1):110-116. https://doi.org/
decreased pancreatic beta-cell function. Diabetes. 2007;56(12): 10.1111/dom.12727.
3101-3104. https://doi.org/10.2337/db07-0634. 145. Park YJ, Woo M. Pancreatic beta cells: gatekeepers of type 2 diabe-
127. Horikoshi M, Hara K, Ito C, Nagai R, Froguel P, Kadowaki T. A tes. J Cell Biol. 2019;218(4):1094-1095. https://doi.org/10.1083/
genetic variation of the transcription factor 7-like 2 gene is associ- jcb.201810097.
ated with risk of type 2 diabetes in the Japanese population. 146. Vana DR, Adapa D, VSS P, Choudhury A, Choudhury A, Ahuja G.
Diabetologia. 2007;50(4):747-751. https://doi.org/10.1007/ Diabetes mellitus types: key genetic determinants and risk assess-
s00125-006-0588-6. ment. Genet Mol Res. 2019;18(2):27.
128. Grarup N, Rose CS, Andersson EA, et al. Studies of association of 147. Saberzadeh-Ardestani B, Karamzadeh R, Basiri M, et al. Type 1 dia-
variants near the HHEX, CDKN2A/B, and IGF2BP2 genes with type betes mellitus: cellular and molecular pathophysiology at a glance.
2 diabetes and impaired insulin release in 10,705 Danish subjects: Cell J. 2018;20(3):294-301. https://doi.org/10.22074/cellj.2018.
validation and extension of genome-wide association studies. Diabe- 5513.
tes. 2007;56(12):3105-3111. https://doi.org/10.2337/db07-0856. 148. Nyaga DM, Vickers MH, Jefferies C, Perry JK, O'Sullivan JM. The
129. Cornelis MC, Zaitlen N, Hu FB, Kraft P, Price AL. Genetic and environ- genetic architecture of type 1 diabetes mellitus. Mol Cell Endocrinol.
mental components of family history in type 2 diabetes. Hum Genet. 2018;477:70-80. https://doi.org/10.1016/j.mce.2018.06.002.
2015;134(2):259-267. https://doi.org/10.1007/s00439-014-1519-0. 149. Storling J, Pociot F. Type 1 diabetes candidate genes linked to pan-
130. Lyssenko V, Groop L, Prasad RB. Genetics of type 2 diabetes: it mat- creatic islet cell inflammation and beta-cell apoptosis. Genes (Basel).
ters from which parent we inherit the risk. Rev Diabet Stud. 2015;12 2017;8(2):1-12. https://doi.org/10.3390/genes8020072.
(3–4):233-242. https://doi.org/10.1900/RDS.2015.12.233. 150. Cerf ME. Beta cell dysfunction and insulin resistance. Front
131. Basile KJ, Johnson ME, Xia Q, Grant SF. Genetic susceptibility to Endocrinol. 2013;4:37. https://doi.org/10.3389/fendo.2013.00037.
type 2 diabetes and obesity: follow-up of findings from genome- 151. Chen C, Cohrs CM, Stertmann J, Bozsak R, Speier S. Human beta
wide association studies. Int J Endocrinol. 2014;2014:769671- cell mass and function in diabetes: recent advances in knowledge
769613. https://doi.org/10.1155/2014/769671. and technologies to understand disease pathogenesis. Mol Metab.
132. Grant SFA. The TCF7L2 Locus: A Genetic Window Into the Patho- 2017;6(9):943-957. https://doi.org/10.1016/j.molmet.2017.06.019.
genesis of Type 1 and Type 2 Diabetes. Diabetes Care. 2019;42(9): 152. Prentki M, Nolan CJ. Islet beta cell failure in type 2 diabetes.
1624-1629. https://doi.org/10.2337/dci19-0001. J Clin Invest. 2006;116(7):1802-1812. https://doi.org/10.1172/
133. Skyler JS, Bakris GL, Bonifacio E, et al. Differentiation of diabetes by JCI29103.
pathophysiology, natural history, and prognosis. Diabetes. 2017;66 153. Meier JJ, Bonadonna RC. Role of reduced beta-cell mass versus
(2):241-255. https://doi.org/10.2337/db16-0806. impaired beta-cell function in the pathogenesis of type 2 diabetes.
134. Medici F, Hawa M, Ianari A, Pyke DA, Leslie RD. Concordance rate Diabetes Care. 2013;36(Suppl 2):S113-S119. https://doi.org/10.
for type II diabetes mellitus in monozygotic twins: actuarial analysis. 2337/dcS13-2008.
Diabetologia. 1999;42(2):146-150. https://doi.org/10.1007/ 154. Fonseca SG, Gromada J, Urano F. Endoplasmic reticulum stress and
s001250051132. pancreatic beta-cell death. Trends Endocrinol Metab. 2011;22(7):
135. Ahlqvist E, Ahluwalia TS, Groop L. Genetics of type 2 diabetes. Clin 266-274. https://doi.org/10.1016/j.tem.2011.02.008.
Chem. 2011;57(2):241-254. https://doi.org/10.1373/clinchem. 155. Cnop M, Welsh N, Jonas JC, Jorns A, Lenzen S, Eizirik DL. Mecha-
2010.157016. nisms of pancreatic beta-cell death in type 1 and type 2 diabetes:
136. Groop L, Forsblom C, Lehtovirta M, et al. Metabolic consequences many differences, few similarities. Diabetes. 2005;54(Suppl 2):S97-
of a family history of NIDDM (the Botnia study): evidence for sex- S107. https://doi.org/10.2337/diabetes.54.suppl_2.s97.
specific parental effects. Diabetes. 1996;45(11):1585-1593. https:// 156. Eizirik DL, Cardozo AK, Cnop M. The role for endoplasmic reticulum
doi.org/10.2337/diab.45.11.1585. stress in diabetes mellitus. Endocr Rev. 2008;29(1):42-61. https://
137. Goran MI, Bergman RN, Avila Q, et al. Impaired glucose tolerance doi.org/10.1210/er.2007-0015.
and reduced beta-cell function in overweight Latino children with a 157. Frau F, Crowther D, Ruetten H, Allebrandt KV. Type-2diabetes-
positive family history for type 2 diabetes. J Clin Endocrinol Metab. associated variants with cross-trait relevance: post-GWAs strategies
2004;89(1):207-212. https://doi.org/10.1210/jc.2003-031402. for biological function interpretation. Mol Genet Metab. 2017;121(1):
138. Abdullah N, Attia J, Oldmeadow C, Scott RJ, Holliday EG. The archi- 43-50. https://doi.org/10.1016/j.ymgme.2017.03.004.
tecture of risk for type 2 diabetes: understanding Asia in the context 158. Adeyemo AA, Tekola-Ayele F, Doumatey AP, et al. Evaluation of
of global findings. Int J Endocrinol. 2014;2014:593982-593921. genome wide association study associated type 2 diabetes suscepti-
https://doi.org/10.1155/2014/593982. bility loci in sub Saharan Africans. Front Genet. 2015;6:335. https://
139. Sanghera DK, Blackett PR. Type 2 diabetes genetics: beyond GWAS. doi.org/10.3389/fgene.2015.00335.
J Diabetes Metab. 2012;3(198):1-12. https://doi.org/10.4172/2155- 159. Scott RA, Scott LJ, Magi R, et al. An expanded genome-wide associa-
6156.1000198. tion study of type 2 diabetes in Europeans. Diabetes. 2017;66(11):
140. McCarthy MI, Zeggini E. Genome-wide association studies in type 2888-2902. https://doi.org/10.2337/db16-1253.
2 diabetes. Curr Diab Rep. 2009;9(2):164-171. https://doi.org/10. 160. Karaderi T, Drong AW, Lindgren CM. Insights into the genetic sus-
1007/s11892-009-0027-4. ceptibility to type 2 diabetes from genome-wide association studies
141. Cirillo E, Kutmon M, Gonzalez Hernandez M, et al. From SNPs to of obesity-related traits. Curr Diab Rep. 2015;15(10):83. https://doi.
pathways: biological interpretation of type 2 diabetes (T2DM) org/10.1007/s11892-015-0648-8.
genome wide association study (GWAS) results. PLoS ONE. 2018;13 161. Turki A, Al-Zaben GS, Mtiraoui N, Marmmuoch H, Mahjoub T,
(4):e0193515. https://doi.org/10.1371/journal.pone.0193515. Almawi WY. Transcription factor-7-like 2 gene variants are
142. Visscher PM, Wray NR, Zhang Q, et al. 10 years of GWAS discovery: strongly associated with type 2 diabetes in Tunisian Arab subjects.
biology, function, and translation. Am J Hum Genet. 2017;101(1):5- Gene. 2013;513:244-248. https://doi.org/10.1016/j.gene.2012.
22. https://doi.org/10.1016/j.ajhg.2017.06.005. 10.086.
SIRDAH AND READING 19

162. Chandak GR, Janipalli CS, Bhaskar S, et al. Common variants in the 179. Tabassum R, Chauhan G, Dwivedi OP, et al. Genome-wide associa-
TCF7L2 gene are strongly associated with type 2 diabetes mellitus tion study for type 2 diabetes in Indians identifies a new susceptibil-
in the Indian population. Diabetologia. 2007;50:63-67. https://doi. ity locus at 2q21. Diabetes. 2013;62(3):977-986. https://doi.org/10.
org/10.1007/s00125-006-0502-2. 2337/db12-0406.
163. Huang Z, Liao Y, Huang R, Chen J, Sun H. Possible role of TCF7L2 in 180. Yamauchi T, Hara K, Maeda S, et al. A genome-wide association
the pathogenesis of type 2 diabetes mellitus. Biotechnol Biotechnol study in the Japanese population identifies susceptibility loci for
Equip. 2018;32(4):5. type 2 diabetes at UBE2E2 and C2CD4A-C2CD4B. Nat Genet.
164. Hayashi T, Iwamoto Y, Kaku K, Hirose H, Maeda S. Replication study 2010;42(10):864-868. https://doi.org/10.1038/ng.660.
for the association of TCF7L2 with susceptibility to type 2 diabetes 181. Scott LJ, Mohlke KL, Bonnycastle LL, et al. A genome-wide associa-
in a Japanese population. Diabetologia. 2007;50:980-984. https:// tion study of type 2 diabetes in Finns detects multiple susceptibility
doi.org/10.1007/s00125-007-0618-z. variants. Science (80). 2007;316(5829):1341-1345. https://doi.org/
165. Rees SD, Bellary S, Britten AC, et al. Common variants of the 10.1126/science.1142382.
TCF7L2 gene are associated with increased risk of type 2 diabetes 182. Saxena R, Voight BF, Lyssenko V, et al. Genome-wide association
mellitus in a UK-resident south Asian population. BMC Med Genet. analysis identifies loci for type 2 diabetes and triglyceride levels. Sci-
2008;9(8):1-6. https://doi.org/10.1186/1471-2350-9-8. ence (80). 2007;316:1331-1336. https://doi.org/10.1126/science.
166. Al Ali M, El hajj Chehadeh S, Osman W, et al. Investigating the asso- 1142358.
ciation of rs7903146 of TCF7L2 gene, rs5219 of KCNJ11 gene, 183. Kooner JS, Saleheen D, Sim X, et al. Genome-wide association study
rs10946398 of CDKAL1 gene, and rs9939609 of FTO gene with in individuals of south Asian ancestry identifies six new type 2 diabe-
type 2 diabetes mellitus in Emirati population. Meta Gene. 2019;21: tes susceptibility loci. Nat Genet. 2011;43:984-989. https://doi.org/
100600. https://doi.org/10.1016/j.mgene.2019.100600. 10.1038/ng.921.
167. Al-Safar H, Hassoun A, Almazrouei S, Kamal W, Afandi B, Rais N. 184. Sandhu MS, Weedon MN, Fawcett KA, et al. Common variants in
Association of the genetic polymorphisms in transcription factor WFS1 confer risk of type 2 diabetes. Nat Genet. 2007;39(8):951-
7-like 2 and peroxisome proliferator-activatedreceptors- γ 2 with 953. https://doi.org/10.1038/ng2067.
type 2 diabetes mellitus and its interaction with obesity status in 185. Lyssenko V, Jonsson A, Almgren P, et al. Clinical risk factors, DNA
Emirati population. J Diabetes Res. 2015;2015:1-8. https://doi.org/ variants, and the development of type 2 diabetes. N Engl J Med.
10.1155/2015/129695. 2008;359:2220-2232. https://doi.org/10.1056/NEJMoa0801869.
168. Gloyn AL, Braun M, Rorsman P. Type 2 diabetes susceptibility gene 186. Cho YS, Chen CH, Hu C, et al. Meta-analysis of genome-wide associa-
TCF7L2 and its role in beta-cell function. Diabetes. 2009;58(4):800- tion studies identifies eight new loci for type 2 diabetes in east Asians.
802. https://doi.org/10.2337/db09-0099. Nat Genet. 2011;44(1):67-72. https://doi.org/10.1038/ng.1019.
169. Scott LJ, Bonnycastle LL, Willer CJ, et al. Association of transcription 187. Mahajan A, Wessel J, Willems SM, et al. Refining the accuracy of
factor 7-like 2 (TCF7L2) variants with type 2 diabetes in a Finnish validated target identification through coding variant fine-mapping
sample. Diabetes. 2006;55(9):2649-2653. https://doi.org/10.2337/ in type 2 diabetes article. Nat Genet. 2018;50:559-571. https://doi.
db06-0341. org/10.1038/s41588-018-0084-1.
170. Tong Y, Lin Y, Zhang Y, Yang J, Liu H, Zhang B. Association between 188. Hara K, Fujita H, Johnson TA, et al. Genome-wide association study
TCF7L2 gene polymorphisms and susceptibility to type 2 diabetes identifies three novel loci for type 2 diabetes. Hum Mol Genet. 2014;
mellitus: a large human genome epidemiology (HuGE) review and 23:239-246. https://doi.org/10.1093/hmg/ddt399.
meta-analysis. BMC Med Genet. 2009;10:15. https://doi.org/10. 189. Huyghe JR, Jackson AU, Fogarty MP, et al. Exome array analysis
1186/1471-2350-10-15. identifies new loci and low-frequency variants influencing insulin
171. Grant SF, Thorleifsson G, Reynisdottir I, et al. Variant of transcrip- processing and secretion. Nat Genet. 2013;45(2):197-201. https://
tion factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes. doi.org/10.1038/ng.2507.
Nat Genet. 2006;38(3):320-323. https://doi.org/10.1038/ng1732. 190. Zeggini E, Weedon MN, Lindgren CM, et al. Multiple type 2 diabetes
172. Zeggini E, Scott LJ, Saxena R, et al. Meta-analysis of genome-wide susceptibility genes following genome- wide association scan in UK
association data and large-scale replication identifies additional sus- samples. Science. 2007;316(5829):1336-1341. https://doi.org/10.
ceptibility loci for type 2 diabetes. Nat Genet. 2008;40:638-645. 1126/science.1142364.
https://doi.org/10.1038/ng.120. 191. Go MJ, Hwang JY, Park TJ, et al. Genome-wide association study
173. Mahajan A, Go MJ, Zhang W, et al. Genome-widetrans-ancestry- identifies two novel loci with sex-specific effects for type 2 diabe-
meta-analysis provides insight into the genetic architecture of type tes mellitus and glycemic traits in a Korean population. Diabetes
2 diabetes susceptibility. Nat Genet. 2014;46(3):234-244. https:// Metab J. 2014;38:375-387. https://doi.org/10.4093/dmj.2014.38.
doi.org/10.1038/ng.2897. 5.375.
174. Kwak SH, Park KS. Recent progress in genetic and epigenetic 192. Shu XO, Long J, Cai Q, et al. Identification of new genetic risk vari-
research on type 2 diabetes. Exp Mol Med. 2016;48:e220. https:// ants for type 2 diabetes. PLoS Genet. 2010;6(9):e1001127. https://
doi.org/10.1038/emm.2016.7. doi.org/10.1371/journal.pgen.1001127.
175. Cook JP, Morris AP. Multi-ethnicgenome-wide association study 193. Li H, Gan W, Lu L, et al. A genome-wide association study identifies
identifies novel locus for type 2 diabetes susceptibility. Eur J Hum GRK5 and RASGRP1 as type 2 diabetes loci in Chinese Hans. Diabe-
Genet. 2016;24:1175-1180. https://doi.org/10.1038/ejhg.2016.17. tes. 2013;62(1):291-298. https://doi.org/10.2337/db12-0454.
176. Dupuis J, Langenberg C, Prokopenko I, et al. New genetic loci implicated 194. Tsai FJ, Yang CF, Chen CC, et al. A genome-wide association study
in fasting glucose homeostasis and their impact on type 2 diabetes risk. identifies susceptibility variants for type 2 diabetes in Han Chinese.
Nat Genet. 2010;42:105-116. https://doi.org/10.1038/ng.520. PLoS Genet. 2010;6(2):e1000847. https://doi.org/10.1371/journal.
177. Morris AP, Voight BF, Teslovich TM, et al. Large-scale association pgen.1000847.
analysis provides insights into the genetic architecture and patho- 195. Johnson AM, Olefsky JM. The origins and drivers of insulin resis-
physiology of type 2 diabetes. Nat Genet. 2012;44(9):981-990. tance. Cell. 2013;152(4):673-684. https://doi.org/10.1016/j.cell.
https://doi.org/10.1038/ng.2383. 2013.01.041.
178. Voight BF, Scott LJ, Steinthorsdottir V, et al. Twelve type 2 diabetes 196. Kahn BB. Type 2 diabetes: when insulin secretion fails to compen-
susceptibility loci identified through large-scale association analysis. sate for insulin resistance. Cell. 1998;92(5):593-596. https://doi.org/
Nat Genet. 2010;42(7):579-589. https://doi.org/10.1038/ng.609. 10.1016/s0092-8674(00)81125-3.
20 SIRDAH AND READING

197. Ormazabal P, Scazzocchio B, Vari R, et al. Effect of protocatechuic 216. Do R, Bailey SD, Desbiens K, et al. Genetic variants of FTO influence
acid on insulin responsiveness and inflammation in visceral adipose adiposity, insulin sensitivity, leptin levels, and resting metabolic rate
tissue from obese individuals: possible role for PTP1B. Int J Obes. in the Quebec family study. Diabetes. 2008;57(4):1147-1150.
2018;42(12):2012-2021. https://doi.org/10.1038/s41366-018- https://doi.org/10.2337/db07-1267.
0075-4. 217. Kotnik P, Knapic E, Kokosar J, et al. Identification of novel alleles
198. Czech MP. Insulin action and resistance in obesity and type 2 diabe- associated with insulin resistance in childhood obesity using pooled-
tes. Nat Med. 2017;23(7):804-814. https://doi.org/10.1038/nm. DNAgenome-wide association study approach. Int J Obes. 2018;42
4350. (4):686-695. https://doi.org/10.1038/ijo.2017.293.
199. Weir GC. Can we make surrogate beta-cells better than the original? 218. Brown AE, Walker M. Genetics of insulin resistance and the meta-
Semin Cell Dev Biol. 2004;15(3):347-357. https://doi.org/10.1016/j. bolic syndrome. Curr Cardiol Rep. 2016;18(8):75. https://doi.org/10.
semcdb.2004.02.004. 1007/s11886-016-0755-4.
200. Kahn SE. Clinical review 135: the importance of beta-cell failure in 219. Ma J, Li Y, Zhou F, Xu X, Guo G, Qu Y. Meta-analysis of association
the development and progression of type 2 diabetes. J Clin between the Pro12Ala polymorphism of the peroxisome
Endocrinol Metab. 2001;86(9):4047-4058. https://doi.org/10.1210/ proliferator-activatedreceptor-gamma2 gene and diabetic retinopa-
jcem.86.9.7713. thy in Caucasians and Asians. Mol Vis. 2012;18:2352-2360. http://
201. Shanik MH, Xu Y, Skrha J, Dankner R, Zick Y, Roth J. Insulin resis- www.ncbi.nlm.nih.gov/pubmed/22993484.
tance and hyperinsulinemia: is hyperinsulinemia the cart or the 220. Galbete C, Toledo E, Martinez-Gonzalez MA, Martinez JA, Guillen-
horse? Diabetes Care. 2008;31(Suppl 2):S262-S268. https://doi.org/ Grima F, Marti A. Pro12Ala variant of the PPARG2 gene increases
10.2337/dc08-s264. body mass index: an updated meta-analysis encompassing 49,092
202. Taylor R. Insulin resistance and type 2 diabetes. Diabetes. 2012;61 subjects. Obesity (Silver Spring). 2013;21(7):1486-1495. https://doi.
(4):778-779. https://doi.org/10.2337/db12-0073. org/10.1002/oby.20150.
203. Patil PD, Mahajan UB, Patil KR, et al. Past and current perspective 221. Ghoussaini M, Meyre D, Lobbens S, et al. Implication of the
on new therapeutic targets for type-II diabetes. Drug Des Devel Pro12Ala polymorphism of the PPAR-gamma 2 gene in type 2 diabe-
Ther. 2017;11:1567-1583. https://doi.org/10.2147/DDDT. tes and obesity in the French population. BMC Med Genet. 2005;6:
S133453. 11. https://doi.org/10.1186/1471-2350-6-11.
204. DeFronzo RA, Triplitt CL, Abdul-Ghani M, Cersosimo E. Novel 222. Tonjes A, Scholz M, Loeffler M, Stumvoll M. Association of Pro12Ala
agents for the treatment of type 2 diabetes. Diabetes Spectr. 2014; polymorphism in peroxisome proliferator-activated receptor gamma
27(2):100-112. https://doi.org/10.2337/diaspect.27.2.100. with pre-diabetic phenotypes: meta-analysis of 57 studies on
205. Henry RR. Insulin resistance: from predisposing factor to therapeutic nondiabetic individuals. Diabetes Care. 2006;29(11):2489-2497.
target in type 2 diabetes. Clin Ther. 2003;25(Suppl B):B47-B63. https://doi.org/10.2337/dc06-0513.
https://doi.org/10.1016/s0149-2918(03)80242-4. 223. Kruzliak P, Haley AP, Starcevic JN, Gaspar L, Petrovic D. Polymor-
206. Cheng Z, Schmelz EM, Liu D, Hulver MW. Targeting mitochondrial phisms of the peroxisome proliferator-activatedreceptor-gamma
alterations to prevent type 2 diabetes- -evidence from studies of (rs1801282) and its coactivator-1 (rs8192673) are associated with
dietary redox-active compounds. Mol Nutr Food Res. 2014;58(8): obesity indexes in subjects with type 2 diabetes mellitus. Cardiovasc
1739-1749. https://doi.org/10.1002/mnfr.201300747. Diabetol. 2015;14:42. https://doi.org/10.1186/s12933-015-
207. Eckel RH. Obesity. Circulation. 2005;111(15):e257-e259. https:// 0197-0.
doi.org/10.1161/01.CIR.0000163653.38992.E5. 224. Chan KH, Niu T, Ma Y, et al. Common genetic variants in peroxi-
208. DeFronzo RA. Dysfunctional fat cells, lipotoxicity and type 2 diabe- some proliferator-activatedreceptor-gamma(PPARG) and type 2 dia-
tes. Int J Clin Pr Suppl. 2004;143:9-21. https://doi.org/10.1111/j. betes risk among Women's health initiative postmenopausal
1368-504x.2004.00389.x. women. J Clin Endocrinol Metab. 2013;98(3):E600-E604. https://doi.
209. Tagi VM, Giannini C, Chiarelli F. Insulin resistance in children. Front org/10.1210/jc.2012-3644.
Endocrinol. 2019;10:342. https://doi.org/10.3389/fendo.2019.00342. 225. Qi L, Cornelis MC, Kraft P, et al. Genetic variants at 2q24 are associ-
210. Martinez LC, Sherling D, Holley A. The screening and prevention of ated with susceptibility to type 2 diabetes. Hum Mol Genet. 2010;19
diabetes mellitus. Prim Care. 2019;46(1):41-52. https://doi.org/10. (13):2706-2715. https://doi.org/10.1093/hmg/ddq156.
1016/j.pop.2018.10.006. 226. Garant MJ, Linda Kao WH, Brancati F, et al. SNP43 of CAPN10 and
211. Lentferink YE, Elst MAJ, Knibbe CAJ, van der Vorst MMJ. Predictors the risk of type 2 diabetes in African-Americans: the atherosclerosis
of insulin resistance in children versus adolescents with obesity. risk in communities study. Diabetes. 2002;51:231-237. https://doi.
J Obes. 2017;2017:3793868. https://doi.org/10.1155/2017/ org/10.2337/diabetes.51.1.231.
3793868. 227. Li Y, Ge Gong HG, Yang Z, et al. CAPN10 SNP43 G>a gene poly-
212. Ling JC, Mohamed MN, Jalaludin MY, Rampal S, Zaharan NL, morphism and type 2 diabetes mellitus in the Asian population: a
Mohamed Z. Determinants of high fasting insulin and insulin resis- meta-analysis of 9353 participants. Endocr J. 2015;62(2):183-194.
tance among overweight/obese adolescents. Sci Rep. 2016;6:36270. 228. Song Y, You NC, Hsu YH, et al. Common genetic variation in
https://doi.org/10.1038/srep36270. calpain-10 gene (CAPN10) and diabetes risk in a multi-ethnic cohort
213. Okura T, Nakamura R, Fujioka Y, et al. Body mass index ≥23 is a risk of American postmenopausal women. Hum Mol Genet. 2007;16:
factor for insulin resistance and diabetes in Japanese people: a brief 2960-2971. https://doi.org/10.1093/hmg/ddm256.
report. PLoS ONE. 2018;13:e0201052. https://doi.org/10.1371/ 229. Weedon MN, Schwarz PEH, Horikawa Y, et al. Meta-analysis and a
journal.pone.0201052. large association study confirm a role for calpain-10 variation in type
214. Scott RA, Fall T, Pasko D, et al. Common genetic variants highlight 2 diabetes susceptibility. Am J Hum Genet. 2003;73:1208-1212.
the role of insulin resistance and body fat distribution in type 2 dia- https://doi.org/10.1086/379285.
betes, independent of obesity. Diabetes. 2014;63(12):4378-4387. 230. Saxena R, Elbers CC, Guo Y, et al. Large-scalegene-centricmeta-analysis
https://doi.org/10.2337/db14-0319. across 39 studies identifies type 2 diabetes loci. Am J Hum Genet. 2012;
215. Klimentidis YC, Zhou J, Wineinger NE. Identification of allelic het- 90(3):410-425. https://doi.org/10.1016/j.ajhg.2011.12.022.
erogeneity at type-2 diabetes loci and impact on prediction. PLoS 231. Kim SH, Reaven G. Obesity and insulin resistance: an ongoing saga.
ONE. 2014;9(11):e113072. https://doi.org/10.1371/journal.pone. Diabetes. 2010;59(9):2105-2106. https://doi.org/10.2337/db10-
0113072. 0766.
SIRDAH AND READING 21

232. Noakes TD. So what comes first: the obesity or the insulin resis- 251. Loos RJ, Yeo GS. The bigger picture of FTO: the first GWAS-
tance? And which is more important? Clin Chem. 2018;64(1):7-9. identified obesity gene. Nat Rev Endocrinol. 2014;10(1):51-61.
https://doi.org/10.1373/clinchem.2017.282962. https://doi.org/10.1038/nrendo.2013.227.
233. Ye J. Mechanisms of insulin resistance in obesity. Front Med. 2013;7 252. Speliotes EK, Willer CJ, Berndt SI, et al. Association analyses of
(1):14-24. https://doi.org/10.1007/s11684-013-0262-6. 249,796 individuals reveal 18 new loci associated with body mass
234. Kahn BB, Flier JS. Obesity and insulin resistance. J Clin Invest. 2000; index. Nat Genet. 2010;42(11):937-948. https://doi.org/10.1038/
106(4):473-481. https://doi.org/10.1172/JCI10842. ng.686.
235. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to 253. Frayling TM. Genome-wide association studies provide new insights
insulin resistance and type 2 diabetes. Nature. 2006;444(7121):840- into type 2 diabetes aetiology. Nat Rev Genet. 2007;8(9):657-662.
846. https://doi.org/10.1038/nature05482. https://doi.org/10.1038/nrg2178.
236. Bhurosy T, Jeewon R. Overweight and obesity epidemic in develop- 254. Scuteri A, Sanna S, Chen WM, et al. Genome-wide association scan
ing countries: a problem with diet, physical activity, or socioeco- shows genetic variants in the FTO gene are associated with obesity-
nomic status? ScientificWorldJournal. 2014;2014:964236-964237. related traits. PLoS Genet. 2007;3(7):e115. https://doi.org/10.1371/
https://doi.org/10.1155/2014/964236. journal.pgen.0030115.
237. Hu FB, Malik VS. Sugar-sweetened beverages and risk of obe- 255. Dina C, Meyre D, Gallina S, et al. Variation in FTO contributes to
sity and type 2 diabetes: epidemiologic evidence. Physiol Behav. childhood obesity and severe adult obesity. Nat Genet. 2007;39(6):
2010;100(1):47-54. https://doi.org/10.1016/j.physbeh.2010. 724-726. https://doi.org/10.1038/ng2048.
01.036. 256. Fawcett KA, Barroso I. The genetics of obesity: FTO leads the way.
238. Hossain P, Kawar B, El Nahas M. Obesity and diabetes in the devel- Trends Genet. 2010;26(6):266-274. https://doi.org/10.1016/j.tig.
oping world- -a growing challenge. N Engl J Med. 2007;356(3):213- 2010.02.006.
215. https://doi.org/10.1056/NEJMp068177. 257. Schwenk RW, Vogel H, Schurmann A. Genetic and epigenetic con-
239. Worldwide trends in diabetes since 1980: a pooled analysis of trol of metabolic health. Mol Metab. 2013;2(4):337-347. https://doi.
751 population-based studies with 4.4 million participants. Lancet. org/10.1016/j.molmet.2013.09.002.
2016;387(10027):1513-1530. https://doi.org/10.1016/S0140- 258. Sykiotis GP, Kalliolias GD, Papavassiliou AG. Pharmacogenetic prin-
6736(16)00618-8. ciples in the Hippocratic writings. J Clin Pharmacol. 2005;45:1218-
240. Magi R, Manning S, Yousseif A, et al. Contribution of 32 GWAS- 1220. https://doi.org/10.1177/0091270005281091.
identified common variants to severe obesity in European adults 259. DLV FM, Bustamante CD, Leal SM. Genome-wide association map-
referred for bariatric surgery. PLoS ONE. 2013;8(8):e70735. https:// ping and rare alleles: from population genomics to personalized
doi.org/10.1371/journal.pone.0070735. medicine - session introduction. Pac Symp Biocomput. 2011;74-75.
241. Locke AE, Kahali B, Berndt SI, et al. Genetic studies of body mass https://doi.org/10.1142/9789814335058_0008.
index yield new insights for obesity biology. Nature. 2015;518 260. Suwinski P, Ong C, Ling MHT, Poh YM, Khan AM, Ong HS. Advanc-
(7538):197-206. https://doi.org/10.1038/nature14177. ing personalized medicine through the application of whole exome
242. Xi B, He D, Hu Y, Zhou D. Prevalence of metabolic syndrome and its sequencing and big data analytics. Front Genet. 2019;10:49. https://
influencing factors among the Chinese adults: the China health and doi.org/10.3389/fgene.2019.00049.
nutrition survey in 2009. Prev Med. 2013;57(6):867-871. https:// 261. Rossbach SJ, Rossbach M. An economic perspective on personalized
doi.org/10.1016/j.ypmed.2013.09.023. medicine. HUGO J. 2013;7:6. https://doi.org/10.1186/1877-6566-
243. Grarup N, Sandholt CH, Hansen T, Pedersen O. Genetic susceptibil- 7-1.
ity to type 2 diabetes and obesity: from genome-wide association 262. Mathur S, Sutton J. Personalized medicine could transform
studies to rare variants and beyond. Diabetologia. 2014;57(8):1528- healthcare. Biomed Rep. 2017;7(1):3-5. https://doi.org/10.3892/br.
1541. https://doi.org/10.1007/s00125-014-3270-4. 2017.922.
244. Wang K, Li WD, Zhang CK, et al. A genome-wide association study 263. Kasztura M, Richard A, Bempong NE, Loncar D, Flahault A. Cost-
on obesity and obesity-related traits. PLoS ONE. 2011;6(4):e18939. effectiveness of precision medicine: a scoping review. Int J Public
https://doi.org/10.1371/journal.pone.0018939. Health. 2019;64(9):1261-1271. https://doi.org/10.1007/s00038-
245. Sandholt CH, Hansen T, Pedersen O. Beyond the fourth wave of 019-01298-x.
genome-wide obesity association studies. Nutr Diabetes. 2012;2: 264. Ginsburg GS, Phillips KA. Precision medicine: from science to value.
e37. https://doi.org/10.1038/nutd.2012.9. Health Aff. 2018;37(5):694-701. https://doi.org/10.1377/hlthaff.
246. Kong X, Xing X, Hong J, Zhang X, Yang W. Genetic variants associ- 2017.1624.
ated with lean and obese type 2 diabetes in a Han Chinese popula- 265. Louca S. Personalized medicine- -a tailored health care system: chal-
tion: a case-control study. Medicine. 2016;95(23):e3841. https://doi. lenges and opportunities. Croat Med J. 2012;53(3):211-213. https://
org/10.1097/MD.0000000000003841. doi.org/10.3325/cmj.2012.53.211.
247. Al-Goblan AS, Al-Alfi MA, Khan MZ. Mechanism linking diabetes 266. Olden K, Freudenberg N, Dowd J, Shields AE. Discovering how envi-
mellitus and obesity. Diabetes Metab Syndr Obes. 2014;7:587-591. ronmental exposures alter genes could lead to new treatments for
https://doi.org/10.2147/DMSO.S67400. chronic illnesses. Health Aff. 2011;30(5):833-841. https://doi.org/
248. Lorenzo C, Wagenknecht LE, D'Agostino RB Jr, Rewers MJ, 10.1377/hlthaff.2011.0078.
Karter AJ, Haffner SM. Insulin resistance, beta-cell dysfunction, and 267. Rappaport SM. Genetic factors are not the major causes of chronic
conversion to type 2 diabetes in a multiethnic population: the insulin diseases. PLoS ONE. 2016;11(4):e0154387. https://doi.org/10.
resistance atherosclerosis study. Diabetes Care. 2010;33(1):67-72. 1371/journal.pone.0154387.
https://doi.org/10.2337/dc09-1115. 268. Mohan V, Radha V. Precision diabetes is slowly becoming a reality.
249. Kahn SE. The relative contributions of insulin resistance and beta- Med Princ Pract. 2019;28:1-9. https://doi.org/10.1159/000497241.
cell dysfunction to the pathophysiology of type 2 diabetes. 269. Kavvoura FK. Monogenic diabetes. Owen KR. Med (United King-
Diabetologia. 2003;46(1):3-19. https://doi.org/10.1007/s00125- dom): Monogenic diabetes; 2019. https://doi.org/10.1016/j.
002-1009-0. mpmed.2018.10.007.
250. Montanya E. Insulin resistance compensation: not just a matter of 270. Kleinberger JW, Pollin TI, Medicine G. Personalized medicine in dia-
β-cells? Diabetes. 2014;63:832-834. https://doi.org/10.2337/db13- betes mellitus: current opportunities and future prospects. Ann N Y
1843. Acad Sci. 2015;1346(1):45-56. https://doi.org/10.1111/nyas.12757.
22 SIRDAH AND READING

271. Pearson ER, Liddell WG, Shepherd M, Corrall RJ, Hattersley AT. Sen- 290. Ordelheide AM, Hrabe de Angelis M, Haring HU, Staiger H.
sitivity to sulphonylureas in patients with hepatocyte nuclear factor- Pharmacogenetics of oral antidiabetic therapy. Pharmacogenomics.
1α gene mutations: evidence for pharmacogenetics in diabetes. 2018;19(6):577-587. https://doi.org/10.2217/pgs-2017-0195.
Diabet Med. 2000;17:543-545. https://doi.org/10.1046/j.1464- 291. Läll K, Mägi R, Morris A, Metspalu A, Fischer K. Personalized risk pre-
5491.2000.00305.x. diction for type 2 diabetes: the potential of genetic risk scores. Genet
272. Hattersley AT, Patel KA. Precision diabetes: learning from mono- Med. 2017;19:322-329. https://doi.org/10.1038/gim.2016.103.
genic diabetes. Diabetologia. 2017;60:769-777. https://doi.org/10. 292. Weedon MN, McCarthy MI, Hitman G, et al. Combining information
1007/s00125-017-4226-2. from common type 2 diabetes risk polymorphisms improves disease
273. Gloyn AL, Drucker DJ. Precision medicine in the management of prediction. PLoS Med. 2006;3:e374. https://doi.org/10.1371/
type 2 diabetes. Lancet Diabetes Endocrinol. 2018;6:891-900. journal.pmed.0030374.
https://doi.org/10.1016/S2213-8587(18)30052-4. 293. Layton J, Li X, Shen C, et al. Type 2 diabetes genetic risk scores are
274. Marin-Penalver JJ, Martin-Timon I, Sevillano-Collantes C, Del associated with increased type 2 diabetes risk among African Ameri-
Canizo-Gomez FJ. Update on the treatment of type 2 diabetes cans by cardiometabolic status. Clin Med Insights Endocrinol Diabetes.
mellitus. World J Diabetes. 2016;7(17):354-395. https://doi.org/10. 2018;11:117955141774894. https://doi.org/10.1177/
4239/wjd.v7.i17.354. 1179551417748942.
275. Nathan DM. Progress in diabetes research- -what's next. JAMA. 294. Multhaup ML, Kita R, Eriksson N, et al. 304-OR: polygenic risk score
2009;301(15):1599-1601. https://doi.org/10.1001/jama.2009.509. predicts type 2 diabetes susceptibility in a diverse consumer genetic
276. Chitre MM, Burke S. Treatment algorithms and the pharmacological database. Diabetes. 2019;68:304-3OR. https://doi.org/10.2337/
management of type 2 diabetes. Diabetes Spectr. 2006;19(4):7-255. db19-304-or.
https://doi.org/10.2337/diaspect.19.4.249. 295. Ahlqvist E, Storm P, Käräjämäki A, et al. Novel subgroups of adult-
277. Garber AJ, Abrahamson MJ, Barzilay JI, et al. Consensus statement onset diabetes and their association with outcomes: a data-driven
by the American Association of Clinical Endocrinologists and Ameri- cluster analysis of six variables. Lancet Diabetes Endocrinol. 2018;6:
can College of endocrinology on the comprehensive type 2 diabetes 361-369. https://doi.org/10.1016/S2213-8587(18)30051-2.
management algorithm - 2019 executive summary. Endocr Pr. 2019; 296. Tam V, Patel N, Turcotte M, Bossé Y, Paré G, Meyre D. Benefits
25(1):69-100. https://doi.org/10.4158/CS-2018-0535. and limitations of genome-wide association studies. Nat Rev
278. Knowler WC, Barrett-Connor E, Fowler SE, et al. Reduction in the Genet. 2019;20:467-484. https://doi.org/10.1038/s41576-019-
incidence of type 2 diabetes with lifestyle intervention or metfor- 0127-1.
min. N Engl J Med. 2002;346(6):393-403. https://doi.org/10.1056/ 297. Flannick J, Mercader JM, Fuchsberger C, et al. Exome sequencing of
NEJMoa012512. 20,791 cases of type 2 diabetes and 24,440 controls. Nature. 2019;
279. Gallo M, Mannucci E, De Cosmo S, et al. Algorithms for personalized 570:71-76. https://doi.org/10.1038/s41586-019-1231-2.
therapy of type 2 diabetes: results of a web-based international sur- 298. Small KS, Todorčevic M, Civelek M, et al. Regulatory variants at
vey. BMJ Open Diabetes Res Care. 2015;3(1):e000109. https://doi. KLF14 influence type 2 diabetes risk via a female-specific effect on
org/10.1136/bmjdrc-2015-000109. adipocyte size and body composition. Nat Genet. 2018;50:572-580.
280. Hu C, Jia W. Diabetes in China: epidemiology and genetic risk fac- https://doi.org/10.1038/s41588-018-0088-x.
tors and their clinical utility in personalized medication. Diabetes. 299. Tramunt B, Smati S, Grandgeorge N, et al. Sex differences in meta-
2018;67(1):3-11. https://doi.org/10.2337/dbi17-0013. bolic regulation and diabetes susceptibility. Diabetologia. 2020;63:
281. Peter PR, Lupsa BC. Personalized Management of Type 2 diabetes. 453-461. https://doi.org/10.1007/s00125-019-05040-3.
Curr Diab Rep. 2019;19(11):115. https://doi.org/10.1007/s11892- 300. Verma A, Bang L, Miller JE, et al. Human-disease phenotype map
019-1244-0. derived from PheWAS across 38,682 individuals. Am J Hum Genet.
282. Subramanian S, Hirsch IB. Personalized diabetes management: mov- 2019;104:55-64. https://doi.org/10.1016/j.ajhg.2018.11.006.
ing from algorithmic to individualized therapy. Diabetes Spectr. 301. Hebbring SJ. The challenges, advantages and future of phenome-
2014;27(2):87-91. https://doi.org/10.2337/diaspect.27.2.87. wide association studies. Immunology. 2014;141:157-165. https://
283. Raz I. Guideline approach to therapy in patients with newly diag- doi.org/10.1111/imm.12195.
nosed type 2 diabetes. Diabetes Care. 2013;36(Suppl 2):S139-S144. 302. Robinson JR, Denny JC, Roden DM, Van Driest SL. Genome-wide
https://doi.org/10.2337/dcS13-2035. and phenome-wide approaches to understand variable drug actions
284. Pollastro C, Ziviello C, Costa V, Ciccodicola A. Pharmacogenomics of in electronic health records. Clin Transl Sci. 2018;11:112-122.
drug response in type 2 diabetes: toward the definition of tailored https://doi.org/10.1111/cts.12522.
therapies? PPAR Res. 2015;2015:415149-415110. https://doi.org/ 303. Safarova MS, Satterfield BA, Fan X, et al. A phenome-wide associa-
10.1155/2015/415149. tion study to discover pleiotropic effects of PCSK9, APOB, and
285. Costa V, Federico A, Pollastro C, et al. Computational analysis of sin- LDLR. NPJ Genom Med. 2019;4:3. https://doi.org/10.1038/s41525-
gle nucleotide polymorphisms associated with altered drug respon- 019-0078-7.
siveness in type 2 diabetes. Int J Mol Sci. 2016;17(7):1-14. https:// 304. Li YR, Keating BJ. Trans-ethnicgenome-wide association studies:
doi.org/10.3390/ijms17071008. advantages and challenges of mapping in diverse populations.
286. Maruthur NM, Gribble MO, Bennett WL, et al. The Genome Med. 2014;6:91. https://doi.org/10.1186/s13073-014-
pharmacogenetics of type 2 diabetes: a systematic review. Diabetes 0091-5.
Care. 2014;37(3):876-886. https://doi.org/10.2337/dc13-1276. 305. Medina-Gomez C, Felix JF, Estrada K, et al. Challenges in conducting
287. Becker ML, Pearson ER, Tkac I. Pharmacogenetics of oral anti- genome-wide association studies in highly admixed multi-ethnic
diabetic drugs. Int J Endocrinol. 2013;2013:686315-686310. https:// populations: the generation R study. Eur J Epidemiol. 2015;30:317-
doi.org/10.1155/2013/686315. 330. https://doi.org/10.1007/s10654-015-9998-4.
288. Pearson ER. Diabetes: is there a future for pharmacogenomics 306. McGuire AL, Basford M, Dressler LG, et al. Ethical and practical chal-
guided treatment? Clin Pharmacol Ther. 2019;106(2):329-337. lenges of sharing data from genome-wide association studies: the
https://doi.org/10.1002/cpt.1484. eMERGE consortium experience. Genome Res. 2011;21:1001-1007.
289. Singh S, Usman K, Banerjee M. Pharmacogenetic studies update in https://doi.org/10.1101/gr.120329.111.
type 2 diabetes mellitus. World J Diabetes. 2016;7(15):302-315. 307. Du Y, Xie J, Chang W, Han Y, Cao G. Genome-wide association
https://doi.org/10.4239/wjd.v7.i15.302. studies: inherent limitations and future challenges. Front Med
SIRDAH AND READING 23

China. 2012;6:444-450. https://doi.org/10.1007/s11684-012- heterogeneity. J Diabetes Res. 2018;2018:1-12. https://doi.org/10.


0225-3. 1155/2018/3061620.
308. Brothers KB, Rothstein MA. Ethical, legal and social implications of
incorporating personalized medicine into healthcare. Pers Med.
2015;12:43-51. https://doi.org/10.2217/pme.14.65.
309. Geneviève LD, Martani A, Shaw D, Elger BS, Wangmo T. Struc- How to cite this article: Sirdah MM, Reading NS. Genetic
tural racism in precision medicine: leaving no one behind. BMC predisposition in type 2 diabetes: A promising approach
Med Ethics. 2020;21:17. https://doi.org/10.1186/s12910-020-
toward a personalized management of diabetes. Clinical
0457-8.
310. Bowman P, Flanagan SE, Hattersley AT. Future roadmaps for preci-
Genetics. 2020;1–23. https://doi.org/10.1111/cge.13772
sion medicine applied to diabetes: rising to the challenge of

You might also like