Diabetes Methods in Molecular Biology Stocker 2020
Diabetes Methods in Molecular Biology Stocker 2020
Diabetes Methods in Molecular Biology Stocker 2020
Type 2
Diabetes
Methods and Protocols
Second Edition
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK
Second Edition
Edited by
Claire J. Stocker
Medical School, University of Buckingham, Buckingham, Buckinghamshire, UK
Editor
Claire J. Stocker
Medical School
University of Buckingham
Buckingham, Buckinghamshire, UK
This Humana imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer
Nature.
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Preface
Diabetes is now reaching pandemic proportions, and the associated complications are often
life-threatening and disabling. Diabetes and its comorbidities are considered among the
biggest killers of UK society and by many estimate a worse threat than cancer. Over the last
decade, both clinical and basic researchers have increased our understanding of the physio-
logical and molecular mechanisms involved in the progression of diabetes and importantly
the relative roles played by both environmental and genetic factors.
The primary purpose of this edition is to provide up-to-date and relevant explanations of
commonly used protocols and analyses used in diabetes research. Specifically, the chapters
start with a series of overarching reviews and will provide its readers with simple explanations
of practical techniques or models in a short succinct format, thus allowing research scientist
and early career clinicians with quick and easy practical information to address a particular
question. There are key practical notes at the end of each chapter, as well as numerous
helpful figures and tables.
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
1 Sixty Years of Drug Discovery for Type 2 Diabetes:
Where Are We Now? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
John C. Clapham
2 Practical Considerations for In Vivo Mouse Studies . . . . . . . . . . . . . . . . . . . . . . . . . 31
Edward T. Wargent
3 Nutritional Models of Type 2 Diabetes Mellitus . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
Beverly Sara Mühlh€ a usler, Carla Toop, and Sheridan Gentili
4 Cheminformatics in the Identification of Drug Classes
for the Treatment of Type 2 Diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 71
Paul W. Finn
5 Whole-Exome Sequencing (WES) for Illumina Short Read Sequencers
Using Solution-Based Capture. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
Milind C. Mahajan and Andrew S. McLellan
6 Gene Expression Mining in Type 2 Diabetes Research. . . . . . . . . . . . . . . . . . . . . . . 109
Donald R. Dunbar
7 Pathways Enrichment Analysis of Gene Expression Data
in Type 2 Diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
Maysson Ibrahim
8 Diagnostic Genetic Testing for Monogenic Diabetes and Congenital
Hyperinsulinemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Jayne A. L. Houghton
9 Isolation and Purification of Rodent Pancreatic Islets of Langerhans . . . . . . . . . . 179
Jacqueline F. O’Dowd and Claire J. Stocker
10 Characterization of Islet Leukocyte Populations in Human
and Murine Islets by Flow Cytometry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
Matthew J. Butcher, Michelle B. Trevino, Yumi Imai,
and Elena V. Galkina
11 Analysis of Histone Modifications in Rodent Pancreatic
Islets by Native Chromatin Immunoprecipitation . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
Ionel Sandovici, Lisa M. Nicholas, and Laura P. O’Neill
12 Quantification of Pancreatic Islets: Using Image Analysis Tools. . . . . . . . . . . . . . . 215
Parvathy E. Harikumar
13 Measurement of Calcium Signaling in Beta-Cell Lines
Using Epifluorescence and Confocal Microscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . 231
Joanne L. Selway
14 Nitric Oxide and Redox State Measurements in Pancreatic Beta Cells . . . . . . . . . 241
Rodrigo Carlessi, Vinicius Cruzat, Younan Chen,
and Philip Newsholme
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 299
Contributors
ix
x Contributors
CHLOE L. RACKHAM Diabetes Research Group, School of Life Course Sciences, King’s College
London, London, UK
IONEL SANDOVICI Metabolic Research Laboratories, Medical Research Council Metabolic
Diseases Unit, Department of Obstetrics and Gynaecology, University of Cambridge,
Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development
and Neuroscience, University of Cambridge, Cambridge, UK
JOANNE L. SELWAY Medical School, University of Buckingham, Buckingham, UK
CLAIRE J. STOCKER Medical School, University of Buckingham, Buckingham,
Buckinghamshire, UK
CARLA TOOP Sansom Institute for Health Research, School of Pharmacy and Medical
Sciences, University of South Australia, Adelaide, SA, Australia
MICHELLE B. TREVINO Department of Internal Medicine, Strelitz Diabetes Center, Eastern
Virginia Medical School, Norfolk, VA, USA
EDWARD T. WARGENT Buckingham Institute of Translational Medicine, University of
Buckingham, Buckingham, UK
MOHAMED S. ZAIBI Buckingham Institute of Translational Medicine, University of
Buckingham, Buckingham, UK
Chapter 1
Abstract
Today, excluding insulin, there are eight classes of anti-diabetic medicines that have been added to the
pharmacy since the introduction of metformin in the mid-1950s; the sulfonylureas, biguanides, thiazoli-
dinediones, α-glucosidase inhibitors, meglitinides, incretins, and sodium glucose transport 2 inhibitors.
Does the fact that metformin is still first-line treatment suggest that our drug discovery efforts over the past
60 years have not been good enough? Or does it suggest that diabetes is such a complex disorder that no
single treatment, other than gastric bypass surgery, can affect true normalization of not only blood sugar
but also the underlying pathologies? Our understanding of the disease has most definitely improved which
may bring hope for the future in terms of science, but for it to be beneficial, this science has to be translated
into better drug treatments for the disease. In this review, I have examined the eight classes of anti-diabetes
drugs from a drug discovery perspective.
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_1, © Springer Science+Business Media, LLC, part of Springer Nature 2020
1
2 John C. Clapham
discovery in this area has been slow with relatively few new entrants
to the market in the 60 years since the introduction of today’s first-
line drug treatment, metformin.
Maintenance of a normal blood glucose level involves a
dynamic interplay between glucose absorption, glucose produc-
tion, and glucose utilization. These in turn are controlled by inter-
actions between circulating hormones (primarily insulin, though
there are many others) and the cellular processes involved in insulin
(and other hormone) signaling, glucose uptake, and glucose dis-
posal. The principal tissues involved in this interplay are the liver,
brain, skeletal muscle, and adipose tissue which in turn differ in
their patterns of substrate utilization, production, and recycling
which are critical to maintaining normal blood glucose levels in
the range of 3.8–6.1 mmol/L (68.4–109.8 mg/dL). Following a
meal, the degree to which blood glucose increases is a function of
the amount of glucose absorbed, the pancreatic insulin secretory
response, suppression of hepatic glucose output and increased glu-
cose uptake by insulin-sensitive (skeletal muscle, liver, adipose tis-
sue—65–70%) and insulin-insensitive organs (brain, kidney—30%)
[5–7].
Type 2 diabetes is the pathologic consequence of two concur-
rent and interacting conditions of insulin resistance and relative
insulin deficiency. On the one hand, insulin’s ability to suppress
hepatic glucose output and stimulate glucose uptake and utilization
is impaired (resulting in chronically raised insulin levels), and on the
other hand, the capacity of the pancreatic β-cell to maintain this
hyperinsulinemic state also begins to fail [8, 9]. Type 2 diabetic
patients are rarely hypoinsulinemic when compared to nondiabetic
individuals [8]. Another twist in the natural history of type 2 diabe-
tes story is that there is also a degenerative process that affects the
normal healthy existence of the pancreatic β-cell. In a landmark
study, it has been shown that after a point, a relatively small decrease
in β-cell mass is all that is required to have profound effects on
fasting blood glucose levels [10].
The Diabetes Control and Complications Trial (DCCT) and
the UK Prospective Diabetes Study (UKPDS) have shown us that
maintaining blood glucose levels to near-normal levels significantly
improves type 2 diabetes and its ensuing complications
[11–13]. The American Diabetes Association has recommended
that treatment should reduce HbA1c levels to less than 7%
[14, 15]. This is a relaxation of earlier guidelines which recom-
mended reducing HbA1c to 6.5% or below [16] as such aggressive
treatment was found to be associated at best with no additional
cardiovascular benefit but at worst with increased mortality
[17, 18]. However, in practice, HbA1c of 7% or above should
initiate treatment regimens. Regulatory guidelines for new drug
treatments usually stipulate an endpoint for a clinically meaningful
and statistically significant reduction in HbA1c, usually at least 1%
Drug Discovery in Type 2 Diabetes 3
block opening of KATP channels [26, 27]. They are termed insulin
secretagogues.
A first-generation sulfonylurea is tolbutamide. Tolbutamide
originated from a war-effort search for antibiotics and the unpleas-
ant side effect of blackouts resulting from hypoglycemia; while
unacceptable for an antibiotic, it turned out to be a new therapeutic
approach for the treatment of diabetes. Tolbutamide, and an ana-
log, carbutamide, were launched in the mid-1950s [28]. Some
studies have suggested that tolbutamide, because of a short dura-
tion of action, could have a place today in the treatment of elderly
type 2 diabetics [29]. The elderly type 2 diabetic patient is particu-
larly vulnerable to the consequences of hypoglycemic episodes that
can make them prone to falls [30] and cardiovascular issues [29]. In
practice, however, tolbutamide is rarely prescribed today and it
carries an FDA warning regarding cardiovascular mortality.
3 Biguanides
4.1 Failure Although the meta-analysis reassures us that they are effective in
in Efficacy lowering HbA1c, both as monotherapy or on top of other agents,
the issue with sulfonylureas with regard to efficacy is durability. In
other words, while reasonable efficacy is seen at the start of treat-
ment, it declines with continued use [64, 65] in a phenomenon
called “secondary failure.” Secondary failure in response to sulfo-
nylurea appears to be specific as long-term treatment with sulfony-
lureas can selectively reduce the insulin secretory response to an
acute dose of another sulfonylurea but not to glucagon [66]. The
exact mechanism is not known though sulfonylureas appear to
induce β-cell apoptosis in cultured human islets [67].
4.2 Failure in Safety The other issue for the sulfonylureas is the increased risk of cardio-
vascular side effects, an issue that has been debated since the 1970s.
The Sulfonylurea glyburide was shown to have a greater risk of
cardiovascular mortality than metformin [68] in a study that raised
the question about the manner in which blood glucose can be safely
reduced. Since then, several studies, using metformin as a compar-
ator, have reported that a number second-generation sulfonylureas
share this cardiovascular risk [64, 65]. There also appears to be
differences between different sulfonylureas on the degree of risk
[67]. Increased cardiovascular risk with these agents is probably
beyond doubt now and has serious implications for developing
economies that rely on cheap drugs [69] as preferred treatments.
The increased cardiovascular risk is probably due to the inter-
action with cardiac KATP channels [70]. In patients with already
increased risk of cardiovascular disease, sulfonylureas acting on
cardiac KATP could mask ST-segment elevation causing opportu-
nities for life-saving interventions to be missed [71].
5 Thiazolidinediones
But the pain did not end there. Pioglitazone sparked some
further concern surrounding increased risk of bladder cancer
[90]. Thiazolidinediones are also associated with fractures, particu-
larly in postmenopausal women [91] and macular oedema [92],
which appears to be reversible on discontinuation of therapy
[93]. Thiazolidinediones are available in their generic form but
really only as third-line therapies.
6 α-Glucosidase Inhibitors
7 Meglitinides
8 Incretins
8.2 DPPIV Inhibitors An alternative strategy to the very challenging approach of produc-
ing Class-B GPCR agonists is to extend the half-life of the endoge-
nous hormones; this has been achieved by inhibiting the DPPIV
enzyme. This is generally an easier option for the drug hunter and
pharmaceutical industry.
DPPIV is a 766-amino acid glycoprotein and is well conserved
across species, which is very useful from a drug discovery perspec-
tive to reduce the number of screens required. It is a serine prote-
ase, so called due to the presence of a serine residue in a catalytic
triad comprising serine-624, aspartic acid-702, and histidine-734.
As a serine protease, DPPIV removes a dipeptide from the
N-terminus of peptide hormones of around 30 amino acids in
Drug Discovery in Type 2 Diabetes 15
length and recognizes its substrates via amino acid motifs. For
DPPIV, peptides which have proline or alanine as the penultimate
amino acid are favored substrates (i.e., N2H-x-Pro-). Although well
conserved across species, DPPIV lacks similarity to the classical
serine proteases such as chymotrypsin [164]. However, there are
other peptidases that favor the N2H-x-Pro- motif, DPPVIII, and
DPPIX, where inhibition of these enzymes, as an off-target effect,
could result in severe adverse effects [165].
DPPIV is a type 2 cell surface protein in terms of its spatial
arrangement; most of the heavily glycosylated molecule is exposed
to the extracellular space and is anchored to the cell membrane by
virtue of a transmembrane helix anchor [166]. A soluble form of
DPPIV, which lacks the anchor, was discovered in human plasma
[167]. In the context of its therapeutic role, its most important
substrates are GIP and GLP-1. However, neuropeptide Y (NPY),
peptide YY, Substance P, and chemokines such as RANTES are also
substrates; indeed, we have found that DPPIV inhibition enhances
the antilipolytic activity of NPY in human adipose tissue [168]. The
crystal structure of DPPIV was solved in 2003 [169] and subse-
quently many structures have been published co-crystallizing
DPPIV with small molecule ligands and peptides [170]. This is a
hugely valuable resource in drug discovery as it increases confidence
and facilitates rational drug design.
Therapeutic inhibitors of DPPIV fall broadly into three types:
reversible substrate analogs (no example on market), covalently
bound substrate analogs such as vildagliptin and saxagliptin and
reversible nonpeptidic heterocycles such as sitagliptin [171]. The
development path for launch of sitagliptin is worthy of mention.
Sitaglitptin (MK-0431) was nominated as the lead candidate for
development in January 2002 and by only 2006 was approved by
the FDA and by the EMEA the following year; this was a spectacu-
lar achievement for Merck and was an example to the industry of
what clear planning and crisp decision-making looks like. Sitagliptin
was the first DPPIV inhibitor to market.
This drug class has been extensively reviewed (for example, see
[165, 172–175]). All marketed DPP-IV inhibitors show augmen-
tation of GIP and GLP-1 levels and produce broadly similar reduc-
tions in HbA1c of around 1–1.5%, close to that produced by
metformin. However, metformin is superior with respect to fasting
plasma glucose levels. DPP-IV inhibitors have a low risk for hypo-
glycemia in monotherapy and are weight-neutral. All DPPIV inhi-
bitors can be given with other oral hypoglycemic agents, and as the
diabetes advances, they can be given with insulin in the late stages of
the disease where perhaps combination with metformin fails to
produce the desired level of glucose control [176].
As expected for agents working in a common pathway, similar
effects of DPP-IV inhibitors to GLP-1 mimetics on β-cell biology
have been reported. Clinical studies in type 2 diabetics also show an
16 John C. Clapham
9 SGLT2 Inhibitors
was cloned in 1992 [189] and is expressed at very high levels in the
early proximal tubule [190], very suggestive of a bulk
transport role.
Over the course of 24 h, the kidneys of a normal individual will
have reabsorbed over 160 g of glucose [191] which equates to
“energy conservation” of the order of 2560 kJ (600 kcal) per day.
This degree of reabsorption is well within the maximum renal
absorptive capacity for a normal individual and gives the system
resilience. However, when plasma glucose levels exceed
10.0 mmol/L (180.0 mg/dL), as seen in diabetes, glucose starts
appearing in the urine, a condition known as glucosuria.
Before significant investment is committed to initiating and
progressing drug discovery programs, various levels of comfort
are required. The highest level is of course when a first-in-class
drug is successfully launched; but then it really should be too late
for anyone else to follow because of the decade-long lead-time.
Commonly, in drug discovery projects, some form of human target
validation is sought early in the projects lifetime, perhaps even
before a formal investment decision. For SGLT2, there was luck
in two respects. First, a form of human target validation presented
itself in the form of loss of function mutations (human gene knock-
outs if you will) that exist and result in glucosuria [192, 193]. Sec-
ond, there was a competitive inhibitor that has been known to
science since the mid-1880s: phlorizin, a naturally occurring dihy-
drochalcone glucoside found in the bark of pear, apple, and cherry
trees. In a study conducted in the 1930s, phlorizin was reported to
promote glucosuria and reduce plasma glucose levels in humans
[194]. In a reversal of the normal linear process, 50 years elapsed in
order to find a study in animals where phlorizin was shown to
ameliorate hyperglycemia and, interestingly, improve insulin sensi-
tivity in partially pancreatectomized diabetic rats [195]. Phlorizin
was subsequently found to be a competitive inhibitor of SGLT2
with a Ki of around 220 nM [187]. For a project start-up, this is
“gold dust”!
However, phlorizin has a number of issues: [1] it is poorly
absorbed, [2] it is nonselective with regard to other SGLTs, [3] it
is poorly bioavailable, and it needs to get to its target, and [4] has
unpleasant gastric side effects probably due to point 2. These are all
features that can be fixed by a decent medicinal chemistry approach
and indeed the currently approved SGLT2 inhibitors used phlor-
izin as a molecular starting point. Early attempts were not so
successful since they, like phlorizin itself, were O-linked glycosides
and very prone to rapid metabolism [196]. This was solved by
changing the O-linked bond to a C-glucosidic bond to yield cana-
gliflozin [197], dapagliflozin [198], and empagliflozin [199] which
gained FDA approval in the order cited here between late 2013 and
autumn of 2014. The structural parameters in order to mass-
produce SGLT2 inhibitors are well defined [200] as evidenced by
18 John C. Clapham
References
1. Sugiyama Y (2005) Druggability: selecting 8. Reaven GM (2014) Pathophysiology of insu-
optimized drug candidates. Drug Discov lin resistance in human disease. Physiol Rev
Today 10:1577–1579 75:473–486
2. Allen MJ, Carey AH (2004) Target identifica- 9. Reaven GM (1988) Role of insulin resistance
tion and validation through genetics. Drug in human disease. Diabetes 37:1595–1607
Discov Today TARGETS 3:183–190 10. Ritzel RA, Butler AE, Rizza RA, Veldhuis JD,
3. Sams-Dodd F (2005) Target-based drug dis- Butler PC (2006) Relationship between b-cell
covery: is something wrong? Drug Discov mass and fasting blood glucose concentration
Today 10:139–147 in humans. Diabetes Care 29:717–718
4. Paul SM, Mytelka DS, Dunwiddie CT, Per- 11. The Diabetes Control and Complications
singer CC, Munos BH, Lindborg SR, Schact Trial Research Group (1993) The effect of
AL (2010) How to improve R&D productiv- intensive treatment of diabetes on the devel-
ity: the pharmaceutical industry’s grand chal- opment and progression of long-term compli-
lenge. Nat Rev Drug Discov 9:203–214 cations in insulin-dependent diabetes
5. Bouzakri K, Koistinen HA, Zierath JR (2005) mellitus. N Engl J Med 329:977–986
Molecular mechanisms of skeletal muscle 12. UK Prospective Diabetes Study (UKPDS)
insulin resistance in type 2 diabetes. Curr Dia- Group (1998) Effect of intensive blood-
betes Rev 1:167–174 glucose control with metformin on complica-
6. Gerich JE, Mitrakou A, Kelley D, tions in overweight patients with type 2 diabe-
Mandarino L, Nurjhan N, Reilly J, tes (UKPDS 34). Lancet 352:854–865
Jenssen T, Veneman T, Consoli A (1990) 13. UK Prospective Diabetes Study (UKPDS)
Contribution of impaired muscle glucose Group (1998) Intensive blood-glucose con-
clearance to reduced postabsorptive systemic trol with sulphonylureas or insulin compared
glucose clearance in NIDDM. Diabetes with conventional treatment and risk of com-
39:211–216 plications in patients with type 2 diabetes
7. Consoli A (1992) Role of liver in pathophysi- (UKPDS 33). Lancet 352:837–853
ology of NIDDM. Diabetes Care
15:430–441
Drug Discovery in Type 2 Diabetes 21
14. American Diabetes Association (2011) Exec- 25. Gribble FM, Tucker SJ, Seino S, Ashcroft FM
utive summary: standards of medical care in (1998) Tissue specificity of sulfonylureas:
diabetes - 2011. Diabetes Care 34:S4–S10 studies on cloned cardiac and beta-cell K
15. Nathan DM, Buse JB, Davidson MB, (ATP) channels. Diabetes 47:1412–1418
Ferrannini E, Holman RR, Sherwin R, Zin- 26. Zünkler BJ, Lenzen S, M€anner K, Panten U,
man B (2009) Medical management of hyper- Trube G (2014) Concentration-dependent
glycemia in type 2 diabetes: a consensus effects of tolbutamide, meglitinide, glipizide,
algorithm for the initiation and adjustment glibenclamide and diazoxide on
of therapy: a consensus statement of the ATP-regulated K+ currents in pancreatic
American Diabetes Association and the b-cells. Naunyn Schmiedebergs Arch Pharma-
European Association for the Study of Diabe- col 337:225–230
tes. Diabetes Care 32:193–203 27. Sturgess NC, Kozlowski RZ, Carrington CA,
16. Nathan DM, Buse JB, Davidson MB, Heine Hales CN, Ashford ML (1988) Effects of sul-
RJ, Holman RR, Sherwin R, Zinman B phonylureas and diazoxide on insulin secre-
(2006) Management of hyperglycemia in tion and nucleotide-sensitive channels in an
type 2 diabetes: a consensus algorithm for insulin-secreting cell line. Br J Pharmacol
the initiation and adjustment of therapy: a 95:83–94
consensus statement from the American Dia- 28. Splitter S, Brown FR, Friskey RW, Grindell L,
betes Association and the European Associa- Kinsell LW (1956) Treatment of diabetic
tion for the study of diabetes. Diabetes Care patients-observations on the use of carbuta-
29:1963–1972 mide and tolbutamide. Calif Med
17. The ADVANCE Collaborative Group (2008) 85:285–288
Intensive blood glucose control and vascular 29. Graal MB, Wolffenbuttel BHR (1999) The
outcomes in patients with type 2 diabetes. N use of sulphonylureas in the elderly. Drugs
Engl J Med 358:2560–2572 Aging 15:471–481
18. The Action to Control Cardiovascular Risk in 30. Malabu UH, Vangaveti VN, Kennedy LR
Diabetes Study Group (2008) Effects of (2014) Disease burden evaluation of fall-
intensive glucose lowering in type 2 diabetes. related events in the elderly due to hypoglyce-
N Engl J Med 358:2545–2559 mia and other diabetic complications: a clini-
19. Herman WH, Dirani RG, O’Neill MC, cal review. Clin Epidemiol 6:287–294
Kravitz B, Heise A, Bakst A, Freed.M.I. 31. Bailey CJ, Day C (2004) Metformin: its
(2005) Reduction in use of healthcare services botanical background. Pract Diabetes Int
with combination sulfonylurea and rosiglita- 21:115–117
zone: findings from the Rosiglitazone Early vs 32. Bailey CJ (1992) Biguanides and NIDDM.
SULfonylurea Titration (RESULT) study. Diabetes Care 15:755–772
Am J Manag Care 11:273–278
33. Tucker GT, Casey C, Phillips PJ, Connor H,
20. Proks P, Reimann F, Green N, Gribble F, Ward JD, Woods HF (1981) Metformin
Ashcroft F (2002) Sulfonylurea stimulation kinetics in healthy subjects and in patients
of insulin secretion. Diabetes 51:S368–S376 with diabetes mellitus. Br J Clin Pharmacol
21. Nichols CG (2006) KATP channels as molec- 12:235–246
ular sensors of cellular metabolism. Nature 34. Zhou M, Xia L, Wang J (2007) Metformin
440:470–476 transport by a newly cloned proton-
22. Ashcroft FM (2007) ATP-sensitive K channels stimulated organic cation transporter (Plasma
and disease: from molecule to malady. Am J Membrane Monoamine Transporter)
Physiol Endocrinol Metab 293:E880–E889 expressed in human intestine. Drug Metab
23. Inagaki N, Gonoi T, Clement JP, Wang CZ, Dispos 35:1956–1962
Aguilar-Bryan L, Bryan J, Seino S (1996) A 35. Gong L, Goswami S, Giacomini KM, Altman
family of sulfonylurea receptors determines RB, Klein TE (2012) Metformin pathways:
the pharmacological properties of pharmacokinetics and pharmacodynamics.
ATP-sensitive K+ channels. Neuron Pharmacogenet Genomics 22:820–827
16:1011–1017 36. Graham GG, Punt J, Arora M, Day RO, Doo-
24. Aittoniemi J, Fotinou C, Craig TJ, de Wet H, gue MP, Duong J, Furlong TJ, Greenfield JR,
Proks P, Ashcroft FM (2009) SUR1: a unique Greenup LC, Kirkpatrick CM, Ray JE,
ATP-binding cassette protein that functions Timmins P, Williams KM (2011) Clinical
as an ion channel regulator. Philos Trans R pharmacokinetics of metformin. Clin Phar-
Soc B 364:257–267 macokinet 50:81–98
22 John C. Clapham
37. Strack T (2008) Metformin: a review. Drugs intake in obese subjects. Eur J Clin Invest
Today 44:303–314 28:441–446
38. Campbell RK, White JR, Saulie BA (1996) 52. Bruijstens LA, van Luin M, Buscher-
Metformin: a new oral biguanide. Clin Ther Jungerhans PM, Bosch FH (2008) Reality of
18:360–371 severe metformin-induced lactic acidosis in
39. DeFronzo RA, Goodman AM (1995) Efficacy the absence of chronic renal impairment.
of metformin in patients with non-insulin- Neth J Med 66:185–190
dependent diabetes mellitus. N Engl J Med 53. Carter D, Howlett HCS, Wiernsperger NF,
333:541–549 Bailey CJ (2003) Differential effects of met-
40. Bailey CJ, Turner RC (1996) Metformin. N formin on bile salt absorption from the jeju-
Engl J Med 334:574–579 num and ileum. Diabetes Obes Metab
41. Stumvoll M, Nurjhan N, Perriello G, 5:120–125
Dailey G, Gerich JE (1995) Metabolic effects 54. Davidson J, Howlett H (2004) New
of metformin in non-insulin-dependent dia- prolonged-release metformin improves gas-
betes mellitus. N Engl J Med 333:550–554 trointestinal tolerability. Br J Diabetes Vasc
42. Hundal RS, Krssak M, Dufour S, Laurent D, Dis 4:273–277
Lebon V, Chandramouli V, Inzucchi SE, 55. Mallick S (2004) Metformin induced acute
Schumann WC, Petersen KF, Landau BR, pancreatitis precipitated by renal failure. Post-
Shulman GI (2000) Mechanism by which grad Med J 80:239–240
metformin reduces glucose production in 56. Alsubaie S, Almalki MH (2013) Metformin
type 2 diabetes. Diabetes 49:2063–2069 induced acute pancreatitis. Derm Endocrinol
43. Klip A, Leiter LA (1990) Cellular mechanism 5:317–318
of action of metformin. Diabetes Care 57. Oprea TI, Bauman JE, Bologa CG,
13:696–704 Buranda T, Chigaev A, Edwards BS, Jarvik
44. Fryer LGD, Parbu-Patel A, Carling D (2002) JW, Gresham HD, Haynes MK, Hjelle B,
The anti-diabetic drugs rosiglitazone and Hromas R, Hudson L, Mackenzie DA, Muller
metformin stimulate AMP-activated protein CY, Reed JC, Simons PC, Smagley Y,
kinase through distinct signaling pathways. J Strouse J, Surviladze Z, Thompson T,
Biol Chem 277:25226–25232 Ursu O, Waller A, Wandinger-Ness A, Winter
45. Hardie DG (2008) AMPK: a key regulator of SS, Wu Y, Young SM, Larson RS, Willman C,
energy balance in the single cell and the whole Sklar LA (2011) Drug repurposing from an
organism. Int J Obes Relat Metab Disord 32: academic perspective. Drug Discov Today
S7–S12 Ther Strat 8:61
46. Ruderman N, Prentki M (2004) AMP-kinase 58. Kasznicki J, Sliwinska A, Drzewoski J (2014)
and malonyl-CoA: targets for therapy of the Metformin in cancer prevention and therapy.
metabolic syndrome. Nat Rev Drug Discov Ann Transl Med 2:57
3:340–351 59. Diamanti-Kandarakis E, Christakou CD,
47. Owen MR, Doran E, Halestrap AP (2000) Kandaraki E, Economou FN (2010) Metfor-
Evidence that metformin exerts its anti- min: an old medication of new fashion: evol-
diabetic effects through inhibition of complex ving new molecular mechanisms and clinical
1 of the mitochondrial respiratory chain. Bio- implications in polycystic ovary syndrome.
chem J 348:607–614 Eur J Endocrinol 162:193–212
48. Sakamoto K, Göransson O, Hardie DG, Alessi 60. Scarpello JHB, Howlett HCS (2008) Metfor-
DR (2004) Activity of LKB1 and AMPK- min therapy and clinical uses. Diabetes Vasc
related kinases in skeletal muscle: effects of Dis Res 5:157–167
contraction, phenformin, and AICAR. Am J 61. Hirst JA, Farmer AJ, Dyar A, Lung TWC,
Physiol Endocrinol Metab 287:E310–E317 Stevens RJ (2013) Estimating the effect of
49. Golay A (2007) Metformin and body weight. sulfonylurea on HbA1c in diabetes: a system-
Int J Obes Relat Metab Disord 32:61–72 atic review and meta-analysis. Diabetologia
56:973–984
50. Clapham JC, Arch JR (2008) Influencing
energy expenditure and substrate utilisation. 62. Rydberg T, Jönsson A, Karlsson MO, Melan-
In: Pharmacotherapy of obesity, Milestones in der A (1997) Concentration-effect relations
drug therapy. Birkh€auser, Basel, pp 101–115 of glibenclamide and its active metabolites in
man: modelling of Pharmacokinetics and
51. Paolisso G, Amato L, Eccellente R, Pharmacodynamics. Br J Clin Pharmacol
Gambardella A, Tagliamonte MR, 43:373–381
Varricchio G, Carella C, Giugliano D, D’ono-
frio F (1998) Effect of metformin on food
Drug Discovery in Type 2 Diabetes 23
63. Kola I, Landis J (2004) Can the pharmaceuti- and therapeutic possibilities. Prog Drug Res
cal industry reduce attrition rates? Nat Rev 51:33–94
Drug Discov 3:711–716 75. Henry RR (1997) Thiazolidinediones. Endo-
64. Rosengren A, Jing X, Eliasson L, Renstrøm E crinol Metab Clin 26:553–573
(2008) Why treatment fails in type 2 diabetes. 76. Gale EAM (2006) Troglitazone: the lesson
PLoS Med 5:e215 that nobody learned? Diabetologia 49:1–6
65. Kahn SE, Haffner SM, Heise MA, Herman 77. Barnett AH (2009) Redefining the role of
WH, Holman RR, Jones NP, Kravitz BG, thiazolidinediones in the management of
Lachin JM, O’Neill MC, Zinman B, Viberti type 2 diabetes. Vasc Health Risk Manag
G (2006) Glycemic durability of rosiglita- 5:141–151
zone, metformin, or glyburide monotherapy. 78. Phillips LS, Grunberger G, Miller E,
N Engl J Med 355:2427–2443 Patwardhan R, Rappaport EB, Salzman A
66. Karam JH, Sanz N, Salamon E, Nolte MS (2001) Once- and twice-daily dosing with
(1986) Selective unresponsiveness of pancre- rosiglitazone improves glycemic control in
atic b-cells to acute sulfonylurea stimulation patients with type 2 diabetes. Diabetes Care
during sulfonylurea therapy in NIDDM. Dia- 24:308–315
betes 35:1314–1320 79. Fonseca V, Rosenstock J, Patwardhan R, Salz-
67. Pantalone KM, Kattan MW, Yu C, Wells BJ, man A (2000) Effect of metformin and rosi-
Arrigain S, Jain A, Atreja A, Zimmerman RS glitazone combination therapy in patients
(2012) Increase in overall mortality risk in with type 2 diabetes mellitus: a randomized
patients with type 2 diabetes receiving glipi- controlled trial. JAMA 283:1695–1702
zide, glyburide or glimepiride monotherapy 80. Poitout V (2004) b-cell lipotoxicity: burning
versus metformin: a retrospective analysis. fat into heat? Endocrinology 145:3563–3565
Diabetes Obes Metab 14:803–809
81. Nolan CJ, Prentki M (2008) The islet b-cell:
68. Rustenbeck I, Wienbergen A, Bleck C, Jörns fuel responsive and vulnerable. Trends Endo-
A (2004) Desensitization of insulin secretion crinol Metab 19:285–291
by depolarizing insulin secretagogues. Diabe-
tes 53:S140–S150 82. Buckingham RE, Al-Barazanji KA, Toseland
CDN, Slaughter M, Connor SC, West A,
69. Sadikot SM, Mogensen CE (2008) Risk of Bond B, Turner NC, Clapham JC (1998)
coronary artery disease associated with initial Peroxisome proliferator-activated receptor-g
sulphonylurea treatment of patients with type agonist, rosiglitazone, protects against
2 diabetes: a matched case-control study. Dia- nephropathy and pancreatic islet abnormal-
betes Res Clin Pract 82:391–395 ities in Zucker fatty rats. Diabetes
70. Lefer DJ, Nichols CG, Coetzee WA (2009) 47:1326–1334
Sulfonylurea receptor 1 subunits of 83. Nissen SE, Wolski K (2007) Effect of rosigli-
ATP-sensitive potassium channels and myo- tazone on the risk of myocardial infarction
cardial Ischemia/reperfusion Injury. Trends and death from cardiovascular causes. N
Cardiovasc Med 19:61–67 Engl J Med 356:2457–2471
71. Bell DSH (2006) Do sulfonylurea drugs 84. Singh S, Loke YK, Furberg CD (2007) Thia-
increase the risk of cardiac events? Can Med zolidinediones and heart failure: a teleo-
Assoc J 174:185–186 analysis. Diabetes Care 30:2148–2153
72. Grygiel-Górniak B (2014) Peroxisome 85. Bloomgarden ZT (2007) The avandia debate.
proliferator-activated receptors and their Diabetes Care 30:2401–2408
ligands: nutritional and clinical implications -
a review. Nutr J 13:17 86. Misbin RI (2007) Lessons from the avandia
controversy: a new paradigm for the develop-
73. Glosli H, Gudbrandsen OA, Mullen AJ, ment of drugs to treat type 2 diabetes. Diabe-
Halvorsen B, Røst TH, Wergedahl H, tes Care 30:3141–3144
Prydz H, Aukrust P, Berge RK (2005)
Down-regulated expression of PPARa target 87. Bhatt DL, Chew DP, Grines C, Mukherjee D,
genes, reduced fatty acid oxidation and Leesar M, Gilchrist IC, Corbelli JC, Blanken-
altered fatty acid composition in the liver of ship JC, Eres A, Steinhubl S, Tan WA, Resar
mice transgenic for hTNFa. Biochim Biophys JR, Al Mahameed A, Abdel-Latif A, Tang
Acta 1734:235–246 HW, Brennan D, McErlean E, Hazen SL,
Topol EJ (2007) Peroxisome proliferator-
74. Turner NC, Clapham JC (1998) Insulin resis- activated receptor g agonists for the preven-
tance, impaired glucose tolerance and non- tion of adverse events following percutaneous
insulin-dependent diabetes, pathologic coronary revascularizationG-results of the
mechanisms and treatment: current status PPAR study. Am Heart J 154:137–143
24 John C. Clapham
88. McAfee AT, Koro C, Landon J, Ziyadeh N, 101. Hanefeld M, Fischer S, Schulze J,
Walker AM (2007) Coronary heart disease Spengler M, Wargenau M, Schollberg K,
outcomes in patients receiving antidiabetic Fücker K (1991) Therapeutic potentials of
agents. Pharmacoepidem Drug Safe acarbose as first-line drug in NIDDM insuffi-
16:711–725 ciently treated with diet alone. Diabetes Care
89. Home PD, Pocock SJ, Beck-Nielsen H, 14:732–737
Gomis R, Hanefeld M, Dargie H, 102. Salman S, Salman F, Satman I, Yilmaz Y,
Komajda M, Gubb J, Biswas N, Jones NP Özer E, Sengül A, Özer H, Demirel HO,
(2005) Rosiglitazone evaluated for cardiac Karsidag K, Dinççag N, Yilmaz MT (2001)
outcomes and regulation of glycaemia in dia- Comparison of acarbose and gliclazide as first-
betes (RECORD): study design and protocol. line agents in patients with type 2 diabetes.
Diabetologia 48:1726–1735 Curr Med Res Opin 16:296–306
90. Hillaire-Buys D, Faillie JL, Montastruc JL 103. Wang G, Liu J, Yang N, Gao X, Fan H, Xu Y,
(2011) Pioglitazone and bladder cancer. Lan- Yang W (2014) MARCH2:comparative
cet 378:1543–1544 assessment of therapeutic effects of acarbose
91. Grey A (2009) Thiazolidinedione-induced and metformin in newly diagnosed type 2 dia-
skeletal fragility - mechanisms and implica- betes patients. PLoS One 9:e105698
tions. Diabetes Obes Metab 11:275–284 104. Dabhi AS, Bhatt NR, Shah MJ (2013) Vogli-
92. Oshitari T, Asaumi N, Watanabe M, bose: an alpha glucosidase inhibitor. J Clin
Kumagai K, Mitamura Y (2008) Severe mac- Diagn Res 7:3023–3027
ular edema induced by pioglitazone in a 105. Standl E, Theodorakis MJ, Erbach M,
patient with diabetic retinopathy: a case Schnell O, Tuomilehto J (2014) On the
study. Vasc Health Risk Manag 4:1137–1140 potential of acarbose to reduce cardiovascular
93. Liazos E, Broadbent DM, Beare N, Kumar N disease. Cardiovasc Diabetol 13:81
(2008) Spontaneous resolution of diabetic 106. Ceriello A, Taboga C, Giacomello R, Stel L,
macular oedema after discontinuation of thia- Motz E, Pirisi M (1996) Post-meal coagula-
zolidenediones. Diabet Med 25:860–862 tion activation in diabetes mellitus: the effect
94. Cordain L, Eaton SB, Sebastian A, Mann N, of acarbose. Diabetologia 39:469–473
Lindeberg S, Watkins BA, O’Keefe JH, 107. Kawano H, Motoyama T, Hirashima O,
Brand-Miller J (2005) Origins and evolution Hirai N, Miyao Y, Sakamoto T, Kugiyama K,
of the Western diet: health implications for Ogawa H, Yasue H (1999) Hyperglycemia
the 21st century. Am J Clin Nutr 81:341–354 rapidly suppresses flow-mediated endothe-
95. Prentice AM, Jebb SA (1995) Obesity in Brit- lium-dependent vasodilation of brachial
ain: gluttony or sloth? Br Med J 311:437–439 artery. J Am Coll Cardiol 34:146–154
96. Röder PV, Geillinger KE, Zietek TS, 108. Heitzer T, Schlinzig T, Krohn K, Meinertz T,
Thorens B, Koepsell H, Hannelore D (2014) Münzel T (2001) Endothelial dysfunction,
The role of SGLT1 and GLUT2 in intestinal oxidative stress, and risk of cardiovascular
glucose transport and sensing. PLoS One 9: events in patients with coronary artery dis-
e89977 ease. Circulation 104:2673–2678
97. Caspary WF (1992) Physiology and patho- 109. Caumo A, Luzi L (2004) First-phase insulin
physiology of intestinal absorption. Am J secretion: does it exist in real life? Considera-
Clin Nutr 55:299S–308S tions on shape and function. Am J Physiol
98. Chiba S (1997) Molecular mechanism in Endocrinol Metab 287:E371–E385
alpha-glucosidase and glucoamylase. Biosci 110. Lewis GF, Zinman B, Groenewoud Y,
Biotechnol Biochem 61:1233–1239 Vranic M, Giacca A (1996) Hepatic glucose
99. Göke B, Fuder H, Wieckhorst G, Theiss U, production is regulated both by direct hepatic
Stridde E, Littke T, Kleist P, Arnold R, Lücker and extrahepatic effects of insulin in Humans.
PW (1995) Voglibose (AO-128) is an efficient Diabetes 45:454–462
a-glucosidase inhibitor and mobilizes the 111. Calles-Escandon J, Robbins DC (1987) Loss
endogenous GLP-1 reserve. Digestion of early phase of insulin release in humans
56:493–501 impairs glucose tolerance and blunts thermic
100. Standl E, Schernthaner G, Rybka J, effect of glucose. Diabetes 36:1167–1172
Hanefeld M, Raptis SA, Naditch L (2001) 112. Del Prato S, Miccoli R, Penno G (2005) The
Improved glycaemic control with miglitol in importance of effective early phase insulin
inadequately-controlled type 2 diabetics. Dia- secretion. Br J Diabetes Vasc Dis 5:198–202
betes Res Clin Pract 51:205–213 113. Gerich JE (2002) Is reduced first-phase insu-
lin release the earliest detectable abnormality
Drug Discovery in Type 2 Diabetes 25
acting human glucagon-like peptide 1 ana- Ippolito DE, Kim D, Lyons KA, Ok HO,
logue, improves human islet survival in cul- Patel RA, Petrov AN, Pryor KA, Qian X,
ture. Transpl Int 23:259–265 Reigle L, Woods A, Wu JK, Zaller D,
155. Bregenholt S, Møldrup A, Blume N, Karlsen Zhang X, Zhu L, Weber AE, Thornberry
AE, Friedrichsen BN, Tornhave D, Knudsen NA (2005) Dipeptidyl peptidase IV inhibi-
LB, Petersen JS (2005) The long-acting glu- tion for the treatment of type 2 diabetes:
cagon-like peptide-1 analogue, liraglutide, potential importance of selectivity over dipep-
inhibits b-cell apoptosis in vitro. Biochem tidyl peptidases 8 and 9. Diabetes
Biophys Res Commun 330:577–584 54:2988–2994
156. Butler AE, Janson J, Bonner-Weir S, Ritzel R, 166. Lambeir A-M, Durinx C, Scharpé S, De Mee-
Rizza RA, Butler PC (2003) b-Cell deficit and ster I (2013) Dipeptidyl-peptidase IV from
Increased β-cell apoptosis in humans with bench to bedside: an update on structural
type 2 diabetes. Diabetes 52:102–110 properties, functions, and clinical aspects of
157. Bosco D, Armanet M, Morel P, Niclauss N, the enzyme DPPIV. Crit Rev Clin Lab Sci
Sgroi A, Muller YD, Giovannoni L, 40:209–294
Parnaud G, Berney T (2010) Unique arrange- 167. Durinx C, Lambeir AM, Bosmans E, Fal-
ment of α- and β-cells in human Islets of magne JB, Berghmans R, Haemers A,
Langerhans. Diabetes 59:1202–1210 Scharpé S, De Meester I (2000) Molecular
158. Cabrera O, Berman DM, Kenyon NS, characterization of dipeptidyl peptidase activ-
Ricordi C, Berggren PO, Caicedo A (2006) ity in serum. Eur J Biochem 267:5608–5613
The unique cytoarchitecture of human pan- 168. Kos K, Baker AR, Jernas M, Harte AL, Clap-
creatic islets has implications for islet cell func- ham JC, O’Hare JP, Carlsson L, Kumar S,
tion. Proc Natl Acad Sci U S A McTernan PG (2009) DPP-IV inhibition
103:2334–2339 enhances the antilipolytic action of NPY in
159. Steiner DJ, Kim A, Miller K, Hara M (2010) human adipose tissue. Diabetes Obes Metab
Pancreatic islet plasticity: interspecies compar- 11:285–292
ison of islet architecture and composition. 169. Engel M, Hoffmann T, Wagner L,
Islets 2:135–145 Wermann M, Heiser U, Kiefersauer R,
160. Butler PC, Elashoff M, Elashoff R, Gale EAM Huber R, Bode W, Demuth HU, Brandstetter
(2013) A critical analysis of the clinical use of H (2003) The crystal structure of dipeptidyl
incretin-based therapies: are the GLP-1 thera- peptidase IV (CD26) reveals its functional
pies safe? Diabetes Care 36:2118–2125 regulation and enzymatic mechanism. Proc
Natl Acad Sci U S A 100:5063–5068
161. Perfetti R, Zhou J, Doyle ME, Egan JM
(2000) Glucagon-like peptide-1 induces cell 170. Aertgeerts K, Ye S, Tennant MG, Kraus ML,
proliferation and pancreatic-duodenum Rogers J, Sang BC, Skene RJ, Webb DR,
homeobox-1 expression and increases endo- Prasad GS (2004) Crystal structure of
crine cell mass in the pancreas of old, glucose human dipeptidyl peptidase IV in complex
intolerant rats. Endocrinology with a decapeptide reveals details on substrate
141:4600–4605 specificity and tetrahedral intermediate for-
mation. Protein Sci 13:412–421
162. Nauck MA, Friedrich N (2013) Do GLP-1-
based therapies increase cancer risk? Diabetes 171. Sebokova E, Christ AD, Boehringer M, Miz-
Care 36:S245–S252 rahi J (2015) Dipeptidyl peptidase IV inhibi-
tors: the next generation of new promising
163. Noel RA, Braun DK, Patterson RE, Bloomg- therapies for the management of type 2 diabe-
ren GL (2009) Increased risk of acute pancre- tes. Curr Top Med Chem 7:547–555
atitis and biliary disease observed in patients
with type 2 diabetes: a retrospective cohort 172. Shubrook J, Colucci R, Guo A, Schwartz F
study. Diabetes Care 32:834–838 (2011) Saxagliptin: a selective DPP-4 inhibi-
tor for the treatment of type 2 diabetes melli-
164. David F, Bernard AM, Pierres M, Marguet D tus. Clin Med Insights Endocrinol Diab
(1993) Identification of serine 624, aspartic 4:1–12
acid 702, and histidine 734 as the catalytic
triad residues of mouse dipeptidyl-peptidase 173. Toth PP (2015) Overview of saxagliptin effi-
IV (CD26). A member of a novel family of cacy and safety in patients with type 2 diabetes
nonclassical serine hydrolases. J Biol Chem and cardiovascular disease or risk factors for
268:17247–17252 cardiovascular disease. Vasc Health Risk
Manag 11:9–23
165. Lankas GR, Leiting B, Roy RS, Eiermann GJ,
Beconi MG, Biftu T, Chan CC, 174. Banerjee M, Younis N, Soran H (2009) Vil-
Edmondson S, Feeney WP, Huaibing H, dagliptin in clinical practice: a review of
28 John C. Clapham
literature. Expert Opin Pharmacother 186. Wright EM, Loo DDF, Hirayama BA (2011)
10:2745–2757 Biology of human sodium glucose transpor-
175. Dhillon S (2010) Sitagliptin: a review of its ters. Physiol Rev 91:733–794
use in the management of type 2 diabetes 187. Wright EM (2001) Renal Na+-glucose
mellitus. Drugs 70:489–512 cotransporters. Am J Physiol Renal Physiol
176. Ahrén B (2014) Insulin plus incretin: a 280:F10–F18
glucose-lowering strategy for type 188. Mackenzie B, Loo DDF, Panayotova-
2-diabetes. World J Diabetes 5:40–51 Heiermann M, Wright EM (1996) Biophysi-
177. Pratley RE, Schweizer A, Rosenstock J, Foley cal characteristics of the pig kidney Na+/Glu-
JE, Banerji MA, Pi-Sunyer FX, Mills D, Deja- cose cotransporter SGLT2 reveal a common
ger S (2008) Robust improvements in fasting mechanism for SGLT1 and SGLT2. J Biol
and prandial measures of b-cell function with Chem 271:32678–32683
vildagliptin in drug-naı̈ve patients: analysis of 189. Wells RG, Pajor AM, Kanai Y, Turk E, Wright
pooled vildagliptin monotherapy database. EM, Hediger MA (1992) Cloning of a human
Diabetes Obes Metab 10:931–938 kidney cDNA with similarity to the sodium-
178. Del Prato S, Barnett AH, Huisman H, glucose cotransporter. Am J Physiol Renal
Neubacher D, Woerle HJ, Dugi KA (2011) Physiol 263:F459–F465
Effect of linagliptin monotherapy on glycae- 190. Kanai Y, Lee WS, You G, Brown D, Hediger
mic control and markers of b-cell function in MA (1994) The human kidney low affinity Na
patients with inadequately controlled type +/glucose cotransporter SGLT2. Delineation
2 diabetes: a randomized controlled trial. Dia- of the major renal reabsorptive mechanism for
betes Obes Metab 13:258–267 D-glucose. J Clin Invest 93:397–404
179. Ahrén B, Pacini G, Tura A, Foley JE, Schwei- 191. DeFronzo RA, Hompesch M,
zer A (2007) Improved meal-related insulin Kasichayanula S, Liu X, Hong Y, Pfister M,
processing contributes to the enhancement of Morrow LA, Leslie BR, Boulton DW,
b-cell function by the DPP-4 inhibitor vlda- Ching A, LaCreta FP, Griffen SC (2013)
gliptin in patients with type 2 diabetes. Horm Characterization of renal glucose reabsorp-
Metab Res 39:826–829 tion in response to dapagliflozin in healthy
180. Yeom JA, Kim ES, Park HS, Ham DS, Sun K, subjects and subjects with type 2 diabetes.
Kim JW, Cho JH, Yoon KH (2011) Both Diabetes Care 36:3169–3176
sitagliptin analogue & pioglitazone preserve 192. Calado J, Soto K, Clemente C, Correia P,
the beta-cell proportion in the islets with dif- Rueff J (2004) Novel compound heterozy-
ferent mechanism in non-obese and obese gous mutations in SLC5A2 are responsible
diabetic mice. BMB Rep 44:713–718 for autosomal recessive renal glucosuria.
181. Girgis CM, Champion BL (2011) Hum Genet 114:314–316
Vildagliptin-induced acute pancreatitis. 193. van den Heuvel L, Assink K, Willemsen M,
Endocr Pract 17:e48–e50 Monnens L (2002) Autosomal recessive renal
182. Lee CF, Sun MS, Tai YK (2014) Saxagliptin- glucosuria attributable to a mutation in the
induced recurrent acute pancreatitis. Intern sodium glucose cotransporter (SGLT2).
Med 53:1351–1354 Hum Genet 111:544–547
183. Chang CH, Lin JW, Chen ST, Lai MS, Cuang 194. Gallo LA, Wright EM, Vallon V (2015) Prob-
LM, Chang YC (2016) Dipeptidyl peptidase- ing SGLT2 as a therapeutic target for diabe-
4 inhibitor use is not associated with acute tes: basic physiology and consequences.
pancreatitis in high-risk type 2 diabetic Diabetes Vasc Dis Res 12:78–89
patients: a nationwide cohort study. Medicine 195. Rossetti L, Smith D, Shulman GI,
95:e2603–e2609 Papachristou D, DeFronzo RA (1987) Cor-
184. Karagiannis T, Bekiari E, Boura P, Tsapas A rection of hyperglycemia with phlorizin nor-
(2015) Cardiovascular risk with DPP-4 inhi- malizes tissue sensitivity to insulin in diabetic
bitors: latest evidence and clinical implica- rats. J Clin Invest 79:1510–1515
tions. Ther Adv Drug Saf 7:36–38 196. Isaji M (2011) SGLT2 inhibitors: molecular
185. Brunton S (2014) GLP-1 receptor design and potential differences in effect. Kid-
agonists vs. DPP-4 inhibitors for type 2 diabe- ney Int 79:S14–S19
tes: is one approach more successful or prefer- 197. Nomura S, Sakamaki S, Hongu M,
able than the other? Int J Clin Pract Kawanishi E, Koga Y, Sakamoto T,
68:557–567 Yamamoto Y, Ueta K, Kimata H,
Nakayama K, Tsuda-Tsukimoto M (2010)
Discovery of canagliflozin, a novel
Drug Discovery in Type 2 Diabetes 29
C-glucoside with thiophene ring, as sodium- 208. Anderson SL (2014) Dapagliflozin efficacy
dependent glucose cotransporter 2 inhibitor and safety: a perspective review. Ther Adv
for the treatment of type 2 diabetes Mellitus. J Drug Saf 5:242–254
Med Chem 53:6355–6360 209. Bolinder J, Ljunggren Ö, Kullberg J,
198. Meng W, Ellsworth BA, Nirschl AA, McCann Johansson L, Wilding J, Langkilde AM,
PJ, Patel M, Girotra RN, Wu G, Sher PM, Sugg J, Parikh S (2011) Effects of dapagliflo-
Morrison EP, Biller SA, Zahler R, Deshpande zin on body weight, total fat mass, and
PP, Pullockaran A, Hagan DL, Morgan N, regional adipose tissue distribution in patients
Taylor JR, Obermeier MT, Humphreys WG, with type 2 diabetes mellitus with inadequate
Khanna A, Discenza L, Robertson JG, glycemic control on metformin. J Clin Endo-
Wang A, Han S, Wetterau JR, Janovitz EB, crinol Metabol 97:1020–1031
Flint OP, Whaley JM, Washburn WN (2008) 210. Nauck MA, Del Prato S, Meier JJ, Durán-
Discovery of dapagliflozin: a potent, selective Garcı́a S, Rohwedder K, Elze M, Parikh SJ
renal sodium-dependent glucose cotranspor- (2011) Dapagliflozin versus glipizide as
ter 2 (SGLT2) inhibitor for the treatment of add-on therapy in patients with type 2 diabetes
type 2 diabetes. J Med Chem 51:1145–1149 who have inadequate glycemic control with
199. Neumiller JJ (2014) Empagliflozin: a new metformin: a randomized, 52-week, double-
sodium-glucose co-transporter 2 (SGLT2) blind, active-controlled noninferiority trial.
inhibitor for the treatment of type 2 diabetes. Diabetes Care 34:2015–2022
Drugs Context 3:212262 211. Strojek K, Yoon KH, Hruba V, Sugg J,
200. Hummel CS, Lu C, Liu J, Ghezzi C, Hir- Langkilde A, Parikh S (2014) Dapagliflozin
ayama BA, Loo DDF, Kepe V, Barrio JR, added to glimepiride in patients with type
Wright EM (2011) Structural selectivity of 2 diabetes mellitus sustains glycemic control
human SGLT inhibitors. Am J Physiol Cell and weight loss Over 48-weeks: a rando-
Physiol 302:C373–C382 mized, double-blind, parallel-group,
201. Valentine V (2015) The role of the kidney and placebo-controlled trial. Diabetes Ther
sodium-glucose cotransporter-2 inhibition in 5:267–283
diabetes management. Clin Diab 30:151–155 212. Geerlings S, Fonseca V, Castro-Diaz D, List J,
202. Kalra S (2014) Sodium glucose co-transpor- Parikh S (2014) Genital and urinary tract
ter-2 (SGLT2) inhibitors: a review of their infections in diabetes: impact of
basic and clinical pharmacology. Diabetes pharmacologically-induced glucosuria. Dia-
Ther 5:355–366 betes Res Clin Pract 103:373–381
203. Dietrich E, Powell J, Taylor JR (2013) Cana- 213. Sonesson C, Johansson PA, Johnsson E,
gliflozin: a novel treatment option for type Gause-Nilsson I (2016) Cardiovascular
2 diabetes. Drug Des Devel Ther effects of dapagliflozin in patients with type
7:1399–1408 2 diabetes and different risk categories: a
204. Aylsworth A, Dean Z, VanNorman C, Okere meta-analysis. Cardiovasc Diabetol 15:37
AN (2014) Dapagliflozin for the treatment of 214. MacIsaac RJ, Jerums G, Ekinci EI (2016)
type 2 diabetes mellitus. Ann Pharmacother Cardio-renal protection with empagliflozin.
48:1202–1208 Ann Transl Med 4:409–412
205. Heise T, Seman L, Macha S, Jones P, 215. Liakos A, Karagiannis T, Bekiari E, Boura P,
Marquart A, Pinnetti S, Woerle HJ, Dugi K Tsapas A (2014) Update on long-term effi-
(2013) Safety, tolerability, pharmacokinetics, cacy and safety of dapagliflozin in patients
and pharmacodynamics of multiple rising with type 2 diabetes mellitus. Ther Adv Endo-
doses of empagliflozin in patients with type crinol Metab 6:61–67
2 diabetes mellitus. Diabetes Ther 4:331–345 216. Sosale B, Sosale A, Bhattacharyya A (2016)
206. Kasichayanula S, Liu X, LaCreta F, Griffen Clinical effectiveness and impact on insulin
SC, Boulton DW (2014) Clinical pharmaco- therapy cost after addition of dapagliflozin to
kinetics and pharmacodynamics of dapagliflo- patients with uncontrolled type 2 diabetes.
zin, a selective inhibitor of sodium-glucose Diabetes Ther 7:765–776
co-transporter type 2. Clin Pharmacokinet 217. Issandou M, Bouillot A, Brusq J, Forest M,
53:17–27 Grillot D, Guillard R, Martin S, Michiels C,
207. Ghezzi C, Hirayama BA, Gorraitz E, Loo Sulpice T, Daugan A (2009) Pharmacological
DDF, Liang Y, Wright EM (2014) SGLT2 inhibition of Stearoyl-CoA Desaturase
inhibitors act from the extracellular surface 1 improves insulin sensitivity in insulin-
of the cell membrane. Physiol Rep 2:e12058 resistant rat models. Eur J Pharmacol
618:28–36
30 John C. Clapham
218. Lepifre F, Christmann-Franck S, Roche D, drugs and targets for type 2 diabetes: review-
Leriche C, Carniato D, Charon C, Bozec S, ing the evidence. Am Health Drug Benefits
Doare L, Schmidlin F, Lecomte M, Valeur E 7:452–463
(2009) Discovery and structure-guided drug 223. Cornell S (2015) Continual evolution of type
design of inhibitors of 11b-hydroxysteroid- 2 diabetes: an update on pathophysiology and
dehydrogenase type I based on a spiro- emerging treatment options. Ther Clin Risk
carboxamide scaffold. Bioorg Med Chem Manag 11:621–632
Lett 19:3682 224. Morgan S, Grootendorst P, Lexchin J,
219. Birch AM, Buckett LK, Turnbull AV (2010) Cunningham C, Greyson D (2011) The cost
DGAT1 inhibitors as anti-obesity and anti- of drug development: a systematic review.
diabetic agents. Curr Opin Drug Discov Health Policy 100:4–17
Devel 13:489–496 225. Paul SM, Mytelka DS, Dunwiddie CT, Per-
220. Kaiser D, Oetjen E (2014) Something old, singer CC, Munos BH, Lindborg SR, Schacht
something new and something very old: AL (2010) How to improve R&D productiv-
drugs for treating type 2 diabetes. Br J Phar- ity: the pharmaceutical industry’s grand chal-
macol 171:2940–2950 lenge. Nat Rev Drug Discov 9:203–214
221. Ichimura A, Hasegawa S, Kasubuchi M, 226. Herper M (2012) The truly staggering cost of
Kimura I (2014) Free fatty acid receptors as inventing new drugs. Forbes. https://www.
therapeutic targets for the treatment of diabe- forbes.com/sites/matthewherper/2012/
tes. Front Pharmacol 5:236 02/10/the-truly-staggering-cost-of-
222. Miller BR, Nguyen H, Hu CJH, Lin C, inventing-new-drugs/
Nguyen QT (2014) New and emerging
Chapter 2
Abstract
Obesity and type 2 diabetes are serious conditions that have reached pandemic proportions. The underlying
physiology is complex with multiorgan interactions involved and, consequently, multiorgan approaches are
necessary for researchers to elucidate and find treatments. As such, in vivo models are an invaluable resource
for these studies and mice are, for many reasons, by far the most common species used. The use of animals
comes with responsibilities to ensure their welfare and well-being: primarily for the sake of the mice
themselves, but also to ensure quality of data. Physiological stress responses, such as adrenalin (epinephrine)
and corticosterone release among others, have major consequences on metabolism. Additionally, behavioral
stress responses are also a source of data variance. This chapter looks at the main in vivo procedures
incorporated in mouse obesity/diabetes protocols and considers the practical factors involved that can be
considered to minimize animal stress and improve study data quality.
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_2, © Springer Science+Business Media, LLC, part of Springer Nature 2020
31
32 Edward T. Wargent
remaining for the treatment of obesity (the only one licensed in the
UK, for example). However, the effectiveness of orlistat is slim with
an average weight loss of only 2.9 kg (or about 2.3 kg for the over-
the-counter dose). There are more therapeutic options for type
2 diabetes, but the older drugs merely slow the advance of the
disease by a few years (at most), and the newer ones are either
suspected of having a poor benefit-to-risk ratio, or their ability to
halt the advance of the disease has yet to be demonstrated. There is
a need for better treatments for diabetes and obesity in the form of
drugs, nutraceuticals (since much of the diabetes epidemic is occur-
ring in countries and populations that cannot afford Western
drugs), diets or lifestyle modifications. To achieve this, we need to
understand better the causes of the diseases, identify potential
therapeutic targets, and investigate whether and how potential
interventions work. Control of metabolism, growth, and body
composition involve the interaction of many body organs including
brain, liver, adipose tissue, muscle, and endocrine organs. Neither
cell or tissue cultures nor nonvertebrate organisms can replicate
such mammalian whole body systems. Computational methods are
still in a developmental stage, requiring further experimental data
before they can reliably replace whole animals.
The use of animals in scientific research comes with responsi-
bilities to ensure the welfare and ethical treatment of those animals.
It is every in vivo researcher’s duty to explore ways to replace
animals with other means to answer the questions postulated; to
reduce the numbers of animals used; and the refine the procedures
employed to reduce harm and distress experienced by experimental
animals. When studying conditions such as obesity and diabetes,
the reduction of stress is also necessary as it impacts greatly upon
metabolism [2]. Consequently, reducing stress is the single most
important factor to be considered when designing reliable and
reproducible experiments and reducing the variation of the data
generated.
For manifold reasons, including finance, ethics, genetics, time
scales, etc., rodents and mice are by far the most widely employed
species in obesity and diabetes studies and this chapter describes the
typical in vivo procedures used in a manner intended to minimize
stress by exposing the rodents to no more than transient pain,
distress or harm. When carried out in the ways described, each
procedure can be classified as mild. However, animals should be
monitored before and after each procedure to ensure no compound
suffering from consecutive procedures. The recovery times detailed
below are minimal and the health of each animal should be moni-
tored carefully during and after each procedure.
Practical Considerations for In Vivo Mouse Studies 33
2.1 Obesity Body weight and composition may be determined in vivo by quan-
titative magnetic resonance (QMR) or dual-energy X-ray absorpti-
ometry (DEXA) scanning. QMR measurements can be made
without the need for anesthesia. In this procedure, mice are unre-
strained in a plastic cylinder with freedom to turn about but limited
vertical movement and placed in the scanner. Measurement is com-
pleted within 1–2 min. DEXA scanning requires animals to be
under light general anesthesia to prevent movement.
2.2 Insulin Blood levels of markers of insulin resistance and type 2 diabetes in
Resistance either the fed state or fasted state include glucose, insulin, lipids,
and Glucose Tolerance incretins, and pro- and anti-inflammatory molecules such as cyto-
kines and chemokines. Blood samples will typically be taken from a
2.2.1 Blood Levels superficial blood vessel or by removing the tip of the tail (~1 mm).
of Markers of Insulin Repeat samples can then be collected by removing the scab and
Resistance gently massaging the tail. Not more than 10% blood volume should
be taken in a single day and not more than 15% in a 28-day period.
On average, mice have around 58.5 mL of blood per kg of body-
weight and this approximation should be used for the above calcu-
lation, e.g., a 20.0 g mouse will be estimated to have 1.17 mL
blood and therefore blood sampling should be no more than
117 μL in a day or 175 μL in any 28-day period.
2.2.2 Glucose Tolerance GTTs assess the disposal of a glucose load administered via oral
Test (GTT) gavage or intraperitoneal/intravenous injection. The response to a
GTT is determined by insulin secretion and insulin resistance and is
impaired in obesity-related diabetes. Mice are fasted for 5–6 h to
ensure steady-state blood glucose levels. An aqueous glucose load is
administered in a volume of 10 mL/kg and blood glucose is
measured over a span of 2–3 h. The glucose load is typically
2–3 g/kg and may be adjusted based on lean body mass body
composition data if available due to muscle being the main tissue
responsible for glucose clearance in a GTT (the liver second). A
glucose load of 3 g/kg is sufficient to cause a doubling of blood
glucose concentration after 30 min in normal mice. Insulin-
resistant mice require less glucose to effectuate a doubling in
blood glucose concentration: 2.5 g/kg is recommended in diet-
induced obese mice and 2 g/kg in more extreme models of insulin
resistance such as ob/ob or db/db mice. A doubling in blood
34 Edward T. Wargent
2.2.3 Sensitivity This is the gold standard technique for measuring insulin sensitiv-
to Insulin, by Euglycemic/ ity. Mice are fasted for 4–6 h to prevent the appearance of meal-
Hyperinsulinemic Clamp derived glucose, but not deplete liver glycogen stores. Mice then
receive a constant infusion of insulin to achieve a steady-state
hyperinsulinemia above fasting insulin level. A variable glucose
infusion is administered to maintain euglycemia. Infusions are
administered via a previously implanted indwelling catheter. The
glucose infusion rate is determined by measuring blood glucose at
regular intervals, typically every 10 min, and adjusting the glucose
infusion accordingly. This variable rate of glucose infusion indicates
whole-body insulin action, as mice with enhanced insulin action
require a greater infusion of glucose. Isotopic tracer infusions can
be applied to assess sites where insulin action is affected.
Practical Considerations for In Vivo Mouse Studies 35
2.2.5 Real-Time Minute- Under in vivo conditions, the study of physiological and pharma-
by-Minute Changes cological functions of an organ is difficult due to whole-body
in Insulin Secretion interactions with the organ. In vitro perfusion of isolated pancreas
is required for physiologic and response studies, including the
investigation of endocrine function and secretory responsiveness
under a variety of diabetes-associated conditions and treatments.
Animals are terminally anesthetized, and the pancreas isolated from
the connecting spleen, stomach, and duodenum and transferred to
a pre-warmed chamber, where it is perfused in isolation from all
other organs and the perfusate collected for analysis of insulin
secretion.
36 Edward T. Wargent
2.2.6 Blood Pressure Hypertension is associated with hyperinsulinemia, and the same
programming conditions that cause increased risk of obesity and
diabetes also increase the susceptibility to hypertension. Therefore,
blood pressure is an important measure and can be done satisfacto-
rily by the noninvasive tail cuff method. Systolic blood pressure
equal to or greater to 140 mmHg is considered to be hypertensive.
A positive outcome of treatment would be to reduce systolic blood
pressure below 130 mmHg.
2.3 Energy Balance (a) Measurement of food intake and measurement of overall daily
energy expenditure using indirect calorimetry measuring the
amount of oxygen consumed and carbon dioxide produced in
expired air.
(b) Measurement of basal metabolic rate by measuring the utiliza-
tion of glucose and release of carbon dioxide in animals in a
thermoneutral environment over 8 h. The only difference
from normal measurements of energy expenditure is that the
ambient temperature raised to 28–30 C and measurements
are limited to 8 h during the light period.
(c) Measurement of brown adipose tissue activity after adminis-
tration of a thermogenic agent (such as a beta 3-adrenoceptor
agonist) by measuring oxygen consumption and carbon diox-
ide output and blood pressure indirectly by the tail cuff
method. Brown adipose tissue thermogenic capacity will also
be assessed ex vivo by determining the expression of uncou-
pling protein-1.
3.1 Strain Many strains of mice are available for the study of obesity and type
2 diabetes and the question of a “right model” per se a complex,
and possibly unanswerable, one [3]. Genetically modified mice will
usually be generated on a 129 background and thereafter trans-
ferred to the strain of choice, usually C57Bl/6 mice because of
their susceptibility to environmentally induced obesity and insulin
resistance. However, the strain employed will depend on the ques-
tion being posed by the study. For example, studies into browning
of adipose tissue may well be suited to being on the 129 strain
because they exhibit a propensity for browning of white adipose
tissue and so have a window to both upregulate and downregulate
this physiological effect. Experimental consequences will occur
with all strain, e.g., in the case of 129 mice, the propensity for
browning might make them suitable to study promoters of brown-
ing, but will make them resistant to the effects of a high-fat diet and
so require a longer period before they develop insulin resistance and
require a 60% fat diet rather than a 40% fat diet. The complex
Practical Considerations for In Vivo Mouse Studies 37
3.2 Single Housing Animals should be group housed wherever possible. For some
studies, it may be necessary to house animals singly so that individ-
ual food intake can be measured, or to remove any effect of hierar-
chy or aggression on feeding behavior.
3.3 Food Modified diets should be adequate with respect to all known nutri-
and Feeding ents including vitamins and minerals. As such, commercial rodent
feeds are preferable to handcrafted feeds, and these are also consis-
3.3.1 Feeding
tent in constitution. Diets should be stored under appropriate
a Modified Diet
conditions to prevent deterioration and be given a defined shelf-
life. Feeding high-fat diets may reduce wear of the teeth. Over-
growth of teeth will reduce the animal’s ability to feed and drink
resulting in loss of weight. All experimental animals should have
their teeth checked for malocclusion, with the weighing times a
good time to do this. Where possible, animals should be provided
with environmental enrichment, such as wooden sticks, to help
increase teeth wear. Advice on treatment or culling should be
sought from a veterinary surgeon if teeth become overgrown.
Likewise, diets with a high sugar intake may increase the risk of
dental caries, which may cause pain and a reduction in food intake.
Mice on these diets should have their teeth inspected weekly (when
weighed would be ideal). If evidence of dental caries is observed,
the advice on tooth management should be sought from a veteri-
nary surgeon.
3.3.2 Pair-Feeding In the event of an experimental group having reduced food intake,
it may be necessary to pair-feed a group of control animals so that
they receive the same amount of food. A reduction in food intake
results in an improvement in insulin sensitivity and pair-feeding
allows differentiation between the insulin sensitizing effects of
reduced food intake and the primary mechanistic actions being
investigated in the study. This reduction in food intake should be
no less than 40% of normal on any one occasion (up to a maximum
of 72 h) and no more than 25% over the period of an experiment to
minimize the stress imposed on an animal. Body weight must be
measured to assess the impact of pair-feeding on the control ani-
mals and should not drop below 80% of the weight of normal mice
on standard diet.
38 Edward T. Wargent
3.3.3 Fasting The withholding of food from rodents is stressful but some studies
require deprivation of food for a defined period. The minimal
period of food withdrawal commensurate with the objectives of
the experiment should therefore always be used and should never
be greater than 18 h, which may be required if it is necessary to
deplete the primary nutrient stores of glycogen and mobilize lipid
stores from adipose tissue. Measurements of lipid dysregulation are
more likely to be seen if the animal is fasted for 18 h in mice
weighing 20 g or more and 5 h in mice less than 20 g. Animals
should have a minimum of 3 days with continuous availability of
food between any periods of fasting. There is no evidence that mice
that are hyperphagic show an enhanced level of fasting stress.
For some studies, animals may need to be deprived of food for a
defined period. Glucose tolerance and insulin sensitivity tests
require there to be no uncontrolled glucose appearance into the
blood and require a fasting period (typically 5 h) that is long
enough to ensure that the gut is empty, but short enough not to
stimulate hepatic gluconeogenesis. This is typically 5 h in mice.
3.4 Temperature The topic of appropriate housing temperature for mice is currently
under debate. With appropriate nesting material, mouse housing
temperatures are generally maintained 1–2 C above the room
temperature. Thermoneutrality (the temperature with the mini-
mum of physiological thermoregulation) is achieved with a room
temperature of 28–30 C. Below this zone, mice start to generate
their own heat by increased skeletal muscle and brown adipose
tissue activity, resulting in increased fat catabolism. Therefore,
studies designed to study the adverse effects of fat accumulation
(particularly those investigating chemical or genetic resistance to
these effects) would benefit from higher temperatures, e.g., high-
fat diet-induced obesity and insulin resistance is promoted at tem-
peratures close to thermoneutrality (with a tendency toward
reduced variation in data generated). However, mouse aggression
also increases with temperature and mice housed at thermoneutral-
ity tend to fight more and for this reason, mice should only be
housed at thermoneutrality when necessary. C57Bl/6 mice, a strain
commonly employed in metabolic studies because of its physiolog-
ical suitability, are naturally aggressive and an alternative strain
should be considered if housing at thermoneutrality for extended
periods be intended. Behavioral studies have shown that mice
prefer temperatures higher than the usual housing conditions of
around 20–24 C; however, temperatures in the thermoneutral
range are only preferred during periods of rest. Taking the meta-
bolic and behavioral aspects into consideration, the ethically recom-
mended temperature range for in vivo mice protocols is 24–26 C.
Procedures investigating energy expenditure at thermoneutrality
should be performed during the light phase for a maximum period
of 8 h.
Practical Considerations for In Vivo Mouse Studies 39
3.5 Blood Sampling Blood sampling would preferably be taken either from a superficial
blood vessel or from the tip of the tail after removal of no more than
0.5 mm of the tip of the tail on the first occasion and on subsequent
occasions after removal of the scab. Blood sampling from a superfi-
cial vessel will be as noninvasive as possible. Pain from tail tipping
for blood samples can be controlled by local anesthetic. The volume
of blood taken should not exceed 10% of total blood volume on a
single occasion or 15% in a 28-day period.
Blood sampling may be facilitated by placing the animals in a
warm environment for the period of the sampling, but this will not
have any adverse effects on the animal. Animals should be checked
for bleeding at 1 and 12 h after the procedure and more frequently
if required. Any other than minor bleeding after blood sampling
should be controlled by the temporary application of pressure and a
clotting agent. A veterinary surgeon should be consulted for advice
on wound healing if any animal showing signs of bleeding more
than 24 h after the last sampling. Any animal showing signs of
excessive blood loss such as torpor lethargy or anemia should be
culled.
Repeat sampling from the tip of the tail may result in bruising if
too much pressure is applied when massaging. If hematomas and
localized inflammation are noted at the tip of the tail, then animals
should be carefully monitored. Localized inflammation and redness
should recede within 24 h. If prolonged redness is observed or is
accompanied by swelling or darkening of the tail, the animal should
be removed from any further procedures. Any animal showing signs
of necrosis, infection or delayed healing more than 24 h after the
last sampling should be culled.
Long-term studies should have a maximum of five tolerance
tests each requiring repeat blood sampling during the procedure.
Animals should be assessed prior to each test to ensure the animal
has healed fully from the previous procedure, and if there is any sign
of hyperalgesia or abnormal healing, the animal should not be used.
with implanted pumps should not be kept more than 7 days after
the expected exhaustion of the pump, nor should they undergo
measurement of body composition by QMR.
3.6.2 Administration This is the first choice of route for chronic dosing as no discomfort
of Test Agents in the Food or distress is expected from feeding compounds in the diet and
or Water should be employed if compounds can be homogenously
distributed with no degradation. If this is not possible, or if taste
aversion occurs, another method will need to be considered. Waste
measurement for accurate assessment of dosage is also a consider-
ation. Leaky water bottles, “shredding” of solid diets and dispersal
of powdered diets out of the food tray are complications that may
arise. There are no fool proof methods for overcoming these issues,
e.g., trays with a wire top tend to accumulate urinary and fecal
waste and wire-bottomed cages with a waste tray underneath can
cause weight loss in the mice. Another disadvantage to this route is
that this is not a method that would be used in humans, and so
some studies, by necessity, require a bolus dosing route.
3.6.3 Oral Gavage Dosing by oral gavage in mice should be at a maximum of 20 mL/
kg, and preferably 10 mL/kg. Some minor discomfort and tran-
sient distress is expected on each occasion, and on rare occasions,
the esophagus becomes damaged or a small volume may inadver-
tently enter the lungs. Animals should be observed immediately
after dosing. Any animal showing signs of mis-dosing or damage
such as by coughing/choking or collapsing after administration of
substances should be culled. A mis-dose directly into the lungs is
extremely unlikely and results in immediate collapse shortly fol-
lowed by death. Should collapse occur, the mouse should be imme-
diately culled. More likely than a direct dose into the lungs is the
event of a small amount of fluid ending up in the lungs. This
happens when the dosing tube is inserted into the esophagus rather
than the stomach, combined with too rapid a dosing. Detection is
by careful listening immediately after dosing—a rasping sound will
indicate this has happened. If this happens, the animal should be
immediately culled, as death will occur slowly within 2 days and
cause pain and distress.
References
1. Bessesen DH, Van Gaal LF (2018) Progress and 3. Leiter EH (2009) Selecting the “right” mouse
challenges in anti-obesity pharmacotherapy. model for metabolic syndrome and type 2 diabe-
Lancet Diabetes Endocrinol 6:237–248 tes research. Methods Mol Biol 560:1–17
2. Rabassa C, Dickson SL (2016) Impact of stress
on metabolism and energy balance. Curr Opin
Behav Sci 9:71–77
Chapter 3
Abstract
In order to better understand the events that precede and precipitate the onset of type 2 diabetes (T2DM),
several nutritional animal models have been developed. These models are generated by manipulating the
diet of either the animal itself, or its mother during her pregnancy, and in comparison to traditional genetic
and knock out models, have the advantage that they more accurately reflect the etiology of human T2DM.
This chapter will discuss some of the most widely used nutritional models of T2DM: Diet-induced obesity
(DIO) in adult rodents, and studies of offspring of mothers fed a low-protein, high-fat and/or high-sugar
diet during pregnancy and/or lactation. Several common mechanisms have been identified through which
these nutritional manipulations can lead to metabolic disease, including pancreatic beta-cell dysfunction,
impaired insulin signaling in skeletal muscle, and the excess accumulation of visceral adipose tissue and
consequent deposition of nonesterified fatty acids in peripheral tissues. In addition, there is an emerging
concept that obesity/poor quality diets result in increased production and release of pro-inflammatory
cytokines from adipose tissue leading to a state of chronic low-grade inflammation, and that this is likely to
represent an important link between obesity/diet and metabolic dysfunction. The following chapter will
discuss the most common nutritional models of T2DM in experimental animals, their application, and
relationship to human etiology, and will highlight the important insights these models have provided into
the pathogenesis of T2DM.
Key words Type 2 diabetes, Obesity, Insulin resistance, Animal models, Nutrition, High-fat diet,
Programming
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_3, © Springer Science+Business Media, LLC, part of Springer Nature 2020
43
44 €usler et al.
Beverly Sara Mühlha
2 Diet-Induced Obesity
2.1 Diet-Induced The DIO rodent model involves a regimen in which healthy, non-
Obesity: Rats and Mice obese mice or rats are provided with ad libitum access to a highly
palatable high-fat, high-energy diet. C57BL/6J mice were origi-
nally selected for this model because in early studies, these mice
developed clear-cut diabetes more rapidly than other strains fed the
same high-fat diet, suggesting a genetic predisposition to T2DM in
this strain [7]. In this model, male C57BL/6J (B6) mice are main-
tained on these diets for 8–12 weeks, and as a result, become obese,
mildly to moderately hyperglycemic, and develop impaired glucose
tolerance [7]. Studies where the animals are maintained on the diets
for longer periods have also demonstrated that after 16 weeks on a
high-fat diet, C57BL/6 mice exhibit adipocyte hyperplasia and
hypertrophy, and are insulin resistant [8]. For similar reasons, that
Models of T2DM 45
2.1.1 High-Fat Diets The majority of high-fat diets used in rodents provide between 40%
and 60% energy as fat, as compared to ~10% energy from fat in
standard laboratory diets [14]. Historically, the major source of fat
in these diets has been lard, and therefore, most studies to date have
predominately increased the dietary saturated fat content. Feeding
rodents diets containing high levels of saturated fat has consistently
been shown to promote fat deposition, increase the production of
pro-inflammatory cytokines, and result in the development of insu-
lin resistance, hypertriglyceridemia, hyperphagia, hypertension,
and nonalcoholic fatty liver disease [14–16].
Over recent years, it has also become increasingly clear, how-
ever, that different types of dietary fats can have profoundly differ-
ent physiological effects. Consequently, not all high-fat diets
provide a suitable nutritional approach for inducing insulin resis-
tance/T2DM. This is particularly true of diets which contain high
levels of the omega-3 long-chain polyunsaturated fatty acids (n 3
LCPUFA), eicosapentaenoic acid (EPA) and docosahexaenoic acid
(DHA), since these n 3 LCPUFA, unlike saturated and omega-6
polyunsaturated fats, have been reported to have both antilipo-
genic/adipogenic and insulin-sensitizing effects in vitro and
in vivo [17, 18].
The insulin sensitizing and anti-obesity actions of n 3
LCPUFA have been attributed to both direct effects on insulin-
sensitive tissues, and the effects of the bioactive derivatives of this
class of fatty acids. Studies in vitro and in adult rodents have shown
that both EPA and DHA act to suppress expression of genes
involved in preadipocyte proliferation/differentiation, thereby
inhibiting the expansion of adipose depots by hyperplasia [17]. In
addition, these fatty acids also suppress expression/activity of the
46 €usler et al.
Beverly Sara Mühlha
2.1.2 High-Sucrose In recent years, there has been an increasing focus on the contribu-
and High-Fructose Diets tion of added sugars in foods/beverages to the current epidemic of
obesity and T2DM in human populations worldwide. This interest
has been heightened by data from epidemiological studies, which
have demonstrated that consumption of one or more sugar-
sweetened beverage per day is associated with an elevated risk of
developing high blood pressure, metabolic syndrome [25], and a
22% increase in the risk of developing T2DM [26]. These data have
resulted in the American Heart Association advising that daily
added sugar intake be limited to 100 cal and 150 cal for women
and men respectively [27], which is equivalent to approximately
5–10% of daily energy intake. More recently, the World Health
Models of T2DM 47
2.1.3 Studies of Obesity- It is now widely accepted that the susceptibility to developing
Prone and Obesity- obesity and T2DM varies between individuals, and there is consid-
Resistant Animals erable interest in understanding the genetic and physiological basis
of this difference. This question has been addressed using an adap-
tation of the DIO model in which adult Sprague-Dawley rats are
fed a purified diet with a moderately high fat content. The rats are
subsequently selected as either obesity-prone or obesity-resistant
according to their response (i.e., degree of weight gain and increase
in percentage body fat) on the high-energy diet [41]. The obesity-
prone rats typically eat approximately 16% more calories over the
first 30 days compared to the obesity-resistant strain and exhibit a
phenotype comparable to human metabolic syndrome, including
glucose intolerance, hyperinsulinemia, and T2DM [41, 42]. Selec-
tive breeding has enabled these researchers to generate obesity-
resistant and obesity-prone strains, and to study these two popula-
tions in order to determine factors which may underlie an increased
propensity to obesity [43, 44]. This is a useful model, in that it
provides a means of distinguishing between the effects due to high
dietary fat content as distinct from those related to the effects of
excess body fat.
2.1.4 Summary: Diet- Irrespective of the precise nature of the diet, diet-induced obesity
Induced Obesity as a Model results in many of the same changes as seen in human obesity,
of Human T2DM including the development of central and peripheral insulin and
leptin resistance [9], increased lipid accumulation in peripheral
tissues, a pro-inflammatory state and in the altered expression of
Models of T2DM 49
Diet-induced Obesity
6) Reduced glucose
uptake
Insulin Resistance
Hyperglycemia
Type 2 Diabetes
source (i.e., animal vs. plant), fatty acid composition (i.e., the
content of saturated, monounsaturated, and polyunsaturated
fats), the balance of n 6 and n 3 PUFA and the level of trans
fats also varies between studies [1, 14]. Similarly, high-sucrose
Models of T2DM 51
and/or high-fructose diets vary in their sugar content and the form
in which the sugar is delivered, as well as the background fat and
protein content of the diet. While there is no clear indication of
which type of nutritional treatment represents the best model of the
metabolic disturbances seen in human obesity, two publications
focused on various types of diets employed in these experiments
concluded that a “cafeteria diet” approach, in which rats/mice were
provided with a selection of high-fat/high-sugar “junk foods,”
produced the metabolic phenotype that was most similar to that
seen in human diet-induced obesity [1, 50]. However, it is also
important to note that the composition of “cafeteria diets” is highly
variable between laboratories and their effects variable between
breeds. Therefore, it is important to critically evaluate the extent
to which the diet used in nutritional models of human T2DM
reflects the typical “poor quality western style diet,” including the
proportion of energy derived from fat/sugar and fat composition,
and the precise phenotype of the strain under investigation when
interpreting results and extrapolating these to humans.
2.2 Diet-Induced The sand rat (Psammomys obesus) has been another popular model
Obesity: Psammomys for studying the degradation of metabolic function associated with
Obesus obesity. This is an attractive model because the sand rat rapidly
develops obesity when fed a standard laboratory chow in standard
animal housing conditions, which is considerably less expensive
than custom-made high-fat diets. Many of the pathophysiological
changes found in the obese sand rat are similar to those seen in
human type 2 diabetic patients [51], and this model therefore
appears to provide an appropriate nutritional model of human
T2DM. When provided with ad libitum access to laboratory
chow, the adult sand rat progress through from normoglycemia
and normoinsulinemia, to hyperinsulinemia with marked insulin
resistance, followed by pronounced hyperinsulinemia with hyper-
glycemia, and after a further 6–10 weeks of chow feeding, gradual
beta-cell degradation and disappearance of beta-cell insulin which
eventually results in severe insulin deficiency and overt diabetes
[52]. The obese sand rat exhibits profound insulin resistance in
both skeletal muscle and liver, and provides an attractive model for
studying the mechanisms which underlie the development of dia-
betes in human obesity [53]. Studies utilizing this model have
reported that GLUT4 protein content is reduced in skeletal muscle,
resulting in a reduced peripheral glucose uptake and hyperglycemia
[52]. In addition, hepatic PEPCK activity is increased, suggesting
that the ability of insulin to inhibit hepatic glucose production is
impaired [54]. Extensive use has been made of the obese sand rats
for the purpose of testing potential T2DM drugs, including tyro-
sine phosphatase inhibitors and glucagon like peptide-1 (GLP-1)
analogues [52].
52 €usler et al.
Beverly Sara Mühlha
The major limitations of this model are that the sand rat exhi-
bits insulin resistance even when fed on a low energy diet, suggest-
ing that the physiology of this desert-adapted animal may not be
entirely comparable with humans. The link between weight gain
and the development of insulin resistance in this model has yet to be
clearly defined; however, the phenotype is normalized by dietary
restriction. Nevertheless, this model has been utilized extensively
for the study of obesity-induced diabetes, and continues to provide
important insights into the cellular defects, which contribute to the
development of central and peripheral insulin resistance.
3.1 The Maternal The maternal low-protein model is one of the most extensively
Low-Protein Model studied models of fetal growth restriction. In this model, pregnant
dams are fed a diet containing approximately 8% of energy as
protein, compared to approximately 20% in controls, and this is
associated with low birth weight in the offspring, followed by
accelerated postnatal growth [68, 69]. The period of rapid postna-
tal growth is due to enhanced insulin sensitivity in the period
immediately after birth; however, this is not maintained beyond
the early postnatal period, and the offspring develop insulin resis-
tance by the age of 15 months, frank T2DM by 17 months
[65, 70], and the deterioration of metabolic function is accelerated
by a postnatal high-fat diet [71]. The phenotype of the low-protein
offspring has many similarities to that of human type 2 diabetics,
including insulin resistance, altered regulation of hepatic glucose
output and pancreatic dysfunction [68]. As a result, this model has
been used extensively to explore and characterize the cellular
defects within the insulin signaling pathway which may contribute
to reduced insulin sensitivity. These studies have demonstrated that
prenatal exposure to a low-protein diet results in defects in several
peripheral insulin-sensitive tissues, which are central to the mainte-
nance of glucose homeostasis. In the pancreas, the islets of Langer-
hans are smaller and exhibit a reduced insulin secretion in response
to amino acid stimulation [72]. While there is no obvious defect in
glucose-stimulated insulin release in offspring fed a control chow
diet postweaning, a reduction in the capacity of glucose to stimulate
insulin release emerges if these offspring are fed on a high-fat diet
postweaning. The liver of the low-protein offspring is unresponsive
to the action of glucagon, and insulin stimulates, rather than sup-
presses, hepatic glucose output [73]. Furthermore, adipose cells
exhibit increased basal and insulin-stimulated glucose uptake, lead-
ing to an increased accumulation of fat in the visceral compartment
54 €usler et al.
Beverly Sara Mühlha
4) Beta cell
1) Increased 2) Impaired insulin 3) Hepatic insulin dysfunction
Glucose uptake signalling resistance
Hyperglycemia
Insulin resistance
Type 2 Diabetes
Fig. 2 Summary of mechanisms proposed to contribute to the programming of type 2 diabetes of offspring in
the maternal low-protein model. Prenatal exposure to a low-protein diet results in (1) an increased capacity for
glucose uptake by visceral adipocytes (2) impaired insulin signaling in skeletal muscle, (3) hepatic insulin
resistance and (4) impaired pancreatic function. The increased glucose uptake by visceral adipocytes results
in an increased accumulation of visceral adipose tissue, resulting in elevated plasma concentrations of
nonesterified free fatty acids (NEFAs) (5) which are deposited in peripheral tissues (liver and skeletal muscle),
further reducing insulin sensitivity. This reduced insulin sensitivity is associated with impaired glucose uptake
by skeletal muscle (6), increased glucose output by the liver (7) and reduced insulin secretion by the pancreas
(8), resulting in peripheral hyperglycemia, insulin resistance and type 2 diabetes
and ectopic fat storage in liver and skeletal muscle, which further
contributes to the peripheral insulin resistance [74] (Fig. 2).
Given that muscle represents the major site of postprandial
glucose disposal, it is not surprising that changes in the functional
characteristics of muscle fibers during the perinatal period are
important in the programming of insulin resistance and diabetes
(Fig. 2). Isolated muscle strips from these low-protein animals
exhibit enhanced basal and insulin-stimulated glucose uptake, and
this increase in insulin sensitivity is associated with a twofold
Models of T2DM 55
3.2 Prenatal There is increasing evidence that prenatal exposure to a high plane
Overnutrition, of nutrition is also associated with an increased risk of obesity and
Postnatal Obesity, T2DM in postnatal life [61]. The rat and sheep represent the two
and T2DM main animal models that have been utilized thus far for investigat-
ing the effects of prenatal overnutrition on the offspring, and there
is increasing evidence pointing to the negative impact of intrauter-
ine exposure to maternal obesity and/or high-fat/high-sugar diets
on insulin signaling in the offspring [61, 79]. Studies to date have
suggested that the deterioration of metabolic function in these
individuals is associated with altered development of pancreatic
beta cells, mitochondrial dysfunction and programming of the
central appetite-regulating network [11, 80–82]. Several studies
have also suggested that the deterioration of metabolic function
may be secondary to the increased accumulation of adipose tissue in
these offspring (Fig. 3) [82, 83]; however, it is not clear whether
this is the case in all models of prenatal overnutrition.
56 €usler et al.
Beverly Sara Mühlha
Prenatal
Overnutrition
Obesity
(2) (3) β-cell dysfunction
Inflammation
LIVER SKELETAL
MUSCLE
(5) Increased (6) Reduced
glucose output glucose uptake
Insulin resistance
Hyperglycemia
Type 2 Diabetes
Fig. 3 Potential mechanisms contributing to the programming of type 2 diabetes following prenatal overnutri-
tion. It has been demonstrated that prenatal overnutrition is associated with an increased lipogenic capacity of
adipose cells (1), which results in increased fat accumulation after birth (2) and predisposes these offspring to
obesity in later life. In addition, prenatal high-fat diets result in impaired pancreatic development, and beta-
cell dysfunction in postnatal life (3) and increased inflammation in key metabolic tissues, both of which
contribute to increased insulin resistance. The increased accumulation of body fat further contributes to
metabolic dysfunction by elevating plasma nonesterified free fatty acid (NEFA) concentrations (4) which are
Models of T2DM 57
Fig. 3 (continued) deposited in skeletal muscle and liver, resulting in insulin resistance in these tissues
(5) and (6). Epigenetic changes which occur before birth (7) may also play a role in metabolic programming
after prenatal overnutrition. Together, these changes result in peripheral insulin resistance and, ultimately,
type 2 diabetes
58 €usler et al.
Beverly Sara Mühlha
3.4 Maternal High- The adverse metabolic effects associated with excess consumption
Sucrose/High- of additional sweeteners in adults have led to a growing interest in
Fructose Diets the potential role of these substances in the maternal diet in the
and Metabolic programming of insulin resistance and T2DM. In epidemiological
Programming studies, excess consumption of sugar-sweetened beverages prior to
pregnancy has been associated with an increased risk of developing
gestational diabetes [95], which is known to be a risk factor for
obesity and T2DM in the offspring [79]. The consumption of these
sugar-sweetened beverage has also been linked to an increased birth
weight [96]. Thus far, however, there have been relatively few
experimental studies which have attempted to address the effects
of sucrose/fructose in the maternal diet at levels typically encoun-
tered in Western diets on the long-term metabolic health of the
offspring.
There is evidence from rodent studies, however, that maternal
consumption of high levels of fructose or fructose containing
sugars during pregnancy is associated with metabolic dysfunction
in the mother, including increased adiposity, weight gain and
hepatic insulin resistance, and adverse effects on placental and
fetal development [97–103]. As discussed above, the metabolic
effects of fructose in combination with glucose are different from
fructose alone; however, few studies have investigated the separate
effects of sucrose or fructose during pregnancy on maternal and
Models of T2DM 59
4.1 The Adipocyte In large animal models, there is evidence that exposure to prenatal
overnutrition or undernutrition can act to permanently alter the
function of adipocytes, and result in an increased lipogenic capacity
in adipose depots in postnatal life. In sheep, which have a similar
profile of adipose cell development to humans, exposure to
increased glucose concentrations in late fetal life increases the
expression of genes within adipose cells that are responsible for
promoting lipid storage and forming new adipocytes [105]. This
is associated with increased adipose tissue mass by the end of the
first month of life, due primarily to an increase in adipocyte cell size
[82]. In pigs, adipocytes exposed to high glucose levels before birth
also exhibit a dramatic increase in their capacity for lipogenesis
[106] and this precedes the development of obesity in the piglets
[107]. These findings have since been supported by work in
rodents, in which maternal junk food feeding during pregnancy
and lactation in rats and an obesogenic diet during this same period
in mice were each associated with increased adiposity and increased
expression of lipogenic genes and insulin-independent glucose
transporters in the perirenal adipose depot [83, 108]. It would
therefore appear that fat cells exposed to an excess substrate supply
during critical windows in their development have an increased
capacity for storing lipid in postnatal life. This enhanced lipogenic
capacity would render these individuals more likely to store excess
energy in the form of fat, and increase their susceptibility to weight
gain, obesity and, consequently, to the excess deposition of fatty
acids in liver and skeletal muscle, resulting in peripheral insulin
resistance, hyperglycemia, and ultimately, T2DM [109] (Fig. 3).
In individuals exposed to low nutrition levels before birth,
adipocyte development is initially sacrificed in favor of “essential”
organs [57, 110]. If an in utero “restricted” individual is born into
a postnatal environment where nutrient supply is no longer con-
strained, a period of “catch-up” fat deposition ensues, mainly in the
visceral adipose depot [111]. These individuals are thus at increased
risk of visceral obesity [112], and consequently, to the development
of insulin resistance and T2DM [113–115]. It is the increased
60 €usler et al.
Beverly Sara Mühlha
4.2 Mitochondrial Mitochondria play a central role in the regulation of cellular energy
Biogenesis metabolism, and impaired mitochondrial function and reduction in
mitochondrial oxidative capacity in skeletal muscle have been asso-
ciated with the onset of insulin resistance and T2DM both in
experimental animal models and in human subjects [120]. In exper-
imental animal studies, both diet-induced obesity and prenatal
undernutrition have been associated with impaired mitochondrial
biogenesis and reduced abundance of oxidative enzymes in skeletal
muscle. Importantly, there is evidence that the reduction of mito-
chondrial biogenesis in adult rats exposed to prenatal undernutri-
tion precedes the development of insulin resistance and glucose
intolerance in this model [77, 78], therefore, implicating impaired
mitochondrial function in the causal pathway linking prenatal
undernutrition to later metabolic disease. The role of mitochon-
drial dysfunction in metabolic programming has yet to be explored
in large animal models, and the potential role of mitochondrial
dysfunction in the development of T2DM remains an important
area for future investigation.
4.4 Epigenetics The concept that changes in phenotype could be elicited by modi-
fication of the DNA in the absence of changes in DNA sequence,
that is, epigenetic changes, provides a new basis for understanding
of phenotypic programming. The two most well-characterized epi-
genetic modifications include DNA methylation and histone acety-
lation, which both act to suppress gene expression
[125, 126]. Recent evidence has demonstrated that exposure to
an inappropriate nutrient supply during early development may
result in epigenetic modifications in the fetus which have the
potential to permanently alter the function of important metabolic
systems. Waterland and colleagues demonstrated that when mice
with a genetic tendency toward obesity were fed a standard diet
during pregnancy, the degree of obesity in the offspring increased
progressively with each generation. This effect was, however,
completely abolished if mice were fed a diet high in methyl groups
(which increases DNA methylation) during pregnancy, and the
offspring remained lean. These findings suggested that reduced
DNA methylation in genes which play a role in the regulation of
energy balance and fat storage, as a result of early nutritional
programming, may contribute to the later development of obesity
and metabolic disease [127]. The early work of Waterland has since
been supported by a large number of subsequent experimental
animal studies, and the importance of epigenetics in mediating
metabolic programming is now well established. There is also
increasing evidence that exposure to a suboptimal nutrient supply
before birth can also alter methylation of a series of genes involved
in growth and energy homeostasis, and that this may explain the
link between both fetal growth restriction and maternal nutritional
imbalance and later metabolic dysfunction. In rodent studies,
Burdge and Lilycrop have reported altered methylation of a num-
ber of key hepatic genes, including those involved in carbohydrate
metabolism, in offspring exposed to maternal undernutrition in
utero [128]. Importantly, these studies have provided the evidence
of transgenerational programming effects, such that epigenetic
alterations persist into the F2 generation [129]. There is also
emerging evidence of epigenetic programming in human studies,
including a study reporting altered Igf2 methylation in adults
whose mothers were exposed to the Dutch Winter Hunger Famine
during pregnancy [130]. Overall, the role of epigenetics in the
developmental origins of metabolic disease is certainly an exciting
area of research, and has been the subject of a number of excellent
reviews [131–134].
62 €usler et al.
Beverly Sara Mühlha
5 Summary
Acknowledgments
PANCREAS
7) Epigenetic
changes
8) Mitochondrial
dysfunction
3) Adipocyte
hypertrophy
2) Reduced Insulin
secretion SKELETAL MUSCLE
6) Increased glucose
output
5) Reduced glucose
uptake
Hyperglycemia
Type 2 Diabetes
Fig. 4 Summary of common mechanisms identified in all nutritional models of type 2 diabetes. Both diet-
induced obesity and prenatal nutritional interventions are associated with pancreatic dysfunction (1) and
impaired insulin secretion (2), which contributes to peripheral hyperglycemia. In addition, adipocyte hypertro-
phy, in particular within the visceral compartment (3), is associated with increased circulating concentrations
of nonesterified free fatty acids (NEFA) (4) and increased production and release of pro-inflammatory cytokines
(5). The infiltration of NEFA and pro-inflammatory cytokines liver and skeletal muscle results in development of
insulin resistance in these tissues, and consequently, reduced glucose uptake by skeletal muscle (5) and
increased hepatic glucose output (6). In addition, epigenetic changes and mitochondrial dysfunction in liver
and skeletal muscle may also contribute to development of metabolic disorders
64 €usler et al.
Beverly Sara Mühlha
References
1. Buettner R, Scholmerich J, Bollheimer LC 12. Ong ZY, Wanasuria AF, Lin MZP, Hiscock J,
(2007) High-fat diets: modeling the meta- Muhlhausler BS (2013) Chronic intake of a
bolic disorders of human obesity in rodents. cafeteria diet and subsequent abstinence.
Obesity 15:798–808 Sex-specific effects on gene expression in the
2. Portha B, Blondel O, Serradas P, McEvoy R, mesolimbic reward system. Appetite
Giroix MH, Kergoat M, Bailbe D (1989) The 65:189–199
rat models of non-insulin dependent diabetes 13. Reichelt AC, Morris MJ, Westbrook RF
induced by neonatal streptozotocin. Diabete (2014) Cafeteria diet impairs expression of
Metab 15:61–75 sensory-specific satiety and stimulus-outcome
3. Bray GA (2004) Medical consequences of learning. Front Psychol 5:852
obesity. J Clin Endocrinol Metab 14. Hariri N, Thibault L (2010) High-fat diet-
89:2583–2589 induced obesity in animal models. Nutr Res
4. Goralski KB, Sinal CJ (2007) Type 2 diabetes Rev 23:270–299
and cardiovascular disease: getting to the fat 15. Flanagan AM, Brown JL, Santiago CA, Aad
of the matter. Can J Physiol Pharmacol PY, Spicer LJ, Spicer MT (2008) High-fat
85:113–132 diets promote insulin resistance through cyto-
5. Smith SR, Lovejoy JC, Greenway F, Ryan D, kine gene expression in growing female rats. J
deJonge L, de la Bretonne J, Volafova J, Bray Nutr Biochem 19:505–513
GA (2001) Contributions of total body fat, 16. Barbosa-da-Silva S, da Silva NC, Aguila MB,
abdominal subcutaneous adipose tissue com- Mandarim-de-Lacerda CA (2013) Liver dam-
partments, and visceral adipose tissue to the age is not reversed during the lean period in
metabolic complications of obesity. Metabo- diet-induced weight cycling in mice. Hepatol
lism 50:425–435 Res 18:12138
6. Ravussin E, Smith SR (2002) Increased fat 17. Flachs P, Rossmeisl M, Bryhn M, Kopecky J
uptake, impaired fat oxidation, and failure of (2009) Cellular and molecular effects of n-3
fat cell proliferation result in ectopic fat stor- polyunsaturated fatty acids on adipose tissue
age, insulin resistance, and type 2 diabetes. biology and metabolism. Clin Sci (Lond)
Ann N Y Acad Sci 967:363–378 116:1–16
7. Surwit R, Kuhn C, Cochrane C, McCubbin J, 18. Muhlhausler BS, Cook-Johnson R, James M,
Feinglos M (1988) Diet-induced type II dia- Miljkovic D, Duthoit E, Gibson R (2010)
betes in C57BL/6J mice. Diabetes Opposing effects of omega-3 and omega-6
37:1163–1167 long chain polyunsaturated fatty acids on the
8. Black BL, Croom J, Eisen EJ, Petro AE, expression of lipogenic genes in omental and
Edwards CL, Surwit RS (1998) Differential retroperitoneal adipose depots in the rat. J
effects of fat and sucrose on body composi- Nutr Metab
tion in A/J and C57BL/6 mice. Metabolism 19. Ruzickova J, Rossmeisl M, Prazak T, Flachs P,
47:1354–1359 Sponarova J, Veck M, Tvrzicka E, Bryhn M,
9. Roberts CK, Berger JJ, Barnard RJ (2002) Kopecky J (2004) Omega-3 PUFA of marine
Long-term effects of diet on leptin, energy origin limit diet-induced obesity in mice by
intake, and activity in a model of diet-induced reducing cellularity of adipose tissue. Lipids
obesity. J Appl Physiol 93:887–893 39:1177–1185
10. Bayol SA, Farrington SJ, Stickland NC (2007) 20. Massiera F, Guesnet P, Ailhaud G (2006) The
A maternal ‘junk food’ diet in pregnancy and crucial role of dietary n-6 polyunsaturated
lactation promotes an exacerbated taste for fatty acids in excessive adipose tissue develop-
‘junk food’ and a greater propensity for obe- ment: relationship to childhood obesity. Nes-
sity in rat offspring. Br J Nutr 98:843–851 tle Nutr Workshop Ser Pediatr Program
11. Taylor PD, McConnell J, Khan IY, 57:235–242
Holemans K, Lawrence KM, Asare-Anane H, 21. Massiera F, Saint-Marc P, Seydoux J,
Persaud SJ, Jones PM, Petrie L, Hanson MA, Murata T, Kobayashi T, Narumiya S,
Poston L (2005) Impaired glucose homeosta- Guesnet P, Amri E-Z, Negrel R, Ailhaud G
sis and mitochondrial abnormalities in off- (2003) Arachidonic acid and prostacyclin sig-
spring of rats fed a fat-rich diet in pregnancy. naling promote adipose tissue development: a
Am J Physiol Regul Integr Comp Physiol 288: human health concern? J Lipid Res
R134–R139 44:271–279
Models of T2DM 65
22. Semple RK, Chatterjee VK, O’Rahilly S 34. Tran LT, Yuen VG, McNeill JH (2009) The
(2006) PPAR gamma and human metabolic fructose-fed rat: a review on the mechanisms of
disease. J Clin Invest 116:581–589 fructose-induced insulin resistance and hyper-
23. De Caterina R, Basta G (2001) n-3 Fatty acids tension. Mol Cell Biochem 332:145–159
and the inflammatory response — biological 35. Pagliassotti MJ, Prach PA, Koppenhafer TA,
background. Eur Heart J Suppl 3:D42–D49 Pan DA (1996) Changes in insulin action,
24. Chandalia M, Abate N (2007) Metabolic triglycerides, and lipid composition during
complications of obesity: inflated or inflamed? sucrose feeding in rats. Am J Physiol 271:
J Diabetes Complications 21:128–136 R1319–R1326
25. Malik VS, Popkin BM, Bray GA, Despres JP, 36. Sheludiakova A, Rooney K, Boakes RA
Hu FB (2010) Sugar-sweetened beverages, (2012) Metabolic and behavioural effects of
obesity, type 2 diabetes mellitus, and cardio- sucrose and fructose/glucose drinks in the
vascular disease risk. Circulation rat. Eur J Nutr 51:445–454
121:1356–1364 37. Chicco A, D’Alessandro ME, Karabatas L,
26. Romaguera D, Norat T, Wark PA, Vergnaud Pastorale C, Basabe JC, Lombardo YB
AC, Schulze MB, van Woudenbergh GJ, (2003) Muscle lipid metabolism and insulin
Drogan D, Amiano P, Molina-Montes E, San- secretion are altered in insulin-resistant rats
chez MJ, Balkau B, Barricarte A, Beulens JW, fed a high sucrose diet. J Nutr 133:127–133
Clavel-Chapelon F, Crispim SP, Fagherazzi G, 38. Oliveira LS, Santos DA, Barbosa-da-Silva S,
Franks PW, Grote VA, Huybrechts I, Key TJ, Mandarim-de-Lacerda CA, Aguila MB
Khaw KT, Nilsson P, Overvad K, Palli D, (2014) The inflammatory profile and liver
Panico S, Quiros JR, Rolandsson O, damage of a sucrose-rich diet in mice. J Nutr
Sacerdote C, Sieri S, Slimani N, Spijkerman Biochem 25:193–200
AM, Tjonneland A, Tormo MJ, Tumino R, 39. Catena C, Giacchetti G, Novello M,
van den Berg SW, Wermeling PR, Zamara- Colussi G, Cavarape A, Sechi L (2003) Cellu-
Ros R, Feskens EJ, Langenberg C, Sharp SJ, lar mechanisms of insulin resistance in rats
Foroughi NG, Riboli E, Wareham NJ (2013) with fructose-induced hypertension. Am J
Consumption of sweet beverages and type Hypertens 16:973–978
2 diabetes incidence in European adults: 40. Kazumi T, Odaka H, Hozumi T, Ishida Y,
results from EPIC-InterAct. Diabetologia Amano N, Yoshino G (1997) Effects of die-
56:1520–1530 tary fructose or glucose on triglyceride pro-
27. Johnson RK, Appel LJ, Brands M, Howard duction and lipogenic enzyme activities in the
BV, Lefevre M, Lustig RH, Sacks F, Steffen liver of Wistar fatty rats, an animal model of
LM, Wylie-Rosett J (2009) Dietary sugars NIDDM. Endocr J 44:239–245
intake and cardiovascular health: a scientific 41. Levin BE, Hogan S, Sullivan AC (1989) Initi-
statement from the American Heart Associa- ation and perpetuation of obesity and obesity
tion. Circulation 120:1011–1020 resistance in rats. Am J Physiol Regul Integr
28. Te Morenga L, Mallard S, Mann J (2012) Comp Physiol 256:R766–R771
Dietary sugars and body weight: systematic 42. Clegg DJ, Benoit SC, Reed JA, Woods SC,
review and meta-analyses of randomised con- Dunn-Meynell A, Levin BE (2005) Reduced
trolled trials and cohort studies. BMJ 346: anorexic effects of insulin in obesity-prone
e7492 rats fed a moderate-fat diet. Am J Physiol
29. Willett WC, Ludwig DS (2013) Science sour- Regul Integr Comp Physiol 288:R981–R986
ing on sugar. BMJ 346:e8077 43. Levin BE, Dunn-Meynell AA, Balkan B, Kee-
30. McLennan W, Podger A (1998) National sey RE (1997) Selective breeding for diet-
nutrition survey nutrient intakes and physical induced obesity and resistance in Sprague-
measurements 1995. Canberra, ACT, Dawley rats. Am J Physiol Regul Integr
Australian Bureau of Statistics Comp Physiol 273:R725–R730
31. Tappy L, Le KA (2010) Metabolic effects of 44. Tkacs NC, Levin BE (2004) Obesity-prone
fructose and the worldwide increase in obe- rats have preexisting defects in their counter
sity. Physiol Rev 90:23–46 regulatory response to insulin-induced hypo-
32. Douard V, Ferraris RP (2013) The role of glycemia. Am J Physiol Regul Integr Comp
fructose transporters in diseases linked to Physiol 287:R1110–R1115
excessive fructose intake. J Physiol 45. Woods SC, Seeley RJ, Rushing PA,
591:401–414 D’Alessio D, Tso P (2003) A controlled
33. Lustig RH (2013) Fructose: it’s “alcohol high-fat diet induces an obese syndrome in
without the buzz”. Adv Nutr 4:226–235 rats. J Nutr 133:1081–1087
66 €usler et al.
Beverly Sara Mühlha
46. Qiu L, List EO, Kopchick JJ (2005) Differen- Bleker OP (1998) Glucose tolerance in adults
tially expressed proteins in the pancreas of after prenatal exposure to famine. Lancet
diet-induced diabetic mice. Mol Cell Proteo- 351:173–177
mics 4:1311–1318 60. Pettit DJ, Knowler WC (1998) Long-term
47. Corbett SW, Stern JS, Keesey RE (1986) effects of the intrauterine environment, birth
Energy expenditure in rats with diet-induced weight, and breast-feeding in Pima Indians.
obesity. Am J Clin Nutr 44:173–180 Diabetes Care 21:B138–B141
48. Yaqoob P, Sherrington EJ, Jeffery NM, 61. Rkhzay-Jaf J, O’Dowd JF, Stocker CJ (2012)
Sanderson P, Harvey DJ, Newsholme EA, Maternal obesity and the fetal origins of the
Calder PC (1995) Comparison of the effects metabolic syndrome. Curr Cardiovasc Risk
of a range of dietary lipids upon serum and Rep 6:487–495
tissue lipid composition in the rat. Int J Bio- 62. Muhlhausler BS, Ong ZY (2011) The fetal
chem Cell Biol 27:297–310 origins of obesity: early origins of altered
49. Dandona P, Aljada A, Bandyopadhyay A food intake. Endocr Metab Immune Disord
(2004) Inflammation: the link between insu- Drug Targets 11:189–197
lin resistance, obesity and diabetes. Trends 63. Bavdekar A, Yajnik C, Fall C, Bapat S,
Immunol 25:4–7 Pandit A, Deshpande V, Bhave S,
50. Higa TS, Spinola AV, Fonseca-Alaniz MH, Kellingray S, Joglekar C (1999) Insulin resis-
Evangelista FSA (2014) Comparison between tance syndrome in 8-year-old Indian children:
cafeteria and high-fat diets in the induction of small at birth, big at 8 years, or both? Diabetes
metabolic dysfunction in mice. Int J Physiol 48:2422–2429
Pathophysiol Pharmacol 6:47–54 64. McMillen IC, Adam CL, Muhlhausler BS
51. Wassink AM, Olijhoek JK, Visseren FL (2005) Early origins of obesity: programming
(2007) The metabolic syndrome: metabolic the appetite regulatory system. J Physiol
changes with vascular consequences. Eur J 565:9–17
Clin Invest 37:8–17 65. Hales CN, Barker DJP (2001) The thrifty
52. Shafrir E, Ziv E, Kalman R (2006) Nutrition- phenotype hypothesis: type 2 diabetes. Br
ally induced diabetes in desert rodents as Med Bull 60:5–20
models of type 2 diabetes: acomys cahirinus 66. Holemans K, Verhaeghe J, Dequeker J, Van
(spiny mice) and Psammomys obesus (desert Assche FA (1996) Insulin sensitivity in adult
gerbil). ILAR J 47:212–224 female rats subjected to malnutrition during
53. Kaiser N, Nesher R, Donath MY, Fraenkel M, the perinatal period. J Soc Gynecol Investig
Behar V, Magnan C, Ktorza A, Cerasi E, Lei- 3:71–77
bowitz G (2005) Psammomys obesus, a 67. Thompson NM, Norman AM, Donkin SS,
model for environment-gene interactions in Shankar RR, Vickers MH, Miles JL, Breier
type 2 diabetes. Diabetes 54:S137–S144 BH (2007) Prenatal and postnatal pathways
54. Maislos M, Medvedovskv V, Sztarkier I, to obesity: different underlying mechanisms,
Yaari A, Sikuler E (2006) Psammomys obesus different metabolic outcomes. Endocrinol
(sand rat), a new animal model of Metab Clin North Am 148:2345–2354
non-alcoholic fatty liver disease. Diabetes 68. Ozanne SE (2001) Metabolic programming
Res Clin Pract 72:1–5 in animals: type 2 diabetes. Br Med Bull
55. Barker DJP, Bull AR, Osmond C, Simmonds 60:143–152
SJ (1990) Fetal and placental size and risk of 69. Ozanne SE, Jensen CB, Tingey KJ,
hypertension in adult life. Br Med J Storgaard H, Madsbad S, Vaag AA (2005)
301:259–262 Low birthweight is associated with specific
56. Hales CN, Barker DJP (2001) The thrifty changes in muscle insulin-signalling protein
phenotype hypothesis. Br Med Bull 60:5–20 expression. Diabetologia 48:547–552
57. McMillen IC, Robinson JS (2005) Develop- 70. Hales CN, Desai M, Ozanne SE, Crowther
mental origins of the metabolic syndrome: NJ (1996) Fishing in the stream of diabetes:
prediction, plasticity, and programming. from measuring insulin to the control of fetal
Physiol Rev 85:571–633 organogenesis. Biochem Soc Trans
58. Ravelli AC, van der Meulen JH, Osmond C, 24:341–350
Barker DJ, Bleker OP (1999) Obesity at the 71. Petry CJ, Ozanne SE, Wang CL, Hales CN
age of 50 y in men and women exposed to (1997) Early protein restriction and obesity
famine prenatally. Am J Clin Nutr independently induce hypertension in
70:811–816 1-year-old rats. Clin Sci (Lond) 93:147–152
59. Ravelli ACJ, van der Meulen JHP, Michels 72. Snoeck A, Remacle C, Reusens B, Hoet JJ
RPJ, Osmond C, Barker DJP, Hales CN, (1990) Effect of a low protein diet during
Models of T2DM 67
pregnancy on the fetal rat endocrine pancreas. 83. Samuelsson A-M, Matthews PA, Argenton M,
Biol Neonate 57:107–118 Christie MR, McConnell JM, Jansen EHJM,
73. Ozanne SE, Smith GD, Tikerpae J, Hales CN Piersma AH, Ozanne SE, Twinn DF,
(1996) Altered regulation of hepatic glucose Remacle C, Rowlerson A, Poston L, Taylor
output in the male offspring of protein- PD (2008) Diet-induced obesity in female
malnourished rat dams. Am J Physiol Endo- mice leads to offspring hyperphagia, adiposity,
crinol Metab 270:E559–E564 hypertension, and insulin resistance: a novel
74. Ozanne SE, Nave BT, Wang CL, Shepherd murine model of developmental program-
PR, Prins J, Smith GD (1997) Poor fetal ming. Hypertension 51:383–392
nutrition causes long-term changes in expres- 84. Muhlhausler BS, Roberts CT, McFarlane JR,
sion of insulin signaling components in adi- Kauter KG, McMillen IC (2002) Fetal leptin
pocytes. Am J Physiol Endocrinol Metab 273: is a signal of fat mass independent of maternal
E46–E51 nutrition in ewes fed at or above maintenance
75. Ozanne SE, Wang CL, Coleman N, Smith energy requirements. Biol Reprod
GD (1996) Altered muscle insulin sensitivity 67:493–499
in the male offspring of protein-malnourished 85. Bruce KD, Cagampang FR, Argenton M,
rats. Am J Physiol Endocrinol Metab 271: Zhang J, Ethirajan PL, Burdge GC, Bateman
E1128–E1134 AC, Clough GF, Poston L, Hanson MA,
76. Ozanne SE, Olsen GS, Hansen LL, Tingey McConnell JM, Byrne CD (2009) Maternal
KJ, Nave BT, Wang CL, Hartil K, Petry CJ, high-fat feeding primes steatohepatitis in
Buckley AJ, Mosthaf-Seedorf L (2003) Early adult mice offspring, involving mitochondrial
growth restriction leads to down regulation of dysfunction and altered lipogenesis gene
protein kinase C zeta and insulin resistance in expression. Hepatology 50:1796–1808
skeletal muscle. J Endocrinol 177:235–241 86. Srinivasan M, Aalinkeel R, Song F, Mitrani P,
77. Wadley GD, Siebel AL, Cooney GJ, McConell Pandya JD, Strutt B, Hill DJ, Patel MS
GK, Wlodek ME, Owens JA (2008) Utero- (2006) Maternal hyperinsulinemia predis-
placental insufficiency and reducing litter size poses rat fetuses for hyperinsulinemia, and
alters skeletal muscle mitochondrial biogene- adult-onset obesity and maternal mild food
sis in a sex-specific manner in the adult rat. Am restriction reverses this phenotype. Am J
J Physiol Endocrinol Metab 294:E861–E869 Physiol Endocrinol Metab 290:E129–E134
78. Siebel AL, Mibus A, De Blasio MJ, Westcott 87. Cerf ME, Williams K, Chapman CS, Louw J
KT, Morris MJ, Prior L, Owens JA, Wlodek (2007) Compromised beta-cell development
ME (2008) Improved lactational nutrition and beta-cell dysfunction in weanling off-
and postnatal growth ameliorates impairment spring from dams maintained on a high-fat
of glucose tolerance by uteroplacental insuffi- diet during gestation. Pancreas 34:347–353
ciency in male rat offspring. Endocrinology 88. McCurdy CE, Bishop JM, Williams SM,
149:3067–3076 Grayson BE, Smith MS, Friedman JE, Grove
79. Poston L (2011) Intergenerational transmis- KL (2009) Maternal high-fat diet triggers
sion of insulin resistance and type 2 diabetes. lipotoxicity in the fetal livers of nonhuman
Prog Biophys Mol Biol 106:315–322 primates. J Clin Invest 119:323–335
80. Srinivasan M, Katewa SD, Palaniyappan A, 89. Ailhaud G, Massiera F, Weill P, Legrand P,
Pandya JD, Patel MS (2006) Maternal high- Alessandri JM, Guesnet P (2006) Temporal
fat diet consumption results in fetal malpro- changes in dietary fats: role of n-6 polyunsat-
gramming predisposing to the onset of meta- urated fatty acids in excessive adipose tissue
bolic syndrome-like phenotype in adulthood. development and relationship to obesity. Prog
Am J Physiol Endocrinol Metab 291: Lipid Res 45:203–236
E792–E799 90. Muhlhausler BS, Ailhaud GP (2013) Omega-
81. Cerf ME, Muller CJ, Du Toit DF, Louw J, 6 polyunsaturated fatty acids and the early
Wolfe-Coote SA (2006) Hyperglycaemia and origins of obesity. Curr Opin Endocrinol Dia-
reduced glucokinase expression in weanling betes Obes 20:56–61
offspring from dams maintained on a high- 91. Massiera F, Barbry P, Guesnet P, Joly A,
fat diet. Br J Nutr 95:391–396 Luquet S, Moreilhon-Brest C, Mohsen-
82. Muhlhausler BS, Adam CL, Findlay PA, Duf- Kanson T, Amri E-Z, Ailhaud G (2010) A
field JA, McMillen IC (2006) Increased Western-like fat diet is sufficient to induce a
maternal nutrition alters development of the gradual enhancement in fat mass over genera-
appetite-regulating network in the brain. tions. J Lipid Res 51:2352–2361
FASEB J 20:1257–1259 92. Muhlhausler BS, Gibson RA, Makrides M
(2011) The effect of maternal omega-3
68 €usler et al.
Beverly Sara Mühlha
long-chain polyunsaturated fatty acid (n-3 affects maternal and fetal leptin signaling. J
LCPUFA) supplementation during preg- Nutr Biochem 24:1709–1716
nancy and/or lactation on body fat mass in 103. Samuelsson AM, Matthews PA, Jansen E,
the offspring: a systematic review of animal Taylor PD, Poston L (2013) Sucrose feeding
studies. Prostaglandins Leukot Essent Fatty in mouse pregnancy leads to hypertension,
Acids 85:83–88 and sex-linked obesity and insulin resistance
93. Muhlhausler BS, Gibson RA, Makrides M in female offspring. Front Physiol 4:14
(2010) Effect of long-chain polyunsaturated 104. Regnault TR, Gentili S, Sarr O, Toop CR,
fatty acid supplementation during pregnancy Sloboda DM (2013) Fructose, pregnancy
or lactation on infant and child body compo- and later life impacts. Clin Exp Pharmacol
sition: a systematic review. Am J Clin Nutr Physiol 40:824–837
92:857–863 105. Muhlhausler BS, Duffield JA, McMillen IC
94. Muhlhausler BS, Miljkovic D, Fong L, Xian (2007) Increased maternal nutrition stimu-
CJ, Duthoit E, Gibson RA (2011) Maternal lates peroxisome proliferator activated
omega-3 supplementation increases fat mass receptor-{gamma} (PPAR{gamma}), adipo-
in male and female rat offspring. Front Genet nectin and leptin mRNA expression in adi-
2:48 pose tissue before birth. Endocrinology
95. Chen L, Hu FB, Yeung E, Willett W, Zhang C 148:878–885
(2009) Prospective study of pre-gravid sugar- 106. Kasser TR, Martin RJ, Allen CE (1981) Effect
sweetened beverage consumption and the risk of gestational alloxan diabetes and fasting on
of gestational diabetes mellitus. Diabetes Care fetal lipogenesis and lipid deposition in pigs.
32:2236–2241 Biol Neonate 40:105–112
96. Phelan S, Hart C, Phipps M, Abrams B, 107. Ezekwe MO, Martin RJ (1980) The effects of
Schaffner A, Adams A, Wing R (2011) Mater- maternal alloxan diabetes on body composi-
nal behaviors during pregnancy impact off- tion, liver enzymes and metabolism and
spring obesity risk. Exp Diabetes Res serum metabolites and hormones of fetal
985139:26 pigs. Horm Metab Res 12:136–139
97. Toop CR, Muhlhausler BS, O’Dea K, Gentili 108. Bayol SA, Simbi BH, Bertrand JA, Stickland
S (2014) Consumption of sucrose, but not NC (2008) Offspring from mothers fed a
high fructose corn syrup, leads to increased ‘junk food’ diet in pregnancy and lactation
adiposity and dyslipidaemia in the pregnant exhibit exacerbated adiposity that is more
and lactating rat. J Dev Orig Health Dis pronounced in females. J Physiol
19:1–9 586:3219–3230
98. Goran MI, Dumke K, Bouret SG, Kayser B, 109. Muhlhausler B, Smith SR (2009) Early-life ori-
Walker RW, Blumberg B (2013) The obeso- gins of metabolic dysfunction: role of the adi-
genic effect of high fructose exposure during pocyte. Trends Endocrinol Metab 20:51–57
early development. Nat Rev Endocrinol 110. Padoan A, Rigano S, Ferrazzi E, Beaty BL,
9:494–500 Battaglia FC, Galan HL (2004) Differences in
99. Sloboda DM, Li M, Patel R, Clayton ZE, fat and lean mass proportions in normal and
Yap C, Vickers MH (2014) Early life exposure growth-restricted fetuses. Am J Obstet Gyne-
to fructose and offspring phenotype: implica- col 191:1459–1464
tions for long term metabolic homeostasis. J 111. Crescenzo R, Samec S, Antic V, Rohner-
Obesity 203474:23 Jeanrenaud F, Seydoux J, Montani J-P, Dul-
100. Vickers MH, Clayton ZE, Yap C, Sloboda loo AG (2003) A role for suppressed thermo-
DM (2011) Maternal fructose intake during genesis favoring catch-up fat in the
pregnancy and lactation alters placental pathophysiology of catch-up growth. Acta
growth and leads to sex-specific changes in Paediatr 52:1090–1097
fetal and neonatal endocrine function. Endo- 112. Ibanez L, Ong K, Dunger DB, de Zegher F
crinology 152:1378–1387 (2006) Early development of aiposity and
101. Mukai Y, Kumazawa M, Sato S (2013) Fruc- insulin resistance after catch-up weight gain
tose intake during pregnancy up-regulates the in small-for-gestational-age children. J Clin
expression of maternal and fetal hepatic sterol Endocrinol Metab 91:2153–2158
regulatory element-binding protein-1c in 113. Jaquet D, Gaboriau A, Czernichow P, Levy-
rats. Endocrine 44:79–86 Marchal C (2000) Insulin resistance early in
102. Rodriguez L, Panadero MI, Roglans N, adulthood in subjects born with intrauterine
Otero P, Alvarez-Millan JJ, Laguna JC, growth retardation. J Clin Endocrinol Metab
Bocos C (2013) Fructose during pregnancy 85:1401–1406
Models of T2DM 69
114. De Blasio MJ, Gatford KL, McMillen IC, 126. Gallou-Kabani C, Junien C (2005) Nutri-
Robinson JS, Owens JA (2006) Placental tional epigenomics of metabolic syndrome:
restriction of fetal growth increases insulin new perspective against the epidemic. Diabe-
action, growth and adiposity in the young tes 54:1899–1906
lamb. Endocrinology 148:1350–1358 127. Waterland RA, Travisano M, Tahiliani KG,
115. De Blasio MJ, Gatford KL, Robinson JS, Rached MT, Mirza S (2008) Methyl donor
Owens JA (2007) Placental restriction of supplementation prevents transgenerational
fetal growth reduces size at birth and alters amplification of obesity. Int J Obes (Lond)
postnatal growth, feeding activity, and adipos- 32:1373–1379
ity in the young lamb. Am J Physiol Regul 128. Burdge GC, Hanson MA, Slater-Jefferies JL,
Integr Comp Physiol 292:R875–R886 Lillycrop KA (2007) Epigenetic regulation of
116. Ozanne SE (2001) Metabolic programming transcription: a mechanism for inducing var-
in animals. Br Med Bull 60:143–152 iations in phenotype (fetal programming) by
117. Kind KL, Clifton PM, Grant PA, Owens PC, differences in nutrition during early life? Br J
Sohlstrom A, Roberts CT, Robinson JS, Nutr 97:1036–1046
Owens JA (2003) Effect of maternal feed 129. Burdge GC, Slater-Jefferies J, Torrens C,
restriction during pregnancy on glucose tol- Phillips ES, Hanson MA, Lillycrop KA
erance in the adult guinea pig. Am J Physiol (2007) Dietary protein restriction of preg-
Regul Integr Comp Physiol 284:R140–R152 nant rats in the F0 generation induces altered
118. Alexander G (1978) Quantitative develop- methylation of hepatic gene promoters in the
ment of adipose tissue in foetal sheep. Aust J adult male offspring in the F1 and F2 genera-
Biol Sci 31:489–503 tions. Br J Nutr 97:435–439
119. Merklin RJ (1973) Growth and distribution of 130. Tobi EW, Slagboom PE, van Dongen J,
human fetal brown fat. Anat Res 178:637–646 Kremer D, Stein AD, Putter H, Heijmans
120. Højlund K, Mogensen M, Sahlin K, Beck- BT, Lumey LH (2012) Prenatal famine and
Nielsen H (2008) Mitochondrial dysfunction genetic variation are independently and addi-
in type 2 diabetes and obesity. Endocrinol tively associated with DNA methylation at
Metab Clin North Am 37:713–731 regulatory loci within IGF2/H19. PLoS
One 7:30
121. Howie GJ, Sloboda DM, Reynolds CM, Vick-
ers MH (2013) Timing of maternal exposure 131. Hanley B, Dijane J, Fewtrell M, Grynberg A,
to a high fat diet and development of obesity Hummel S, Junien C, Koletzko B, Lewis S,
and hyperinsulinemia in male rat offspring: Renz H, Symonds M, Gros M,
same metabolic phenotype, different develop- Harthoorn L, Mace K, Samuels F, van Der
mental pathways? J Nutr Metab 517384:13 Beek EM (2010) Metabolic imprinting, pro-
gramming and epigenetics – a review of pres-
122. Palmer AC (2011) Nutritionally mediated ent priorities and future opportunities. Br J
programming of the developing immune sys- Nutr 104:S1–S25
tem. Adv Nutr 2:377–395
132. Heerwagen MJR, Miller MR, Barbour LA,
123. Zhu MJ, Du M, Nathanielsz PW, Ford SP Friedman JE (2010) Maternal obesity and
(2010) Maternal obesity up-regulates inflam- fetal metabolic programming: a fertile epige-
matory signaling pathways and enhances netic soil. Am J Physiol 299:R711–R722
cytokine expression in the mid-gestation
sheep placenta. Placenta 31:387–391 133. Li CC, Maloney CA, Cropley JE, Suter CM
(2010) Epigenetic programming by maternal
124. Murabayashi N, Sugiyama T, Zhang L, nutrition: shaping future generations. Epige-
Kamimoto Y, Umekawa T, Ma N, Sagawa N nomics 2:539–549
(2013) Maternal high-fat diets cause insulin
resistance through inflammatory changes in 134. Li CC, Young PE, Maloney CA, Eaton SA,
fetal adipose tissue. Eur J Obstet Gynecol Cowley MJ, Buckland ME, Preiss T, Hen-
Reprod Biol 169:39–44 stridge DC, Cooney GJ, Febbraio MA, Mar-
tin DI, Cropley JE, Suter CM (2013)
125. Junien C, Gallou-Kabani C, Vigé A, Gross Maternal obesity and diabetes induces latent
MS (2005) Nutritional epigenomics: conse- metabolic defects and widespread epigenetic
quences of unbalanced diets on epigenetics changes in isogenic mice. Epigenetics 8:602
processes of programming during lifespan
and between generations. Ann Endocrinol 135. Haslam DW, James WP (2005) Obesity. Lan-
(Paris) 66:S19–S28 cet 366:1197–1209
Chapter 4
Abstract
Computer-Aided Drug Design has developed into a powerful suite of methods that complement experi-
mental approaches to the identification of new pharmacologically active compounds. In particular, virtual
screening has become a standard tool for lead identification. Diverse examples of the application of virtual
screening applied to T2DM target proteins have been reported. While several of these indicate successful
identification of new lead compounds from synthetic chemical and natural product databases, many of them
have been performed on a small scale and with limited validation. Careful study design and collaboration
with cheminformaticians and computational chemists will enable these approaches to fulfil their potential
for T2DM.
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_4, © Springer Science+Business Media, LLC, part of Springer Nature 2020
71
72 Paul W. Finn
Fig. 1 A typical virtual screening pipeline. An input database, which may contain tens of millions of unique
chemical structures, is searched against either a ligand or structure-based model of activity. Typically, the
initial rank-ordered list of “hits” is further processed by additional cheminformatic criteria and by manual
selection to identify the compounds to be tested experimentally. The final numbers chosen range from a
handful to several hundreds or thousands
Fig. 2 Since the mid-1990s there has been continual growth in the number of new protein structures that can
be used as input to computational methods
Table 1
Tools and databases for virtual screening
2 Virtual Screening
3 Natural Products
4 Polypharmacology
5 Conclusions
Fig. 3 Chemical structures of selected natural product PPAR modulators reported in the literature
References
1. Sterling T, Irwin JJ (2015) ZINC 15 – ligand by means of cheminformatics. Drug Des Devel
discovery for everyone. J Chem Inf Model Ther 7:279–288
55:2324–2337 6. Wang X-J, Zhang J, Wang S-Q et al (2014)
2. Pence HE, Williams A (2010) ChemSpider: an Identification of novel multitargeted PPARα/
online chemical information resource. J Chem γ/δ pan agonists by core hopping of rosiglita-
Educ 87:1123–1124 zone. Drug Des Devel Ther 8:2255–2262
3. Gaulton A, Bellis LJ, Bento AP et al (2011) 7. Gee VM, Wong FS, Ramachandran L et al
ChEMBL: a large-scale bioactivity database for (2014) Identification of novel peroxisome
drug discovery. Nucleic Acids Res 40: proliferator-activated receptor-gamma
D1100–D1107 (PPARγ) agonists using molecular modeling
4. Ma Y, Wang S-Q, Xu W-R et al (2012) Design method. J Comput Aided Mol Des
novel dual agonists for treating type-2 diabetes 28:1143–1151
by targeting peroxisome proliferator-activated 8. Tanwar O, Tanwar L, Shaquiquzzaman M et al
receptors with core hopping approach. PLoS (2014) Structure based virtual screening of
One 7:e38546. https://doi.org/10.1371/ MDPI database: discovery of structurally
journal.pone.0038546 diverse and novel DPP-IV inhibitors. Bioorg
5. Liu L, Ma Y, Wang R-L et al (2013) Find novel Med Chem Lett 24:3447–3451
dual-agonist drugs for treating type 2 diabetes
New Diabetes Drugs from Cheminformatics 83
35. Gu J, Chen L, Yuan G et al (2013) A drug- 38. Priyadarsini KI (2013) Chemical and structural
target network-based approach to evaluate the features influencing the biological activity of
efficacy of medicinal plants for type II diabetes curcumin. Curr Pharm Des 19:2093–2100
mellitus. Evid Based Complement Alternat 39. Ingólfsson HI et al (2014) Phytochemicals per-
Med 2013:203614 turb membranes and promiscuously alter pro-
36. Tian S, Li Y, Li D et al (2013) Modelling tein function. ACS Chem Biol 9:1788–1798
compound-target interaction network of tradi- 40. Weidner C et al (2012) Amorfrutins are potent
tional Chinese medicines for type II diabetes antidiabetic dietary natural products. Proc Natl
mellitus: insight for polypharmacology and Acad Sci U S A 109:7257–7262
drug design. J Chem Inf Model 53:1787–1803 41. Zhou W, Hevener KE et al (2009) A statistical
37. Baell JB, Holloway GA (2010) New substruc- framework to evaluate virtual screening. BMC
ture filters for removal of pan assay interference Bioinformatics 10:225
compounds (PAINS) from screening libraries 42. Scior T, Bender A et al (2012) Recognizing
and for their exclusion in bioassays. J Med pitfalls in virtual screening: a critical review. J
Chem 53:2719–2740 Chem Inf Model 52:867–881
Chapter 5
Abstract
Next-generation sequencing (NGS) is transforming clinical research and diagnostics, vastly enhancing our
ability to identify novel disease-causing genetic mutations and perform comprehensive diagnostic testing in
the clinic. Whole-exome sequencing (WES) is a commonly used method which captures the majority of
coding regions of the genome for sequencing, as these regions contain the majority of disease-causing
mutations. The clinical applications of WES are not limited to diagnosis; the technique can be employed to
help determine an optimal therapeutic strategy for a patient considering their mutation profile. WES may
also be used to predict a patient’s risk of developing a disease, e.g., type 2 diabetes (T2D), and can therefore
be used to tailor advice for the patient about lifestyle choices that could mitigate those risks. Thus, genome
sequencing strategies, such as WES, underpin the emerging field of personalized medicine. Initiatives also
exist for sharing WES data in public repositories, e.g., the Exome Aggregation Consortium (ExAC)
database. In time, by mining these valuable data resources, we will acquire a better understanding of the
roles of both single rare mutations and specific combinations of common mutations (mutation signatures)
in the pathology of complex diseases such as diabetes.
Herein, we describe a protocol for performing WES on genomic DNA extracted from blood or saliva.
Starting with gDNA extraction, we document preparation of a library for sequencing on Illumina instru-
ments and the enrichment of the protein-coding regions from the library using the Roche NimbleGen
SeqCap EZ Exome v3 kit; a solution-based capture method. We include details of how to efficiently purify
the products of each step using the AMPure XP System and describe how to use qPCR to test the efficiency
of capture, and thus determine finished library quality.
Key words NGS, WES, Exome-seq, Sequencing, AMPure XP, NEBNext, Illumina, qPCR
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_5, © Springer Science+Business Media, LLC, part of Springer Nature 2020
85
86 Milind C. Mahajan and Andrew S. McLellan
datasets annotated and placed into the public domain, the more
accurate predictions of pathogenicity become.
WES is commonly performed using short-read sequencing
using Illumina platforms (NextSeq, HiSeq series, and NovaSeq
series), which use the sequencing by synthesis (SBS) method
[22, 23]. Sequencing may be up to 150 bp, but can be performed
“paired-end” meaning that both ends of a DNA fragment are
sequenced, aiding alignment. Illumina are the market leader in
short read NGS technologies [22, 23], but there are other plat-
forms available which have their strengths and weaknesses. For
example, short read sequencing has limited use in de-novo assem-
bly or for resolving large structural variants and complex or repeti-
tive regions. Long-read sequencing platforms, e.g., the Pacific
BioSciences RSII and Sequel, excel in this space, being able to
sequence regions 8–20 kb in length [22, 23].
We herein provide a working protocol for the preparation of a
gDNA library followed by exome enrichment using the Roche
NimbleGen SeqCap v3 kit to obtain a final WES library designed
for sequencing on the Illumina family of sequencers. A diagram of
the entire workflow is shown in Fig. 1. This protocol is by no means
a promotion of the reagents and kits used, but is intended as a
general practical guide that can be used for any WES library prepa-
ration kit of choice. Increasingly, kits for library preparation are
becoming available that provide faster turnaround time with
reduced amplification cycles and which combine several reactions
into one step. We have chosen the NEBNext kit, which yields
excellent quality WES libraries compatible with the Illumina
HiSeq and NovaSeq sequencers (we currently use the NovaSeq).
This protocol is designed to be used for the preparation of WES
libraries from 10 to 12 DNA samples simultaneously using Eppen-
dorf tubes. However, the protocol can easily be adapted for high-
throughput WES library preparation in a 96-well format, using
liquid handlers and automation.
2 Materials
2.2 Preparation 1. NEBNext DNA Library Prep Master Mix Set for Illumina
of the gDNA (NEB, Ipswich, MA).
Sequencing Library 2. KAPA HiFi HotStart ReadyMix (KAPA Biosystems, Wilming-
ton, MA, USA): KAPA HiFi HotStart DNA Polymerase in a
proprietary reaction buffer, dNTPs (0.3 mM of each dNTP at
1), MgCl2 (2.5 mM at 1) and stabilizers.
3. Qubit dsDNA BR Assay Kit (ThermoFisher Scientific, Wal-
tham, MA, USA).
3 Methods
3.1 Purification Purification and size selection of DNA can be carried out by con-
and Size Selection ventional agarose gel chromatography. However, it is a
of DNA During WES low-throughput and time-consuming process that requires a large
Library Preparation amount of input DNA and the gel extraction process results in the
Steps loss of a large proportion of the gel-isolated fraction. Thus, Solid
Phase Reversible Immobilisation (SPRI) beads are used to achieve
efficient purification and size selection of nanogram to microgram
levels of DNA [24]. SPRI beads are paramagnetic, which prevents
them from clumping and falling out of solution. Each bead is made
of polystyrene surrounded by a layer of magnetite and is coated
with carboxyl molecules that are variably protonated/deprotonated
depending on pH. These carboxylic groups reversibly bind DNA in
the presence of polyethylene glycol (PEG) and salt (20% PEG,
2.5 M NaCl) binding buffer and can be eluted by 10–20 mM
Tris-HCl pH 8–8.5. This system is ideally suited to purifying
from low concentrations of DNA, is easy to use, and is amenable
to automation, facilitating high-throughput DNA purification and
size selection (see Note 1).
92 Milind C. Mahajan and Andrew S. McLellan
3.1.1 Random Classically, fragmentation of gDNA has been achieved via physical
Fragmentation of Genomic shearing of the DNA, which yields unbiased random DNA frag-
DNA by Sonication ments representing the whole genome in the final library. Recently,
however, several vendors including Kapa Biosystems and NEB have
developed enzymatic shearing protocols, using endonucleases to
create double-stranded breaks in the DNA. The advantage of enzy-
matic fragmentation is that it does not require any specialized
equipment; however, efficacy, consistency, and reliability of enzy-
matic shearing needs to be established. Hence, we describe Covaris-
based ultrasonic shearing of DNA for the preparation of WES
libraries.
The Covaris sonicator transmits focused acoustic energy waves
to randomly fragment DNA (by physical shearing). Prior to use, the
sonicator must be set up (see Note 2).
1. Transfer 0.5–3 μg of gDNA in 120 μL of nuclease-free water
into Covaris MicroTubes.
2. Vortex MicroTubes containing DNA samples briefly, then cen-
trifuge to remove air bubbles (see Note 3). If air bubbles
remain, a second centrifugation step may be necessary.
3. Place the MicroTubes in the MicroTube rack (see Note 4), then
place the rack in the sonication chamber. We use the following
parameters to obtain a DNA size range of 150–400 base pairs,
with mean peak size in the range of 200–250 base pairs:
time ¼ 600 s, duty ¼ 5%, intensity ¼ 2, cycle per burst ¼ 50
(see Note 5).
4. After fragmentation is complete, remove the shearing plate
from the sonicator, wipe the water from the outside of the
tubes and pulse spin in a centrifuge.
5. For each sample, transfer the sheared DNA to a 1.5 mL
Eppendorf tube.
6. Assess the quality of the sheared genomic DNA using an Agi-
lent 2100 Bioanalyzer: Run sheared DNA on the DNA HS
chip using the Bioanalyzer to determine the size of sheared
DNA (Fig. 2).
7. You may pause the protocol at this point (see Note 6).
Fig. 2 Determining optimal shearing of DNA using the Bioanalyzer. (a) Bioanalyzer trace of the input DNA,
showing size and integrity. (b) Optimally sheared gDNA. (c) Poorly sheared gDNA
3.2 Preparation The NEB Next™ DNA Sample Prep kit is used for constructing the
of the Whole Genome gDNA library from the fragmented DNA. This kit contains master
DNA (gDNA) mixes of enzymes and reagents for end repair of fragmented DNA,
Sequencing Library dA-tailing, ligation of synthetic oligonucleotide adaptors and
amplification of the adaptor-ligated DNA, using adaptor-specific
barcoded PCR primers, to obtain the final gDNA library. After each
of these four steps, DNA is purified using AMPure beads (see Note
9), selecting a binding buffer with the appropriate concentration of
PEG and NaCl (as detailed above).
94 Milind C. Mahajan and Andrew S. McLellan
3.2.1 End Repair Bead-bound purified sheared DNA from step 11 above (Subhead-
of Sheared DNA ing 3.1.2) is used to create blunt-ended DNA by end repair.
1. For each sample, set up the end repair reaction mix as follows:
5 μL of NEB Next End Repair Reaction Buffer (10), 2.5 μL
of NEBNext End Repair Enzyme Mix, 42.5 μL of AMPure
bead-bound sheared DNA (50 μL total).
2. Incubate in a thermocycler for 30 min at 20 C to complete the
end repair reaction.
3.2.2 Cleaning Up Prior to using the end-repaired DNA, it must first be purified as
End-Repaired DNA follows:
1. Add 90 μL of SPRI binding buffer (20% PEG-2.5 M NaCl)
solution to the 50 μL of end repair reaction mix to bind the
DNA back to the beads.
2. Pipette up and down 20 times to resuspend the beads and
incubate at room temperature for 5 min.
3. Spin the tube for 1 min at 3000 g in a microcentrifuge. Place
the tube on the magnetic stand for 3 min or until the solution is
clear.
4. Without disturbing the beads, remove 135 μL of supernatant
and discard. A small volume of supernatant will be left in
each well.
5. Continue to keep the tube on the magnetic stand and add
200 μL of freshly prepared 70% ethanol, incubate for 30 s
then discard the ethanol.
6. Repeat step 5 for a total of two ethanol washes.
7. Dry the samples at room temperature for 10 min or until the
residual ethanol completely evaporates.
8. Add 42 μL of SPRI elution buffer and pipette up and down
15 times to resuspend the beads. The bead-bound DNA is now
ready for dA-tailing.
9. You may pause the protocol at this point to continue next day
or proceed to the dA-tailing step described below in Subhead-
ing 3.2.3 (see Note 8).
3.2.3 Addition 1. Prepare the dA-tailing reaction mix using reagents from the
of Deoxyadenine to the 30 NEBNext DNA Library Prep Master Mix Set for Illumina as
End of Blunt-Ended DNA follows: 5 μL of NEBNext dA-Tailing Reaction Buffer (10),
(dA-Tailing) 3 μL of Klenow Fragment (30 ! 50 exo), 42 μL of DNA-bead
mix after end repair step (total of 50 μL).
2. Incubate the reaction mix at 37 C for 30 min to obtain DNA
fragments with dA-overhangs on the 30 end.
Whole Exome Sequencing (WES) for Illumina HiSeq 95
3.2.5 Ligation of Illumina 1. Prepare the Adaptor Ligation reaction mix as follows: 10 μL of
Adaptors to dA-Tailed DNA NEBNext Quick Ligation Reaction Buffer (5), 10 μL of
DNA Multiplex Adaptor (10 μM), 5 μL of Quick T4 DNA
Ligase, 25 μL of Quick T4 DNA Ligase (total volume of
50 μL). Adaptor sequences are shown in Table 1.
2. Incubate for 60 min at 20 C to allow ligation to occur.
Table 1
Illumina Adaptor sequences ligated to dA-tailed gDNA during library preparation (rows 1 and 2)
3.2.7 Amplification The quantity of adaptor-ligated gDNA library is currently too small
of the gDNA Library to proceed with exome capture by solution hybridization. It must
first be amplified by a few PCR cycles as necessary to obtain a yield
of 500 ng.
1. Prepare the PCR mix in a pre-PCR hood (to reduce the risk of
contamination with aerosolized PCR products) as follows:
25 μL KAPA HiFi HotStart ReadyMix, 2.5 μL of forward
PCR primer (10 μM), 2.5 μL of indexed reverse primer
(10 μM), 20 μL of adaptor-ligated DNA (50 μL total volume).
PCR primer sequences are shown in Table 1.
2. Set up the PCR reaction on a thermocycler using the following
program: 98 C for 5 min (denaturation), followed by 8 cycles
of 98 C for 1 min, 60 C for 30 s and 72 C for 45 s (amplifi-
cation), then finish with 72 C for 10 min (extension). Hold at
4 C until ready for the next stage.
3. Clean up 50 μL of PCR amplified DNA using 60 μL of AMPure
XP beads as described in Subheading 3.2.6 (see Note 12).
Whole Exome Sequencing (WES) for Illumina HiSeq 97
Fig. 3 A Bioanalyzer trace showing the profile of an optimally prepared genomic DNA library
3.2.8 Removal of High 1. Remove a stock tube of AMPure XP beads from storage at 4 C
Molecular Weight DNA and allow to equilibrate to room temperature.
from the gDNA Library 2. Centrifuge the sample plate at 300 g for 30 s to ensure all
components are collected at the bottom of each well.
3. To 30 μL of PCR amplified WES library, add 19.5 μL of
AMPure beads. This is a 0.65 ratio of beads to sample
98 Milind C. Mahajan and Andrew S. McLellan
Fig. 4 A Bioanalyzer trace obtained before removal of high molecular weight DNA from a genomic DNA library
3.3 Enrichment The gDNA library prepared as described above comprises sequence
of Exonic Content content from the entire genome out of which only ~2% belongs to
of the gDNA Library the coding portion of the genome. To isolate/enrich this relatively
(Exome Enrichment) tiny fraction of the total gDNA library, a solution hybridization-
based capture method can be employed. There is an assortment of
vendors producing kits for generating capture libraries for WES and
most use tiled or overlapping biotinylated DNA or RNA capture
probes (baits) and streptavidin-coated magnetic beads to isolate the
fragments containing exonic sequence. Of these, the most popular
kits are produced by Illumina, Roche NimbleGen, Agi-
lent, Integrated DNA Technologies (IDT) and Twist Bioscience.
Illumina offers three kits based on the Nextera Rapid Capture
enrichment method. The Nextera Rapid Capture Exome kit targets
all coding sequence (~37 megabases), the Nextera Rapid Capture
Expanded Exome kit also captures non-coding regions, UTRs and
miRNAs (~62 megabases) and the TrueSight One kit captures a
reduced set of exons (~12 megabases) that have been implicated in
at least one disease. Roche NimbleGen provides a similar selection
with its SeqCap EZ Exome Library v3.0 (~64 megabases), SeqCap
EZ Exome + UTR Library (~96 megabases), and SeqCap EZ
HGSC VCRome (~45 megabases) solution-based capture kits.
Agilent’s SureSelectXT Human All Exon V6 (~58 megabases),
SureSelectXT Human All Exon V6 + UTR (size not specified),
and SureSelectXT Focused Exon Capture (~12 megabases) are
equivalent. IDT offers the xGen Exome Research Panel (~39Mb),
and several disease specific and custom targeted panels, boasting a
comprehensively high on-target rate and uniform coverage, even
across GC-rich regions. Twist Bioscience is the latest entrant in the
growing list of vendors offering whole exome and targeted capture
solutions. It offers the Human Core Exome Kit (~33 Mb of highly
conserved protein-coding regions) and custom panels. The com-
pany has developed high-fidelity double stranded oligos which
are uniquely designed to capture both DNA strands, providing
enhanced specificity and uniformity across targeted regions without
unexpected dropouts. Some of these manufacturers also allow for
the addition of custom probes to the exome panel. While there are
widely reported advantages and disadvantages to specific kits
[17, 25] in general they appear to perform similarly and it is
100 Milind C. Mahajan and Andrew S. McLellan
Table 2
Preparation of wash buffers for washing and recovery of hybridized samples using Streptavidin
Dynabeads
3.3.2 Wash and Recovery 1. Dilute stock Stringent Wash Buffer, SC Wash Buffers (I, II, and
of Hybridized Samples III), and Bead Wash Buffer to create 1 working solutions as
shown in Table 2. The values in the table are for a single exome
Step 1. Prepare Sequence capture. For more exome captures, simply multiply each vol-
Capture Wash Buffers ume by the number of samples (see Notes 15–17).
2. Preheat 1 Stringent Wash Buffer and one-third of the volume
of 1 SC Wash Buffer I to 47 C in a water bath.
Step 3. Bind DNA 1. Transfer the ~15 μL of amplified sample library/COT DNA/
to the Streptavidin PE-HE Oligos/Hybridization cocktail (which should have
Dynabeads been hybridized for 64–72 h) to the Streptavidin Dynabeads
prepared in step 2.
2. Mix thoroughly by pipetting up and down 10 times.
3. Bind the captured sample to the beads by placing the tubes
containing the beads and DNA in a thermocycler at 47 C for
45 min. Mix the samples by vortexing for 3 s every 15 min to
ensure that the beads remain in suspension (see Note 20).
Step 4. Wash 1. If more than eight captures are being washed, transfer the
the Streptavidin Dynabeads entire contents (~115 μL) of each tube to a 96-deep well
Plus Bound DNA plate. If less than eight captures are being washed, samples
may remain in strip tubes.
2. Place the strip tube or 96-deep well plate on the magnet.
Remove the supernatant immediately once clear.
3. Add 100 μL of SC Wash Buffer I preheated to 47 C (see Note
21).
4. Cap tubes or seal plate and mix by vortexing for 10 s.
5. Place the tubes or plate on the magnet to bind the beads.
Remove and discard the liquid immediately once clear.
6. Remove the tubes or plate from the magnet and add 200 μL of
Stringent Wash Buffer preheated to 47 C. Pipette up and
down 5 times to mix (see Note 21).
7. Incubate at 47 C for 5 min. If captures are in a 96-deep well
plate, use an incubation chamber. If samples are in a strip tube,
use a PCR block with heated lid.
8. Repeat steps 5–7 for a total of two washes with Stringent Wash
Buffer heated to 47 C.
Whole Exome Sequencing (WES) for Illumina HiSeq 103
9. Place the tubes or plate on the magnet to bind the beads. After
5 min remove and discard the supernatant.
10. Add 200 μL of room temperature SC Wash Buffer I and mix by
vortexing for 2 min. If liquid has collected in the tube’s cap,
pulse-spin in a microcentrifuge to collect the liquid into the
tube’s bottom before continuing to the next step.
11. Place the tubes or plate on the magnet to bind the beads.
Remove and discard the liquid once clear.
12. Add 200 μL of room temperature SC Wash Buffer II and mix
by vortexing for 1 min. Pulse-spin in a microcentrifuge if
necessary.
13. Place the tubes or plate on the magnet to bind the beads.
Remove and discard the liquid once clear.
14. Add 200 μL of room temperature SC Wash Buffer III and mix
by vortexing for 30 s. Pulse-spin in a microcentrifuge if
necessary.
15. Place the tubes or plate on the magnet to bind the beads.
Remove and discard the liquid once clear.
16. Remove the tubes or plate from the magnet and add 20 μL of
PCR grade water to each tube of bead-bound exome-captured
library (see Note 22).
3.4 Final The quantity of exome enriched library is usually less than 1 ng,
Amplification which is too low to be used for sequencing on the Illumina
of Captured Library sequencer directly and must be amplified using a limited number
of PCR cycles.
1. Generate a PCR Master Mix by adding the following compo-
nents: 25 μL of KAPA HiFi HotStart ReadyMix, 2.5 μL of IS
5 Primer (10 μM), 2.5 μL of IS 6 Primer (10 μM), 20 μL of
exome captured library with beads (total volume of 50 μL). See
Table 1 for primer sequences.
2. Run the PCR reaction on a thermocycler using the following
program: 98 C for 10 min (denaturation), followed by
12 cycles of 98 C for 1 min, 60 C for 30 s and 72 C for
45 s, then finish with 72 C for 10 min. Hold at 4 C until
ready for the next stage.
3. Clean up the post-capture PCR product using AMPure XP
beads as shown in Subheading 3.2.6.
4. Elute WES library DNA in a final volume of 30 μL of nuclease-
free water.
5. Assess library size on a Bioanalyzer and quantify using the
Qubit dsDNA BR Assay Kit. On the Bioanalyzer, the trace
should look as shown in Fig. 5.
104 Milind C. Mahajan and Andrew S. McLellan
3.5 Assessing To assess if the exonic regions of the gDNA library were success-
the Quality of Exonic fully enriched, qPCR can be performed using exon-specific primers
Enrichment in a WES for a couple of genes. Significant enrichment of targeted exonic
Library by qPCR regions in a WES library over the original gDNA library indicates a
successful outcome of the exome enrichment process described
above. Roche NimbleGen recommends verifying two exonic
regions, and also two intronic regions as controls. When running
qPCR for verification of exon enrichment, it is necessary to include
both the input gDNA Seq library (prepared in Subheading 3.2) and
the amplified capture library (prepared in Subheading 3.4). Every
qPCR assay must include standard curves for all four amplicons
(two exons and two introns) using a dilution series of wild-type
human genomic DNA, and a no template control (NTC) must be
included. The dissociation curves for all four amplicons must be
reviewed to ensure that no unexpected PCR products are gener-
ated. Compare the ratios of the results (Ct values) from qPCR of all
the exonic and intronic regions in the capture libraries vs. the input
libraries to determine whether enrichment of exonic regions has
been successful.
1. Determine the concentration of each of the samples using
either the Qubit or Bioanalyzer. Each 15 μL of qPCR reaction
will contain 0.5 ng of library DNA. Make working dilutions of
all libraries to a concentration of 0.1 ng/μL.
2. For quantitation of amplicon enrichment for a given qPCR
reaction, each qPCR assay must include a standard curve gen-
erated using a dilution series of normal human genomic DNA
Whole Exome Sequencing (WES) for Illumina HiSeq 105
Table 3
qPCR results for quality control analysis of an example WES library “WES-Seq library-1”
Table 4
qPCR results for quality control analysis of an example WES library “WES-Seq library-2”
4 Notes
References
1. Prasad RB, Groop L (2015) Genetics of type mellitus using next-generation sequencing of
2 diabetes—pitfalls and possibilities. Genes 6 the whole exome. PLoS One 5(10):e13630
(1):87–123 15. Shaer N, Khan JJ, Dallol A, Abuzenadah A
2. Murea M, Ma L, Freedman BI (2012) Genetic (2014) Diabetes personalized: the use of
and environmental factors associated with type whole-exome sequencing in order to identify
2 diabetes and diabetic vascular complications. familial type 2 diabetes mellitus susceptibility
Rev Diabet Stud 9(1):6–22 factors. BMC Genomics 15(Suppl 2):P51
3. Lander ES, Linton LM, Birren B et al (2001) 16. Ellingford JM, Barton S, Bhaskar S et al (2016)
Initial sequencing and analysis of the human Whole genome sequencing increases molecular
genome. Nature 409(6822):860–921 diagnostic yield compared with current diag-
4. Muir P, Li S, Lou S et al (2016) The real cost of nostic testing for inherited retinal disease.
sequencing: scaling computation to keep pace Ophthalmology 123(5):1143–1150
with data generation. Genome Biol 17:53 17. Meienberg J, Zerjavic K, Keller I et al (2015)
5. Wetterstrand KA (2016) DNA sequencing New insights into the performance of human
costs: data from the NHGRI genome sequenc- whole-exome capture platforms. Nucleic Acids
ing program (GSP). www.genome.gov/ Res 43(11):e76
sequencingcostsdata. Accessed 27 Feb 2017 18. Rabbani B, Tekin M, Mahdieh N (2014) The
6. Dorajoo R, Liu J, Boehm BO (2015) Genetics promise of whole-exome sequencing in medi-
of type 2 diabetes and clinical utility. Genes cal genetics. J Hum Genet 59(1):5–15
6:372–384 19. Kwak SH, Jung CH, Ahn CH et al (2016)
7. Dickson SP, Wang K, Krantz I, Hakonarson H Clinical whole exome sequencing in early
et al (2010) Rare variants create synthetic onset diabetes patients. Diabetes Res Clin
genome-wide associations. PLoS Biol 8: Pract 122:71–77
e1000294. https://doi.org/10.1371/journal. 20. Lyssenko V, Laakso M (2013) Genetic screen-
pbio.1000294 ing for the risk of type 2 diabetes: worthless or
8. Fuchsberger C, Flannick J, Teslovich TM et al valuable? Diabetes Care 36(Suppl 2):
(2016) The genetic architecture of type 2 dia- S120–S126
betes. Nature 536(7614):41–47 21. Exome Aggregation Consortium, Lek M,
9. Katsanis SH, Katsanis N (2013) Molecular Karczewski KJ, Minikel EV et al (2016) Analy-
genetic testing and the future of clinical geno- sis of protein-coding genetic variation in
mics. Nat Rev Genet 14(6):415–426 60,706 humans. Nature 536(7616):285–291
10. Ellard S, Lango Allen H, De Franco E et al 22. Goodwin S, McPherson JD, McCombie WR
(2013) Improved genetic testing for mono- (2016) Coming of age: ten years of next-
genic diabetes using targeted next-generation generation sequencing technologies. Nat Rev
sequencing. Diabetologia 56:1958 Genet 17(6):333–351
11. Warr A, Robert C, Hume D et al (2015) 23. Levy SE, Myers RM (2016) Advancements in
Exome sequencing: current and future per- next-generation sequencing. Annu Rev Geno-
spectives. G3 5(8):1543–1550 mics Hum Genet 17:95–115
12. Samorodnitsky E, Jewell BM, Hagopian R et al 24. DeAngelis MM, Wang DG, Hawkins TL
(2015) Evaluation of hybridization capture (1995) Solid-phase reversible immobilization
versus amplicon‐based methods for whole‐ for the isolation of PCR products. Nucleic
exome sequencing. Hum Mutat 36 Acids Res 23(22):4742–4743
(9):903–914 25. Chilamakuri CSR, Lorenz S, Madoui M-A,
13. Lelieveld SH, Spielmann M, Mundlos S et al Vodák D, Sun J, Hovig E, Myklebost O,
(2015) Comparison of exome and genome Meza-Zepeda LA (2014) Performance com-
sequencing technologies for the complete cap- parison of four exome capture systems for
ture of protein‐coding regions. Hum Mutat 36 deep sequencing. BMC Genomics 15(1):449
(8):815–822 26. Roche Sequencing (2017) SeqCap EZ System.
14. Bonnefond A, Durand E, Sand O et al (2010) http://sequencing.roche.com/products/
Molecular diagnosis of neonatal diabetes nimblegen-seqcap-target-enrichment/seqcap-
ez-system.html. Accessed 27 Feb 2017
Chapter 6
Abstract
Gene expression analysis by microarray and more recently by next-generation sequencing has become a core
part of biomedical research and its value can be seen in thousands of research papers. A successful gene
expression experiment needs to be augmented by specialized data mining techniques if the data are to be
fully exploited. Here, tools that concentrate on three areas—gene enrichment analysis, literature mining,
and transcription factor binding site analysis—are described for the novice user of microarray and next
generation sequencing technologies. The focus of this chapter is on free, publicly available, web-based
tools.
Key words Bioinformatics, Microarray, Next generation sequencing, Gene expression, Data mining,
Gene enrichment, Literature mining, Transcription factor binding site
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_6, © Springer Science+Business Media, LLC, part of Springer Nature 2020
109
110 Donald R. Dunbar
1.1 Gene Enrichment Gene enrichment analysis uses statistical procedures to discover
Analysis overrepresented features within a dataset. Given a gene list and
some biological information, these tools can identify categories
(e.g., biological processes and molecular functions from the Gene
Ontology consortium or molecular pathways from KEGG) that
appear more often in the gene set that they would by chance in a
random gene set of the same size. If a category is overrepresented,
then this might have biological significance and point to that part of
biology being perturbed in the experiment [1]. Gene enrichment
analysis with the online tool DAVID (Database for Annotation,
Visualization, and Integrated Discovery) will be described [2]. A
tool with a broader and complementary scope than DAVID,
Enrichr, has similar functionality to look for enriched biological
functions [3, 4] and is used in a very similar way to DAVID. Enrichr
gives access to libraries of data including transcription, pathways,
ontologies, diseases, and drug and is very intuitive to explore.
1.2 Literature Mining Literature Mining probes the huge body of information available to
biomedical scientists, for example in the Medline database [5]. If
two or more genes or proteins occur in the same paper, it is possible
that they have some sort of association. Two main problems face
the biologist when mining the literature, however. First, because
there are often hundreds of genes of interest, looking for pairwise
associations using manual web-based searching (e.g., through the
PubMed web interface) is unfeasible. Computationally, however,
these searches are trivial. The PubMatrix text mining tool will be
described [6]. Second, genes often have multiple synonyms that are
used in the literature. For example, the human gene transcription
Microarrays in T2D 111
1.3 Transcription Transcription factor binding site (TFBS) analysis can identify bind-
Factor Binding Site ing sites that are statistically overrepresented in the sequences of a
(TFBS) Analysis gene list. Finding such enriched sites can give an idea of the factors
that are driving gene expression locally at the cellular level in the
experimental model. TFBSs often bind to several similar sequences
and computational tools that access TFBS databases can use
sequence models to predict binding sites in gene sequences
[8]. In addition, if a binding site is conserved between species, it
is more likely to be functionally important. Whole Genome RVista
allows for identification of conserved (between pairs of species)
statistically overrepresented TFBSs in specified regions (e.g., 5 kb
upstream) of groups of genes [9].
2 Materials
3 Methods
and 3.5; gene symbols and names for Subheading 3.3) for further
use. Other identifiers are available and documentation in each of
the tools will describe the appropriate one. Data should come to the
user in the form of a spreadsheet or a tab delimited text file.
3.1 Gene List 1. Open the file containing gene expression data with spreadsheet
Preparation software.
2. Copy the gene identifiers.
3. Paste into a new spreadsheet.
4. Sort the identifiers.
5. Remove any duplicate entries (see Note 1).
6. Save as a plain text file with appropriate name, e.g.,
“exp1_up_2fold_AffyIDs.txt.”
3.2 Gene Enrichment 1. Open the gene list text file (see Note 2).
Analysis with DAVID 2. Copy the identifiers, noting their type (e.g., Affymetrix ID,
Entrez Gene IDs).
3. Go to DAVID website: https://david.ncifcrf.gov/.
4. Click “Start Analysis” in the menu bar.
5. Paste the gene list into “Step 1: Enter Gene List” box.
6. Select the type of identifier (e.g., AFFY_ID) in “Step 2: Select
Identifier”.
7. Select “Gene List” from “Step 3: List Type.”
8. Submit List (this will take you to the analysis wizard).
9. Click “Rename,” rename the list in the dialog box, click “OK.”
10. The “Current Background” will be automatically selected (see
Note 3).
11. If your data are Affymetrix IDs, click “Background” in the
navigation bar.
12. Click the “+” beside “Affymetrix.” Choose the appropriate
GeneChip.
13. Click “Functional Annotation Tool” in the analysis wizard.
14. Click “Functional Annotation Chart.”
15. Click the “+” for “Options.” Check “Fold Enrichment” then
“Re-run Using Options.”
16. Explore the results (see Note 4).
4 Notes
References
1. Curtis RK, Oresic M, Vidal-Puig A (2005) Path- 6. Becker KG, Hosack DA, Dennis G, Lempicki
ways to the analysis of microarray data. Trends RA, Bright TJ, Cheadle C et al (2003) PubMa-
Biotechnol 23(8):429–435 trix: a tool for multiplex literature mining. BMC
2. Huang DW, Sherman BT, Lempicki RA (2009) Bioinformatics 4:61
Systematic and integrative analysis of large gene 7. Hoffmann R, Valencia A (2004) A gene network
lists using DAVID bioinformatics resources. Nat for navigating the literature. Nat Genet 36
Protoc 4(1):44–57 (7):664
3. Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, 8. Elnitski L, Jin VX, Farnham PJ, Jones SJM
Meirelles GV et al (2013) Enrichr: interactive (2006) Locating mammalian transcription factor
and collaborative HTML5 gene list enrichment binding sites: a survey of computational and
analysis tool. BMC Bioinformatics 14:128 experimental techniques. Genome Res 16
4. Kuleshov MV, Jones MR, Rouillard AD, Fernan- (12):1455–1464
dez NF, Duan Q, Wang Z et al (2016) Enrichr: a 9. Zambon AC, Zhang L, Minovitsky S, Kanter JR,
comprehensive gene set enrichment analysis web Prabhakar S, Salomonis N et al (2005) Gene
server 2016 update. Nucleic Acids Res 44(W1): expression patterns define key transcriptional
W90–W97 events in cell-cycle regulation by cAMP and pro-
5. Jensen LJ, Saric J, Bork P (2006) Literature tein kinase A. Proc Natl Acad Sci U S A 102
mining for the biologist: from information (24):8561–8566
retrieval to biological discovery. Nat Rev Genet
7(2):119–129
Chapter 7
Abstract
Profiling genome-wide transcriptional changes with advanced high-throughput transcriptional profiling
techniques has led to a revolution in biomedical science. It has been challenging to handle the massive data
generated by these techniques and draw meaningful conclusions from it. Therefore, computational biolo-
gists have developed a number of innovative methods of varying complexity and effectiveness to analyze
such complex data. Over the past decade, rich information in pathway repositories has attracted and
motivated researchers to incorporate such existing biological knowledge into computational analysis tools
to develop what is known as pathway enrichment analysis tools. This chapter describes a new sophisticated
pathway enrichment tool that exploits topology of pathway as well as expression of significantly changed
genes to identify biologically significant pathways for high-dimensional gene expression datasets. Also, we
demonstrate the use of this tool to analyze gene expression data from a type 2 diabetes dataset to identify a
list of significantly enriched metabolic pathways.
Key words Pathway analysis, Metabolic pathway, Gene expression, Type 2 diabetes, Microarray
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_7, © Springer Science+Business Media, LLC, part of Springer Nature 2020
119
120 Maysson Ibrahim
2 Materials
3 Methods
3.1 Software 1. If you do not have MATLAB on your PC, you should initially
Installation install the MCR (MATLAB Compiler Runtime) as explained in
the tool webpage (http://www.buckingham.ac.uk/research/
clore-laboratory-diabetes-obesity-and-metabolic-research/
staff/maysson-al-haj-ibrahim/prs-tool/).
2. Click on the “Topology-based Pathway Analysis Tool” hyper-
link in the tool webpage to download the .zip file.
3. Right-click on the .zip format file to extract all relevant files,
and then run the PRS_interface file.
3.2 Data 1. Prior to the analysis of gene expression data, some information
Preprocessing should be checked such as the array platform, number of
samples per group, species, and whether data is raw or
normalized.
2. The tool does not provide a filter to normalize data. Therefore,
users should normalize the data before using the tool for
analysis (see Note 1).
3. Data must be saved in a form of simple Excel spreadsheet, in
which the first column should be a probe ID, and the following
columns are normalized gene expression values from control
and test groups (see Note 2).
122 Maysson Ibrahim
3.3 Pathways 1. Right-click on the main file to open the GUI (see Fig. 2).
Enrichment with PRS 2. Select the type of species (e.g., Human, Mouse, Rat) from the
drop-down menu.
3. Click the “browse” button to select the input data file (.xls
format) pre-prepared in step 3 of Subheading 3.2.
4. Enter the number of samples in group 1 and 2 (i.e., group1
might refer to control or test group based on which one comes
first in the input file).
5. Enter the fold-change threshold for selecting highly changed
genes (see Note 3).
6. Enter the p-value threshold for selecting significantly changed
genes (see Note 4).
7. Select the normalization method (e.g., RMA/GCRMA, MAS)
used for normalizing your data.
8. Select the platform (e.g., Affy HG U133A, Affy HG U133B,
Affy HG U133 Plus 2.0, Affy HG U95A, Agilent) that is
compatible with your data.
9. Finally, select which group of pathways (signaling or metabolic
pathways, or both) you would like to map your data onto to
run the analysis.
10. Press the “Analyze” button to start the analysis and see the
results in the main interface (see Note 5).
3.4 Statistical 1. Click the “browse” button to select a results’ file (.mat format)
Evaluation that was generated from a previous analysis (see Note 6).
Pathways Analysis 123
3.5 Visualizing 1. In the main interface, activate the “Select a pathway to visua-
Enriched Pathway Map lize” checkbox option.
2. From the results’ table in the main interface, click on the
pathway that you want to visualize.
3. Click the “Apply” button and wait for the pathway map web-
page to open (see Note 9).
4 Notes
Fig. 3 “Arachidonic acid metabolism pathway” enriched in T2D dataset (significant genes are colored in red)
126 Maysson Ibrahim
Table 1
Input data information for pathways analysis and significance evaluation in T2D and pancreatic islets
dataset
PRS analysis
Species Human
# of samples in group1 9
# of samples in group2 54
Fold-change threshold 1.3
p-Value threshold 0.05
Normalization method RMA
Platform Affymetrix human gene 1.0 ST
Pathway type Both (metabolic and signaling)
Statistical significance evaluation
Number of permutations 1000
p-Value threshold 0.05
Table 2
Top ten pathways ranked by PRS score (T2D and pancreatic islets dataset)
Table 3
Top ten pathways ranked by z-score (T2D and pancreatic islets dataset)
References
1. Bauer-Mehren A, Furlong LI, Sanz F (2009) microarray platforms comparable? Genomics
Pathway databases and tools for their exploita- 83(6):1164–1168
tion: benefits, current limitations and chal- 11. Li C, Wong W (2003) DNA-chip analyzer
lenges. Mol Syst Biol 5:290 (dChip). http://www.springerlink.com/
2. Khatri P, Sirota M, Butte AJ (2012) Ten years index/G5V0232463245R24.pdf
of pathway analysis: current approaches and 12. Pepper SD, Saunders EK, Edwards LE, Wilson
outstanding challenges. PLoS Comput Biol 8 CL, Miller CJ (2007) The utility of MAS5
(2):e1002375 expression summary and detection call algo-
3. Gao S, Wang X (2007) TAPPA: topological rithms. BMC Bioinformatics 8(1):273
analysis of pathway phenotype association. Bio- 13. Do JH, Choi D (2006) Normalization of
informatics 23(22):3100–3102 microarray data: single-labeled and dual-
4. Vert JP, Kanehisa M (2003) Extracting active labeled arrays. Mol Cells 22(3):254
pathways from gene expression data. Bioinfor- 14. Persaud SJ, Muller D, Belin VD, Kitsou-
matics 19(Suppl 2):ii238–ii244 Mylona I, Asare-Anane H, Papadimitriou A
5. Tarca AL, Draghici S, Khatri P, Hassan SS, et al (2007) The role of Arachidonic acid and
Mittal P, Kim J et al (2009) A novel signaling its metabolites in insulin secretion from human
pathway impact analysis. Bioinformatics 25 islets of Langerhans. Diabetes 56(1):197–203
(1):75–82 15. Prentki M, Nolan CJ (2006) Islet cell failure in
6. Ibrahim MA, Jassim S, Cawthorne MA, Lang- type 2 diabetes. J Clin Invest 116
lands K (2014) A MATLAB tool for pathway (7):1802–1812
enrichment using a topology-based pathway reg- 16. Yaney GC, Corkey BE (2003) Fatty acid
ulation score. BMC Bioinformatics 15(1):358 metabolism and insulin secretion in pancreatic
7. Ibrahim MA, Jassim S, Cawthorne MA, Lang- beta cells. Diabetologia 46(10):1297–1312
lands K (2012) A topology-based score for 17. McGarry JD (2002) Banting lecture 2001 Dys-
pathway enrichment. J Comput Biol 19 regulation of fatty acid metabolism in the etiol-
(5):563–573 ogy of type 2 diabetes. Diabetes 51(1):7–18
8. Kanehisa M, Goto S, Kawashima S, Nakaya A 18. Kim H-S, Hwang Y-C, Koo S-H, Park KS, Lee
(2001) The KEGG databases at GenomeNet. M-S, Kim K-W et al (2013) PPAR-γ activation
Nucleic Acids Res 2(1):42–46 increases insulin secretion through the
9. Cheadle C, Vawter MP, Freed WJ, Becker KG up-regulation of the free fatty acid receptor
(2003) Analysis of microarray data using Z GPR40 in pancreatic β-cells. PLoS One 8(1):
score transformation. J Mol Diagn 5(2):73–81 e50128
10. J€arvinen A-K, Hautaniemi S, Edgren H, 19. Sugden MC, Holness MJ (2004) Potential role
Auvinen P, Saarela J, Kallioniemi O-P et al of peroxisome proliferator-activated receptor-α
(2004) Are data from different gene expression in the modulation of glucose-stimulated insu-
lin secretion. Diabetes 53(1):S71–S81
Chapter 8
Abstract
Monogenic diabetes and hyperinsulinism are genetically heterogeneous disorders. The determination of
the genetic etiology defines the diagnostic subtype, predicts prognosis, and importantly can guide clinical
management. This chapter focuses on the processes and methodologies utilized in the diagnostic testing for
monogenic diabetes and congenital hyperinsulinism (i.e., Sanger sequencing and targeted next-generation
sequencing).
Key words Sanger sequencing, Targeted next-generation sequencing, Neonatal diabetes, MODY,
Hyperinsulinism, KCNJ11 and ABCC8
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_8, © Springer Science+Business Media, LLC, part of Springer Nature 2020
129
130 Jayne A. L. Houghton
2 Materials
2.1 DNA Sample DNA is extracted from peripheral blood in EDTA within 5 days of
Preparation venesection using standard laboratory protocols or commercial
DNA extraction and purification kits. High-quality DNA (nonde-
graded, A260/A280 is 1.8–2.0) is essential for reliable PCR amplifi-
cation and sequencing.
Diabetes and Hyperinsulinism Genetic Testing 131
2.4 Sanger Sequencing reagents will vary because they will be specific to the
Sequencing: laboratory’s available sequencing equipment. We have found that
Sequencing Reagents the following produce high-quality data and fit with our automated
laboratory system.
1. 5 BigDye dilution buffer (Life Technologies Cat# 4336697).
2. BigDye terminator v3.1 (Life Technologies Cat# 4337455).
3. dH2O (Baxter (UKF7114).
4. M13 sequencing primers (Stock 100 pmol/μl).
5. POP7 Polymer (Life Technologies Cat#4342759).
6. 10 3730 running buffer (Life Technologies Cat# 4335613).
7. EDTA (0.5 mM).
Primer DNA
PCR concentration concentration Size
Exon method (pmol/μl) (ng/μl) Forward and reverse primer sequences (M13 tailed) chromosome number (bp)
1 MMR 2 (80% MF) 10 ABCC8ex1F_and_ABCC8ex1R agctgcaagggacagagg gagtgaagggatgagctgg 11 417
2 MMR 2 10 ABCC8ex2F_and_ABCC8ex2R GAGGCAACAGAGCAAGACC 422
Jayne A. L. Houghton
ACCCTGGAGCAGATTCACTT 11
3 MMR 2 10 ABCC8ex3F_and_ABCC8ex3R gccctgcagcctataaagtg ctccatgaaggcagggatt 11 390
4 MMR 2 10 ABCC8ex4F_and_ABCC8ex4R AAATGTACACACCCAGGCAC 411
GGGTAAAACAAGCTGATCCC 11
5 MMR 2 10 ABCC8ex5F_and_ABCC8ex5R gtgttggggaatccttttcc ccctttgaggtccctctctg 11 478
6 MMR 2 10 ABCC8ex6F_and_ABCC8ex6R GTTTCCCCAGACAACAGGAG 522
TGGTAGTGACGGTGAGAGGA 11
7 MMR 2 10 ABCC8ex7F_and_ABCC8ex7R CAGGGTGTAAGCAACCTTCC 453
TGAGGATGAATAACACTCATGGAC 11
8 MMR 2 10 ABCC8ex8F_and_ABCC8ex8R AAGTTGGAACGGTGATACAG 435
TGTGAAAGGTACAGGCAAGC 11
9 MMR 2 10 ABCC8ex9F_and_ABCC8ex9R GATAATTTGGAAACCTGGGC 387
TGAAGTGGCCTACTCAAAGTC 11
10 MMR 2 10 ABCC8ex10F_and_ABCC8ex10R TCTGGGAAATGGAGTCAATG 432
GAGTCGGATAATCTCAAGGC 11
11 MMR 2 10 ABCC8ex11F_and_ABCC8ex11R TGCCCCTAGCCTACTGGAG 248
CTGGGCAGCCTGTCACTG 11
12 MMR 2 10 ABCC8ex12F_and_ABCC8ex12R ATGAAGGTGTCTCCAACTAAAAGAT 362
ATCACTCGAGCAAGCCTTG 11
13 MMR 2 10 ABCC8ex13F_and_ABCC8ex13R TTCAGTGTGGGCTTTGTGG 390
GGTGGTTTGGAGGTGAGGA 11
14 MMR 2 10 ABCC8ex14F_and_ABCC8ex14R GCTGTGTCGGACTTCTGCCTTT 266
GCTCCCTCTGGGAGTTGGTG 11
15 MMR 2 10 ABCC8ex15F_and_ABCC8ex15R TTTTGGCTTTCATGGAGGAG 251
TGCAGCTTTGTCTTTTTATCTCTATG 11
16 MMR 2 10 ABCC8ex16F_and_ABCC8ex16R GAGGATGTTGATTTCCAGAAGG 346
TGAGGAGGATGGTTAAAAGGAG 11
17 MMR 2 10 ABCC8ex17F_and_ABCC8ex17R ACAGAGGCCATTTGGAAAC 298
TCTGAAAATATGTAGGCTGCAC 11
18 MMR 2 10 ABCC8ex18F_and_ABCC8ex18R TCTCTATGCAGCATTTGTGG 366
AATGGATGCACAGAAACAGC 11
19 MMR 2 10 ABCC8ex19F_and_ABCC8ex19R AGACCCAGACCTCTCAAACC 439
GGTGCACCATATGGAGAGG 11
20 MMR 2 10 ABCC8ex20F_and_ABCC8ex20R GAGGCCTATTAAAGCCATTGC 350
CATGTTTGACCTTACTGCAGGC 11
21 MMR 2 10 ABCC8ex21F_and_ABCC8ex21R AGGTGAGAAGCAGGCAAAGA 277
GGTGGAGGTGGGCAGTTAG 11
22 MMR 2 10 ABCC8ex22F_and_ABCC8ex22R TCCAAAGCCACACAGCTAAC 404
CCAGTGCTGGTCTCTTATGC 11
23 MMR 2 10 ABCC8ex23F_and_ABCC8ex23R AGGAGTATGTTGGTTGGGGTAG 399
GGGCACTAAGGACAGGAAGA 11
24 MMR 2 10 ABCC8ex24F_and_ABCC8ex24R TGAATGTGTGTCTGTCTGCC 370
CAGAGGGAAGCCATTTAATC 11
25 MMR 2 10 ABCC8ex25F_and_ABCC8ex25R CCCGTTGTCCCCTCAGTAAG 393
CTCAGCCCTTCCCCCATC 11
Diabetes and Hyperinsulinism Genetic Testing
(continued)
Table 1
134
(continued)
Primer DNA
PCR concentration concentration Size
Exon method (pmol/μl) (ng/μl) Forward and reverse primer sequences (M13 tailed) chromosome number (bp)
27 MMR 2 10 ABCC8ex27F_and_ABCC8ex27R TGAATGACTCCAGAGACACTTA 246
AGACAGGAGAAGCCCCCAG 11
Jayne A. L. Houghton
(continued)
135
Table 1
136
(continued)
CGGAAGGGGAAACTGAGG 8
2A AG60 5 10 GATA4ex2AF_and_GATA4ex2AR AAATTGGGATTTTCCGGAGT 477
GTGTGGGCACGTAGACTGG 8
137
(continued)
Table 1
(continued)
138
GGTAGGGGCTGGAGTAGGAG 8
2D AG60 5 10 GATA4ex2DF_and_GATA4ex2DR CTGCGGCCTACAGCAGTG 299
CCTCGACAGGGCTCAAGAC 8
3 AG60 5 10 GATA4ex3F_and_GATA4ex3R AGCCCGAGGTGGTCTTCT 435
TATTATGGGGCTCTCACCCA 8
4 MMR 2 10 GATA4ex4F_and_GATA4ex4R CGCAGGTGACAGGAGAGTTAG 373
AAGGAAGAAGACAAGGGAGGA 8
5 MMR 2 10 GATA4ex5F_and_GATA4ex5R TTGCTTAGGTGTTGCCTTCTC 353
TTTTTGCTGGGCTCTTCATC 8
6 MMR 2 10 GATA4ex6F_and_GATA4ex6R AGCCATCCCTGTGAGAACTG 387
GCTGGCCTCTGGGACTCT 8
7 MMR 2 10 GATA4ex7F_and_GATA4ex7R AGAAGTGCTCCTTGGTCCCT 449
TTCCCAGTTGTTGTTCTGGA 8
PCR Primer concentration DNA concentration Forward and reverse primer sequences (M13 tailed) chromosome Size
Exon method (pmol/μl) (ng/μl) number (bp)
1 MMR 2 10 GCKex1F_and_GCKex1R TCCACTTCAGAAGCCTACTG 195
TCAGATTCTGAGGCTCAAAC 7
2 MMR 2 10 GCKex2F_and_GCKex2R GGGGTCAGAAGACAGAAGGA 511
AGAGGAGCCAAGGGTGAGA 7
3 MMR 2 10 GCKex3F_and_GCKex3R ATATCCGGGCTCAGTCACC 332
CACCTCCCGTCAGGACTAGC 7
4 MMR 2 10 GCKex4F_and_GCKex4R TAGCTTGGCTTGAGGCCGTG 308
TGAAGGCAGAGTTCCTCTGG 7
5 MMR 2 10 GCKex5F_and_GCKex5R tcactcacggcagaagca tggaagccaaggagaaagg 7 634
6 MMR 2 10 GCKex6F_and_GCKex6R TGCGAGACGCTATCAAACG 358
Diabetes and Hyperinsulinism Genetic Testing
GGCTCTGCTCTGACATCACC 7
7 MMR 2 10 GCKex7F_and_GCKex7R ccattgttccagacaaagca caagcccattatctgcaatg 7 436
139
(continued)
Table 1
(continued) 140
PCR Primer concentration DNA concentration Forward and reverse primer sequences (M13 tailed) chromosome Size
Exon method (pmol/μl) (ng/μl) number (bp)
8 MMR 2 10 GCKex8F_and_GCKex8R CATTTCTAAAGCTCTGGCTCATT 634
GTCCTGTCCCAGCCTCCT 7
9 MMR 2 10 GCKex9F_and_GCKex9R ctttggctgggtgaggtg agggggacgagaagaggac 7 574
10 MMR 2 10 GCKex10F_and_GCKex10R CCTCTTCTCGTCCCCCTTG 360
Jayne A. L. Houghton
ATGGAGCCTGGGTGCTGT 7
PROM MMR 2 (80% MF) 10 GCKpromF_and_GCKpromR gctcggcatttcctgct agtcaggctgccaagaggt 7 512
(continued)
Table 1
(continued)
142
DNA
PCR Primer concentration concentration Size
Exon method (pmol/μl) (ng/μl) Forward and reverse primer sequences (M13 tailed) chromosome number (bp)
1 MMR 2 10 HADHex1F_and_HADHex1R CGTGTATACCCGCTCAACG 502
GTGAAAACTCCCTGGTGTCG 4
2 MMR 2 10 HADHex2F_and_HADHex2R TCGGTATTTTGAATGGAATGG 538
Jayne A. L. Houghton
AACCTCAAATCCCACCCAAC 4
3 MMR 2 10 HADHex3F_and_HADHex3R GAACAGATAGGGTAGGCCAAT 436
GCATGGAACAAAACTGCAC 4
4 MMR 2 10 HADHex4F_and_HADHex4R CCCAATGGGTCAGGACAAC 425
TATGAGGCTCAGGGACAACC 4
5 MMR 2 10 HADHex5F_and_HADHex5R TTGTTTCTGGCTTGAGATTCC 439
AACTAGAACAGGAGGCACGG 4
6 MMR 2 10 HADHex6F_and_HADHex6R TGTACAGCTTGATAAATGGGG 363
TCACAAAGCTAGAAAACAGACACTG 4
7 MMR 2 10 HADHex7F_and_HADHex7R CCAAGCCAGAAAGTCTCAGTC 413
GTGAGGCAGGATTTTGGAAG 4
8 MMR 2 10 HADHex8F_and_HADHex8R CACCAGCACAGCCTTCCT 490
TGCTGTAACTGGTGTAATAATCACTTC 4
IVS5 MMR 2 10 HADH_Cryptic_Donor_FandR ttttaggtacagcttggtaaacaca tcatttcagcaactgggatt 4 297
GAGGGTTCCTGGGTCTGTG 17
4 MMR 2 10 HNF1Bex4F_and_HNF1Bex4R actcccaaccaagactgctg 415
GATCCGTGGCAAGAACCA 17
143
(continued)
Table 1
(continued)
144
AATGCCACCACCCACTATTC 19
3 MMR 2 10 INSRex3F_and_INSRex3R TGTTTGGTTGGCTTTCACTG 541
AGTTTTAACAAGCGGCATCG 19
145
(continued)
Table 1
(continued)
146
GGTCCCTCATGCCAAAAAG 19
7 MMR 2 10 INSRex7F_and_INSRex7R TTCCCCCATTCATACTCAGC 367
TGGAGCACAAACGTAGCAAG 19
8 MMR 2 10 INSRex8F_and_INSRex8R TCCTTTCTCTTTGGCTGTTCC 467
AAAGCAAGAGGTCTGATTCACATAC 19
9 MMR 2 10 INSRex9F_and_INSRex9R CATCCTCCCACCAGCTTTC 322
GATAGCTGCTTCCCTAGAGGTG 19
10 MMR 2 10 INSRex10F_and_INSRex10R ATGTGTGTTCAGCCGCAGAG 389
AAGGGCTCCATTCAGACTCC 19
11 MMR 2 10 INSRex11F_and_INSRex11R GATTTCTTCCCCTTCCCTTG 377
AAGCATCTGCTCTCCAGCAC 19
12 MMR 2 10 INSRex12F_and_INSRex12R TAATCCATGTTCCCCATTGC 496
GCACTGCTTAGAGGGTGGAG 19
13 MMR 2 10 INSRex13F_and_INSRex13R TGTGGGATGAGTTTTAGATGTCC 371
GAGGAAGGAGGTACCAAGTGC 19
14 MMR 2 10 INSRex14F_and_INSRex14R AAATTCTGCAATTCTCAAAGTCG 385
ATGTCCCACCATGCTCAGTG 19
15 MMR 2 10 INSRex15F_and_INSRex15R GTGGAAGAGCAGCAGAGAGG 285
CAACTGTTCCCAGCACACC 19
16 MMR 2 10 INSRex16F_and_INSRex16R AGGATTATGGGGATTCTGCTG 249
GAAGGCAAAGGAAGCTGATG 19
17 MMR 2 10 INSRex17F_and_INSRex17R GCATGGGTCCTGGATCAC 467
CTGTGTCCTCTGTCGCTCTG 19
18&19 MMR 2 10 INSRex18&19F_and_INSRex18&19R ATAGACACCAGGGAGGGAGG 546
AAGACTTAACGGCTCATTATAGACAAC 19
20 MMR 2 10 INSRex20F_and_INSRex20R TTACTGGATTTGGCCACCAC 399
CTGCCTTCCTTTCCTTGATG 19
21 MMR 2 10 INSRex21F_and_INSRex21R TTTATGAACTGCAGCATGAATTG 593
GGGCAATACCCTTTCAACG 19
22 MMR 2 10 INSRex22F_and_INSRex22R ACCATCTCTCAGCACCTCTAGC 587
AGGAAAGCGAAAATGGGAAC 19
GTTTCCACCACGCCTTCC 11
1D MMR 2 10 KCNJ11ex1DF_and_KCNJ11ex1DR CTACCATGTCATTGATGC 491
CCACATGGTCCGTGTGTA 11
147
(continued)
Table 1
148
(continued)
GTTAGGTGGGGCCATGGGT 1
3 MMR 2 10 LMNAex3F_and_LMNAex3R atgtgtgaaggggtgcacag ccagccctactccatgagc 1 419
4 MMR 2 10 LMNAex4F_and_LMNAex4R TAAAGTGGGGCTGGTAGTGG 380
TAAGGGTAGGGCTGCCAAG 1
5 MMR 2 10 LMNAex5F_and_LMNAex5R GGGATCAGGCAGATGGTG 385
ACCCTTCTCTGTGGTTGTGG 1
6 MMR 2 10 LMNAex6F_and_LMNAex6R CTGGGGAAGCTCTGATTGC 420
CAGAGGACACTGCCAGCAC 1
7 MMR 2 10 LMNAex7F_and_LMNAex7F gtccctccttccccatactt tctcacagccaaagagtcca 1 813
8 MMR 2 10 LMNAex8F_and_LMNAex8R gttgtcaggaagatgaaagataagg 395
cctccccagagtcccaag 1
9 MMR 2 10 LMNAex9F_and_LMNAex9R ATGGAGATGATCCCTTGCTG 420
tctagaaaggggccctgaat 1
10 MMR 2 10 LMNAex10F_and_LMNAex10R CAGGCCACAAGAAAAGTTGC 388
GTTCAAGGTATAGGGAGGAG 1
11 MMR 2 (80% MF) 10 LMNAex11F_and_LMNAex11R GGGCACAGAACCACACCTT 610
CAGACAAGAGGGGCAGGA 1
12 MMR 2 10 LMNAex12F_and_LMNAex12R GGGAGATGCTACCTCCCTTC 240
GGGTTATTTTTCTTTGGCTTCA 1
Gene: NEUROG3 Accession No.: NM_020999.2
PCR Primer concentration DNA concentration Forward and reverse primer sequences (M13 tailed) chromosome Size
Exon method (pmol/μl) (ng/μl) number (bp)
2A MMR 2 10 NEUROG3ex2AF_and_NEUROG3ex2AR ctattcttttgcgccggtag 499
cagtgccgagttgaggttg 10
2B MMR 2 10 NEUROG3ex2BF_and_NEUROG3ex2BR gagttggcactgagcaagc 475
ccctctcccttacccttagc 10
CACCATAAGGACCGAGGC 20
(continued)
149
Table 1
(continued)
150
CCAGCAAATACAAAGGGACAA 6
11 MMR 2 10 RFX6ex11F_and_RFX6ex11R GGCAGATCTAGGCTGTCTGA 300
TTCCTCAATCTGTCCTTCTCA 6
151
(continued)
Table 1
(continued)
152
GCCTGCCTTATTCGTAGAGC 6
14 MMR 2 10 RFX6ex14F_and_RFX6ex14R TGCCATCTGTCCATTTGTTAG 328
GACAGGAACTGTGACTGAGGA 6
15 MMR 2 10 RFX6ex15F_and_RFX6ex15R TCCCCTTTTTACATTTCACACTG 700
GACATGTTTGGAGAAAAACTCTGA 6
16 MMR 2 10 RFX6ex16F_and_RFX6ex16R TGGGTTTATCTACGAGGAATGTG 357
TGAAGCAACAGGCTTCTCTG 6
17A MMR 2 10 RFX6ex17AF_and_RFX6ex17AR TGAAGCAAGCTGGAAAACAA 495
AGAGGTGGAACGCAATGTCT 6
17B MMR 2 10 RFX6ex17BF_and_RFX6ex17BR AGAGGTGGAACGCAATGTCT 495
TGAAGCAAGCTGGAAAACAA 6
18 MMR 2 10 RFX6ex18F_and_RFX6ex18R TTTTCCTTCCTGAAAGTTCTTTGT 487
TGGAAACATGCTTGCTCAGT 6
19 MMR 2 10 RFX6ex19F_and_RFX6ex19R TGAGTGGCATTTGCAGAGAA 246
CAGCCACTGCATATTCATTTT 6
PCR Primer concentration DNA concentration Forward and reverse primer sequences (M13 tailed) chromosome Size
Exon method (pmol/μl) (ng/μl) number (bp)
1 MMR 2 10 SLC19A2ex1F_and_SLC19A2ex1R gcgtccgctgtgattggtt 606
gagaaaagcgctcgagcc 1
2A MMR 2 10 SLC19A2ex2AF_and_SLC19A2ex2AR agatctttgaggtatttgtagg 534
acacaggtaagagagatgaca 1
2B MMR 2 10 SLC19A2ex2BF_and_SLC19A2ex2BR acagccactgaaattgccta 529
agatctaccaagagggagttt 1
3 MMR 2 10 SLC19A2ex3F_and_SLC19A2ex3R ttcgccagaggggataaatg 596
cctgctccacttgagtactt 1
4 MMR 2 10 SLC19A2ex4F_and_SLC19A2ex4R ccctccataatcttgagctatt 476
ttcctcccatttgcctcattt 1
5 MMR 2 10 SLC19A2ex5F_and_SLC19A2ex5R gttggaaaggcaattgacagt 423
actttacatctgttccctattg 1
6 MMR 2 10 SLC19A2ex6F_and_SLC19A2ex6R ctcaggcagtcaggctttatt 470
gctgctgtgaagtcaagaaat 1
PCR Primer concentration DNA concentration Forward and reverse primer sequences (M13 tailed) chromosome Size
Exon method (pmol/μl) (ng/μl) number (bp)
1 MMR 2 10 SLC2A2_Ex1F_and_SLC2A2_Ex1R ctctccttgctcctcctcct 297
aaacagttcttcaattggcaaag 3
2 MMR 2 10 SLC2A2_Ex2F_and_SLC2A2_EX2R cgatattgaggggaaagaacc 384
gggagtcaaggccaagttatc 3
3 MMR 2 10 SLC2A2_Ex3F_and_SLC2A2_Ex3R tggccaactgtaaacaaatct 518
aaagctattccacaagaagaaaga 3
4 MMR 2 10 SLC2A2_Ex4F_and_SLC2A2_Ex4R ccccaactagcttttacagca 398
tgctattgcctttccctctg 3
5 MMR 2 10 SLC2A2_Ex5F_and_SLC2A2_Ex5R aattcagggaggggctttc 339
gagggacgagatggatgaag 3
Diabetes and Hyperinsulinism Genetic Testing
(continued)
153
Table 1
(continued)
154
PCR Primer concentration DNA concentration Forward and reverse primer sequences (M13 tailed) chromosome Size
Exon method (pmol/μl) (ng/μl) number (bp)
7 MMR 2 10 SLC2A2_Ex7F_and_SLC2A2_Ex7R gaatttccaacacccatgct 432
gacccatgatgccaatacct 3
8 MMR 2 10 SLC2A2_EX8F_and_SLC2A2_Ex8R gcatgccgctatagaaacaa 400
Jayne A. L. Houghton
tgcagtggttggcttattca 3
9 MMR 2 10 SLC2A2_EX9F_and_SLC2A2_EX9R tccccaccttgatctcactc 397
ttatgctgttgctctggcttt 3
10 MMR 2 10 SLC2A2_EX10F_and_SLC2A2_EX10R aaaagttgtatgccgcctgt 436
agcactccagcaaagaggaa 3
11 MMR 2 10 SLC2A2_EX11F_and_SLC2A2_EX11R gcattcagcaattggacctg 482
tgacatttctgatgagagcaca 3
(continued)
Table 1
(continued)
156
AACAGGGTGTTCAGGGTCTC 17
22 MMR 2 10 STAT3_22F_and_STAT3_22R TAAATGAGGGCAGACAACCC 397
TCAAACTCTGGTCTCCAACAG 17
23 MMR 2 10 STAT3_23F_and_STAT3_23R AGCCCCTGGGCTATGTTTAG 428
TCTCTTTTGGAAAGCAAAGCTC 17
24 MMR 2 10 STAT3_24F_and_STAT3_24R TCCAGGGAGGAGGGTAAATC 425
AGCAGATCACCCACATTCAC 17
Primer DNA
PCR concentration concentration Size
Exon method (pmol/μl) (ng/μl) Forward and reverse primer sequences (M13 tailed) chromosome number (bp)
1 MMR 2 10 ZMPSTE24ex1F_and_ZMPSTE24ex1R TCGGAAAGAACGGATATTGC 381
GACCACAAAGACGAGACTGG 1
2 MMR 2 10 ZMPSTE24ex2F_and_ZMPSTE24ex2R ATCAAGTGCAATATTTGTGTAGG 420
TCTGGGACTTGTAAGTGTGG 1
3 MMR 2 10 ZMPSTE24ex3F_and_ZMPSTE24ex3R GATGGGGTCTCAACTCACCT 373
CCTGCCAAGCTAAAAGAAATACC 1
4 MMR 2 10 ZMPSTE24ex4F_and_ZMPSTE24ex4R GATGATATTTCAGACCTTATGTTATCC 432
ATGATCTTTACCATATCTGTAGTTCC 1
5 MMR 2 10 ZMPSTE24ex5F_and_ZMPSTE24ex5R TGCAAGACATTTACCCATTGT 575
TGGTCAGTCAATACTCCTGTGTC 1
6 MMR 2 10 ZMPSTE24ex6F_and_ZMPSTE24ex6R GGAATACCAGAGCAAGTAAGTTCC 471
TAAAAAGCCAAAAGGTAAACTCC 1
7 MMR 2 10 ZMPSTE24ex7F_and_ZMPSTE24ex7R actgctggtggtgaattatgtt aaggagccatggttttcatct 475
1
8 MMR 2 10 ZMPSTE24ex8F_and_ZMPSTE24ex8R TGTCTCATGTGGAGCAGTGG 415
CAGGTAGCCTGGCTGTGG 1
9 MMR 2 10 ZMPSTE24ex9F_and_ZMPSTE24ex9R ACTTTATGGATGCTACTGATCC 406
TTGGGCTTGAATTTTTCTGC 1
10 MMR 2 10 ZMPSTE24ex10F_and_ZMPSTE24ex10R AGGCTCAATTTATAATTTCCTCACC 504
CATCAAGAGCTGGAACATGC 1
Diabetes and Hyperinsulinism Genetic Testing
157
158 Jayne A. L. Houghton
Table 2
NEXTflex primer sequences
Table 3
Nextflex adapter sequences
2.8 Methods: Sanger The PCR and sequencing methods detail primers and PCR condi-
Sequencing tions for Sanger sequencing of all coding regions and intron–exon
boundaries for single gene analysis to detect missense, nonsense,
frameshift, and splicing mutations. For a list of genes, primer pairs,
amplicon sizes, and optimized PCR conditions for each amplicon
see Table 1.
3 Methods
3.1 PCR A PCR is set up for each of the primer pairs using one of the
Amplification following conditions (Tables 4 and 5).
160 Jayne A. L. Houghton
Table 4
MegaMix Royal (MMR) 10 μl PCR reaction per sample
Table 5
AmpliTaq Gold (AG) with dNTP mix 2 and mix 3, 25 μl PCR reaction per sample
3.2 PCR Cycling The following (Tables 5–8) outlines the thermocycler programs for
Conditions using MegaMix Royal and AmpliTaq Gold (see Note 2).
3.3 PCR Product The PCR product is purified prior to sequencing to remove excess
Purification Prior primers, primer dimers, unincorporated dNTPs, salts, and other
to Sanger Sequencing contaminants. There are several commercially available methods for
purifying PCR products. We use the Agencourt AMPure XP PCR
purification system, a solid-phase paramagnetic bead technology
that is highly amenable to automation and high sample numbers.
We use the Biomek NXP (Beckman Coulter) to perform our liquid
handling, though other liquid handling workstations are available.
To summarize, the PCR products are bound to paramagnetic beads
and separated before being washed to remove any contaminants.
The purified PCR products are then eluted from the magnetic
beads ready for the sequencing reaction.
1. Transfer 5 μl of the PCR products to a clean 384-well plate.
2. Briefly centrifuge the plate so that the PCR product is at the
bottom of the well.
Diabetes and Hyperinsulinism Genetic Testing 161
Table 6
MegaMix Royal (MMR) PCR cycling conditions
Table 7
AmpliTaq Gold (AG60) PCR cycling conditions
Table 8
AmpliTaq Gold (AG58) PCR cycling conditions
5. Place the plate onto the magnetic stand for 3–5 min. This step
separates the beads containing the bound PCR products from
the solution.
6. The beads form a spot on the side of the well. Remove the
cleared solution from the plate and discard. This step needs to
be performed while the plate is situated on the magnetic stand
so that the bead pellet is not disturbed.
7. Dispense 30 μl of 85% ethanol to each well of the plate while
placed on the magnetic stand. Incubate for 30 s at room
temperature. Remove the ethanol and discard (see Note 3).
8. Repeat step 6 (total of two washes).
9. Allow the plate to air dry completely.
10. Add 30 μl of elution buffer (dH2O).
11. Transfer the purified PCR products into a new plate.
3.4 Sanger Prepare a sequencing master mix (n + 5) if there are more than
Sequencing Reaction 10 samples. If there are less than 10 samples, make up a sequencing
mix of n + 1. We perform unidirectional sequencing (forward
direction) and all our primers are M13 tailed so that a universal
sequencing master mix can be used. Note: for samples shown to
contain a frameshift mutation or if mosaicism is suspected, the
reverse sequencing is performed using a revere M13 (M13R)
primer so that bidirectional sequence can be analyzed [5].
1. Add the sequencing reaction mix as in Table 9 (4.52 μl) to 2 μl
of each PCR sample and run on the thermocycler using the
program detailed in Table 10.
Table 9
Sequencing reaction
Table 10
Sequencing PCR cycling conditions
3.6 Targeted Next- Targeted next-generation sequencing (also called massive parallel
Generation sequencing, MPS) is a powerful methodology for the analysis of
Sequencing Using multiple genes in a single assay. It enables the surveillance of a
the Agilent SureSelect subset of the genome which contains genes relevant to the pheno-
System type in question. There are many different platforms, chemistries,
and enrichment strategies available. We designed a custom Agilent
SureSelect exon-capture assay with baits for all the diagnostic
monogenic diabetes and hyperinsulinism genes to identify patho-
genic mutations located within 50 bp upstream and 10 bp down-
stream of each exon [4]. Sequencing is performed on an Illumina
HiSeq 2500. This methodology will identify the same types of
mutations detected by Sanger sequencing as well as large deletions
or duplications previously identified by a separate dosage assay.
The majority of the liquid handling in this protocol is amenable
to automation. We use the Bravo liquid handling platform which is
equipped with a ThermoCube thermoelectric and INHECO tem-
perature control system, used to incubate components at defined
temperatures during the procedure, but other systems are available.
Samples for the capture library are individually tagged with unique
barcodes and then combined into 12-plex pools prior to hybridiza-
tion. We process 48 samples at a time.
One library is prepared for all samples, which are individually
tagged with unique barcodes. The process is described below and is
modified from Agilent Technologies SureSelect Automated Library
Prep and Capture System protocol.
3.7 DNA DNA is quantified using the Qubit® dsDNA BR assay kit which is
Quantification designed for use with the Qubit® 2.0 fluorometer. The assay mea-
sures the concentration of double stranded DNA (dsDNA) in the
sample. Used according to the manufacturer’s specific protocols.
4. Make sure all the tube hinges face outward and the tubes are
“flush” and the adaptor is tightened properly. Refer to the
Bioruptor® instrument user’s guide for more details.
5. Place the adaptor head in the Bioruptor® and fragment for
54 min.
6. Briefly centrifuge the samples.
7. Transfer each of the samples (80 μl) to a 96-well skirted Eppen-
dorf plate.
8. Repeat steps 2–8 for all of the samples.
9. Centrifuge the plate briefly to collect the sample at the bottom
of the well and seal the plate (see Note 12).
3.10 Purification This system can be used in manual and automated formats. To
of Sheared DNA Using summarize, samples are cleaned up with 120 μl AMPure beads
the Agencourt AMPure (1.5 volumes) and eluted in 40 μl dH2O.
XP System 1. Allow the AMPure XP beads to equilibrate to room tempera-
ture before use and shake well so that the reagent appears
homogenous and consistent in color.
2. Add 80 μl of the sheared DNA to each well and mix by
pipetting.
3. Add 120 μl of the AMPure XP beads to each well of a Nunc
DeepWell plate.
4. Incubate for 5 min at room temperature.
5. Place the plate onto the magnetic stand to separate the beads
from the solution for 3 min.
6. Remove the cleared liquid from the solution while the plate is
situated on the magnetic stand, being careful not to disturb the
separated beads.
7. Add 70% ethanol to each well, enough to cover the beads,
while the plate is situated on the magnetic stand and incubate
for 30 s at room temperature.
8. Repeat for a total of two washes.
9. Remove the plate from the magnetic stand and add 40 μl of
dH2O.
10. Resuspend the beads by pipetting up and down.
11. Incubate for 1 min.
12. Place the plate onto the magnetic stand and transfer the eluate
to a fresh plate.
3.11 Sample Quality A quality control check on D1000 tapes is performed to ensure that
Check Using Agilent each of the samples are fragmented sufficiently. Set up the Agilent
2200 TapeStation®™ 2200 TapeStation®™ according to the manufacturer’s instructions.
166 Jayne A. L. Houghton
3.12 Pre End-Repair 1. Dilute 350 ng of the fragmented DNA in dH2O to 8.75 ng/μl
Dilutions to a final volume of 40 μl in a 96-well skirted Eppendorf plate.
3.13 End Repair Forty microliters diluted fragmented DNA is converted by the
NEBNext End Repair Module to blunt ended DNA having 50
phosphates and 30 hydroxyl, to a final volume of 50 μl.
1. Make an end-repair master mix on ice (see Table 11) and pipette
into a plate. Add 40 μl of DNA from the previous step.
2. Incubate the samples for 30 min at 20 C (see Note 12).
Table 11
End-repair master mix
Volume (n ¼ 1) (μl)
10 End-repair buffer 5.0
End-repair enzyme mix 2.5
dH2O 2.5
Total volume per sample 10
Diabetes and Hyperinsulinism Genetic Testing 167
3.14 AMPure XP Samples are cleaned up with 90 μl AMPure XP beads (1.8 volumes)
Clean-Up and eluted in 22 μl dH2O
1. Allow the AMPure XP beads to equilibrate to room tempera-
ture before use and shake well so that the reagent appears
homogenous and consistent in color.
2. Add 90 μl of the AMPure XP beads to each well of a Nunc
DeepWell plate.
3. Add the 50 μl sample to each well and mix by pipetting.
4. Incubate for 5 min at room temperature.
5. Place the plate onto the magnetic stand for 3 min to separate
the beads from the solution.
6. Remove the cleared liquid from the solution while the plate is
situated on the magnetic stand, being careful not to disturb the
separated beads.
7. Add 70% ethanol (enough to cover the beads) to each well
while the plate is situated on the magnetic stand and incubate
for 30 s at room temperature.
8. Repeat for a total of two washes.
9. Remove the plate from the magnetic stand and add 22 μl of
dH2O. Resuspend the beads by pipetting.
10. Incubate for 1 min.
11. Place the plate onto the magnetic stand and transfer the elute
to a fresh plate (see Note 12).
Table 12
dA tailing master mix
Volume (n ¼ 1) (μl)
10 Rx buffer 3
Klenow fragment (without exonuclease activity) 1.5
dH2O 4.5
End-repaired DNA 21
Total volume 25
168 Jayne A. L. Houghton
3.16 AMPure Samples are cleaned with 54 μl AMPure beads (1.8 volumes) and
Clean-Up eluted in 12 μl dH2O
1. Allow the AMPure XP beads to equilibrate to room tempera-
ture before use and shake well so that the reagent appears
homogenous and consistent in color.
2. Add 54 μl of the AMPure XP beads to each well of a Nunc
DeepWell plate.
3. Add 50 μl sample to each well and mix by pipetting and
incubate for 5 min at room temperature.
4. Place the plate onto the magnetic stand to separate the beads
from the solution for 3 min.
5. Remove the cleared liquid from the solution while the plate is
situated on the magnetic stand, being careful not to disturb the
separated beads.
6. Add 70% ethanol (enough to cover the beads) to resuspend and
mix by pipetting. Incubate for 1 min at room temperature.
7. Repeat for a total of two washes.
8. Remove the plate from the magnetic stand and add 12 μl of
dH2O to elute the samples. Incubate for 2 min.
9. Place the plate onto the magnetic stand for 2 min and transfer
the eluate to a fresh plate (see Note 12).
Table 13
Ligation master mix
Volume (n ¼ 1) (μl)
Fast-Link DNA ligase 1
10 mM ATP 1
10 buffer 2
Water 0.5
Total volume 4.5
Diabetes and Hyperinsulinism Genetic Testing 169
3.18 AMPure Clean samples with 20 μl AMPure beads (1.0 volumes) and eluted
Clean-Up in 42 μl dH2O (see Note 13).
1. Allow the AMPure XP beads to equilibrate to room tempera-
ture before use and shake well so that the reagent appears
homogenous and consistent in color.
2. Add 20 μl of the AMPure XP beads to each well of a Nunc
DeepWell plate.
3. Add the 20 μl sample to each well and mix by pipetting and
incubate for 5 min at room temperature.
4. Place the plate onto the magnetic stand for 3 min to separate
the beads from the solution.
5. Remove the cleared liquid from the solution while the plate is
situated on the magnetic stand, being careful not to disturb the
separated beads.
6. Add 70% ethanol (enough to cover the beads) and resuspend
and mix by pipetting to each well while the plate is situated on
the magnetic stand and incubate for 1 min at room temperature.
7. Repeat for a total of two washes.
8. Remove the plate from the magnetic stand and add 42 μl of
dH2O to elute the samples.
9. Incubate for 2 min.
10. Place the plate onto the magnetic stand and allow to separate
for 2 min.
11. Transfer the eluate to a fresh plate (see Note 12).
3.19 Pre Hybe PCR Twenty microliter adapter-ligated DNA is amplified in a final PCR
volume of 50 μl.
1. Prepare the PCR master mix (Table 14) and add 30 μl of the
PCR master mix to each adapter-ligated DNA sample.
2. Run the program in Table 15 on the thermocycler (see Note 12).
Table 14
PCR reaction mix
Volume (n ¼ 1) (μl)
5 Herculase II buffer 10
NEXTflex primer mix (12.5 μM each primer) 2.0
dNTP mix (Herculase kit only) 0.5
Herculase II polymerase 1.0
PCR-grade water 16.5
Total volume 30
170 Jayne A. L. Houghton
Table 15
PCR cycling conditions
3.20 AMPure Clean- Samples are cleaned up with 60 μl AMPure beads (1.2 volumes) and
Up eluted in 30 μl dH2O
1. Allow the AMPure XP beads to equilibrate to room tempera-
ture before use and shake well so that the reagent appears
homogenous and consistent in color.
2. Add 60 μl of the AMPure XP beads to each well of a Nunc
DeepWell plate.
3. Add 50 μl sample to each well and mix by pipetting and
incubate for 5 min at room temperature.
4. Place the plate onto the magnetic stand to separate the beads
from the solution for 3 min.
5. Remove the cleared liquid from the solution while the plate is
situated on the magnetic stand, being careful not to disturb the
separated beads.
6. Add 70% ethanol (enough to cover the beads) and resuspend
and mix by pipetting to each well while the plate is situated on
the magnetic stand and incubate for 1 min at room
temperature.
7. Repeat for a total of two washes.
8. Remove the plate from the magnetic stand and add 30 μl of
dH2O to elute the samples.
9. Incubate for 2 min.
10. Place the plate onto the magnetic stand and allow to separate
for 2 min.
11. Transfer the eluate to a fresh plate (see Note 12).
3.21 TapeStation®™ A quality control check on D1000 tapes are performed to ensure
Quality and Quantity adapters have been ligated and there is sufficient sample to continue
Check (see the Agilent 220 TapeStation®™ user manual for the manufac-
turer’s instructions).
Diabetes and Hyperinsulinism Genetic Testing 171
1. To analyze results, set the region from 115 to 700 bp (this will
exclude any adapter dimer peaks [~135 bp] from the analysis).
The average sample peak should be less than 350–400 bp (due
to the adapter ligation, products will be longer by about
120 bp) (see Note 14).
3.22 Hybe Pools Four 12-plex pools each containing 1200 ng total DNA (i.e.,
100 ng per sample) are prepared for hybe reactions.
1. Label 4 Anachem tubes (e.g., A–D).
2. Add 12 samples per tube (100 ng per sample) to form a pool of
1200 ng.
3. Mix well on a vortex mixer and spin in a microcentrifuge for
1 min.
4. Using a vacuum concentrator dry the samples until no liquid is
left in the tubes (~60 min). A little white pellet may be visible
(see Note 12). We use a SpeedVac, although different instru-
ments are available.
5. If a sample is not completely dehydrated after 60 min, continue
dehydration for an additional 5–10 min (see Note 15).
6. Reconstitute the dehydrated samples in 8.2 μl dH2O to bring
the final concentration up to 147 ng/μl (see Note 16).
3.23 Hybridization For each pool, prepare one hybridization and capture 3.4 μl pooled
DNA (500 ng) hybridized with the Capture library in a final
volume of 28 μl. During the following steps, some of the transfers
are carried out while tubes remain on the PCR thermocycler.
During these steps care should be taken to remove and replace
tube lids without disturbing samples. After transfers, lids should
be replaced as quickly as possible to minimize loss by evaporation.
From this point onward, one sample refers to one pooled
library.
1. Label three rows of strip tubes as follows:
(a) Row 1: Pools A–D prepped library.
(b) Row 2: Pools A–D hybridization buffer.
(c) Row 3: Pools A–D SureSelect capture library.
2. Make up the SureSelect block mix (n + 1) as in Table 16 in strip
tubes for each pool (5.6 μl) and add the DNA.
3. Place the tubes on a PCR thermocycler and incubate at 95 C
for 5 min and then hold at 65 C (see Table 17).
4. In separate strip tubes, make up the hybridization buffer (one
for each pooled sample) (see Table 18), and once the PCR block
has reached 65 C from the previous step, place the tubes
containing the hybridization buffer on the PCR thermocycler
set at 65 C for 5 min.
172 Jayne A. L. Houghton
Table 16
SureSelect block mix
Table 17
PCR thermocycler program
Table 18
Hybridization buffer
Table 19
RNase block dilution
5. Dilute the RNase blocker (Tables 19 and 20) and put on ice.
6. The custom library is stored at 80 C, remove from the
freezer, and defrost quickly and gently (i.e., gloved hand).
7. On ice, add 5 μl of the diluted RNase blocker to the labeled
tubes.
Diabetes and Hyperinsulinism Genetic Testing 173
Table 20
Capture library (for capture size <3.0 Mb)
Volume (n ¼ 1) (μl)
SureSelect library 2
RNase block dilution (see Table 19) 5
Total 7 μl
8. Add 2 μl of the capture library to the same tubes and put on the
thermocycler set at 65 C for 2 min.
9. Add 13 μl of hybridization buffer (on the 65 C block) into the
capture library RNase tubes.
10. Then add all of the SureSelect block mix and DNA to the
capture library tubes and pipette to mix.
11. Close the lids and incubate for 16 h at 65 C on a thermocycler.
3.25 Washing 1. Place the tubes into the magnetic stand for 2 min and remove
the Capture Library the supernatant.
2. Resuspend the beads in 500 μl of SureSelect wash buffer 2 pre-
warmed to 65 C.
174 Jayne A. L. Houghton
3.27 AMPure Samples are cleaned up with 90 μl AMPure beads and eluted in
Clean-Up 30 μl dH2O
1. Allow the AMPure XP beads to equilibrate to room tempera-
ture before use and shake well so that the reagent appears
homogenous and consistent in color.
2. Label four tubes.
3. Transfer the entire PCR product of the labeled tubes.
Table 21
Posthybridization PCR master mix
Volume (n ¼ 1) (μl)
5 Herculase II buffer 10
NEXTflex primer mix (12.5 μM each primer) 2
dNTP mix (Herculase kit only) 0.5
Herculase II polymerase 1.0
PCR-grade water 21.5
Total volume 35
Diabetes and Hyperinsulinism Genetic Testing 175
Table 22
PCR thermocycler conditions
3.30 TapeStation®™ A final quality check is run on HSD1K tape to ensure the library
Quality Check pool has a concentration of 10 nM.
1. Dilute the sample 1:3 using 2 μl sample.
2. Run the diluted sample in triplicate on a HSD1K tape on the
TapeStation®™ according to the manufacturer’s instructions.
The samples are now ready to be sequenced. We use the Illu-
mina HiSeq2500 according to standard procedures (see Note 12).
4 Notes
14. If the samples are too strong and the presence of artifacts
>900 bp long in the TapeStation®™ trace can be seen, repeat
the PCR with fewer cycles. If too weak, repeat with more
cycles.
15. This is not DNA, but precipitated salt from the input sample.
16. The samples should be dried for the minimum time possible,
excessive drying may result in poor resuspension of DNA.
17. The final volume of the pooled library can be adjusted to the
desired concentration. Exact library pool dilutions and proces-
sing can vary based on the cell capacity and analysis pipeline
being used. Refer to appropriate Illumina guide for
instructions.
References
1. Pearson ER, Flechtner I, Njølstad PR, Malecki 3. Flanagan SE, Kapoor RR, Hussain K (2011)
MT, Flanagan SE, Larkin B, Ashcroft FM, Genetic of congenital hyperinsulinemic hypogly-
Klimes I, Codner E, Iotova V, Slingerland AS, cemia. Semin Pediatr Surg 20(1):13–17
Shield J, Robert JJ, Holst JJ, Clark PM, Ellard S, 4. Ellard S, Lango Allen H, De Franco E, Flanagan
Søvik O, Polak M, Hattersley AT, Neonatal Dia- SE, Hysenaj G, Colclough K, Houghton JA,
betes International Collaborative Group (2006) Shepherd M, Hattersley AT, Weedon MN, Cas-
Switching from insulin to oral sulfonylureas in well R (2013) Improved genetic testing for
patients with diabetes due to Kir6.2 mutations. monogenic diabetes using targeted next-
N Engl J Med 355(5):467–477 generation sequencing. Diabetologia 56
2. Shields BM, Hicks S, Shepherd MH, (9):1958–1963
Colclough K, Hattersley AT, Ellard S (2010) 5. Ellard S, Shields B, Tysoe C, Treacy R, Yau S,
Maturity-onset diabetes of the young Mattocks C, Wallace A (2009) Semi-automated
(MODY): how many cases are we missing? Dia- unidirectional sequence analysis for mutation
betologia 53(12):2504–2508 screening in a clinical diagnostic setting. Genet
Test 13(3):381–386
Chapter 9
Abstract
This chapter describes the detailed protocol for the isolation and purification of islets of Langerhans from
rodent pancreas using collagenase digestion. The first step of the process is to separate and isolate the
insulin-producing islets of Langerhans from the rest of the pancreas. The pancreas is excised from the
animal, trimmed of nonpancreatic tissues before being inflated and chopped into small pieces. The
connective tissue is then broken down with a collagenase enzyme solution to selectively digest the bulk
of the exocrine tissue while leaving the endocrine islets intact and separated from their surrounding
non-islet tissue. Once this process is completed, the islets of Langerhans are separated from the remaining
mixture by centrifugation and purified by the means of hand picking. Once isolated, the subsequent islets
can be used for several varied experimental processes, including transplantation, the study of pathophysio-
logical mechanisms in diabetic conditions, and in the screening of novel therapeutic approaches in phar-
macological research.
Key words Islets of Langerhans, Insulin, Isolation, Endocrine pancreas, β-cell, Collagenase
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_9, © Springer Science+Business Media, LLC, part of Springer Nature 2020
179
180 Jacqueline F. O’Dowd and Claire J. Stocker
2 Materials
2.3 Islets Isolation 1. For islets isolated from normal rats (150–180 g) use two ani-
mals per isolation or three normal mice (see Note 2).
2. Just prior to dissection, euthanize the donor rodent by an
appropriate technique.
182 Jacqueline F. O’Dowd and Claire J. Stocker
2.4 Islets Culturing For certain experimental protocols, it may be necessary to culture
the isolated islets. When culturing, it is important to handle the
islets under aseptic conditions and use sterile tubes and flasks.
1. Wash the islets in more than ten volumes of sterile Gey and Gey
solution.
2. Resuspend groups of 200 islets in RPMI-1640 containing
11.1 mM glucose (see Note 7) and transfer to a six-well cell-
culture plate.
3. Incubate the islets in a humidified culture incubator with 5%
CO2; 95% air.
3 Notes
3. If no islets are isolated or isolated islets are not intact, the islets
are over-digested. This can be prevented by decreasing the
collagenase digestion time or collagenase concentration (see
Subheading 2.3 step 10).
4. Normal bore pipettes draw up too much liquid as islets are
picked, so drawn pipettes are preferential. Draw glass pipettes
by heating in a Bunsen flame. Once the glass begins to give,
pull sharply to yield pipettes with a bore of approximately
1 mm.
5. If most of the islets are not discrete and difficult to isolate (see
Subheading 2.3 step 15), the exocrine tissues are under-
digested. In order to overcome this problem, increase the
collagenase digestion time (see Subheading 2.3 step 10)
and/or increase the collagenase concentration (see Subheading
2.3 step 10) and ensure vigorously shaking of the vial to
disrupt the pancreas (see Subheading 2.3 step 11).
6. The yield of islets will vary depending on the age and strain.
Diabetic and older animals will have reduced numbers of viable
islets. Digestion continues as the handpicking selection pro-
ceeds; therefore, speed is important. Islets should be collected
within 15–20 min, and the entire isolation completed within
45 min to 1 h of removal of the organ.
7. The culture medium can be altered depending on the experi-
mental conditions required.
References
1. Hellerstrom C (1984) The life story of the pan- 6. Olack B, Finke E, Scharp DW (1987)
creatic β-cell. Diabetologia 26(6):393–400 Low-temperature culture of human islets by
2. Lifson N, Kramlinger K, Mayrand R, Lender E the distension method and purified with Ficoll
(1980) Blood flow to the rabbit pancreas with or Percoll gradients. Surgery 102(5):869–879
special reference to the islets of Langerhans. 7. Saliba Y, Fares N (2019) Isolation, purification
Gastroenterology 79(3):466–473 and culture of mouse pancreatic islets of Langer-
3. Bonner-Weir S, Orci L (1982) New perspectives hans. Methods Mol Biol 1940:255–265
on the microvasculature of the islet of Langer- 8. Ayenehdeh JM, Niknam B, Hashemi SM,
hans in the rat. Diabetes 31(10):883–889 Rahavi H, Rezaei N, Soleimani M, Tajik N
4. Nolan AL, O’Dowd JF (2009) The measure- (2017) Introducing a new experimental islet
ment of insulin secretion from isolated rodent transplantation model using a biometric hydro-
islets of Langerhans. Methods Mol Biol gel and a simple high yield islet isolation tech-
560:43–51 nique. Iran Biomed J 21(4):218–227
5. Lacy P, Kostianovsky M (1967) Method for the
isolation of intact islets of Langerhans from the
rat pancreas. Diabetes 16(1):35–39
Chapter 10
Abstract
An increasing body of evidence indicates that a local islet immune response is not only limited to type
1 diabetes, but also is associated with islet dysfunction in type 2 diabetes. Recently, the presence of
pancreatic CD68+ macrophages within islet tissues was demonstrated by RT-PCR and immunohistochemi-
cal methods. However, the precise profile and activation status of intraislet leukocytes, which are present in
both murine and human islets, are poorly defined. Here, we describe a detailed flow cytometry protocol
designed to analyze both human and murine islets for intraislet leukocytes and leukocyte subsets. This
approach permits the simultaneous identification of multiple intraislet leukocyte subsets, as well as their
activation statuses. The use of flow cytometry-based approaches will advance the field of islet biology and
help to identify unique changes in the immune cell composition that accompanies pathological islet
inflammation and dysfunction in type 2 diabetes.
Key words Type 2 diabetes, Leukocytes, Human islets, Murine islets, Flow cytometry
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_10, © Springer Science+Business Media, LLC, part of Springer Nature 2020
185
186 Matthew J. Butcher et al.
2 Materials
2.2 Murine Islets: 1. Dissection instruments: Hooked nose forceps, scissors, hemo-
Harvest stat clamps, 30 gauge syringe.
and Preparation 2. Surgical microscope.
for Flow Cytometry
3. 1 Dulbecco’s phosphate-buffered saline (PBS) without cal-
cium or magnesium.
4. 12 75 mm, 5 ml polystyrene round bottom test tubes (FACS
tubes).
5. Collagenase P (Roche).
6. Ficoll PM400.
7. 1 Hank’s balanced salt solution (HBSS).
3 Method
3.1 Human Islets: 1. Upon receiving the islets, culture the islets in a 100 20-mm
Care Post- polystyrene nontreated tissue culture Petri dish in supplemen-
procurement ted CMRL-1066 media overnight (37 C, 5% CO2) to allow
and Preparation the islets to recover from the shipment. 5000–7000 Islet
for Flow Cytometry equivalents (IEQ) of human islets are sufficient for a flow
(See Note 1) cytometry experiment (see Note 2).
2. Prepare a 40-μm nylon mesh cell sieve by wetting it with 4 C
PBS in a 35 10 mm polystyrene nontreated tissue culture
Petri dish, making sure that the entire mesh has been soaked.
Using a pipette, gently collect the islets from culture and
transfer them to the center of a 40-μm cell strainer placed in a
clean 100 20-mm polystyrene nontreated tissue culture Petri
dish. This will remove cellular debris and disintegrated islets,
and only the intact islets will be retained on the cell sieve.
3. Wash the islets retained on the sieve by very gently pipetting
cold PBS over the strainer into the dish. Transfer the strainer
containing intact islets to a new 35 10-mm polystyrene
nontreated tissue culture Petri dish containing 1–2 ml of cold
PBS. Very gently pipette up and down to suspend the islets off
of the sieve and into a 15 ml tube. Repeat by adding 1–2 ml of
cold PBS to the strainer to pick up residual islets. Centrifuge
(800 g, room temperature, 1 min) and aspirate the excess
PBS with a transfer pipette, leaving the islet pellet intact.
4. Resuspend the islets in 0.5 ml PBS by gently pipetting up and
down using low-retention pipette tips to minimize islet loss (see
Note 3).
5. Continue the protocol with Subheading 3.2.
3.2 Murine Islets: 1. After euthanizing the mouse, open the abdominal cavity, locate
Harvest and clamp the common bile duct as it enters the duodenum.
and Preparation Inject 1.4 mg/ml collagenase P solution into the common bile
for Flow Cytometry duct with a 30-gauge needle until the pancreas appears fully
inflated. Transfer to a 15 ml tube containing 1 ml of cold
HBSS. Keep on ice before proceeding to the next step as
collagenase injections are repeated when multiple pancreata
are isolated. Typically, 500–800 islets are used for a single
analysis by flow cytometry. This requires combining pancreata
from three to five mice.
2. Collect the spleen in order to prepare single-color and positive
flow cytometry controls. Place the spleen in a FACS tube with
1–2 ml of PBS and place the tube on ice.
3. Incubate the 15 ml tube containing the pancreas in a 37 C
water bath and mechanically disrupt the pancreatic tissue (see
Notes 4 and 5).
Flow Cytometric Analysis of Islet Leukocytes 189
3.4 Cell Counts 1. Pipette or gently vortex the cell suspensions to ensure that they
are homogenous.
2. Collect a 10- to 20-μl aliquot of each sample and determine the
approximate number of viable leukocytes using trypan blue and
a hemocytometer or an automated cell counter that is capable
of distinguishing between leukocytes and stromal cells.
3. Calculate the approximate number of leukocytes present in the
suspension.
4. Centrifuge cells (400 g, 4 C, 5 min) and decant the super-
natant. Resuspend the pellets in an appropriate volume of PBS
and transfer approximately 1 106 cells to labeled FACS tubes.
For a successful flow cytometry experiment, single-color
controls, positive and negative splenic/lymph node controls,
nonstained islet, isotype islet controls, and experimental islet
tubes are necessary (see Notes 6–8 and Fig. 2).
5. Add 1 ml of PBS to each tube and pellet the cells (400 g,
4 C, 5 min, and aspirate the supernatant). Proceed to Sub-
heading 3.5.
3.6 Intracellular 1. Very gently resuspend the pelleted cells (Subheading 3.5, step
Staining 6) in 100–200 μl of a suitable fixation/permeabilization
reagent (i.e., BD Cytofix/Cytoperm), following the manufac-
turer’s instructions for preparation and use (see Note 9).
The rest of the protocol assumes that BD Cytofix/Cyto-
perm reagents were used to fix and permeabilize the cells. If
other intracellular staining reagents were used, the intracellular
staining protocol should be modified following the manufac-
turer’s instructions.
2. Incubate the samples for 20–30 min at 4 C in the dark to
permeabilize the cells. Intracellular antibody cocktails (100 μl/
tube) should be prepared using BD permeabilization/wash
solution. The appropriate amount of intracellular antibody to
use for one test should be empirically determined (see Note 6).
3. Add 1 ml of BD permeabilization/wash solution to each tube
and pellet the cells by centrifugation (400 g, 4 C, 5 min).
Discard the supernatant.
4. Gently resuspend the samples in 100 μl of the correct intracel-
lular antibody cocktail. Incubate the samples for 20–30 min at
4 C in the dark to stain for intracellular antigens.
5. Add 1 ml of 1 permeabilization/wash solution to each tube
to wash the cells.
6. Gently pellet the cells and decant the supernatant (400 g,
4 C, 5 min).
7. Repeat steps 4 and 5.
8. Gently resuspend the pelleted cells in 100 μl of 2% paraformal-
dehyde in PBS. The tubes may be stored at 4 C in the dark or
immediately analyzed on the flow cytometer.
3.7 Sample 1. Start and set up the flow cytometer and instrument settings
Acquisition according to manufacturer’s instructions and in-house proto-
on the Flow Cytometer cols. The islet samples should be filtered using a 70-μm cell
strainer cap just before the sample acquisition.
2. The splenic unstained and single-color control samples should
be used to set and adjust the gain values on the forward (FSC)
scatter, side (SSC) scatter, and fluorescent channels. Ideal
single-color controls should have a clearly defined positive
and negative population. Once the acquisition settings have
been finalized, the positive control, negative control, and
experimental samples can be acquired.
192 Matthew J. Butcher et al.
4 Notes
Fig. 1 Recommended gating scheme for human and murine islet-infiltrating leukocytes. As multiple cell types
are present within pancreatic islets, including alpha, beta, delta, PP cells, and leukocytes, the initial forward
scatter (FSC) and side scatter (SSC) settings should be set based on a tissue or specimen with clear leukocyte
populations, such as peripheral blood leukocytes, splenocytes, or lymphocytes (not shown). As shown in the
figure, the initial ungated islet cell forward and side scatter profile will have various cell populations that are
dispersed throughout the graph (far left column, a). To specifically examine leukocytes within the islet
suspension, CD45, the common leukocyte marker, can be used (b)—second column first and third rows:
CD45 staining, second and fourth rows: nonstained control). As large granular endocrine cells are more
autofluorescent than leukocytes, islet CD45 staining should be compared with an islet isotype control for each
specimen to place the initial CD45 gate (second column, empty channel). After the CD45 gate has been set,
aggregates of cells should be excluded from the analysis using FSC/FSC-A (c) or FSC/FSC-W (not shown)
plots. Once single CD45+ leukocytes have been gated, a forward versus side scatter should be placed to
exclude debris that are under 40K on the FSC axis and any events that are larger than 250K on the side scatter
axis ((d)—fourth column). Single CD45+ cells ((e)—shown in the fifth column) can be used for subsequent
leukocyte subset, phenotype, and activation analyses. Note: a cell viability dye can also be used to specifically
gate on viable islet-infiltrating leukocytes (not shown). To correctly distinguish between dead cells and viable
cells, an appropriate islet-positive control should be used. HFD High-fat diet, CD Show diet, T2DM
Type 2 diabetes mellitus, Non-DM Nondiabetic, FSC Forward scatter, SSC Side scatter, and FSC-A Forward
scatter-area
Fig. 2 Representative CD3+ T-cell and CD20+ B-cell staining within nondiabetic and type 2 diabetic human
and murine islets. To examine intraislet CD3+ T-cell and CD20+ B-cell content, single CD45+ islet leukocytes
were gated as described in Fig. 1 and analyzed for CD3 (top) and CD20 (bottom) staining. Nondiabetic and type
2 diabetic human islets (first and third rows) and their corresponding isotype controls are shown. In addition,
chow diet fed and high-fat western diet fed C57Bl6 islets (second and fourth rows) and their corresponding
isotype controls are also shown. Islet isotype controls are useful and recommended for initially placing the
gates as nonspecific staining or high autofluorescence can be a confounding factor when analyzing non-
lymphoid cell suspensions by flow cytometry
Flow Cytometric Analysis of Islet Leukocytes 195
Acknowledgments
References
1. Osborn O, Olefsky JM (2012) The cellular and promotes inflammation in type 2 diabetes. J
signaling networks linking the immune system Immunol 186(2):1162–1172
and metabolism in disease. Nat Med 18 4. Zeng C, Shi X, Zhang B, Liu H, Zhang L,
(3):363–374 Ding W, Zhao Y (2012) The imbalance of
2. Imai Y, Dobrian AD, Morris MA, Nadler JL Th17/Th1/Tregs in patients with type 2 dia-
(2013) Islet inflammation: a unifying target for betes: relationship with metabolic factors and
diabetes treatment? Trends Endocrinol Metab complications. J Mol Med (Berl) 90
24(7):351–360 (2):175–186
3. Jagannathan-Bogdan M, McDonnell ME, 5. Jagannathan M, McDonnell M, Liang Y,
Shin H, Rehman Q, Hasturk H, Apovian CM, Hasturk H, Hetzel J, Rubin D, Kantarci A,
Nikolajczyk BS (2011) Elevated proinflamma- Van Dyke TE, Ganley-Leal LM, Nikolajczyk
tory cytokine production by a skewed T cell BS (2010) Toll-like receptors regulate B cell
compartment requires monocytes and
Flow Cytometric Analysis of Islet Leukocytes 197
cytokine production in patients with diabetes. (2008) Isolation of major pancreatic cell types
Diabetologia 53(7):1461–1471 and long-term culture-initiating cells using
6. Ehses JA, Perren A, Eppler E, Ribaux P, Pospi- novel human surface markers. Stem Cell Res 1
silik JA, Maor-Cahn R, Gueripel X, (3):183–194
Ellingsgaard H, Schneider MK, Biollaz G, 13. Pechhold K, Zhu X, Harrison VS, Lee J,
Fontana A, Reinecke M, Homo-Delarche F, Chakrabarty S, Koczwara K, Gavrilova O, Har-
Donath MY (2007) Increased number of lan DM (2009) Dynamic changes in pancreatic
islet-associated macrophages in type 2 diabetes. endocrine cell abundance, distribution, and
Diabetes 56(9):2356–2370 function in antigen-induced and spontaneous
7. Richardson AJ, Willcox A, Bone AJ, Foulis AK, autoimmune diabetes. Diabetes 58
Morgan NG (2009) Islet-associated macro- (5):1175–1184
phages in type 2 diabetes. Diabetologia 14. Nackiewicz D, Dan M, He W, Rosa K, Salmi A,
51:1686–1688 Rutti S, Westwell-Roper C, Cunningham A,
8. Brissova M, Fowler MJ, Nicholson WE, Chu A, Speck M, Shuster-Klein C, Guardiola B,
Hirshberg B, Harlan DM, Powers AC (2005) Maedler K, Ehses JA (2014) TLR2/6 and
Assessment of human pancreatic islet architec- TLR4 activated macrophages contribute to
ture and composition by laser scanning confo- islet inflammation and impair beta cell-insulin
cal microscopy. J Histochem Cytochem 53 gene expression via IL-1 and IL-6. Diabetolo-
(9):1087–1097 gia 57:1645–1654
9. Cabera O, Berman DM, Kenyon NS, 15. Westwell-Roper CY, Ehses JA, Verchere CB
Ricordi C, Berggren PO, Caicedo A (2006) (2013) Resident macrophages mediate islet
The unique cytoarchitecture of human pancre- amyloid polypeptide-induced islet IL-1beta
atic islets has implications for islet cell function. production and beta cell dysfunction. Diabetes
Proc Natl Acad Sci U S A 103:2334–2339 63(5):1698–1711
10. MacDonald MJ, Longacre MJ, Stoker SW, 16. Yin N, Xu J, Ginhoux F, Randolph GJ,
Kendrick M, Thonpho A, Brown LJ, Hasan Merad M, Ding Y, Bromberg JS (2012) Func-
NM, Jitrapakdee S, Fukao T, Hanson MS, Fer- tional specialization of islet dendritic cell sub-
nandez LA, Odorico J (2011) Differences sets. J Immunol 188(10):4921–4930
between human and rodent pancreatic islets: 17. Butcher MJ, Hallinger D, Garcia E, Machida Y,
low pyruvate carboxylase, ATP citrate lyase, Chakrabarti S, Nadler J, Galkina EV, Imai Y
and pyruvate carboxylation and high glucose- (2014) Association of proinflammatory cyto-
stimulated acetoacetate in human pancreatic kines and islet resident leucocytes with islet
islets. J Biol Chem 286(21):18383–18396 dysfunction in type 2 diabetes. Diabetologia
11. Beare A, Stockinger H, Zola H, Nicholson I 57(3):491–501
(2008) Monoclonal antibodies to human cell 18. Ricordi C (1991) Quantitative and qualitative
surface antigens. Curr Protoc Immunol 4:4A standards for islet isolation assessment in
12. Dorrell C, Abraham SL, Lanxon-Cookson humans and large mammals. Pancreas 6
KM, Canaday PS, Streeter PR, Grompe M (2):242–244
Chapter 11
Abstract
The islets of Langerhans are clusters of cells dispersed throughout the pancreas that produce several
hormones essential for controlling a variety of metabolic processes, including glucose homeostasis and
lipid metabolism. Studying the transcriptional control of pancreatic islet cells has important implications for
understanding the mechanisms that control their normal development, as well as the pathogenesis of
metabolic diseases such as diabetes. Histones represent the main protein components of the chromatin
and undergo diverse covalent modifications that are very important for gene regulation. Here we describe
the isolation of pancreatic islets from rodents and subsequently outline the methods used to immunopre-
cipitate and analyze the native chromatin obtained from these cells.
Key words Islets of Langerhans, Histone modifications, Nucleosomes, Native chromatin immuno-
precipitation (NChIP), Antibodies
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_11, © Springer Science+Business Media, LLC, part of Springer Nature 2020
199
200 Ionel Sandovici et al.
2 Materials
3 Methods
Fig. 1 Isolation of pancreatic islets from mature mice. (a) The metallic clamp is first placed on the common
biliary duct near liver, and then the collagenase P solution is slowly injected into the duct under a microscope.
(b) The red dotted line indicates the fully inflated pancreas at the end of the collagenase injection. (c) Isolated
mouse pancreatic islets
Fig. 2 Diagram depicting the principle of NChIP. (a) Isolation of nuclei. (b)
Chromatin digestion. (c) Immunoprecipitation with an antibody that recognizes
a specific chromatin modification (green star). (d) DNA purification. (e) Analysis
by quantitative PCR
Fig. 3 Analysis of native chromatin fractions isolated from pancreatic islets using
agarose gel electrophoresis. Bands corresponding to chromatin fragments of
one nucleosome (mono) to four nucleosomes (tetra) in length are indicated. The
sizes of the chromatin fragments are verified against the 100 bp DNA ladder
(Hyperladder IV—Bioline) loaded on the right side of the gel
3.4 DNA Extraction 1. To each bound and mock fraction, add proteinase K (to a final
and Quantification concentration of 20 μg/ml), then incubate the tubes at 55 C
and 800 rpm (on a Thermomixer Comfort shaker or equiva-
lent) for 1–2 h (see Note 16). The input sample (saved in
Subheading 3.3) should also be used at this stage for DNA
extraction. After the proteinase K digestion is over, briefly spin
the tubes and place them in the magnetic rack for 1 min.
NChIP in Pancreatic Islets 209
3.5 Analysis by 1. Design primers suitable for qPCR (80–150 bp) using freely
Quantitative PCR available software such as Primer3. Before the first use, primers
(qPCR) and Data should be tested for specificity, using input DNA as template.
Interpretation The optimal annealing temperature (no primer dimers or non-
specific PCR products) can be identified using a gradient PCR
machine and by melting curve analysis (ideally this should be
between 58 C and 62 C). Using ten-fold serial dilutions of
input DNA, the efficiency of each primer pair can be calculated
using the formula E ¼ 10(1/slope) 1. The optimal primer
efficiency is 1; however, primer efficiencies between 0.9 and 1.1
are acceptable. A good primer pair should generate Ct values
210 Ionel Sandovici et al.
4 Notes
References
1. Puri S, Hebrok M (2010) Cellular plasticity epigenetic programming of the endocrine pan-
within the pancreas—lessons learned from creas: consequences for type 2 diabetes. Cell
development. Dev Cell 18:342–356 Mol Life Sci 70:1575–1595
2. Carter JD, Dula SB, Corbin KL et al (2009) A 6. Sandovici I, Smith NH, Nitert MD et al (2011)
practical guide to rodent islet isolation and Maternal diet and aging alter the epigenetic
assessment. Biol Proced Online 11:3–31 control of a promoter-enhancer interaction at
3. Ravier MA, Rutter GA (2010) Isolation and the Hnf4a gene in rat pancreatic islets. Proc
culture of mouse pancreatic islets for ex vivo Natl Acad Sci U S A 108:5449–5454
imaging studies with trappable or recombinant 7. Haumaitre C (2013) Epigenetic regulation of
fluorescent probes. Methods Mol Biol pancreatic islets. Curr Diab Rep 13:624–632
633:171–184 8. O’Neill LP, Turner BM (2003) Immunopre-
4. Li F, Jiang X, Li Y et al (2013) Isolation of cipitation of native chromatin: NChIP. Meth-
mouse islet by collagenase perfusion through ods 31:76–82
the splenic vein. Transplantation 96:e88–e89 9. Wagschal A, Delaval K, Pannetier M et al
5. Sandovici I, Hammerle CM, Ozanne SE et al (2007) Chromatin immunoprecipitation
(2013) Developmental and environmental (ChIP) on unfixed chromatin from cells and
NChIP in Pancreatic Islets 213
Abstract
Histological image analysis is becoming an increasingly important tool for research in biological science.
They are important in analyzing biological systems on various scales, from structural details to determina-
tion of number of cells, its area, localization, and concentration. This chapter focuses on analysis of
pancreatic sections stained for insulin and glucagon using a commercially available software.
Key words Histology, Pancreas, Immunohistochemistry, Whole-slide image, Image analysis, Visio-
morphDP, ImageJ
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_12, © Springer Science+Business Media, LLC, part of Springer Nature 2020
215
216 Parvathy E. Harikumar
its constituent pixel values are then used by the software to identify
all similar features in an image. Some of the analyses included in this
chapter are quantification of total pancreas area, total islet area,
alpha and beta cell area, islet number, and size distribution with
reference to specific parameters.
Although the method detailed here is specific for analysis with
VisiomorphDP, the idea is easily transferrable across different
computational image processing systems including imageJ, an
open-source image-processing program. This chapter does not
discuss the algorithms, its implementation, and image processing
techniques in detail as it is beyond the scope. Here, the author
presents a general protocol for detecting and quantifying islets and
its associated measurable features.
2 Materials
2.1 Animals C57/Bl6 mice (Charles River, Manston, UK) were obtained at
5–6 weeks of age. All procedures were conducted in accordance
with the UK Government Animals Scientific Procedures Act 1986,
and approved by the University of Buckingham Ethical Review
Board. The animals were housed in solid-bottomed, sawdust-filled
cages with access to tap water and maintained on a standard chow
diet fed ad libitum (10% kcal fat, 70% kcal carbohydrates, 20% kcal
protein [Beekay Diets, New Brunswick, NJ, USA]). The source of
fat in the chow diet was soybean oil. Mice were maintained at a
controlled temperature of 23 1 C, on a 12 h light–dark cycle. At
12 weeks of age, mice were killed using carbon dioxide inhalation.
2.2 Tissue 1. PBS-T: Phosphate buffered saline (PBS) with 0.1% Triton
Preparation & Staining X-100.
2. IMS: Industrial methylated spirit (Hayman Ltd., Essex, UK).
IMS can be used instead of molecular grade ethanol.
3. 6% hydrogen peroxide: 30% hydrogen peroxide is diluted with
methanol and stored in dark conditions at 4 C until ready to
be used. Use fresh preparations at all times.
4. Acid alcohol: 1% HCl is dissolved in 70% ethanol (v/v).
2.3 Suppliers The following reagents and equipment were purchased from vari-
ous suppliers: Histology cassettes (VWR International Ltd., UK),
automated tissue processor (TP1020, Leica Microsystems, Milton
Keynes, UK), Histo-ClearTM II (National Diagnostics, Hessle,
UK), paraffin wax (Thermo Scientific, Histoplast, UK), microtome
stainless steel blades (Feather S35; pfm medical UK Ltd., Chesire,
UK), microscope slides (Starfrost, Fischer Scientific, UK), VECTA-
SHIELD® HardSet™ Mounting medium (Vector Laboratories,
Peterborough, UK), insulin and glucagon antibody (Dako,
Quantification of Pancreatic Islets: Using Image Analysis Tools 217
3 Methods
3.1 Pancreas 1. Mice were sacrificed using carbon dioxide inhalation, after
Preparation which dissection was carried out to obtain pancreas for
histology.
2. Excess fat and connective tissues bound to the pancreas were
removed using forceps and scissors.
3. Weigh the pancreas and lay each pancreas between two sheets
of filter paper to keep the “head” (pancreas bound to duode-
num) and “tail” (pancreas bound to spleen) orientation and to
maintain the structure of the pancreas.
3.2 Fixation 1. Individual tissues were placed in histology cassettes and fixed in
and Tissue Processing 10% neutral buffered formalin for 17 h in room temperature
(RT) (see Note 1).
2. Specimens were processed using a semi-enclosed benchtop
automated tissue processor. Tissue processing involves dehy-
dration of the samples using graded ethanol (IMS) followed by
clearing in Histo-Clear™ II and finally paraffin wax infiltration
according to the program listed in Table 1.
3.3 Embedding 1. After processing, samples were removed promptly and trans-
and Sectioning ferred to metal cassettes before embedding in paraffin wax at
60 C using a Leica embedding station (model EG1150H) to
form paraffin blocks.
2. The blocks were cooled in ice-cold water for few seconds prior
to sectioning (see Notes 2 and 3).
3. Each block was then placed on a Leica microtome (model
RM2255) set with a clearance angle of 4 to cut 4 μm sections
with stainless steel blades.
4. During sectioning, each pancreas was divided into six different
planes that were separated by an interval of 80 μm, and from
each plane, 10 serial sections of 4 μm thickness were taken (see
Note 4).
5. Sections were then floated in a 37 C water bath and were taken
using positively charged glass slides.
6. Sections were then initially dried for 10 min on hot plate and
then allowed to dry overnight at 35 C in an oven.
7. The resulting slides can be used for various staining purposes as
well as immunohistochemistry.
218 Parvathy E. Harikumar
Table 1
Processing protocol
3.7 Image Capture 1. Whole-slide images of all stained sections were captured with
an Aperio Scanscope CS scanner.
2. Images can be captured at any magnification up to 40.
Whole-slide images were scanned at 20 and 40 original
magnification.
3.8 Image Analysis 1. Development of image analysis protocols was a six-step process
Software and an overview is presented in Fig. 1.
2. Once a protocol was created, it could be run on regions of
interest (ROIs), an entire image, or a batch of whole-slide
220 Parvathy E. Harikumar
Fig. 1 Overview of steps involved in developing image analysis protocols. The specific components of each
step are described for individual protocols
Fig. 2 Training by example. Panel A original image, B training image (blue: background, green: pancreas [both
identified by user] see Table 2) and C final segmented image (blue outline: ROI). Original magnification 10
(A) and 4 (B and C)
images. See Note 9 for an important tip before using images for
analysis.
3. Training by Example: The system was trained to recognize
different image features such as exocrine tissue, islets, blood
vessels, insulin, and glucagon (Figs. 2a and 3b). These were
defined by the operator using a labeling tool to isolate a repre-
sentative area of the feature to be quantified. The new markup
for each object is termed as label and is represented by different
colors.
4. Defining Preprocessing Steps: The purpose of preprocessing was
to allow enhancement of structure of interest by suppressing
noise and nonrelevant image components to improve image
segmentation. This feature combines information such as spec-
tral (RGB or contrast) or morphological information. For a
standard color image, the features are described by red, green,
and blue (RGB) pixel values, where each color band has a
certain intensity at each given pixel location. If the basic input
color band does not provide sufficient robust segmentation
results across multiple images, there are other image processing
filters available to use as required by the user. For this study,
Fig. 3 Islet detection. Islets are shown in green and exocrine are shown in pink. Panel A original image,
B training by example (see Table 3), C preprocessed image with median filter, D segmented image after
Bayesian classification and E final image after postprocessing. Image C shows the outcome of applying a [9, 9]
median filter to an original RGB image. This filter helps to reduce “noise” by smoothing out bright and dark
pixels. Original magnification 40
222 Parvathy E. Harikumar
RGB was insufficient in one case and the use of basic median
filter with a kernel size of 9 9 to RGB color band enhanced
the image by removing noise and preserving the edges. See
Note 10 for the list of all preprocessing filters available in
VisiomorphDP.
5. Defining Image Classification Method: Preprocessed images
undergo segmentation where every pixel sharing a defined set
of characteristics is segmented to allow quantification. There
are many image segmentation methods available, of which
thresholding is the most common. A threshold is defined for
a given feature and a class is assigned to all pixels with a feature
value above or equal to threshold, and remaining pixels are
assigned to another class. A Bayesian classifier can be used
instead of thresholding for image segmentation under two
main reasons. First, colors in specially stained tissues are a
mixture, for which thresholding does not allow for accurate
segmentation. Moreover, Bayesian classification does not
require the user to define an all-inclusive intensity range for
an image class as the representative training set defined will
attribute all pixels in the image to one of the defined classes. See
Note 11 for the list of all classification methods available in
VisiomorphDP.
6. Defining Postprocessing Steps: Following image segmentation,
postprocessing steps can be performed to further refine seg-
mented images based on morphological and contextual infor-
mation. Postprocessing steps allow incorporation of prior
knowledge into the segmentation process, often resulting in
better quantitative results. For example, one can fill holes,
separate objects, and smooth edges of the feature of interest.
Note that these steps create a marked-up image in order that
the user can make a visual assessment of the accuracy of seg-
mentation. By repeating the steps above, one may iteratively
create an effective algorithm. See Note 12 for the list of all
postprocessing operators.
7. Defining Output: The final step is the definition of quantifica-
tion parameters. There are a wide range of output parameters
to choose from to measure objects including number, length,
area, shape, and intensity. See Note 13 for the list of all outputs
available in VisiomorphDP.
3.9 Development 1. The four protocols developed were for (1) tissue detection;
of Image Analysis (2) islets detection; and quantification of (3) islet area and
Protocols (4) alpha and beta cell mass. These protocols were run on
micrographs of pancreas sections and each protocol was run
one after the other sequentially to get the output of interest.
2. Tissue Detection (Protocol 1): As whole-slide images were used
for analysis, non-tissue regions were excluded to decrease the
Quantification of Pancreatic Islets: Using Image Analysis Tools 223
Table 2
Protocol 1 for tissue detection
Table 3
Protocol 2 for islet detection
time required for analysis. Once protocol 1 was run (Table 2), a
ROI was automatically generated to delineate tissue (Fig. 2).
The resulting ROI was then analyzed using protocol 2 for islet
detection.
3. Islet Detection (Protocol 2): This protocol was developed to
identify and mask islets from the surrounding tissue (Fig. 3).
The steps involved in this protocol are detailed in Table 3.
4. Quantification of Islets (Protocol 3): Run this protocol on the
image resulting from protocol 2. The aim is get quantitative
data from the marked-up image; in this case, number of islets,
area of islets, and area of exocrine region (Fig. 4). The steps
involved in this protocol are detailed in Table 4.
224 Parvathy E. Harikumar
Fig. 4 Islet quantification. Panels A marked-up image (final image from protocol 2), B final segmented image
with islets (green) marked as ROI (red) and C output showing the quantitative data processed from the
marked-up image. Output from this analysis includes number of islets, islet area, and exocrine area. Original
magnification 40
Table 4
Protocol 3 for islet quantification
Fig. 5 Alpha and beta cell area quantification. Panels A marked-up image (final image after protocol 3),
B preprocessed image, C image segmented after Bayesian classification and D final image after postproces-
sing showing glucagon-producing alpha cells in blue and insulin-producing beta cells in green. Original
magnification 40
Table 5
Protocol 4 for alpha and beta cell quantification
a 20
db rosi
db control
db exe
15
lslet Number
10
0
9
99
99
99
99
99
-9
-1
-2
-3
-4
-9
0
0
10
20
30
40
50
15.0 db exe
12.5
db control
10.0
7.5
5.0
2.5
0.0
+
0 9
0 9
0 9
0 9
10 - 9 9
00 99
20 - 1 99
25 - 2 99
30 - 2 99
35 - 3 99
40 - 3 99
45 - 4 99
50 - 4 99
55 - 5 9
60 - 5 99
65 - 6 99
70 - 6 99
75 - 7 99
80 - 7 99
85 - 8 99
90 - 8 99
95 - 9 99
-9 9
10 99
0
10 - 9
20 - 19
30 - 29
40 - 39
50 - 49
00 99
00 49
00
00 4
00 9
00 4
00 9
00 4
00 9
00 4
00 4
00 9
00 4
00 9
00 4
00 9
00 4
00 9
9
15 - 1
0
Fig. 6 Example graphs showing distribution of islets based on its size. Such plots are useful for studies
involving drug treatments to show influence of drugs/compounds on islet neogenesis, islet size, and number
Quantification of Pancreatic Islets: Using Image Analysis Tools 227
4 Notes
References
1. Gamble M (2008) The hematoxylins and eosin. other molecules from fixed and processed tis-
In: Theory and practice of histological techni- sues, developmental methods of fixation. J His-
ques. Churchill Livingstone Elsevier, London, totechnol 24:201–210
pp 121–134 4. Al-Habian A∗, Harikumar PE∗, Stocker CJ,
2. Grizzle WE (2009) Special symposium: fixation Langlands K, Selway JL (2014) Histochemical
and tissue processing models. Biotech Histo- and immunohistochemical evaluation of mouse
chem 84:185–193. https://doi.org/10.3109/ skin histology: comparison of fixation with neu-
10520290903039052 tral buffered formalin and alcoholic formalin. J
3. Eltoum I, Fredenburgh J, Grizzle WE (2001) Histotechnol 37:115–124. https://doi.org/10.
Advanced concepts in fixation: effects of fixation 1179/2046023614Y.0000000050. (∗ equal
on immunohistochemistry, reversibility of fixa- contribution)
tion and recovery of proteins, nucleic acid, and
Chapter 13
Abstract
One of the main consequences of glucose action on pancreatic β-cells is the stimulation of Ca2+ entry as well
as Ca2+ release from intracellular compartments. Therefore, one of the cornerstones of any diabetes research
laboratory should be the ability to measure changes in intracellular calcium concentrations within pancre-
atic β-cells. There are a variety of methods available for the measurement of intracellular calcium, from
multiple regions of interest (ROIs) within single cells to observe oscillating calcium, to population-based
96-well applications to enable high-throughput screening of the effects of novel agonists. These methods
allow calcium signaling to be observed in a single cellular assay to look at oscillations at a cellular level, to
view a population response, and to enable high-throughput assays where the mean reflects a single cell
response.
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_13, © Springer Science+Business Media, LLC, part of Springer Nature 2020
231
232 Joanne L. Selway
2 Materials
3 Methods
3.1 Cell Culture 1. MIN6 cells, a cell line derived from murine pancreatic β-cells,
should be used at approximately 80% confluence between pas-
sages 16 and 40.
2. MIN6 cells are grown in DMEM containing 25 mM glucose
supplemented with 15% heat-inactivated FCS, 100 μg/ml
streptomycin, 100 units/ml neomycin 100 units/ml penicillin
sulfate, 40 mM NaHCO3, and 75 μM β-mercaptoethanol,
equilibrated with 5% CO2, 95% air at 37 C.
3. The medium was changed every 2–3 days.
Measurement of Calcium Signaling in Beta-Cell Lines Using Epifluorescence. . . 233
3.2 Ca2+ Imaging 1. MIN6 cells are plated on glass coverslips (size dependent on
Techniques microscopy equipment but typically 25 mm) for approximately
72 h to allow adhesion prior to experimentation (see Note 5).
3.2.1 Epifluorescence
Intracellular Ca2+ Imaging 2. Initially, cells should be washed three times in KRB to remove
DMEM components from the cells by gentle removal of media
and then addition and removal of KRB three times to cover the
coverslip (see Note 6).
3. MIN6 cells are then loaded with 2 μM Fura-2-AM by covering
the coverslip in dye loading buffer for 1 h at 37 C. In addition
to enabling the fluorescent dye to enter the cells, it starves the
cells of glucose and reduce spontaneous oscillations of Ca2+.
4. After an hour, wash the cells three times with KRB supplemen-
ted with 2 mM glucose by gentle removal of loading solution
and then addition and removal of KRB three times, sufficient to
cover the coverslip (see Notes 6 and 7).
5. Coverslips can then be mounted on the stage of an inverted
microscope (Nikon Diaphot inverted epifluorescence micro-
scope) and left in 200 μl KRB buffer (or a volume sufficient
to cover the surface of the coverslip when mounted in a cover-
slip bath) at room temperature (see Notes 6 and 7).
6. To measure [Ca2+]i, the cells must be excited at 340 and
380 nm at 1 s intervals and emissions collected at wavelengths
above 520 nm (see Note 8).
7. A period of recording should occur before agonist stimulation
(at least 30 s) to ensure that the cells have a low basal activity
(see Note 9).
8. The agonist of choice (such as glucose) can now be pipetted
onto the coverslip in a concentrated form to ensure the final
concentration is the required dose. Typically, agonists are
applied at 1.4 times the required concentration to the bath
directly above the coverslip (see Notes 6, 7 and 10).
9. Plot the data by looking at the ratio of 340:380 signal as a
pseudo measure of [Ca2+]i. The baseline readings should be
stable, with a value of 1.
3.3 Single-Cell 1. MIN6 cells should be spilt 1:3 onto 25-mm diameter glass
Confocal Intracellular coverslips and left for a minimum of 72 h before experimenta-
Ca2+ Imaging tion (see Note 5).
234 Joanne L. Selway
3.4 Population Based 1. MIN6 cells should be passaged 1:2 into 96-well plates (200 μl
or High-Throughput total volume) and left to culture for 48 h.
Intracellular Ca2+ 2. Wells should be washed three times with KRB supplemented
Imaging with 2 mM glucose by gentle removal of the media and then
addition and removal of KRB, sufficient to cover the well, three
times (see Notes 6 and 10) and then placed at room
temperature.
3. After 30 min, the KRB is replaced with dye loading solution
containing 2 μM Fluo-4.
4. After an additional 30 min, wash the wells three times with
KRB supplemented with 2 mM glucose by gentle removal of
the media and then addition and removal of KRB, sufficient to
cover the well, three times (see Notes 6 and 10).
5. Cells can then be left with 25% of the total volume of KRB as a
starting point (typically this is 200 μl).
Measurement of Calcium Signaling in Beta-Cell Lines Using Epifluorescence. . . 235
6. At least one well of cells that has not been loaded with Fluo-4-
AM should be included in all experiments as an autofluores-
cence control for the data analysis.
7. For stimulation, solutions should be 1.17 higher than
required with 75% of the total volume added from the solution
plate per well. All measurements must be made at room
temperature.
8. The reader required for the population measurement should
measure fluorescence at 520 nm every second during the exper-
iment (see Note 12).
3.5.2 FRET-Based 1. MIN6 cells need to be starved of glucose and serum for at least
Imaging 1 h in KRB prior to treatment.
2. All experiments must be carried out at room temperature.
3. Images are captured with the 20 objective of an epifluores-
cence microscope with a CCD camera.
4. The emission ratio imaging of the cameleon is accomplished by
using a 436DF20 excitation filter, 450-nm dichroic mirror, and
two emission filters (475/40 for enhanced CFP and 535/25
for citrine) controlled by a Lambda 10–2 filter changer.
5. Exposure times are typically 100–1000 ms and images can be
collected every 8–20 s for the most optimal results.
236 Joanne L. Selway
4 Notes
Table 1
Plate reader parameters to measure fluorescence at 520 nm
References
Abstract
The role of oxidative stress in the pathogenesis of type 2 diabetes (T2D), especially pancreatic β-cell
dysfunction and death, has become apparent in the last two decades. Peroxidase- and catalase-based
antioxidant mechanisms are particularly weak in β-cells and can be easily overwhelmed by excessive
production of reactive oxygen and nitrogen species in the course of pathological processes. Recent research
has attempted to define in detail the mechanistic aspects of oxidative stress-induced β-cell dysfunction.
Here, we describe the procedures for the measurement of various parameters important to assess oxidative
stress in pancreatic β-cells. Detailed protocols for determination of nitric oxide (NO) production, the
glutathione redox status, and general oxidative status in β-cells are presented in this chapter.
Key words Nitric oxide, Glutathione, Oxidative stress, β-cells, Type 2 diabetes mellitus (T2DM)
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_14, © Springer Science+Business Media, LLC, part of Springer Nature 2020
241
242 Rodrigo Carlessi et al.
2 Materials
10. Centrifuge.
11. Ultrapure or distilled water (see Note 4).
3 Methods
3.1 Measurement 1. For each assay series, a nitrite standard curve must be prepared
of Nitric Oxide Free for accurate quantitation of NO2 levels in experimental sam-
Radical (NO) Release ples. Prepare 5 mL of a 100 μM nitrite solution by diluting the
100 mM nitrite stock solution 1 in 1000 in beta cell culture
media (such as RPMI, see Note 5).
2. Label seven 1.5 mL disposable polypropylene tubes with the
following concentrations: 100, 50, 25, 12.5, 6.25, 3.125 and
1.56 μM. Pipette 200 μL of tissue culture media to all tubes,
labeled from 50 μM downwards, leaving the tube labeled
100 μM empty. Add 400 μL of 100 μM nitrite solution to the
tube labeled 100 μM.
3. Perform a serial twofold dilution by pipetting 200 μL from the
100 μM tube into the 50 μM tube. Vortex to mix, then proceed
down the serial dilution as shown in (Fig. 3).
4. Designate columns A, B, and C of the 96-well plate for the
nitrite standard curve. Pipette 50 μL of each nitrite standard
into the wells in rows A–G, starting at A with μM 100 and
down to 1.56 μM at G. Into the row H pipette tissue culture
media only, which will be the blank.
5. Add 50 μL of each experimental sample to wells in triplicate (see
Note 6).
6. Allow the Sulfanilamide Solution and NED Solution to equili-
brate to room temperature.
246 Rodrigo Carlessi et al.
Fig. 4 Representative nitrite standard curve. Trend line and equation are
calculated through linear regression analysis using Microsoft Excel Professional
Plus 2013
3.2 Glutathione 1. After treatment, washβ-cells with ice-cold PBS, and then resus-
Redox State pend cell pellet in 5% MPA (150–200 μL of meta-phosphoric
Measurement acid (MPA) for about 107–108 cells population or 50–100 μL
of MPA for 106–107 cells) in order to break the cell mem-
branes. Gently homogenize with a pipette, then centrifuge the
samples 15,000 g for 5 min at 4 C and the supernatants
(from now on referred to as ‘samples’) are collected and stored
for subsequent analysis (see Note 8).
2. Samples designated to measurement of initial GSSG should be
conjugated with NEM to eliminate all GSH before commence-
ment of the reaction (steps 3–6). Samples designated to total
glutathione assay go directly to step 7.
3. Using polypropylene microcentrifuge tubes (1.5 mL), sam-
ples are mixed with NEM and Pipes/KOH (see Note 9).
4. Subsequently, samples are centrifuged at max speed for 3 min
and supernatants discarded.
5. Pellets are resuspended in 500 μL of ethyl acetate, centrifuged
at max speed for 3 min and supernatants discarded. This step is
repeated two times more (see Note 10).
6. Following the last wash with ethyl acetate, supernatants should
be carefully removed with pipette and dried in a SpeedVac
machine at room temperature. Finally, samples are resuspended
in 50 μL of 5% MPA for determination of GSSG.
7. For the determination of GSH and GSSG, the following
components need to be prepared fresh just before the assay:
Assay buffer, 14.1 mM DTNB, 1.7 mM NADPH and
Glutathione Reductase solution (for details see items 6–9 of
Subheading 2.2).
8. A standard curve for GSH and GSSG must be prepared accord-
ing to Table 1. To start, dilute the stock of 1 mg/mL GSH 1:4
in 5% MPA to get 813.5 μM and 1 mg/mL GSSG 1:2 in 5%
MPA to get 816.0 μM.
9. GSH and GSSG assay running in a polystyrene 96-well plate for
a final volume of 100 μL. Reagents must be added to the wells
of the 96-well plate in the following order (see Note 11).
(a) 10 μL sample in 5% MPA (cold).
(b) 70 μL of DTNB (37 C).
(c) Mix gently (5 s).
(d) 10 μL NADPH at room temperature.
(e) Mix gently (5 s) and incubate at 37 C for 2–3 min.
(f) Pipette 10 μL of GSSG reductase (it is highly recom-
mended to use a multichannel pipette for fast distribution
of GSSG reductase across all wells).
248 Rodrigo Carlessi et al.
Table 1
Series of dilutions for GSH and GSSG standard curves
Dilution GSH (μM) GSH (μL) MPA (μL) GSSG (μM) GSSG (μL) MPA (μL)
75 μL + 813.5 – 0 816.0 – 0
0
GSH
250
200
Δ Abs/min
150
y = (0.2269)x + (26.675)
100 R² = 0.99962
50
0
0 200 400 600 800 1000
Concentration (mM)
Fig. 5 Representative GSH standard curve. Trend line and equation are calcu-
lated through linear regression analysis using Microsoft Excel Professional Plus
2013
11. Using the slope (m) generated by the linear portion of the
standard curve and the formula y ¼ mx + b. Determine average
absorbance value of each experimental sample and estimate its
concentration by comparison to the standard curve. Impor-
tantly, the above calculation will generate the total amount
of GSH and GSSG; however, for final concentrations of
reduced GSH, the following calculation is required.
[GSH] ¼ GSHTOTAL–2 [GSSG].
3.3 General Oxidative 1. Seed the β-cells in 6-well plates, 4 105 cells/well, with 1 mL
Activity RPMI 1640 (10% FBS). Incubate the plate at 37 C overnight
(see Note 12).
2. Incubate the cells in the desired experimental conditions.
3. Reconstitute one vial of CM-H2DCFDA (50 μg) as follows:
dissolve the lyophilized CM-H2DCFDA with appropriate
amount of DMSO to the final concentration of 200 μM
(add 432.7 μL of DMSO to one vial of CM-H2DCFDA).
Keep it protected from light and on ice until ready to use (see
Note 13).
4. Prepare the working solution of CM-H2DCFDA: dilute the
stock CM-H2DCFDA 1:100 (final concentration 2 μM) into
phenol red and FBS free DMEM. The total volume of
CM-H2DCFDA working solution required is 1 mL per well
of 6-well plate. Keep working on ice and protected from light
(see Note 14).
5. Prepare one 15 mL and one 1.5 mL disposable polypropylene
tubes for each sample. Warm up the RPMI 1640 (10% FBS)
and Trypsin at 37 C ahead of time.
6. Collect the supernatants of cell culture into individual 15 mL
tubes (see Note 15).
7. Wash the cells with 0.5 mL PBS, then collet the PBS into the
same 15 mL tube.
8. Harvest the cells: add 0.25 mL of 0.25% Trypsin, and incubate
at 37 C for 5 min. Stop trypsinization by adding 0.5 mL
RPMI (10% FBS). Make sure all the cells are detached by
pipetting several times. Collect the cells to the same previous
15 mL tubes.
9. Centrifuge at 500 g for 5 min to precipitate the cells.
10. Wash the cell pellets by resuspending in 5 mL of PBS per tube,
and centrifuge again under the same conditions.
11. Discard supernatants and resuspend cells with 1 mL of
CM-H2DCFDA working solution. Incubate at 37 C pro-
tected from light for 30 min. Note, the negative control
(unstained sample) should be resuspended with 1 mL of phe-
nol red and FBS-free DMEM.
250 Rodrigo Carlessi et al.
12. Wash the cells with PBS as in steps 7 and 8 (see Note 16).
13. Resuspend the cells with 1 mL of DMEM (phenol red and FBS
free), and transfer to 1.5 mL tubes. For positive control cells,
add 1 U/mL Glucose Oxidase in 1 mL DMEM, and incubate
for additional 30 min at 37 C.
14. Analyze the samples by flow cytometry as follows: (see
Note 17).
15. Set the instrument parameters:
16. CM-H2DCFDA has approximate fluorescence excitation at
492–495 nm and emission at 517–527 nm. Thus, choose
channel BL1 with filter range of 515–545 nm (Fluorescein
isothiocyanate microscopy (FITC)).
17. Set appropriate gates for the cell population of interest: In the
protocol detailed here, we recommend collecting all the cells in
the wells, including floating dead cells, and cells in the process
of apoptosis. However, the profile of ROS generation is differ-
ent between living, dead, and apoptotic cells. By forward (FSC)
and side (SSC) scatter analysis, it is possible to choose different
target populations according to the experimental aims. For
instance, if only living cells are wanted, a gate must be set to
include only the cell population within typical FSC and SSC
values, and exclude dead or apoptotic cells with smaller FSC
but larger SSC values (Fig. 6a). Otherwise, a gate can be set to
include both populations, and ROS generation analyzed in all
cells (Fig. 6b).
a b
8.4
8.4
6
6
SSC-A (106)
SSC-A (106)
4
4 2
2
E1 E1
31.66% 46.00%
0
0
0 2 4 6 8.4 0 2 4 6 8.4
6
FSC-A (106) FSC-A (10 )
Fig. 6 FSC/SSC dot plot of palmitate treated BRIN-BD11 cells. (a) the gate was drawn to include only living
cells. (b) the gate was drawn to include both living and dead cells
Nitric Oxide and Redox State Measurements in Pancreatic Beta Cells 251
Fig. 7 Histogram of FITC fluorescence of BRIN-BD11 cells. The X-axis indicates fluorescence intensity, and the
Y-axis indicates cell count. (a) cells treated with vehicle (BSA) for 24 h. (b) cells treated with 125 μM palmitate
for 24 h. Palmitate treatment promoted elevated levels of ROS (Geometric Mean of fluorescence intensity
9377 vs. 24,920 respectively)
4 Notes
References
1. Halliwell B (1994) Free radicals, antioxidants, beta-cell functional integrity, metabolism and
and human disease: curiosity, cause, or conse- defense from inflammatory challenge. J Endo-
quence? Lancet 344(8924):721–724 crinol 211(1):87–97. https://doi.org/10.
2. Tiedge M, Lortz S, Drinkgern J, Lenzen S 1530/Joe-11-0236
(1997) Relation between antioxidant enzyme 8. Cruzat VF, Keane KN, Scheinpflug AL,
gene expression and antioxidative defense sta- Cordeiro R, Soares MJ, Newsholme P (2015)
tus of insulin-producing cells. Diabetes 46 Alanyl-glutamine improves pancreatic beta-cell
(11):1733–1742. https://doi.org/10.2337/ function following ex vivo inflammatory chal-
diabetes.46.11.1733 lenge. J Endocrinol 224(3):261–271. https://
3. Lenzen S, Drinkgern J, Tiedge M (1996) Low doi.org/10.1530/JOE-14-0677
antioxidant enzyme gene expression in pancre- 9. Kolberg A, Rosa TG, Puhl MT, Scola G, da
atic islets compared with various other mouse Rocha Janner D, Maslinkiewicz A, Lagranha
tissues. Free Radic Biol Med 20(3):463–466 DJ, Heck TG, Curi R, de Bittencourt PI Jr
4. Newsholme P, Cruzat VF, Keane KN, (2006) Low expression of MRP1/GS-X
Carlessi R, de Bittencourt PI Jr (2016) Molec- pump ATPase in lymphocytes of Walker
ular mechanisms of ROS production and oxi- 256 tumour-bearing rats is associated with
dative stress in diabetes. Biochem J 473 cyclopentenone prostaglandin accumulation
(24):4527–4550. https://doi.org/10.1042/ and cancer immunodeficiency. Cell Biochem
BCJ20160503C Funct 24(1):23–39. https://doi.org/10.
5. Rabinovitch A, Suarez-Pinzon WL (1998) 1002/cbf.1290
Cytokines and their roles in pancreatic islet 10. Cruzat VF, Pantaleao LC, Donato J Jr, de Bit-
beta-cell destruction and insulin-dependent tencourt PI Jr, Tirapegui J (2014) Oral supple-
diabetes mellitus. Biochem Pharmacol 55 mentations with free and dipeptide forms of
(8):1139–1149. https://doi.org/10.1016/ L-glutamine in endotoxemic mice: effects on
S0006-2952(97)00492-9 muscle glutamine-glutathione axis and heat
6. Griess P (1879) Bemerkungen zu der Abhan- shock proteins. J Nutr Biochem 25
dlung der HH. Weselsky und Benedikt ?Ueber (3):345–352. https://doi.org/10.1016/j.
einige Azoverbindungen?? Ber Dtsch Chem jnutbio.2013.11.009
Ges 12(1):426–428 11. Akerboom TP, Sies H (1981) Assay of gluta-
7. Krause MS, McClenaghan NH, Flatt PR, de thione, glutathione disulfide, and glutathione
Bittencourt PIH, Murphy C, Newsholme P mixed disulfides in biological samples. Meth-
(2011) L-arginine is essential for pancreatic ods Enzymol 77:373–382
Chapter 15
Abstract
Obesity and its comorbidity insulin resistance lead to the development of chronic metabolic diseases, such
as impaired fasted blood glucose and type 2 diabetes. Adipose tissue plays an important role in whole-body
glucose homeostasis, particularly in obese individuals; therefore, many in vivo models of type 2 diabetes are
obese, such as Lepob/ob and Leprdb/db mice or ZDF rats. Primary adipocytes therefore represent an
attractive in vitro model to study insulin-mediated glucose uptake to investigate the mechanisms of insulin
resistance and explore the potential insulin-sensitizing properties of new antidiabetic drugs.
Primary adipocytes are isolated by collagenase digestion of adipose tissue, Glucose transport is evaluated
by the measurement of intracellular uptake of a tracer (D-[U14C] glucose). The uptake of [U-14 C] glucose
reflects directly glucose transport.
In this chapter, we will describe the protocol for the isolation of primary rodent adipocytes and the
measurement of basal and insulin-stimulated glucose uptake.
Key words Primary adipocytes, Collagenase digestion, Insulin sensitivity, Glucose uptake
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_15, © Springer Science+Business Media, LLC, part of Springer Nature 2020
255
256 Mohamed S. Zaibi
2 Materials
Fig. 1 37 C incubator with rotating system. The 0.5-ml polyethylene microcentrifuge tubes are placed on the
rack and incubated at 37 C for 30 min, under constant rotational movement (30 rpm). Rotating unit (a),
Clamped rack (b), Air flow heating unit (c)
258 Mohamed S. Zaibi
3 Methods
3.1 Adipocytes Fat cells are prepared from different adipose tissue depots by using
Preparation the method described previously by Zaibi et al. [10]. Adipose tissue
(perigonadal, visceral, or subcutaneous) is minced and digested
with collagenase type II in Krebs-Ringer HEPES (KRH) buffer
pH 7.4 and 37 C. Isolated cells are filtered through 250–300 μm
nylon mesh, and the floating layer of adipocytes are washed several
times with a fresh buffer, then concentrated to 40% of final volume
of KRH buffer containing 5% bovine serum albumin (BSA) and
0.3 mM glucose and preincubated for 45 min under 95% O2: 5%
CO2.
1. Prepare Prep/Inc. buffers: 500 ml KRH buffer of stock solu-
tion containing 1.25 ml CaCl2, 5 g BSA (1%), 0.5 g Glucose.
2. Adjust the pH to 7.4.
3. Split into:
(a) Preparation buffer 300 ml containing 30 μl of 2 mM
adenosine (see Note 4) to reduce basal lipolysis).
(b) Incubation buffer 200 ml.
4. Warm both solutions to 37 C in water bath (see Note 5).
5. Animals (mice or rats) are euthanized by a method of killing
animals that are considered to be acceptable and humane (see
Note 6).
6. Collect the adipose tissue (approximately 5 g) in a 50-ml
polypropylene tube containing 30–40 ml of pre-warmed prep-
aration buffer.
7. Weigh the adipose tissue and split into two polypropylene tubes
(approximately 2–3 g of adipose tissue/tube), in 6 ml of prep-
aration buffer containing 1.5–2 mg of collagenase/g of adipose
tissue.
8. Dice the adipose tissue into small pieces (2–3 mm diameter
pieces) is this a note.
9. Incubate at 37 C in a water bath, under constant shaking
(250 cycles/min) until full digestion (approximately 60 min).
Gently swirl the tubes every 15 min and check the degree of
digestion (see Note 7).
10. When digestion is complete, adipocytes are separated by filtra-
tion through the 200–300 μm nylon mesh and the adipocytes
Primary Adipocytes as a Model for Insulin Sensitivity 259
Fig. 2 Procedure for processing frozen microcentrifuge tubes during the transfer of adipocytes layer into
scintillation vials. Frozen microcentrifuge tube (a) containing (from top to bottom) radioactive adipocytes,
dinonyl phthalate oil and radioactive buffer, container with dry ice (b) to maintain the tubes at low temperature,
tubes’ fraction to be transferred to scintillation vial (c)
8. Cut the upper phase which contains adipose cells (Fig. 2), using
a small ‘Resco guillotine’ nail cutter, or a very sharp scissors and
transfer it to the scintillation vial (see Note 15).
9. Add 0.5 ml of distilled water to each vial.
10. To each vial, add 10 ml of scintillation solution Ecosint™ A, or
a best equivalent (see Note 16).
11. Mix each tube for 10–15 s (using the vortex mixer), and leave
overnight.
12. Measure samples using the scintillation counter for 300 s.
13. Disintegrations per min (dpm) are recorded and glucose
uptake is expressed per mg of total cell protein (see Note 17).
4 Notes
Fig. 3 Effect of an insulin sensitizer compound (P) on glucose uptake in primary adipocytes of female wild-type
mice (Lean mice) and insulin-resistant mice model (Obese mice). Adipocytes were treated with insulin,
compound P (3 μM) alone, or a combination with insulin. n ¼ 8 mice per group for all columns. ∗p < 0.05;
∗∗
p < 0.01 for effect of compound P (P) compared to controls at the same concentration of insulin (C).
{
p < 0.05; {{p < 0.01; {{{p < 0.001 for effect of insulin compared to no insulin at the same concentration of
compound P. (Ngala et al. [9])
References
1. Trayhurn P, Beattie JH (2001) Physiological metabolic and genetic abnormalities. Am J
role of adipose tissue: white adipose tissue as Med 113(Suppl A):3S–11S
an endocrine and secretory organ. Proc Nutr 7. Ferrannini E (1995) Physiological and meta-
Soc 60:329–339 bolic consequences of obesity. Metabolism
2. Bailey CJ (1999) Insulin resistance and antidia- 44:15–17
betic drugs. Biochem Pharmacol 8. Walker M (1995) Obesity, insulin resistance,
58:1511–1520. https://doi.org/10.1016/ and its link to non-insulin-dependent diabetes
S0006-2952(99)00191-4 mellitus. Metabolism 44:18–20
3. Vernon RG, Clegg RA (1985) The metabolism 9. Ngala RA, Zaibi MS, Langlands K, Stocker CJ,
of white adipose tissue in vivo and in vitro. In: Arch JRS, Cawthorne MA (2014) Stimulation
Cryer A, Van RLR (eds) New perspectives in of glucose uptake in murine soleus muscle and
adipose tissue: structure, function and develop- adipocytes by 5-(4-phenoxybutoxy) psoralen
ment. Butterworths, London, pp 65–86 (PAP-1) may be mediated by Kv1.5 rather
4. Boden G (2001) Pathogenesis of type 2 diabe- than Kv1.3. PeerJ 2:e614. https://doi.org/
tes. Insulin resistance. Endocrinol Metab Clin 10.7717/peerj.614.
N Am 30:801–815 10. Zaibi MS, Stocker CJ, O’Dowd J, Davies A,
5. Olefsky JM, Ciaraldi TP, Kolterman OG Bellahcene M, Cawthorne MA, Brown AJ,
(1985) Mechanisms of insulin resistance in Smith DM, Arch JR (2010) Roles of GPR41
non-insulin-dependent (type II) diabetes. Am and GPR43 in leptin secretory responses of
J Med 79:12–22 murine adipocytes to short chain fatty acids.
6. LeRoith D (2002) Beta-cell dysfunction and FEBS Lett 584:2381–2386
insulin resistance in type 2 diabetes: role of
Chapter 16
Abstract
Islet transplantation is a potential treatment for Type 1 diabetes; however, improvements need to be made
before it could become clinically widely available. In preclinical studies, the mouse is often used to model
islet transplantation, with most studies aiming to improve transplantation outcome by manipulating the
islets prior to transplantation or by treating the recipient mouse. Here, we describe the process of islet
transplantation in the mouse, including how one can make the mouse diabetic, isolate donor islets, and
transplant the islets into two different sites. Finally, we discuss how to assess the outcome of the transplan-
tation in order to determine whether the experimental intervention has been beneficial.
Key words Islet transplantation, Mouse, Islet isolation, Intraportal islet transplantation, Renal sub-
capsular islet transplantation
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_16, © Springer Science+Business Media, LLC, part of Springer Nature 2020
265
266 Aileen J. F. King and Chloe L. Rackham
1.1 Minimal Mass In minimal mass models, fewer islets are implanted than would be
Models expected to reverse hyperglycemia in all animals [6, 7, 10]. If the
intervention has had a positive effect, more animals will become
normoglycemic. In mice, implanting between 300 and 500 mouse
islets will typically cure all animals. Therefore, the minimal mass
chosen is often between 150 and 250 islets which will usually cure
around 15–50% of the animals in the control group, depending on
site of transplantation, culture period and starting weight of the
mice. Due to the large variation in the blood glucose concentra-
tions of mice implanted with a minimal mass number, at least
10–14 mice per group are often required to see an effect.
1.2 Graft Survival Graft survival designs are most useful for allogeneic or xenogeneic
transplantations where the intervention intends to prevent or delay
immune rejection [2, 5, 11]. In these models, sufficient numbers of
islets should be implanted to initially reverse hyperglycemia in all
animals. Rising blood glucose concentrations can then be used as
an indicator of rejection. In allogeneic mouse models, rejection
occurs in control animals around 10–20 days after implantation
[2, 5, 11]. Alternatively, the grafts can be harvested at specific
time points after implantation to histologically detect rejection by
staining immune cell infiltration of the graft [11].
1.4 Graft Removal If the islets are implanted below the kidney, a nephrectomy of the
graft-bearing kidney can be carried out to demonstrate that the
graft is responsible for normoglycemia. After removing the graft,
the blood glucose concentrations will rise again if it is indeed the
graft that is responsible for maintaining normoglycemia, which
helps to rule out endogenous pancreatic beta cell regeneration
[2, 10]. In addition, the endogenous pancreas can be analyzed for
insulin content or insulin positive cells [16].
1.4.1 Ex Vivo Analysis Depending on the experimental question, the graft can be histo-
logically analyzed. Usually, the graft remains in situ in the implan-
tation organ. Hormone staining can determine the survival and/or
morphology of the islet cells [10], endothelial cell staining can
indicate the revascularization process [10], and staining immune
cells can indicate the involvement of different cells in rejection
[17]. Replication rates of the transplanted islets can be determined
by administering 5-bromo-2-deoxyuridine (BrdU) to the rodents
prior to killing them, which can then be quantified histologically
[18]. Alternatively, Ki-67 staining can detect replicating cells [19].
If more appropriate, the graft can be harvested to measure the
insulin content [10], although it should be noted that in diabetic
animals, the beta cells will be degranulated so it may not form an
accurate basis to determine beta cell mass. There are several steps
involved in measuring islet transplantation outcome in mice which
will be subdivided into the following sections:
1. Inducing diabetes.
2. Isolation of donor islets.
3. Transplantation of islets (under the renal capsule and
intraportally).
4. Posttransplantation monitoring.
5. Graft retrieval.
2 Materials
2.1 Induction 1. Citrate buffer: Dissolve 0.21 g citric acid in 100 ml saline (0.9%
of Diabetes NaCl). Alter the pH to 4.5 and filter through a 0.22 μm filter
into a sterile bottle. Store at 4 C for up to a month or freeze.
2. Streptozotocin solution: Make a 2% streptozotocin solution
immediately before injections by dissolving streptozotocin in
268 Aileen J. F. King and Chloe L. Rackham
2.2 Isolating Islets 1. Suitable cell culture medium for washing the islets (we use
Minimum essential medium (MEM)) supplemented with 10%
Newborn Calf Serum.
2. Collagenase solution: Collagenase type XI dissolved in serum-
free medium (we use MEM) at a concentration of 1 mg/ml.
Approximately 2 ml is needed per mouse.
3. Tools for isolating: one large pair of scissors, one large forceps,
two small forceps, one small pair of scissors, and one bulldog
clamp.
4. Dissecting microscope.
5. 2 ml syringes and 25–30 G needles depending on mouse size.
6. 70% ETOH.
7. Kim wipes.
8. Absorbable paper.
9. Histopaque-1077.
10. Plastic: Sterile 50 ml centrifuge tubes, petri dishes and Eppen-
dorf tubes.
11. Funnel.
12. 10 ml plastic pipettes and autopipettor.
13. 425 micrometer stainless steel mesh.
14. Refrigerated centrifuge with inserts for 50-ml tubes with adjus-
table settings to allow slow acceleration and brake to be
turned off.
15. Phosphate-buffered saline (PBS).
16. Culture medium (RPMI 1640 supplemented with 10% fetal
calf serum, 100 U/ml penicillin, and 100 ng/ml
streptomycin).
9. 70% alcohol.
10. Centrifuge with 15 ml tube inserts.
11. Tools for operation (per mouse): one scalpel, four small for-
ceps, two small pairs of scissors, drapes, needle holder, suture,
23-G needle and cauterizer.
12. Siliconized PE50 tubing (treated with SigmaCote and gas
sterilized).
13. Connector tubing (suitable to connect PE50 tubing to the end
of a P200 pipette tip).
14. P200 pipette and tips (+tips cut to fit the end of the Hamilton
syringe).
2.3.2 Intraportal Islet 1. Anesthetic (we use 1–5% isoflurane, 95% oxygen).
Transplantation 2. Analgesic (we use buprenorphine, 30 μg/kg injected
subcutaneously).
3. Heating pad.
4. Hamilton Syringe (500 μl).
5. Razor.
6. Antiseptic skin scrub.
7. 70% alcohol.
8. 25 G butterfly needle.
9. Tools for operation (per mouse): one scalpel, four small for-
ceps, two small pairs of scissors, alm retractor, Dumont forceps,
needle holder, suture, 23 G needle, cauterizer.
10. Drapes.
11. Absorbable hemostatic gelatin sponge (we use Spongostan).
12. Sterile cotton buds.
13. Sterile swabs (we use Topper 8).
14. Sterile saline.
15. Stereo microscope, preferably on an articulated arm.
3 Methods
3.2 Isolating Islets 1. This procedure can be carried out under anesthesia with exsan-
guination used to kill the mouse at the end of the procedure or
alternatively the mouse can be killed prior to the procedure by
cervical dislocation.
2. Lay mouse in a supine position and wet the fur with 70%
alcohol.
3. Make a large “V” incision in skin and the peritoneum from the
pubic region to the front legs to expose liver and intestines.
4. Rotate the mouse with its head toward you; place a lengthways
folded Kim wipe on the mouse’s chest across the sternum,
depress chest and flip back liver on top of tissue using half of
the tissue to hold the liver in place to expose posterior-dorsal
surface of the liver and the bile duct (see Note 4).
5. Move the intestines to the right side and gently pull the duo-
denum distally so the common bile duct can be visualized and
is taut.
6. Clamp duodenum at the Vater’s ampulla where the common
bile duct empties into the duodenum (see Note 5).
Islet Transplantation in Mice 271
Fig. 1 Pelleted islets in the PE50 tubing ready for transplantation. To transplant,
remove the connector tubing carefully and cut a bevel just in front of the islet
pellet
274 Aileen J. F. King and Chloe L. Rackham
Fig. 2 The correct position of the portal vein prior to intraportal transplantation.
Note the swab to the right is slightly pulling the tissue into the correct position
276 Aileen J. F. King and Chloe L. Rackham
9. Using fine forceps grip the tissue adjacent (operators left hand
side) to the portal vein and cannulate the portal vein.
10. Carefully inject the islets in a volume of approximately 200 μl
by using the screw function of the Hamilton Syringe.
11. Prior to removing the needle from the vein, apply spongostan
gel foam to the point of needle entry using Dumont forceps to
pick up the precut small square of spongostan and ensuring
that the foam is placed flat over the vein to prevent excess loss
of blood (see Note 10).
12. Applying some pressure to the Spongostan using a dry cotton
bud, remove the needle and hold the Spongostan in place for
approximately 2 min.
13. After removing pressure from the Spongostan, observe the vein
to check that there is no bleeding.
14. Leaving the Spongostan in place, carefully remove gauze swabs
and gently replace the intestines into the peritoneal cavity
(using soaked cotton buds).
15. Sew the peritoneum cavity.
16. Sew the skin.
17. Allow the mouse to recover and monitor closely.
3.4 Posttransplan- 1. Blood glucose concentrations and weight together can indicate
tation Monitoring glycemic control and should initially (for the first week) be
measured daily (see Note 11).
3.4.1 Blood Glucose
Monitoring 2. Using a 27-G or 30-G needle, make a small pin prick at the end
of the tail.
3. Apply a small amount of pressure to the tail proximal to the
pinprick and slide fingers toward the pinprick to form a droplet
of blood.
4. Apply the blood drop to a blood glucose meter. Typically, less
than 5 μl is required for most meters.
5. If excessive weight loss is evident, insulin can be administered
to comply with local ethical requirements.
3.5 Graft Retrieval 1. Anesthetize the mouse with isoflurane or another appropriate
anesthetic.
3.5.1 Nephrectomy
to Confirm the Role 2. Shave the fur on the left flank and treat the skin with surgical
of the Graft on Glycemia scrub, followed by 70% ethanol.
3. Administer analgesic.
4. Lay drapes over the mouse, with the operation area exposed
and check the mouse is adequately anesthetized.
5. Using a scalpel, make a 1.5–2 cm incision in the skin and using
scissors cut a 1-cm incision in the peritoneum over the left
kidney.
6. Use scissors to free the kidney from surrounding tissues (being
very careful not to cut the renal artery or vein).
7. Externalize the kidney.
8. Clamp the renal artery, renal vein and ureter close to the kidney
with a hemostatic clamp.
9. Use a scalpel to cut the kidney free. Place the kidney into
appropriate solution for analysis (e.g., formalin for histology
or acid alcohol for insulin content detection).
10. Ligate the renal artery, renal vein and ureter by tying two to
three times with suture.
11. Remove the hemostatic clamp and monitor whether there is
any bleeding (see Note 13).
12. Suture the peritoneum and suture the skin.
13. Monitor the recovery of the mouse.
14. Measure blood glucose daily until stable hyperglycemia has
been reached (at least three consecutive days of blood glucose
concentrations greater than 20 mM). The mouse should then
be killed and if necessary, the pancreas removed for analysis.
278 Aileen J. F. King and Chloe L. Rackham
4 Notes
1. Make sure that all mice are weighed and marked, with the dose
for each mouse calculated, prior to mixing the streptozotocin
with the citrate buffer to allow injection as soon as possible
after reconstitution (we generally try to inject all mice within
10 min of making the streptozotocin [20]).
2. The dose may need to be adjusted due to variation in the
streptozotocin lot and/or mouse strain [20, 21]. As glucose
directly competes with streptozotocin, fasting the mice may
increase percentage of mice from becoming diabetic.
3. Depending on your local ethical guidelines, mice may require
to be treated by insulin (we use 2–3 units daily of Caninsulin)
to prevent too much weight loss (more than 15% within 3 days
or more than 20% overall). If weight loss exceeds this, mice
should be killed. Cages will need to be changed more regularly
due to polyuria. In addition, water supply should be checked
daily as the mice will drink considerably more than normal. If
mice do not become diabetic, they can be re-injected again
with streptozotocin (although it is recommended that this is
only done once).
4. It is easier to flip the liver back if you clip off the xiphysternum.
5. Clamping should be carried out with precision; if the clamping
is not adequate, collagenase will leak into the duodenum. If the
clamp is too far down the pancreas, some ducts will be occluded
and thus the pancreas will not be fully inflated.
6. The connective tissue around the duct can be gently removed
by running the forceps up and down around the duct. This
allows the duct to be easily visualized. The forceps can also be
placed under the duct to form a support for injection. When
getting trained in the technique, it is desirable to initially inject
as closely as possible to the liver end of the duct, allowing the
entire length of the duct to be recannulated if first attempts are
initially not successful.
Islet Transplantation in Mice 279
References
1. Bruni A, Gala-Lopez B, Pepper AR, Abualhas- microcapsules does not improve graft outcome
san NS, Shapiro AJ (2014) Islet cell transplan- at the subcutaneous site. Artif Organs
tation for the treatment of type 1 diabetes: 36:564–570
recent advances and future challenges. Diabe- 4. Estil les E, Tellez N, Escoriza J, Montanya E
tes, Metab Syndr Obes 7:211–223 (2012) Increased beta-cell replication and
2. Zhi ZL, Kerby A, King AJ, Jones PM, Pickup beta-cell mass regeneration in syngeneically
JC (2012) Nano-scale encapsulation enhances transplanted rat islets overexpressing insulin-
allograft survival and function of islets trans- like growth factor II. Cell Transplant
planted in a mouse model of diabetes. Diabe- 21:2119–2129
tologia 55:1081–1090 5. Fernandes JR, Duvivier-Kali VF, Keegan M
3. Kerby A, Bohman S, Westberg H, Jones P, et al (2004) Transplantation of islets trans-
King A (2012) Immunoisolation of islets in duced with CTLA4-Ig and TGFbeta using
high guluronic acid barium-alginate
280 Aileen J. F. King and Chloe L. Rackham
adenovirus and lentivirus vectors. Transpl 14. King A, Bowe J (2016) Animal models for
Immunol 13:191–200 diabetes: understanding the pathogenesis and
6. King A, Lock J, Xu G, Bonner-Weir S, Weir GC finding new treatments. Biochem Pharmacol
(2005) Islet transplantation outcomes in mice 99:1–10
are better with fresh islets and exendin-4 treat- 15. King AJ, Austin AL, Nandi M, Bowe JE (2016)
ment. Diabetologia 48:2074–2079 Diabetes in rats is cured by islet transplanta-
7. McCall M, Toso C, Emamaullee J et al (2011) tion... but only during daytime. Cell Trans-
The caspase inhibitor IDN-6556 plant 26(1):171–172
(PF3491390) improves marginal mass engraft- 16. Mathews CE, Langley SH, Leiter EH (2002)
ment after islet transplantation in mice. Sur- New mouse model to study islet transplanta-
gery 150:48–55 tion in insulin-dependent diabetes mellitus.
8. King AJ, Guo Y, Cai D et al (2013) Sustained Transplantation 73:1333–1336
NF-kappaB activation and inhibition in beta- 17. Hong J, Yeom HJ, Lee E et al (2013)
cells have minimal effects on function and islet Islet allograft rejection in sensitized mice is
transplant outcomes. PLoS One 8:e77452 refractory to control by combination therapy
9. Toso C, Serre-Beinier V, Emamaullee J et al of immune-modulating agents. Transpl Immu-
(2008) The role of macrophage migration nol 28:86–92
inhibitory factor in mouse islet transplantation. 18. Krautz C, Wolk S, Steffen A et al (2013) Effects
Transplantation 86:1361–1369 of immunosuppression on alpha and beta cell
10. Rackham CL, Chagastelles PC, Nardi NB, renewal in transplanted mouse islets. Diabeto-
Hauge-Evans AC, Jones PM, King AJ (2011) logia 56:1596–1604
Co-transplantation of mesenchymal stem cells 19. Chen X, Zhang X, Chen F, Larson CS, Wang
maintains islet organisation and morphology in LJ, Kaufman DB (2009) Comparative study of
mice. Diabetologia 54:1127–1135 regenerative potential of beta cells from young
11. Pawlick R, Gala-Lopez B, Pepper AR, and aged donor mice using a novel islet trans-
McCall M, Ziff O, Shapiro AM (2014) The plantation model. Transplantation
combination of anti-NKG2D and CTLA-4 Ig 88:496–503
therapy prolongs islet allograft survival in a 20. Leiter EH, Schile A (2013) Genetic and phar-
murine model. Am J Transplant macologic models for type 1 diabetes. Curr
14:2367–2374 Protoc Mouse Biol 3:9–19
12. Bohman S, Andersson A, King A (2006) No 21. Sakata N, Yoshimatsu G, Tsuchiya H, Egawa S,
differences in efficacy between noncultured Unno M (2012) Animal models of diabetes
and cultured islets in reducing hyperglycemia mellitus for islet transplantation. Exp Diabetes
in a nonvascularized islet graft model. Diabetes Res 2012:256707
Technol Ther 8:536–545 22. McGuinness OP, Ayala JE, Laughlin MR, Was-
13. Deeds MC, Anderson JM, Armstrong AS et al serman DH (2009) NIH experiment in centra-
(2011) Single dose streptozotocin-induced lized mouse phenotyping: the Vanderbilt
diabetes: considerations for study design in experience and recommendations for evaluat-
islet transplantation models. Lab Anim ing glucose homeostasis in the mouse. Am J
45:131–140 Phys Endocrinol Metab 297:E849–E855
Chapter 17
Abstract
Under in vivo conditions, the study of physiological and pharmacological functions of an organ is difficult
due to whole-body interactions with the organ. Thus, an in vitro technique for the perfusion of isolated
pancreata was developed for physiologic and response studies including the investigation of endocrine
function and secretory responsiveness under a variety of diabetes-associated conditions. The pancreas is
isolated from the connecting spleen, stomach, and duodenum and transferred to a pre-warmed chamber
where it is perfused in isolation from all other organs. A detailed description of the isolation of pancreata
from rats and mice and the perfusion apparatus is described, as well as the measurement of glucose-
stimulated insulin secretion using an in-house-developed radioimmunoassay.
1 Introduction
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1_17, © Springer Science+Business Media, LLC, part of Springer Nature 2020
281
282 Edward T. Wargent
2 Materials
50
A
Turret
15
C
D
10
250
20
120
80
C
B
The turret supports two PVC sheathed copper wires.
It is formed from a tube drilled at 90` to the axis to
accommodate the wires as above. The tube is then
secured with a nylon centre bolt into the top plate.
Fig. 1 Perfusion tank setup. The sheathed copper wires are attached to the two cannulae that are connected to
the celiac artery and hepatic portal vein. The pancreas is housed in the central pool surrounded by perfusion
buffer. (a) inlet and (b) outlet for heated water keeps the central pool at 38 C. (c) inlet and (d) outlet for tubing
transporting the perfusate travels through the heated water re-heating it to perfusion temperature. All
dimensions are expressed in millimeters
13. Glucose.
14. Arginine.
15. Millex HA 0.45 μm filter, (Millipore, Massachusetts, USA).
3 Methods
13. Remove the fat from around the descending aorta between the
diaphragm and aortic bifurcation so that the aorta, caudal vena
cava, and any branching vessels are clearly exposed (see Note 7).
14. Using blunt-ended forceps, separate the entire exposed length
of the aorta from the body wall as well as the initial centimeter of
the celiac, superior mesenteric, and renal arteries (see Note 8).
15. Separate a section of aorta from the vena cava between the renal
and iliac arteries using blunt-ended forceps (see Note 9).
16. Lift the liver to the right and clear the hepatic portal vein of any
connective tissue. Place an untied ligature around the vein and
replace the liver exposing the celiac trunk.
17. Place untied ligatures around the aorta superior to the celiac
artery, inferior to the superior mesenteric artery, and between
the renal and iliac arteries. Leave several millimeters between
the celiac artery and the ligature for ease of manipulation of the
inlet cannula and also to avoid risk of cutting the celiac when
severing the aorta.
18. Place untied ligatures around the celiac, mesenteric, and renal
arteries (see Note 10).
19. Place a serrefine (see Note 11) across the aorta immediately
inferior to the renal arteries and another at the aortic bifurca-
tion. Using spring scissors make a cut into the aorta halfway
between the serrefines.
20. Insert a cannula for connection to a saline-containing syringe
into the aorta up to the superior serrefine and secure by tying
the ligature. For rats, the cannula should be polyethylene
tubing (as described in materials and methods) tapered for
easy insertion. Smaller rodents require a needle—rounded so
it can be inserted into the aorta. With experience, a 25-G
needle is possible, especially, in rodents with a lean body weight
over 30 g. A smaller gauge needle, for example, 27 G, may be
necessary for smaller animals or when learning the technique,
but increases the risk of leaks.
21. Remove the superior serrefine and tie the ligatures around the
renal and mesenteric arteries.
22. Tie the uppermost ligature around the aorta and immediately
perfuse the system with 2.5-ml saline over a period of about
20–30 s. Replace the saline syringe with one containing the
perfusion buffer and perfuse with 2.5 ml over 20–30 s. Imme-
diately, tie the remaining aortic ligature followed by the hepatic
portal ligature. Tie the latter ligature as close to the liver as
possible.
23. Cut the hepatic portal vein on the liver side of the ligature and
cut the aorta on the celiac trunk side of both the top two
ligatures. Lift out the section of abdominal organs, cutting
288 Edward T. Wargent
3.2 Ex Vivo 1. Locate the ligature around the celiac artery and hepatic portal
Preparation vein, lift and replace in the center of the tank with the two
for Pancreas Perfusion blood vessels exposed.
2. Maneuver the inlet cannula next to the celiac trunk so that it is
as almost horizontal. Holding the opening of the aorta adja-
cent to the celiac artery, but on the opposite wall, slip the
cannula into the celiac trunk so that the cannula enters the
celiac artery via the aorta. Secure in place by tying the ligature
around the celiac artery and the cannula. The celiac artery
cannula can be fashioned from a blunted 23-G needle for rats
(or no smaller than 25 G). Celiac cannulas for smaller rodents
should already be in place at the surgery stage and now
connected to the inlet tubing.
3. Maneuver the outlet cannula vertically next to the hepatic
portal vein. The outlet cannula should have an outside diame-
ter of 2–3 mm for rats and 5–6 mm for mice. Glass capillary
tubes with the end tapered and melted work well for rats—just
make sure the insertion angle is as horizontal as possible to
avoid the cannula slipping off. A blunted 25-G or 23-G needle
works well for mice. Lift the vein by the ligature and make an
incision into the vein as close to the ligature as possible (see
Note 13). Holding the thread of the ligature in one hand and
the lip of the aperture in the vein with forceps, slide the cannula
into the vein by almost a centimeter. Tape the ligature threads
to the cannula holder so the vein does not slip off and secure a
ligature around both the vein and cannula (see Note 14).
4. Perfuse the pancreas at a rate of 2 ml per gram pancreas weight
per minute until the perfusate begins to flow out and then
perfuse at 2 ml per gram pancreas weight per minute for
20 min to flush out any secretagogues released into the system
during the set-up period (see Note 15).
3.3 Pancreas 1. Set the perfusion equipment up as shown in Fig. 2. The tem-
Perfusion perature of the water bath and water heater/pump must main-
tain the perfusion buffer and perfusion tank temperature at
38 C. Check the perfusion tank temperature prior to surgery
and monitor throughout the experiment (see Note 16).
2. Glucose is required in the perfusion buffer to maintain the
integrity of the pancreas. The perfusion system can detect an
increase in insulin secretion at 4.4-mM glucose. 2.8-mM glu-
cose is used to maintain a basal level of insulin secretion and
pancreas function. 5.6-mM glucose represents the normal level
Measurement of Insulin Secretion Using Pancreas Perfusion in the Rodent 289
Manometer
Three-way tap
T-junction
Peristaltic
pump Perfusion tank
Perfusion Water bath
buffer
Fig. 2 Perfusion apparatus. The perfusion buffers are heated in a water bath and connected to air traps. Tubing
connects the air traps to a tap, the number and type depending upon the number of different perfusates to be
analyzed. The system powered by a peristaltic pump; immediately after which the perfusion tank is connected.
A T-junction separates the perfusate into two tubes. One is attached to the cannula inserted into the celiac
artery and the other connected to a manometer
100
90
80
% ( Bo- N S B ) / ( T- N S B )
70
60
50
40
30
20
10
0
1000 10000 100000 100000
dilution factor
Fig. 3 Primary antibody titer test. The primary antibody will bind to the hormone
of interest as well as the labeled tracer and standard. The bound fraction of the
assay is that part of the labeled and unlabeled antigen specifically bound to the
primary antibody and counted in the gamma counter. The primary antibody titer
test is a preliminary radioimmunoassay to determine what concentration of
primary antibody should be used. The amounts of tracer and secondary
antibody remain constant between groups of NSB’s and 100% tubes, while
only the primary antibody concentration in the Total tubes varies. Typically, three
totals, three NSBs, and three 100%s for each concentration of primary antibody
are tested. The concentration selected depends on the sensitivity desired in the
assay. The least primary antibody used, the lower the standard curve doses can
be reliable. In this example, the final dilution of primary antisera corresponds to
the dilution of the original stock that precipitates 30% of hot antigen added to the
tubes (minus non-specific precipitation). Here this corresponds to a final dilution
of 1:86,955 of the original stock solution. Note that the primary incubation
contains 100 μl of the standard/sample, 100 μl of label and 100 μl of the
primary antibody so in this example the primary antibody would be diluted
1:28,985 dilution (final dilution 1:86,955)
100
90
80
% (B o- N S B ) / ( T- N S B )
70
60
50
40
30
20
10
0
10000 20000 30000 40000 60000 100000
dilution factor
Fig. 4 Secondary antibody titer test. The secondary antibody binds to the primary
antibody/antigen complex (bound fraction) and after centrifugation, is
precipitated. After the free supernatant is decanted, the assay tube and
precipitate are counted in the gamma counter. A secondary antibody titer test
is a preliminary radioimmunoassay to determine what concentration of
secondary antibody maximally precipitates the bound fraction of the primary
antibody/antigen complex. In this test, the amount of tracer and primary antibody
remain constant while only the secondary antibody concentration varies.
Typically, there are three total count tubes, and for each different amount of
secondary antibody, three NSBs and three 100% tubes. In this example, the final
dilution of secondary antisera corresponding to the lowest concentration that will
still maximally precipitate the label is 1:30,000 would be used
100
75
% Bo
50
25
0
1 2 5 10 20 50 100 200 500
dilution factor
rat insulin dilution factor
perfusate dilution factor
Fig. 5 The parallelism test. The parallelism test is performed to validate the assay or sample material. A serial
dilution of a pooled sample is compared to varying standard curve doses within the same assay. Lines of equal
slope demonstrate no significant proportional analytical error within these ranges. This allows the assay of
unknown samples with the confidence that the dose estimate per/ml will not have a significant error. This is
important when unknowns are run in the same assay at different volumes
110
100
90
80
70
60
% Bo
50
40
30
20
10
0
2 5 10 20 50 100 200 500 1000
[insulin] (fmoles)
Fig. 6 Standard curve for the insulin radioimmunoassay. A calibration or standard curve is set up with
increasing amounts of known antigen, and from this curve, the amount of antigen in the unknown samples can
be calculated. The four basic necessities for a radioimmunoassay are an antiserum to the compound to be
measured, the availability of a radioactively labeled form of the compound, a method whereby antibody-bound
tracer can be separated from unbound tracer, and a standard unlabeled material. Each circle represents a
single data point and a sigmoidal regression line generated
294 Edward T. Wargent
insulin (fmoles/min)
100
10
0
0 10 20 30 40 50 60 70 80 90
time (minutes)
4 Notes
References
1. Sussman KE, Vaughan GD, Timmer RF (1966) Wiedenmann B, Plöckinger U (2008) Orexin-
An in vitro method for studying insulin secretion A inhibits glucagon secretion and gene expres-
in the perfused isolated rat pancreas. Metabolism sion through a Foxo1-dependent pathway.
15:466–476 Endocrinology 149:1618–1626
2. Emilsson V, Liu YL, Cawthorne MA, Morton 5. Egido EM, Hernández R, Marco J, Silvestre RA
NM, Davenport M (1997) Expression of the (2008) Effect of obestatin on insulin, glucagon
functional leptin receptor mRNA in pancreatic and somatostatin secretion in the perfused rat
islets and direct inhibitory action of leptin on pancreas. Regul Pept 152:61–66
insulin secretion. Diabetes 46:313–316 6. Tseng C-C, Zhang X-Y, Wolfe MM (1999)
3. de Heer J, Rasmussen C, Coy DH, Holst JJ Effect of GIP and GLP-1 antagonists on insulin
(2008) Glucagon-like peptide-1, but not release in the rat. Am J Physiol Endocrinol
glucose-dependent insulinotropic peptide, inhi- Metab 276:E1049–E1054
bits glucagon secretion via somatostatin (recep- 7. Eisenman JR, Chew BP (1983) Polyethylene
tor subtype 2) in the perfused rat pancreas. Glycol in conjunction with a secondary antibody
Diabetologia 51:2263–2270 for 24 hour radioimmunoassay of bovine prolac-
4. Göncz E, Strowski MZ, Grötzinger C, Nowak tin and growth hormone. J Dairy Science
KW, Kaczmarek P, Sassek M, Mergler S, 66:1174–1179
El-Zayat BF, Theodoropoulou M, Stalla GK,
INDEX
Claire J. Stocker (ed.), Type 2 Diabetes: Methods and Protocols, Methods in Molecular Biology, vol. 2076,
https://doi.org/10.1007/978-1-4939-9882-1, © Springer Science+Business Media, LLC, part of Springer Nature 2020
299
TYPE 2 DIABETES: METHODS AND PROTOCOLS
300 Index
Microarray .................................. 109–111, 114, 115, 121 R
MIN6 .................................................................... 232–238
Molecular modeling........................................................ 80 Radioimmunoassay .................... 283, 285, 289–294, 296
Mouse ..........................................33, 113, 122, 187, 202, Renal subcapsular islet transplantation ............... 268, 269
227, 265
S
Murine islets ........................................185–192, 195, 196
Sanger sequencing........................................ 86, 130, 131,
N 159–161, 164
Native chromatin immunoprecipitation Sequencing ............................................................ 85, 109,
(NChIP)................. 200, 201, 204, 205, 209–211 130, 209
NEBNext .............................................89, 90, 94–96, 166 SGLT2 inhibitors ......................................................15–20
Neonatal diabetes................................................... 87, 129 Stress .................................5, 10, 32, 34, 37, 38, 42, 185,
Next generation sequencing (NGS) .......................85–89, 241, 242, 279
109–111, 130, 164 Sulfonylureas ..............................3, 5, 7, 8, 10, 11, 13, 18
Nitric oxide (NO) ..............................241–246, 248, 250, Sulphonylureas .......................................... 4, 6, 7, 11, 129
252, 253
T
Nucleosomes ........................................................ 206, 207
Nutrition........................................................... 43–63, 185 Targeted next generation sequencing................. 130, 164
Thiazolidinediones................................ 5, 7–9, 13, 20, 80
O 3Rs ................................................................................. 283
Obesity.............................................. 13, 31, 44, 113, 255 Transcription factor binding site
Oxidative stress...............................................10, 241, 242 (TFBS) ..................................................... 110, 111,
114, 117
P Type 2 diabetes (T2D) ................................. 1–20, 31–33,
43–63, 71–82, 85, 88, 109–117, 119–127, 185,
Pancreas ..........................13, 35, 44, 130, 179, 188, 199, 193, 200, 242, 256, 282
215, 256, 267, 281
Pathway analysis ............................................................ 121 V
Perfusion...................................... 35, 180, 237, 281–287,
289, 290, 292, 294–297 Virtual screening ......................................................71–77,
Pharmaceutical industry ...........................................12, 19 79–82
Primary adipocytes ............................................... 255–262 VisiomorphDP ............................................ 215, 216, 222
Programming ..................................................... 36, 54–61
W
Q Whole exome sequencing (WES)...........................85–107
Quantitative PCR (qPCR).................................. 104–106, Whole slide image ....................................... 215, 219, 220
202, 206, 209, 210