Sie sind auf Seite 1von 22

4.

N
 ew Insights of Islet Biology
and the Pathophysiology of
Type 2 Diabetes
Alvin C. Powers, MD
Roland W. Stein, PhD
Deficits in insulin secretion are a central feature of the pathogenesis of type 2
diabetes mellitus (type 2 DM). As discussed in the chapter by Leahy and Pratley,
reduced insulin secretion results from defects in this process as well as reduced
beta cell number or mass. While there is continuing debate on whether reduced
function or reduced beta mass is more important, many investigators link beta
cell function and beta cell number, often using a term like functional beta cell
mass. This chapter will review the molecular determinants of functional beta
cell mass, including an overview of islet structure, development, and function.
In addition, we discuss how information from genetic and molecular studies
may enhance our understanding of the mechanisms leading to the insulin secretory deficits of type 2 DM. In keeping with the emphasis of the Translational
Endocrinology and Metabolism series, this chapter begins with a case presentation and highlights areas where new discoveries by islet biologists and clinical
investigators are needed (Translational Caveats and Challenges).

Case 4-1
A 28-year-old female is referred for management of diabetes of 13 years
duration. She is overweight with a body mass index (BMI) of 29 and
on a basal-bolus insulin regimen (bedtime lantus insulin; pre-meal is
taking a shorting-acting insulin analogue). She does not have a history
of diabetic ketoacidosis and says that on occasion she has missed her
insulin for 23 days without serious consequence. Her family history
is positive for type 1 DM in her mother. There is no family history of
autoimmune disease. Her physical exam is unremarkable, including normal monofilament testing and normal pedal pulses. Information from
her home glucose monitor shows hyperglycemia throughout the day, but
no hypoglycemia. Laboratory findings include: A1C of 8.1 and a negative
microalbuminuria. Her chemistry, hepatic, and renal profiles are normal.
For case study, see pages 95 and 96.

Translational Endocrinology & Metabolism, Volume 2, Number 1, 2011

95

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

Her Free T4 and thyroid-stimulating hormone (TSH) levels are normal


and her thyroid autoantibodies are negative. She recently had a normal
retinal exam by her optometrist.
A more detailed history finds that her mother was diagnosed with diabetes in her early 20s, and was initially treated with an oral medication,
followed by insulin when told she had type 1 DM. Her mother has a
similar history of insulin use (same type of insulin regimen and occasionally misses her insulin). Because of this new history, additional laboratories were obtained which showed no circulating islet cell and glutamic
acid decarboxylase antibodies and a random C-peptide of 1.0 (normal
0.781.89 ng/ml).

Insight from this patient


Upon further testing, she was found to have a TCF1/HNF-1alpha
mutationa monogenic form of diabetes known as maturity-onset
diabetes of the young (MODY) 3and not type 1 DM. The detectable
C-peptide level and the mothers family history suggested an alternative explanation to autoimmune beta cell destruction.The patient was
switched to sulfonylurea therapy and has improved glycemic control.
Genetic testing was advised for her mother. Most known forms of
monogenic diabetes cause diabetes by impairing insulin secretion.
While such mutations are a relatively rare (< 5%), this chapter will
discuss how the pathogenesis of these may provide a useful guide for
considering the molecular mechanisms responsible for the reduced
functional beta cell mass in type 2 DM.

Pancreatic IsletsA Complex Mini-Organ


The pancreatic islet is composed of five islet endocrine cell types: alpha cells that
produce glucagon, beta cells that produce insulin, delta cells that produce somatostatin, cells that produce pancreatic polypeptide, and epsilon cells that produce
ghrelin. In the adult human pancreas, the number of islets ranges from 500,000 to
2 million and account for 12% of the pancreatic mass (1, 2). The pancreatic islet
is more than a simple collection of endocrine cellsit is a highly vascularized and
innervated mini-organ that contains critical extracellular components (extracellular matrix, integrins, lamins) (Figure 4-1). The highly specialized intra-islet vasculature is lined by fenestrated endothelial cells. Blood flow changes during hyperglycemia and hypoglycemia conditions (3, 4). Alpha islet cells are downstream
from beta cells in terms of blood flow (511) and beta cell secretory products
like zinc, gamma aminobutyric acid (GABA), and insulin likely impact alpha cell
function (1215). The endothelial cells lining the intra-islet vessels provide criti-

96

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

Endothelial cells

Neurons

Extracellular matrix

Ca2+

K+

ATP-sensitive
K+ channel

Glucose
SUR

Depolarization

GLP-1 receptor
Ca

ATP/ADP

Pyruvate
Glucokinase

++

Nucleus
cAMP

Mitochondria

GPCRs

Islet-enriched
transcription
factors

Incretins

Nutrients
FFA
AA

Glucose
GLUT

Voltage-dependent
Ca2+ channel

Secretory granule

Cytokine
receptors

Glucose
Insulin receptor
Insulin

Growth
factors
and other
peptides

FIG 4-1. Beta cell microenvironment and nutrient-mediated insulin secretion. The schematic
shows a beta cell with its intracellular components for insulin secretion. Stimuli that influence beta
cell function, survival, or proliferation are shown on the right side of the figure. The beta cell also
receives input from blood vessels, endothelial cells, neurons, and components of the extracellular
matrix (upper portion of figure). Abbreviations: AA, arachidonic acid; ADP, adenosine
diphosphate; ATP, adenosine triphosphate; FFA, free fatty acids; GPCR, G protein-coupled
receptor; GLP-1, glucagon-like peptide-1; GLUT, glucose transporter; SUR, sulfonylurea receptor

cal signals for islet cell differentiation and development, but also islet cells produce high levels of angiogenic factors that promote their highly vascularized
state, like vascular endothelial growth factor-A (1621).
Insight into islet cell composition and function has principally come from
studies in rodents, as it is not possible to access human islets in vivo.
However, based on autopsy studies in humans and as human islets for
transplantation have become more available for study, some differences in
rodent and human islets have been noted. For example, the frequency and
distribution of endocrine cell types in human islets is different and more
variable. In human islets, beta cells account for 5274% of endocrine cells
(with most reports in the 5560% range) and there is a corresponding
greater percent of alpha cells (2228) in human islets compared to rodent
islets. In contrast, 7580% of mouse endocrine cells are beta cells.

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

97

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

The cellular composition of human islets also varies greatly depending


on pancreatic region, with islets from the ventral pancreas having a high
proportion of pancreatic polypeptide (PP)-producing cells and few glucagonproducing cells while dorsal pancreatic islets contain the percent of beta and
alpha cells stated above. Moreover, beta and alpha cells are found throughout
the human islet and their arrangement is much less uniform than is the beta
cell core and non-beta cell mantle structure of the rodent. As a consequence,
alpha-beta and beta-beta cell contact is greater in human islets as is both
alpha and beta cells in contact with intra-islet capillaries (29). The human
pancreas also appears to contain a greater number of scattered insulin- and
glucagon-producing cell clusters, and these may make a substantial contribution to beta cell mass (2, 24). Due to differences in primary amino acid
sequence of islet amyloid polypeptide, only humans form amyloid plaques
that contributes to islet dysfunction in type 2 DM (30, 31).

Translational Caveats and Challenges


How do non-islet cells and the islet microenvironment (extracellular
matrix, endothelial cells, etc.) influence beta cell function and survival
in vivo?
How do investigators better translate the large amount of genetic and
molecular information from rodent islets and model systems into
human islet biology? Systems that allow the study of human islets
in vivo, and new experimental approaches to study human islets
are needed. However, the human islets currently available for study
are often highly variable in quality and this leads to difficulties in
interpreting experimental results.

Overview of Insulin Secretion and Factors that


Regulate this Process
Insulin secretion is a complex process by which the beta cell integrates
signals from multiple inputs to secrete insulin when challenged and then
to cease secretion appropriately. As depicted in Figure 4-1, glucose enters
beta cells via a facilitated glucose transporter [in human this is glucose
transporter protein (GLUT) 1 and distinctly GLUT2 in rodents (32, 33)] and
is then phosphorylated by glucokinase. Intracellular glucose metabolism
leads to changes in the adenosine triphosphate (ATP) to adenosine diphosphate(ADP) ratio, which mediates closure of the ATP-sensitive potassium channel and subsequently membrane depolarization and activation
of voltage-dependent calcium channels. A secondary glucose-stimulated

98

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

pathway that is independent of the ATP-sensitive potassium channel is also


operative (3436). The increase in intracellular calcium is the final signal
for insulin secretory granule fusion with the plasma membrane and exocytosis. While there is general agreement on these steps, such a description
and schematic underestimates the complexity of regulated insulin secretion
in vivo. For example, other molecules and processes are important for regulated insulin secretion, with alterations potentially involved in the beta cell
dysfunction in type 2 DM, including (Figure 4-1):
The beta cell is more appropriately termed a nutrient sensor rather than
a glucose sensor since it also responds to amino acids and fatty acids
(37). Excess glucose and lipid impair beta cell function (discussed in
detail in What is Type 2 Diabetes Mellitus? Crucial Role of Maladaptive
Changes in Beta Cell and Adipocyte Biology by Leahy and Pratley).
Islet cells express a large number of G-protein coupled receptors (GPCRs;
most appreciated is the GLP-1 receptor), but the function of most in
islets has not been defined (38). Interestingly, GPCR receptor (GPR) 40
and GPR119 are of considerable interest, as they appear to mediate the
response to lipid-derived stimuli. GPCRs (other than the GLP-1 receptor)
may play important roles in regulation of insulin secretion and/or beta
cell mass, and might also serve as therapeutic targets (38).
Beta cells exist within an islet mini-organ and receive important signals from the extracellular matrix and other cells (Figure 4-1; discussed
above). For example, dispersed beta cells have greater basal insulin
secretion and reduced glucose-stimulated insulin secretion (GSIS)
compared to intact islets and this is at least partially dependent on
E-cadherin-beta cell interactions (39). Integrin-mediated interactions
between beta cells and collagen or vitronectin impact basal insulin secretion and insulin gene transcription (40). Likewise, lamin-beta cell
interactions enhance glucose-stimulated insulin secretion and promote
beta cell survival and proliferation (41). The basement membrane associated with endothelial cells and interactions between cells within the
islet (endocrine cells and endothelial cells) also influence insulin secretion and islet function (4244). The human intra-islet vasculature has
a distinctive double basement membrane consisting of lamins (45).
Islets produce and respond to a variety of cytokines and chemokines
(46). An emerging hypothesis is that cytokines produced by inflammatory processes within the islet contribute to the insulin secretory
dysfunction of type 2 DM. Efforts are needed to integrate this concept
into other pathologic processes known to be detrimental to islet function, including endoplasmic reticulum (ER) stress, glucose/oxidative
stress, lipotoxicity, and amyloid deposition (4750).

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

99

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

Translational Caveats and Challenges


What genes/proteins are critical determinants of beta cell function
in humans?
What is the role of GCPRs (beyond the GLP-1 receptor) in islet function or mass? Are any of these beta cell-expressed GPCRs therapeutic
targets in type 2 diabetes?
Because in vitro studies of isolated islets do not replicate the microenvironment that exists for a beta cell within an islet in vivo, conditions
need to be developed to understand how signals from other cell types
and/or the extracellular matrix contribute to the inadequate insulin
secretion capacity of type 2 DM.

Signals and Transcriptional Regulators Critical for


Islet Development and Function
The pancreas, an endoderm-derived organ, develops from the fusion of the
dorsal and ventral buds that evaginate from the primitive gut tube (51, 52).
All three pancreatic cell types (endocrine, acinar, and duct) arise from a
common embryologic progenitor (Figure 4-2). Extracellular signals for normal pancreatic development involve Hedgehog, Fgf, Wnt, and transforming
growth factor (TGF)-beta/activin responsive pathways (51, 52). Transcription factors induced by these cell-cell and cell-environment interactions
(for example, endothelial cells are required for islet development) are critical for pancreas/islet development, and also serve as developmental markers during pancreas/islet formation (51, 52) (Figure 4-2). Such information
is being used to direct human embryonic stem cell and inducible pleuripotent stem cell development into insulin-producing cells (53, 54). Of the
transcription factors essential to these processes, most are involved in both
islet cell development and function [the reader is referred to comprehensive reviews on this topic (51, 52)]. Some of these transcriptional regulators
are discussed below in the context of monogenic diabetes or determinants
of beta cell mass.

Determinants of Functional Beta Cell Mass


The term functional beta cell mass is an amalgam of individual cells that
secrete robustly (or poorly) and the total number of cells with insulin
secretory capacity. However, the molecular determinants regulating insulin
secretory capacity and beta cell mass are not well understood. For example, beta cells can increase insulin biosynthesis and/or amount of insulin
secreted in response to acute or chronic challenges such as hyperglycemia,

100

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

insulin resistance, or pregnancy. The factors that regulate beta cell mass in
humans are also poorly understood because islet can only be analyzed in
post-mortem samples and not non-invasively.
Most commonly, beta cell mass is inferred from functional tests of insulin
secretion and these estimates appear to correlate well with actual beta cell
number when measured in animal and human islet transplant models (55).
In terms of beta cell mass in humans, information from several autopsy series
have provided extremely valuable information, including (Figure 4-3): 1) beta
cell mass increases during childhood and adolescence, but is stable in adults;
2) the normal beta mass varies widely among individuals (perhaps up to
3-4 fold); 3) beta cell mass is only moderately reduced in type 2 DM; and 4)
beta cell mass is moderately increased in obese individuals (1, 2325, 5662).
These conclusions are from cross-sectional studies of a limited number of
cadaver samples. However, it is notable that there appears to be a marked difference in the ability of rodents to increase beta cell mass in relation to humans
(many-fold greater in rodents). For example, beta cell mass increases during
pregnancy in both rodents and humans, but this is much more limited in
humans and appears to principally reflect only more small islets and insulinpositive cells within ducts. In contrast, there is a marked elevation in both
rodent beta cell proliferation and islet beta cell numbers (6365).

Endocrine
Progenitor
Endoderm
Progenitor

Ngn3

Pdx-1
MafB
Nkx6.1

Arx

MafA

MafB

Ptf1a/Pdx-1

Ptf1a
Exocrine
Progenitor
FIG. 4-2. Development of different cell types in the pancreas. Moving from left to right,
progenitors cells differentiate in lineages that give rise to endocrine cells, ductal cells, and
pancreatic acinar cells. Selected islet-enriched transcription factors expressed principally in a
particular cell linege are shown. For a more detailed description of the transcription factors
involved in pancreas development, see references 51 and 52.

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

101

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

Beta mass in humans appears to increase during childhood and adolescence, peaking by early adulthood (Figure 4-3). As such, this highlights
many important questions and deficits in our knowledge that are quite
relevant to the pathogenesis of type 2 DM. How different is peak beta cell
mass between healthy humans and what determines this? What restricts
beta cell mass with age? How does the in utero environment influence
adult beta cell mass? Based on current information, only age and obesity
have been identified as determinants of beta cell mass in humans, but
there are likely more.
The molecular mechanisms controlling functional beta cell mass are
incompletely defined, yet are complex through involvement of islet-enriched
transcription factors, multiple signaling pathways, signals from the islet
microenvironment, growth factors, and metabolic signals (Figures 4-1 and
4-4). For example, a key islet-enriched transcription factor such as pancreatic
and duodenal homeobox 1 (Pdx-1) may impact functional beta cell mass in
multiple direct and/or indirect mechanisms such as (Figure 4-4):
Cell proliferation
Cell apoptosis
Beta cell function
Islet and/or beta development and differentiation.
Pdx-1 regulates genes encoding proteins critical to beta cell function and
is part of an interacting transcriptional network that regulates proliferation,
function and development capacity (Figure 4-2).

100%

High

?
?
?

cell mass
()

Medium

10

20

30

40

50

cell
proliferative
capacity
( )

Low

Age (years)
FIG. 4-3. Beta cell mass and proliferation in humans. The graph shows a proposed schematic of
beta cell mass and beta cell proliferation as a function of age. Following the achievement of peak
beta cell mass, it is proposed that beta cell mass may be stable or decline at different rates. The
schematic is based on data, concepts, and figures from several sources (1, 23, 24, 56-59, 62).

102

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

Expression of other
islet-enriched
transcription factors
Islet-enriched
transcription
factor

cell proliferation
cell apoptosis
cell differentiation
Hormone expression/
processing/secretion
Proteins involved
in glucose/nutrient
sensing

FIG. 4-4. Possible mechanisms by which an islet-enriched transcription factor may have an
impact on functional beta cell mass.

Pdx-1, which is broadly expressed in the developing pancreas and then


becomes restricted to beta cells and a subset of other endocrine cell types in
adult islets, regulates a broad range of processes critical for functional beta
cell mass, including those essential to glucose-stimulated insulin secretion
(GSIS) (such as GLUT2 and glucokinase), cell survival, replication, and the
response to insulin resistance [(51, 52) and references therein; (6675)].
In contrast, the MafA transcription factor is expressed very late in islet
beta cell development and exclusively in this cell type, but also regulates
genes significant to adult islet function (7684) (Figure 4-2). These include
proteins important for GSIS (insulin, ion channels), proliferation, as well
as the Glucose-6-phosphatase catalytic subunit-2 (also known as IGRP, an
autoantigen in type 1 DM) and the Znt8 zinc transporter [autoantigen in
type 1 DM and also a genetic loci linked by genome-wide association studies
(GWAS) to type 2 DM] (Figure 4-4) (76). When considering transcriptional
networks, it is notable that MafA (or closely related MafB) regulates Pdx-1
expression (85) and Pdx-1 regulates MafA expression (86).

Translational Caveats and Challenges


What genes/proteins are critical determinants of human beta cell
function, mass, and survival in vivo?
Why is the proliferative capacity of human beta cells so different from
rodent? Can this be modified?
How do human islets respond to insulin resistance (in vivo) (Figure 4-5)?

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

103

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

Insulin requirements
(Insulin resistance, Pregnancy)

Insulin secretion

cell size

cell number

Neogenesis

cell
proliferation

Apoptosis

FIG. 4-5. Proposed mechanisms by which beta cells respond to increased insulin requirements.

Genetic Loci Linked to Impaired Insulin Secretion


Based on family history data and the concordance of type 2 DM in identical twins, there is a strong consensus that there is an important genetic
component. However, despite an ever-growing amount of genetic data,
the major genetic loci responsible for the susceptibility continue to be
elusive. This section will review information on two fronts, monogenic
forms of diabetes and GWAS, highlighting selected information relevant
to type 2 DM (Table 4-1). Comprehensive reviews of the genetics of both
of these forms of diabetes have been published recently [(8789) and
references therein].

Information from Studies of Monogenic Diabetes


Monogenic forms of diabetes include those responsible for diabetes onset
either in the neonatal period or early adulthood (Table 4-1) (87, 88, 90).
Even though the associated genetic loci are relatively rare within type 2
DM population, their identification illustrated that islet function was principally affected in these individuals.
Common clinical settings for monogenic diabetes are: neonatal diabetes (<
6 months age at onset), familial diabetes with an affected parent (suggestive
of autosomal inheritance), onset of diabetes in a non-obese individual < 25
years of age, or diabetes associated with extra-pancreatic features such as renal
cysts or hepatic abnormalities. Such a diagnosis can have a profound impact
on an individual thought to have insulin-requiring diabetes since some forms

104

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

TABLE 4-1. Selected Genetic Loci Associated with Diabetes


(not immune-mediated) by impairing beta cell mass and/or function

Gene Locus

Phenotype/Comments

Monogenic Forms of Diabetes (Diagnosed > 1 year of age)A


MODY 3

TCF1/HNF-alpha

Islet-enriched transcription factor; progressive; may


respond to sulfonylurea

MODY 2

Glucokinase

Rate-limiting step in glucose metabolism in beta cells;


mild fasting hyperglycemia (heterozygote mutation)

MODY 1

HNF-4alpha

Islet-enriched transcription factor; may respond to


sulfonylurea

MODY 4

Pdx-1/IPF1

Islet-enriched transcription factor; pancreatic agenesis;


severe diabetes

MODY 5

TCF2/HNF-1beta

Islet-enriched transcription factor; renal (cysts,


dysplasia), uterine, and GU disorders

MODY 6

NEUROD1

Islet-enriched transcription factor

MODY 7

INS

Heterozygote mutations in insulin gene

Monogenic Forms of Diabetes (Neonatal; diagnosed < 1 year of age)B


KCNJ11

Sulfonylurea-associated protein; see Figure 4-1; may


respond to sulfonylurea

ABCC8

Sulfonylurea receptor; see Figure 4-1; may respond to


sulfonylurea

INS

Heterozygote mutations in insulin gene

Glucokinase

Rate-limiting step in glucose metabolism in beta cells;


homozygous mutation

Selected genetic loci associated with greater risk for type 2 diabetes
(diagnosed > 20 years of age)
> 30 loci

Identified by GWAS studies (87)

Listed in order of declining frequency

Listed in order of declining frequency; other genetic loci identified [see (87)], but in more
than 50% genetic loci are still unknown

Abbreviations: GWAS, genome-wide association studies; MODY, maturity onset diabetes


of the young

of monogenic diabetes respond to sulfonylureas, such as the MODY 3 (mutation in TCF1/HNF-1alpha) patient presented at the beginning of this chapter
or neonatal diabetes caused by a ATP-sensitive potassium channel mutation.
For some of the genetic loci in Table 4-1, there is experimental evidence
from clinical studies and animal models indicating how beta cell function
is affected and diabetes results in afflicted individuals. Mutations in the

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

105

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

glucokinase gene lead to adult (heterozygote mutation) or neonatal diabetes (homozygous mutation) because of its critical role in GSIS and glucose
homeostasis (91-94). The most common forms of neonatal diabetes arises
from mutations in the ATP-sensitive potassium channel or its associated
sulfonylurea receptor protein leading to impaired insulin secretion, or from
an insulin gene mutation resulting in defective proinsulin processing/folding, which causes beta cell death and reduced beta cell mass (95-97). Other
forms of monogenic diabetes result from mutations in key islet-enriched
transcription factors leading to impaired beta cell function or survival. For
example, IPF1/PDX1 mutations causes pancreatic agenesis (homozygous
mutation) and diabetes (homozygous and heterozygote mutations) as
would be expected from its role in early pancreatic development as well as
adult beta cell function (66, 88, 98, 99) (Figure 4-2).

How the GWAS Identified Loci Potentially Impact Functional


Beta Cell Mass and Type 2 Diabetes
Large amounts of information about type 2 DM or its related features such
as BMI and obesity continue to emerge from GWAS efforts (87). How the
large number of linked loci that convey a significant, but relatively low
relative risk for type 2 DM, will be integrated into the physicians diagnosis/management or into efforts to create new therapeutic agents is uncertain.
Also uncertain is how to use this genetic information to advance our understanding of the molecular pathogenesis of type 2 DM. Clinical studies of individuals carrying the TCF7L2 allele, the most robust GWAS-associated locus,
have shown reduced insulin secretion following an oral glucose tolerance test
and a reduced incretin effect (100103). Islets with siRNA-mediated reduction in TCF7L2 have reduced basal and glucose-stimulated insulin secretion
(100107), as a result of regulating islet genes that encode proteins involved in
insulin secretory granule formation and exocytosis (105, 107). While these findings strongly suggest that the risk associated with the TCF7L2 allele increases the
risk of type 2 DM by (at least) impairing insulin secretion, this widely expressed
protein may also increase diabetes risk by effects in peripheral tissues. Moreover,
there is little experimentally derived information regarding how other GWASidentified alleles increase the risk of type 2 DM.
Two recent studies suggest an additional mechanism for how genetic loci
associated with type 2 DM may lead to impaired insulin secretion (108, 109).
For example, Gaulton and colleagues determined whether the chromatin state
of DNA in human pancreatic islets around the regulatory regions of genes
was open (meaning not protected by nucleosomes) or closed (covered by
nucleosomes and hence, not likely to be accessible to DNA-binding transcrip-

106

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

tion factors that regulate gene expression). In normal human islets, the investigators found that the regulatory region of genes expressed in islets (those
encoding Pdx-1, Pax6, GLP-1 receptor, Znt8) had an open chromatin configuration. Interestingly, islet DNA from individuals with a high-risk TCF7L2 allele
had a different chromatin pattern than DNA from individuals without this
allele, suggesting in this case that TCF7L2 gene transcription was increased as
a result of changes in chromatin (108, 110) (Figure 4-6).

Translational Caveats and Challenges


For many of the monogenic forms of diabetes discussed above (Table 4-1),
the mechanism leading to the impaired functional beta cell mass appears
fairly certain. For targeted treatment, how do physicians readily identify
such patients in the general type 2 DM population?
How do the genetic loci identified by GWAS affect insulin secretion or
beta cell mass and survival (or insulin action)? In contrast to genes
responsible for monogenic diabetes, most genes suggested via GWAS
are expressed in multiple tissues and are not yet confirmed as being
the gene definitively linked to the increased risk of type 2 DM.

Nucleosome

Repressor factor complex

Histone tail
A
Allele
associated
with diabetes

Altered
transcription
Histone

DNA

B
Non-diabetes
allele

Normal
transcription

FIG. 4-6. Possible mechanism by which open or closed chromatin may produce epigenetic
changes involved in functional beta cell mass or type 2 diabetes. The transcriptional complex
involving DNA, nucleosomes, DNA-binding transcription factors, and histone/chromatin is
shown. Chromatin differences allows a DNA motif to be open for a DNA-binding protein
(panel A), but closed to the same DNA-binding protein (panel B).

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

107

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

In vivo studies in humans may identify a defect in insulin action or


insulin secretion in individuals with a particular locus, but these
may not easily translate into a defined molecular mechanism.
Furthermore, how to design studies using genetically modified mice
to define mechanisms by which a human polymorphism or singlenucleotide polymorphism (SNP) may be confounded by the sequence
differences between humans and rodents (in regions other than the
SNP or polymorphism)? If a study with a genetically modified mouse
finds that a protein is involved in a certain pathway (by a knockout or overexpression leading to a change in beta cell functional
mass), this suggests, but does not prove, that the SNP leads to a
defect in this pathway in humans (in whom the gene is not missing
or overexpressed, but whose expression or function has been altered
only slightly by the polymorphism).
How do epigenetic and changes in chromatin patterning affect genes
important for beta cell function, mass, and survival?

Summary and Future Directions


Despite a large of amount of in vivo data about the insulin secretory dysfunction in type 2 DM (discussed in What is Type 2 Diabetes Mellitus?
Crucial Role of Maladaptive Changes in Beta Cell and Adipocyte Biology)
and the increasing mechanistic information about the factors involved in
rodent islet cell formation and function, our understanding of the molecular defects in human islets or human beta cells in type 2 DM is poor. This
lack of knowledge is preventing efforts to understand the pathogenesis
of type 2 DM, to develop better ways of prevention and new therapies to
improve functional beta cell mass. Since the purpose of this Translational
Endocrinology and Metabolism series is to encourage discussion on how to
move concepts from the laboratory to the clinic and back, it is instructive
to consider possible mechanisms of beta cell dysfunction in the context of
this chapters discussion of islet biology (Figure 4-7). Possible mechanisms
for reduced functional beta cell mass in type 2 DM include:
Reduced beta cell mass because of a failure to attain sufficient
beta cell mass in childhood or early adolescence. This beta cell
defect could arise because of reduced beta cell proliferation
in utero, in childhood, or during adolescence or increased beta loss
during adulthood.
Impaired ability of the human beta cell to respond to the demands
of insulin resistance. Possible defects could include a failure of
proliferation, inability to sufficiently increase insulin biosynthesis,

108

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

Normal

Insulin
Secretion

Baseline

Cell
Mass

Baseline

IFG/IGT

Early stages
type 2 DM

Mid- and
late stages
type 2 DM

cell adaptation and compensation


Modulating factors:
Peak cell mass
Genetics
In utero environment

Glucose and lipid toxicity


Amyloid deposition and inflammation
cell loss

FIG. 4-7. Timeline of islet dysfunction associated with type 2 diabetes. The schematic shows
beta cell mass (lower graph) and insulin secretion (upper graph) beginning with normal beta cell
function (far left) and progressing through impaired fasting glucose (IFG) and impaired glucose
tolerance (IGT) to type 2 diabetes. Possible adaptations, compensations, and decompensations
are shown below the graphs. The curve for beta cell mass is speculative and beta cell mass may
not increase before declining or may begin to decline during the IFG/IGT phase.

or increased susceptibility to ER stress associated with elevated


insulin biosynthesis.
Increased susceptibility to glucose or lipid toxicity may lead to reduced beta cell function or survival.
Genetic differences (either monogenic or polygenic) may contribute to
any of these possible mechanisms of reduced functional beta cell mass.
References
1. Rahier J, Guiot Y, Goebbels RM, Sempoux C, Henquin JC 2008 Pancreatic beta-cell
mass in European subjects with type 2 diabetes. Diabetes Obes Metab 10 Suppl
4:32-42
2. Int Veld P, Marichal M 2010 Microscopic anatomy of the human islet of Langerhans.
Adv Exp Med Biol 654:1-19
3. Jansson L, Andersson A, Bodin B, Kallskog O 2007 Pancreatic islet blood flow
during euglycaemic, hyperinsulinaemic clamp in anaesthetized rats. Acta Physiol
(Oxf) 189:319-324
4. Nyman LR, Ford E, Powers AC, Piston DW 2010 Glucose-dependent blood flow
dynamics in murine pancreatic islets in vivo. Am J Physiol Endocrinol Metab
298:E807-814

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

109

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

5. Brunicardi FC, Stagner J, Bonner-Weir S, Wayland H, Kleinman R, Livingston


E, Guth P, Menger M, McCuskey R, Intaglietta M, Charles A, Ashley S, Cheung
A, Ipp E, Gilman S, Howard T, Passaro E, Jr. 1996 Microcirculation of the islets
of Langerhans. Long Beach Veterans Administration Regional Medical Education
Center Symposium. Diabetes 45:385-392
6. Samols E, Stagner JI 1996 Intra-islet cell-cell interactions and insulin secretion.
Diabetes Reviews 4:207-223
7. Marks V, Tan KS, Stagner JI, Samols E 1990 Intra-islet cellular interrelationships.
Biochemical Society Transactions 18:103-104
8. Stagner JI, Samols E 1992 The vascular order of islet cellular perfusion in the
human pancreas. Diabetes 41:93-97
9. Weir GC, Bonner-Weir S 1990 Islets of Langerhans: the puzzle of intraislet
interactions and their relevance to diabetes. J Clin Invest 85:983-987
10. Stagner JI, Samols E 1986 Retrograde perfusion as a model for testing the relative
effects of glucose versus insulin on the A cell. J Clin Invest 77:1034-1037
11. Nyman LR, Wells KS, Head WS, McCaughey M, Ford E, Brissova M, Piston DW,
Powers AC 2008 Real-time, multidimensional in vivo imaging used to investigate
blood flow in mouse pancreatic islets. J Clin Invest 118:3790-3797
12. Franklin IK, Wollheim CB 2004 GABA in the endocrine pancreas: its putative role
as an islet cell paracrine-signalling molecule. J Gen Physiol 123:185-190
13. Slucca M, Harmon JS, Oseid EA, Bryan J, Robertson RP 2010 ATP-Sensitive K+
Channel Mediates the Zinc Switch-Off Signal for Glucagon Response During
Glucose Deprivation. Diabetes 59:128-134
14. Zhou H, Tran PO, Yang S, Zhang T, LeRoy E, Oseid E, Robertson RP 2004 Regulation
of alpha-cell function by the beta-cell during hypoglycemia in Wistar rats: the
switch-off hypothesis. Diabetes 53:1482-1487
15. Gromada J, Franklin I, Wollheim CB 2007 Alpha-cells of the endocrine pancreas:
35 years of research but the enigma remains. Endocr Rev 28:84-116
16. Lammert E, Cleaver O, Melton D 2003 Role of endothelial cells in early pancreas
and liver development. Mech Dev 120:59-64.
17. Cleaver O, Melton DA 2003 Endothelial signaling during development. Nat Med
9:661-668.
18. Lammert E, Cleaver O, Melton D 2001 Induction of pancreatic differentiation by
signals from blood vessels. Science 294:564-567
19. Yoshitomi H, Zaret KS 2004 Endothelial cell interactions initiate dorsal
pancreas development by selectively inducing the transcription factor Ptf1a.
Development 21:21
20. Matsumoto K, Yoshitomi H, Rossant J, Zaret KS 2001 Liver organogenesis promoted
by endothelial cells prior to vascular function. Science 294:559-563
21. Brissova M, Shostak A, Shiota M, Wiebe PO, Poffenberger G, Kantz J, Chen Z, Carr
C, Jerome WG, Chen J, Baldwin HS, Nicholson W, Bader DM, Jetton T, Gannon M,
Powers AC 2006 Pancreatic islet production of vascular endothelial growth factor-A is essential for islet vascularization, revascularization, and function. Diabetes
55:2974-2985
22. Sakuraba H, Mizukami H, Yagihashi N, Wada R, Hanyu C, Yagihashi S 2002
Reduced beta-cell mass and expression of oxidative stress-related DNA damage in
the islet of Japanese Type II diabetic patients. Diabetologia 45:85-96
23. Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC 2003 Beta-cell
deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes
52:102-110

110

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

24. Bonner-Weir S, OBrien TD 2008 Islets in Type 2 Diabetes: In Honor of Dr. Robert
C. Turner. Diabetes 57:2899-2904
25. Yoon KH, Ko SH, Cho JH, Lee JM, Ahn YB, Song KH, Yoo SJ, Kang MI, Cha BY,
Lee KW, Son HY, Kang SK, Kim HS, Lee IK, Bonner-Weir S 2003 Selective beta-cell
loss and alpha-cell expansion in patients with type 2 diabetes mellitus in Korea. J
Clin Endocrinol Metab 88:2300-2308
26. Brissova M, Fowler MJ, Nicholson WE, Chu A, Hirshberg B, Harlan DM, Powers
AC 2005 Assessment of human pancreatic islet architecture and composition by
laser scanning confocal microscopy. J Histochem Cytochem 53:1087-1097
27. Cabrera O, Berman DM, Kenyon NS, Ricordi C, Berggren PO, Caicedo A 2006 The
unique cytoarchitecture of human pancreatic islets has implications for islet cell
function. Proc Natl Acad Sci U S A 103:2334-2339
28. Pisania A, Weir GC, ONeil JJ, Omer A, Tchipashvili V, Lei J, Colton CK, BonnerWeir S 2010 Quantitative analysis of cell composition and purity of human
pancreatic islet preparations. Lab Invest 90:1661-1675
29. Bosco D, Armanet M, Morel P, Niclauss N, Sgroi A, Muller YD, Giovannoni L,
Parnaud G, Berney T 2010 Unique arrangement of alpha- and beta-cells in human
islets of Langerhans. Diabetes 59:1202-1210
30. Haataja L, Gurlo T, Huang CJ, Butler PC 2008 Islet amyloid in type 2 diabetes, and
the toxic oligomer hypothesis. Endocr Rev 29:303-316
31. Kahn SE, Zraika S, Utzschneider KM, Hull RL 2009 The beta cell lesion in type
2 diabetes: there has to be a primary functional abnormality. Diabetologia
52:1003-1012
32. De Vos A, Heimberg H, Quartier E, Huypens P, Bouwens L, Pipeleers D, Schuit F
1995 Human and rat beta cells differ in glucose transporter but not in glucokinase
gene expression. J Clin Invest 96:2489-2495
33. Ferrer J, Benito C, Gomis R 1995 Pancreatic islet GLUT2 glucose transporter mRNA
and protein expression in humans with and without NIDDM. Diabetes 44:1369-1374
34. Straub SG, James RF, Dunne MJ, Sharp GW 1998 Glucose activates both K(ATP)
channel-dependent and K(ATP) channel-independent signaling pathways in
human islets. Diabetes 47:758-763
35. Henquin JC, Dufrane D, Nenquin M 2006 Nutrient control of insulin secretion in
isolated normal human islets. Diabetes 55:3470-3477
36. Henquin JC, Nenquin M, Ravier MA, Szollosi A 2009 Shortcomings of current models
of glucose-induced insulin secretion. Diabetes Obes Metab 11 Suppl 4:168-179
37. Nolan CJ, Prentki M 2008 The islet beta-cell: fuel responsive and vulnerable.
Trends Endocrinol Metab 19:285-291
38. Ahren B 2009 Islet G protein-coupled receptors as potential targets for treatment
of type 2 diabetes. Nat Rev Drug Discov 8:369-385
39. Jaques F, Jousset H, Tomas A, Prost AL, Wollheim CB, Irminger JC, Demaurex N,
Halban PA 2008 Dual effect of cell-cell contact disruption on cytosolic calcium and
insulin secretion. Endocrinology 149:2494-2505
40. Kaido T, Yebra M, Cirulli V, Rhodes C, Diaferia G, Montgomery AM 2006 Impact
of defined matrix interactions on insulin production by cultured human beta-cells:
effect on insulin content, secretion, and gene transcription. Diabetes 55:2723-2729
41. Parnaud G, Hammar E, Ribaux P, Donath MY, Berney T, Halban PA 2009 Signaling
pathways implicated in the stimulation of beta-cell proliferation by extracellular
matrix. Mol Endocrinol 23:1264-1271
42. Kragl M, Lammert E 2010 Basement membrane in pancreatic islet function. Adv
Exp Med Biol 654:217-234

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

111

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

43. Eberhard D, Kragl M, Lammert E 2010 Giving and taking: endothelial and betacells in the islets of Langerhans. Trends Endocrinol Metab 21:457-463
44. Jain R, Lammert E 2009 Cell-cell interactions in the endocrine pancreas. Diabetes
Obes Metab 11 Suppl 4:159-167
45. Virtanen I, Banerjee M, Palgi J, Korsgren O, Lukinius A, Thornell LE, Kikkawa
Y, Sekiguchi K, Hukkanen M, Konttinen YT, Otonkoski T 2008 Blood vessels of
human islets of Langerhans are surrounded by a double basement membrane.
Diabetologia 51:1181-1191
46. Donath MY, Boni-Schnetzler M, Ellingsgaard H, Halban PA, Ehses JA 2010
Cytokine production by islets in health and diabetes: cellular origin, regulation
and function. Trends Endocrinol Metab 21:261-267
47. El-Assaad W, Buteau J, Peyot ML, Nolan C, Roduit R, Hardy S, Joly E, Dbaibo G,
Rosenberg L, Prentki M 2003 Saturated fatty acids synergize with elevated glucose
to cause pancreatic beta-cell death. Endocrinology 144:4154-4163
48. Marchetti P, Bugliani M, Lupi R, Marselli L, Masini M, Boggi U, Filipponi F, Weir
GC, Eizirik DL, Cnop M 2007 The endoplasmic reticulum in pancreatic beta cells
of type 2 diabetes patients. Diabetologia 50:2486-2494
49. Tanaka Y, Tran PO, Harmon J, Robertson RP 2002 A role for glutathione peroxidase
in protecting pancreatic beta cells against oxidative stress in a model of glucose
toxicity. Proc Natl Acad Sci U S A 99:12363-12368
50. Potter KJ, Abedini A, Marek P, Klimek AM, Butterworth S, Driscoll M, Baker R,
Nilsson MR, Warnock GL, Oberholzer J, Bertera S, Trucco M, Korbutt GS, Fraser PE,
Raleigh DP, Verchere CB 2010 Islet amyloid deposition limits the viability of human
islet grafts but not porcine islet grafts. Proc Natl Acad Sci U S A 107:4305-4310
51. Ackermann AM, Gannon M 2007 Molecular regulation of pancreatic beta-cell mass
development, maintenance, and expansion. J Mol Endocrinol 38:193-206
52. Oliver-Krasinski JM, Stoffers DA 2008 On the origin of the beta cell. Genes Dev
22:1998-2021
53. Guo T, Hebrok M 2009 Stem cells to pancreatic beta-cells: new sources for diabetes
cell therapy. Endocr Rev 30:214-227
54. Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H,
Richardson M, Smart NG, Cunningham J, Agulnick AD, DAmour KA, Carpenter
MK, Baetge EE 2008 Pancreatic endoderm derived from human embryonic stem
cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol
26:443-452
55. Robertson RP 2007 Estimation of cell mass by metabolic tests: necessary, but how
sufficient? Diabetes 56:2420-2424
56. Meier JJ, Butler AE, Saisho Y, Monchamp T, Galasso R, Bhushan A, Rizza RA,
Butler PC 2008 Beta-cell replication is the primary mechanism subserving the
postnatal expansion of beta-cell mass in humans. Diabetes 57:1584-1594
57. Butler PC, Meier JJ, Butler AE, Bhushan A 2007 The replication of beta cells in
normal physiology, in disease and for therapy. Nat Clin Pract Endocrinol Metab
3:758-768
58. Butler AE, Galasso R, Meier JJ, Basu R, Rizza RA, Butler PC 2007 Modestly increased
beta cell apoptosis but no increased beta cell replication in recent-onset type 1
diabetic patients who died of diabetic ketoacidosis. Diabetologia 50:2323-2331
59. Bonner-Weir S, Li WC, Ouziel-Yahalom L, Guo L, Weir GC, Sharma A 2010 Beta-cell
growth and regeneration: replication is only part of the story. Diabetes 59:2340-2348

112

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

60. Schrader H, Menge BA, Schneider S, Belyaev O, Tannapfel A, Uhl W, Schmidt


WE, Meier JJ 2009 Reduced pancreatic volume and beta-cell area in patients with
chronic pancreatitis. Gastroenterology 136:513-522
61. Meier JJ, Menge BA, Breuer TG, Muller CA, Tannapfel A, Uhl W, Schmidt WE,
Schrader H 2009 Functional assessment of pancreatic beta-cell area in humans.
Diabetes 58:1595-1603
62. Meier JJ 2008 Beta cell mass in diabetes: a realistic therapeutic target? Diabetologia
51:703-713
63. Butler AE, Cao-Minh L, Galasso R, Rizza RA, Corradin A, Cobelli C, Butler PC 2010
Adaptive changes in pancreatic beta cell fractional area and beta cell turnover in
human pregnancy. Diabetologia 53:2167-2176
64. Van Assche FA, Aerts L, De Prins F 1978 A morphological study of the endocrine
pancreas in human pregnancy. Br J Obstet Gynaecol 85:818-820
65. Rieck S, Kaestner KH 2010 Expansion of beta-cell mass in response to pregnancy.
Trends Endocrinol Metab 21:151-158
66. Johnson JD, Ahmed NT, Luciani DS, Han Z, Tran H, Fujita J, Misler S, Edlund
H, Polonsky KS 2003 Increased islet apoptosis in Pdx1+/- mice. J Clin Invest
111:1147-1160.
67. Shih DQ, Heimesaat M, Kuwajima S, Stein R, Wright CV, Stoffel M 2002 Profound
defects in pancreatic beta-cell function in mice with combined heterozygous mutations
in Pdx-1, Hnf-1alpha, and Hnf-3beta. Proc Natl Acad Sci U S A 99:3818-3823
68. Kushner JA, Ye J, Schubert M, Burks DJ, Dow MA, Flint CL, Dutta S, Wright CV,
Montminy MR, White MF 2002 Pdx1 restores beta cell function in Irs2 knockout
mice. J Clin Invest 109:1193-1201
69. Brissova M, Shiota M, Nicholson WE, Gannon M, Knobel SM, Piston DW, Wright
CV, Powers AC 2002 Reduction in pancreatic transcription factor PDX-1 impairs
glucose-stimulated insulin secretion. J Biol Chem 277:11225-11232
70. Ahlgren U, Jonsson J, Jonsson L, Simu K, Edlund H 1998 Beta-cell-specific
inactivation of the mouse Ipf1/Pdx1 gene results in loss of the beta-cell phenotype
and maturity onset diabetes. Genes & Development 12:1763-1768
71. Watada H, Kajimoto Y, Miyagawa J, Hanafusa T, Hamaguchi K, Matsuoka T,
Yamamoto K, Matsuzawa Y, Kawamori R, Yamasaki Y 1996 PDX-1 induces insulin
and glucokinase gene expressions in alphaTC1 clone 6 cells in the presence of
betacellulin. Diabetes 45:1826-1831
72. Kulkarni RN, Jhala US, Winnay JN, Krajewski S, Montminy M, Kahn CR 2004
PDX-1 haploinsufficiency limits the compensatory islet hyperplasia that occurs in
response to insulin resistance. J Clin Invest 114:828-836
73. Brissova M, Blaha M, Spear C, Nicholson W, Radhika A, Shiota M, Charron MJ,
Wright CV, Powers AC 2005 Reduced PDX-1 expression impairs islet response to
insulin resistance and worsens glucose homeostasis. Am J Physiol Endocrinol
Metab 288:E707-714
74. Sachdeva MM, Stoffers DA 2009 Minireview: Meeting the demand for insulin:
molecular mechanisms of adaptive postnatal beta-cell mass expansion. Mol
Endocrinol 23:747-758
75. Pound LD, Hang Y, Sarkar SA, Wang Y, Milam LA, Oeser JK, Printz RL, Lee
CE, Stein R, Hutton JC, OBrien RM 2010 The pancreatic islet beta cell-enriched
transcription factor Pdx-1 regulates Slc30a8 gene transcription through an intronic
enhancer. Biochem J 433:95-105

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

113

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

76. Artner I, Hang Y, Mazur M, Yamamoto T, Guo M, Lindner J, Magnuson MA, Stein
R 2010 MafA and MafB regulate genes critical to beta-cells in a unique temporal
manner. Diabetes 59:2530-2539
77. Wang H, Brun T, Kataoka K, Sharma AJ, Wollheim CB 2007 MAFA controls genes
implicated in insulin biosynthesis and secretion. Diabetologia 50:348-358
78. Nishimura W, Kondo T, Salameh T, El Khattabi I, Dodge R, Bonner-Weir S, Sharma
A 2006 A switch from MafB to MafA expression accompanies differentiation to
pancreatic beta-cells. Dev Biol 293:526-539
79. Artner I, Le Lay J, Hang Y, Elghazi L, Schisler JC, Henderson E, Sosa-Pineda B,
Stein R 2006 MafB: an activator of the glucagon gene expressed in developing islet
alpha- and beta-cells. Diabetes 55:297-304
80. Zhao L, Guo M, Matsuoka TA, Hagman DK, Parazzoli SD, Poitout V, Stein R 2005
The islet beta cell-enriched MafA activator is a key regulator of insulin gene
transcription. J Biol Chem 280:11887-11894
81. Zhang C, Moriguchi T, Kajihara M, Esaki R, Harada A, Shimohata H, Oishi H, Hamada
M, Morito N, Hasegawa K, Kudo T, Engel JD, Yamamoto M, Takahashi S 2005 MafA is
a key regulator of glucose-stimulated insulin secretion. Mol Cell Biol 25:4969-4976
82. Matsuoka TA, Artner I, Henderson E, Means A, Sander M, Stein R 2004 The MafA
transcription factor appears to be responsible for tissue-specific expression of
insulin. Proc Natl Acad Sci U S A 101:2930-2933
83. Matsuoka TA, Zhao L, Artner I, Jarrett HW, Friedman D, Means A, Stein R 2003
Members of the large Maf transcription family regulate insulin gene transcription
in islet beta cells. Mol Cell Biol 23:6049-6062
84. Kataoka K, Han SI, Shioda S, Hirai M, Nishizawa M, Handa H 2002 MafA is a
glucose-regulated and pancreatic beta-cell-specific transcriptional activator for the
insulin gene. J Biol Chem 277:49903-49910
85. Vanhoose AM, Samaras S, Artner I, Henderson E, Hang Y, Stein R 2008 MafA
and MafB regulate Pdx1 transcription through the Area II control region in
pancreatic beta cells. J Biol Chem 283:22612-22619
86. Raum JC, Gerrish K, Artner I, Henderson E, Guo M, Sussel L, Schisler JC,
Newgard CB, Stein R 2006 FoxA2, Nkx2.2, and PDX-1 regulate islet beta-cellspecific mafA expression through conserved sequences located between base
pairs -8118 and -7750 upstream from the transcription start site. Mol Cell Biol
26:5735-5743
87. Bonnefond A, Froguel P, Vaxillaire M 2010 The emerging genetics of type 2
diabetes. Trends Mol Med 16:407-416
88. Vaxillaire M, Froguel P 2008 Monogenic diabetes in the young, pharmacogenetics
and relevance to multifactorial forms of type 2 diabetes. Endocr Rev 29:254-264
89. McCarthy MI, Hattersley AT 2008 Learning from molecular genetics: novel
insights arising from the definition of genes for monogenic and type 2 diabetes.
Diabetes 57:2889-2898
90. Greeley SAW, Tucker SE, Naylor RN, Bell GI, Philipson LH 2010 Neonatal
diabetes mellitus: A model for personalized medicine. Trends in Endocrinology
& Metabolism 21:464-472
91. Matschinsky FM, Glaser B, Magnuson MA 1998 Pancreatic beta cell
glucokinase. Closing the gap between theoretical concepts and experimental
realities. Diabetes 47:307-315

114

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

92. Postic C, Shiota M, Niswender KD, Jetton TL, Chen Y, Moates JM, Shelton KD,
Lindner J, Cherrington AD, Magnuson MA 1999 Dual roles for glucokinase in
glucose homeostasis as determined by liver and pancreatic beta cell-specific
gene knock-outs using Cre recombinase. J Biol Chem 274:305-315
93. Polonsky KS, Sturis J, Bell GI 1996 Non-insulin-dependent diabetes mellitusa
genetically programmed failure of the beta cell to compensate for insulin
resistance. New England Journal of Medicine 334:777-783
94. Polonsky KS 1995 Lilly Lecture 1994. The beta-cell in diabetes: from molecular
genetics to clinical research. Diabetes 44:705-717
95. Colombo C, Porzio O, Liu M, Massa O, Vasta M, Salardi S, Beccaria L, Monciotti
C, Toni S, Pedersen O, Hansen T, Federici L, Pesavento R, Cadario F, Federici
G, Ghirri P, Arvan P, Iafusco D, Barbetti F, Early Onset Diabetes Study Group
of the Italian Society of Pediatric Endocrinology and Diabetes 2008 Seven
mutations in the human insulin gene linked to permanent neonatal/infancyonset diabetes mellitus. J Clin Invest 118:2148-2156
96. Stoy J, Edghill EL, Flanagan SE, Ye H, Paz VP, Pluzhnikov A, Below JE, Hayes
MG, Cox NJ, Lipkind GM, Lipton RB, Greeley SA, Patch AM, Ellard S, Steiner
DF, Hattersley AT, Philipson LH, Bell GI, Neonatal Diabetes International
Collaborative Group 2007 Insulin gene mutations as a cause of permanent
neonatal diabetes. Proc Natl Acad Sci U S A 104:15040-15044
97. Garin I, Edghill EL, Akerman I, Rubio-Cabezas O, Rica I, Locke JM, Maestro
MA, Alshaikh A, Bundak R, del Castillo G, Deeb A, Deiss D, Fernandez
JM, Godbole K, Hussain K, OConnell M, Klupa T, Kolouskova S, Mohsin F,
Perlman K, Sumnik Z, Rial JM, Ugarte E, Vasanthi T, Johnstone K, Flanagan
SE, Martinez R, Castano C, Patch AM, Fernandez-Rebollo E, Raile K, Morgan N,
Harries LW, Castano L, Ellard S, Ferrer J, Perez de Nanclares G, Hattersley AT
2010 Recessive mutations in the INS gene result in neonatal diabetes through
reduced insulin biosynthesis. Proc Natl Acad Sci U S A 107:3105-3110
98. Clocquet AR, Egan JM, Stoffers DA, Muller DC, Wideman L, Chin GA, Clarke WL,
Hanks JB, Habener JF, Elahi D 2000 Impaired insulin secretion and increased
insulin sensitivity in familial Maturity-onset diabetes of the young 4 (insulin
promoter factor 1 gene). Diabetes 49:1856-1864
99. Stoffers DA, Thomas MK, Habener JF 1997 Homeodomain protein idx-1: a
master regulator of pancreas development and insulin gene expression. Trends in
Endocrinology and Metabolism 8:145-151
100. Villareal DT, Robertson H, Bell GI, Patterson BW, Tran H, Wice B, Polonsky KS
2010 TCF7L2 variant rs7903146 affects the risk of type 2 diabetes by modulating
incretin action. Diabetes 59:479-485
101. Florez JC 2008 Newly identified loci highlight beta cell dysfunction as a key cause
of type 2 diabetes: where are the insulin resistance genes? Diabetologia 51:1100-1110
102. Cauchi S, Froguel P 2008 TCF7L2 genetic defect and type 2 diabetes. Curr Diab
Rep 8:149-155
103. Hattersley AT 2007 Prime suspect: the TCF7L2 gene and type 2 diabetes risk. J Clin
Invest 117:2077-2079
104. Heni M, Ketterer C, Thamer C, Herzberg-Schafer SA, Guthoff M, Stefan N,
Machicao F, Staiger H, Fritsche A, Haring HU 2010 Glycemia determines the effect
of type 2 diabetes risk genes on insulin secretion. Diabetes 59:3247-3252
105. da Silva Xavier G, Loder MK, McDonald A, Tarasov AI, Carzaniga R, Kronenberger
K, Barg S, Rutter GA 2009 TCF7L2 regulates late events in insulin secretion from
pancreatic islet beta-cells. Diabetes 58:894-905

New Insights of Islet Biology and the Pathophysiology of Type 2 Diabetes

115

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

106. Shu L, Sauter NS, Schulthess FT, Matveyenko AV, Oberholzer J, Maedler K 2008
Transcription factor 7-like 2 regulates beta-cell survival and function in human
pancreatic islets. Diabetes 57:645-653
107. Lyssenko V, Lupi R, Marchetti P, Del Guerra S, Orho-Melander M, Almgren P,
Sjogren M, Ling C, Eriksson KF, Lethagen AL, Mancarella R, Berglund G, Tuomi T,
Nilsson P, Del Prato S, Groop L 2007 Mechanisms by which common variants in
the TCF7L2 gene increase risk of type 2 diabetes. J Clin Invest 117:2155-2163
108. Gaulton KJ, Nammo T, Pasquali L, Simon JM, Giresi PG, Fogarty MP, Panhuis
TM, Mieczkowski P, Secchi A, Bosco D, Berney T, Montanya E, Mohlke KL, Lieb
JD, Ferrer J 2010 A map of open chromatin in human pancreatic islets. Nat Genet
42:255-259
109. Stitzel ML, Sethupathy P, Pearson DS, Chines PS, Song L, Erdos MR, Welch R,
Parker SC, Boyle AP, Scott LJ, Margulies EH, Boehnke M, Furey TS, Crawford GE,
Collins FS 2010 Global epigenomic analysis of primary human pancreatic islets
provides insights into type 2 diabetes susceptibility loci. Cell Metab 12:443-455
110. Groop L 2010 Open chromatin and diabetes risk. Nat Genet 42:190-192

116

Translational Endocrinology & Metabolism: Type 2 Diabetes Update

The Endocrine Society. Downloaded from press.endocrine.org by [${individualUser.displayName}] on 02 July 2015. at 16:13 For personal use only. No other uses without permission. . All rights reserved.

Das könnte Ihnen auch gefallen