ArticlePDF AvailableLiterature Review

New insights into the architecture of the islet of Langerhans: a focused cross-species assessment

Authors:

Abstract

The human genome project and its search for factors underlying human diseases has fostered a major human research effort. Therefore, unsurprisingly, in recent years we have observed an increasing number of studies on human islet cells, including disease approaches focusing on type 1 and type 2 diabetes. Yet, the field of islet and diabetes research relies on the legacy of rodent-based investigations, which have proven difficult to translate to humans, particularly in type 1 diabetes. Whole islet physiology and pathology may differ between rodents and humans, and thus a comprehensive cross-species as well as species-specific view on islet research is much needed. In this review we summarise the current knowledge of interspecies islet cytoarchitecture, and discuss its potential impact on islet function and future perspectives in islet pathophysiology research.
REVIEW
New insights into the architecture of the islet of Langerhans:
a focused cross-species assessment
Rafael Arrojo e Drigo
1
&Yusuf Ali
1
&Juan Diez
1
&Dinesh Kumar Srinivasan
1
&
Per-Olof Berggren
1,2,3
&Bernhard O. Boehm
1,2,4
Received: 8 March 2015 /Accepted: 26 June 2015/ Published online: 28 July 2015
#Springer-Verlag Berlin Heidelberg 2015
Abstract The human genome project and its search for
factors underlying human diseases has fostered a major
human research effort. Therefore, unsurprisingly, in recent
years we have observed an increasing number of studies on
human islet cells, including disease approaches focusing
on type 1 and type 2 diabetes. Yet, the field of islet and
diabetes research relies on the legacy of rodent-based
investigations, which have proven difficult to translate to
humans, particularly in type 1 diabetes. Whole islet phys-
iology and pathology may differ between rodents and
humans, and thus a comprehensive cross-species as well
as species-specific view on islet research is much needed.
In this review we summarise the current knowledge of
interspecies islet cytoarchitecture, and discuss its potential
impact on islet function and future perspectives in islet
pathophysiology research.
Keywords Cross-species assessment .Intra-islet signalling .
Islet cell architecture .Islets of Langerhans .Non-human
primates .Review
Abbreviations
ACE Anterior chamber of the eye
AMPA α-Amino-3-hydroxy-5-methyl-4-
isoxazolepropionic acid
[Ca
2+
]
i
Intracellular free calcium
CNS Central nervous system
GABA γ-Aminobutyric acid
GLP-1 Glucagon-like peptide-1
IR Insulin receptor
NHP Non-human primate
NMDA N-methyl-D-aspartate
VEGF-A Vascular endothelial growth factor-A
It is the pervading law of all things organic and inor-
ganic, of all things physical and metaphysical, of all
things human and all things superhuman, of all true
manifestations of the head, of the heart, of the soul, that
the life is recognizable in its expression, that form ever
follows function. This is the law.
Louis H. Sullivan (1896) [1]
Introduction
In his 1869 doctoral thesis, now a medical classic, the 22-year-
old German physician Paul Langerhans reported the
Rafael Arrojo e Drigo and Yusuf Ali contributed equally to this work.
*Per-Olof Berggren
Per-Olof.Berggren@ki.se
*Bernhard O. Boehm
Bernhard.boehm@ntu.edu.sg
1
Lee Kong Chian School of Medicine, Nanyang Technological
University, 50 Nanyang Drive, Research Techno Plaza, Level 4,
637 553 Singapore, Singapore
2
Imperial College London, London, UK
3
The Rolf Luft Research Center for Diabetes and Endocrinology,
Karolinska University Hospital L1, Karolinska Institutet,
SE-171 76 Stockholm, Sweden
4
Department of Internal Medicine 1, Ulm University Medical Centre,
Ulm, Germany
Diabetologia (2015) 58:22182228
DOI 10.1007/s00125-015-3699-0
microscopic appearance of scattered small groups of cells
among the acinar glandular cells in the pancreas of the rabbit
[2]. In 1893 Edouard Laguesse named these cell clusters the
islets of Langerhans, and first postulated an endocrine role
for these clusters in the control of blood glucose levels [3].
Successful extractions of insulin and glucagon and the for-
mal experimental proof that these key hormones regulate glu-
cose levels firmly established the islets of Langerhans as key
mini-organs in the diabetes field. Research on the biology and
pathophysiology of islet cells burgeoned as a result of the
emergence of novel technologies and the application of vari-
ous diabetes animal models as a surrogate for studies on hu-
man islets from healthy and diabetic individuals [414].
Preclinical research on rodent models offers several logis-
tical, methodological and financial advantages. But this
approach may fall short in terms of reflecting critical aspects
of human islet pathophysiology [15,16]. Because islets of
non-human primate (NHP) or human origin are increasingly
becoming available (albeit in limited numbers), there is an
important and creeping increase in the number of studies on
primate islets. Although still in their infancy, such studies have
highlighted key structurefunction differences between
rodents and NHP or human islets, with evidence pointing
towards a high degree of similarity between NHP and human
islets [8,14,17,18]. Moreover, mouse models may not reflect
the complexity of human pathophysiology [19], such as islet
amyloid deposition, underscoring the need for a better animal
model (i.e. NHP) that truly mirrors human islet biology in
health and disease [20].
In this review we will summarise the results on islet archi-
tecture and structurefunction relationships between rodents,
NHPs and humans. In addition, we will briefly highlight the
plasticity of islet cell architecture observed under diabetic
conditions (reviewed in detail in [21]). As noted earlier,
assumptions on human islet function and regulation based
on rodent islets are not always true. Therefore, we would like
to propose, based on data on interspecies islet cytoarchitecture
and functional differences, a paradigm shift whereby investi-
gations of the NHP islet can serve as a successful diabetes
translational platform.
Cellular architecture and composition of islets
of Langerhans is heterogeneous
Islet morphology has been extensively studied using histolo-
gy, immunohistology and electron microscopy. In addition,
different in vivo and in vitro model systems have been used
to address islet cell function [22,23]. Islets contain a variety of
cell types, the most abundant being the insulin-producing
beta cells, the glucagon-producing alpha cells and the
somatostatin-producing delta cells [24](Fig.1). Furthermore,
ghrelin-producing epsilon cells and pancreatic polypeptide
(PP) cells have been described in the mammalian islet, albeit
in smaller numbers [25,26]. Besides hormone-producing
cells, the islet also houses immune cells, autonomic nerve
system endings (reviewed in detail in [27]) and is served by
an abundant vascular network [8,28,29].
Together, the different cells are architecturally arranged to
allow for a fine-tuned functional interrelationship between all
islet cells, possibly orchestrated by autocrine, paracrine and
endocrine signalling cascades, like a true mini-organ (Fig. 2).
With its unique cell organisation, mammalian islets success-
fully accomplish their ultimate physiological role, to adapt to
constant changes in metabolic demands and to maintain
optimal circulating glucose levels throughout life.
In the following section we will highlight the differences
that separate rodent from primate islet physiology.
Rodents Small animal research has been the cornerstone of
islet research for over a century, pioneered by Langerhans
himself in his description of the rabbit islet [2]. Today, mouse
and rat islets are widely studied, with consensus that these
islets are mainly comprised of an insulin-producing beta cell
core surrounded by a mantle of intermingled alpha and delta
cells (Fig. 1).
The islet vasculature is responsible for supplying the islet
cells not only with oxygen, but also with key metabolic cues
(i.e. glucose) and other nutrients, incretin hormones such as
glucagon-like peptide-1 (GLP-1) and cytokines [29,30]. This
vast vascular network develops early in islet embryogenesis,
providing essential developmental signals [31], and is driven
primarily by the secretion of vascular endothelial growth
factor-A (VEGF-A) from beta cells [32,33]. The islet vascular
bed consistsof both large and small fenestrated capillaries [11,
29]. The islet blood flow arrives from peripheral vascular net-
works, where the blood flow moves from the beta cell-rich
inner core to the outer alpha and delta cells [29,34]. The islet
blood flow pattern and speed has been suggested to be depen-
dent on blood glucose levels, with hyperglycaemia leading to
better islet perfusion with faster flow speeds compared with
states of hypoglycaemia [7]. Recent evidence suggests a
degree of heterogeneity of vascularisation within islets in a
single pancreas [35,36]. Such differences result in distinct
islet subpopulations with significant functional implications,
with highly perfused islets displaying an increased beta cell
proliferation rate and enhanced function [36]. Whether such a
phenotype is found in human islets remains unclear.
Furthermore, islet vasculature has been suggested to play a
crucial role in maintaining islet function in ageing [37].
The location of beta cells within the core of the rodent
islet also has important and direct functional consequences.
The biological activity between the majority of rodent beta cells
within any given islet, commonly measured by changes in
intracellular free calcium concentration ([Ca
2+
]
i
), is highly
synchronised, creating an integrated functional syncytium
Diabetologia (2015) 58:22182228 2219
[38]. This synchronisation is dependent on gap junctions made
of Connexin-36 channels between different beta cells [39].
Humans and NHPs Although human islets have been inten-
sively studied over the last decade, their cellular arrangement
is still a matter of debate, largely because of different experi-
mental methodologies. Earlier morphological analysis of
different regions of human and NHP pancreases showed that
the different islet cell types are intermingled, with beta and
alpha cells present in similar numbers with a higher degree of
heterotypic islet cell contacts [14,18]. Thus, human islets
have been perceived to be characterised by an intermingled
majority of beta cells, followed by alpha and delta cells [14,
18,24](Fig.1). However, other groups have provided
evidence of a non-random arrangement of human beta cells
within an islet. They have observed that despite the higher
percentages of alpha cells within the human islet, beta cells
still prefer to remain in contact with one another, in a similar
way to rodent beta cells [4042]. With homotypic cell contacts
predominant, paracrine influences within human islets may be
limited. In a separate study in which three-dimensional recon-
structions of islets were created from serial human pancreatic
sections, the islets were reported to consist of a beta cell core
with a surrounding mantle of alpha cells and vessels that do
not penetrate the islet [43]. However, in the same paper, the
authors go on to suggest that, similar to rodents, human islet
cells are purposely arranged in a complex trilaminar epithelial
plate fold that favours heterologous contacts and paracrine
signalling between islet cells [43]. These varied concepts of
human beta cell distribution need to be resolved to allow a
thorough appreciation of the importance of paracrine interac-
tions within an intact human islet.
Regardless of heterotypic cell arrangements, human islet
cell composition is set early in human life, stemming from a
high proliferative phase (mainly beta cells) in the first 2 years
of life followed by a life-long low-proliferative state [44,45].
Strikingly, unlike rodents, not much is known about the
human (intra)islet vasculature anatomy and flow rate [37,
46], with the exception of early reports that the perfusion of
human and NHP islets is similar to that of rodent islets [47,
48]. We have shown that the vasculature of human islets
becomes inflamed and fibrotic with ageing, which is also
observed in islets from type 2 diabetic patients [37]. In addi-
tion, a unique double basement membrane between islet cells
and vascular cells has been observed in human islets [49], the
function of which is as yet unknown.
The functional consequences of the unique heterogeneous
cellular arrangement of the human islet are even less clear.
Some functional differences between rodent, NHP and human
islets can be attributed to differences in islet cytoarchitecture
Sympathetic
fibres
Blood
vessels
Parasympathetic
fibres
Primate
Rodent
~80%
~5%
~65%
~30%
~5%
Islet cell composition
Alpha cell
Beta cell
Delta cell
~15%
Fig. 1 The cytoarchitecture of islets of Langerhans of rodents and pri-
mates is different. In rodents, islets are composed of beta cells located in
the core of the islet, which are surrounded by a mantle of alpha and delta
cells. Sympathetic fibres surround the islet, while parasympathetic fibres
are distributed throughout the core of the islet, supplying acetylcholine to
beta cells. In contrast, NHP and human islets have a heterogeneous dis-
tribution of beta, alpha and delta cells. Here, sympathetic fibres target the
islet vasculature and parasympathetic fibres are rare, with alpha cells in
charge of the acetylcholine supply to beta and delta cells
2220 Diabetologia (2015) 58:22182228
[14]. By monitoring changes in islet [Ca
2+
]
i
, the authors dem-
onstrate that primate islet cells are activated by changes in low
glucose concentrations (from 3 mmol/l to 1 mmol/l), whereas
mouse islets are not [14]. Moreover, prolonged exposure
of whole mouse islets to 11 mmol/l glucose triggers a
synchronised oscillatory [Ca
2+
]
i
behaviour, an effect that is
absent in primate islets but has been seen in isolated primate
beta cells. [14]. However, such [Ca
2+
]
i
oscillatory behaviour
seen in isolated primate beta cells needs to be verified in an
in vivo setting.
Intra-islet signalling is important for human islet function
Rodent islet function relies on homotypic cellular communi-
cations between beta cells,configuring a functional syncytium
with coordinated islet activity [39,50]. On the other hand, the
cytoarchitecture of the human islet, rich in heterotypic islet
cell contacts, suggests that intra-islet paracrine signalling is
perhaps less straightforward [14,24,43](Fig.2). Although
reports indicate a rodent-like perfusion pattern of the human
islet [47], lending weight to the concept of limited intra-islet
paracrine signalling, the trilaminar-fold hypothesis coupled
with regulated (and directed) blood flow by vascular sphinc-
ters within the human islet suggest that paracrine signalling
may yet be important for islet function [8,51].
Intra-islet communication between different islet cells can
be established by the secretion of signalling molecules into
islet microvasculature or within the islets interstitial space.
Once these signalling molecules find their respective receptors
on target cells they trigger complex autocrine or paracrine
regulatory loops. In addition to hormones, islet cells secrete
an array of signalling molecules, including acetylcholine
[17,52], glutamate [53], ATP [54,55]andγ-aminobutyric
acid (GABA) [5658], all of which are important for islet cell
function (Fig. 2).
Acetylcholine Acetylcholine, the first neurotransmitter to be
discovered, is an important modulator of beta cell activity and
insulin secretion in rodents [59]aswellasinhumans[17].
Although acetylcholine has a similar function in the islets of
Insulin
Insulin
ATP
Insulin gene
expression
Insulin
secretion
Glucagon gene
expression
Glucagon
secretion
Ach
Glutamate
Glucagon
Ach
Somatostatin
secretion
Somatostatin
secretion
llec atleDllec ahplA
Beta cell IR-A/-B
P2X/Y receptor
AMPA/NMDA
receptor
Muscarinic
receptors 1, 3, 5
Insulin
Glucagon/glutamate
Somatostatin
Acetylcholine
SSTR 1-5
Glucagon receptor
GABA
GABAA/B
receptor
Upregulation Downregulation
GABA
Fig. 2 Paracrine and autocrine signalling networks in mammalian islet of
Langerhans. In response to an increase in blood glucose levels, beta cells
secrete insulin. In turn, insulin binds to insulin receptors A and B (IR-A
and IR-B) in beta and alpha cells, regulating insulin gene expression and
insulin and glucagon exocytosis. ATP is secreted together with insulin
and acts on purinergic P2X/Y receptors in beta cells. Beta cells also
secrete GABA, which acts on beta cells in an autocrine manner through
GABA
A
and GABA
B
receptors. When blood glucose levels are low,
alpha cells secrete glucagon, glutamate and acetylcholine
a
. Glucagon
binds to glucagon receptors in beta and alpha cells, regulating insulin
secretion and glucagon gene expression. Similarly, glutamate binds to
AMPA and NMDA receptors in beta and alpha cells, regulating insulin
and glucagon exocytosis. Acetylcholine binds to muscarinic receptors in
beta and delta cells, regulating insulin and somatostatin secretion. At both
high and low blood glucose levels the delta cells secrete somatostatin,
which binds to somatostatin receptors (SSTRs) in beta and alpha cells,
inhibiting insulin and glucagon secretion.
a
In rodent islets, acetylcholine
is secreted by parasympathetic fibres that permeate the beta cell core
(see Fig. 1)
Diabetologia (2015) 58:22182228 2221
both, the source of acetylcholine is different [17,59]. In
rodents, acetylcholine isdelivered through autonomic nervous
system varicosities distributed throughout the islet [8], where-
as in humans islets it is secreted by alpha cells and influences
the activityof neighbouring beta and delta cells [8,17,52,60]
(Fig. 2).
Nonetheless, in both human and rodent islets acetylcholine
secretion is triggered by periods of low circulating levels of
glucose, and serves to sensitise beta cells to future rises in
blood glucose concentration. The shift from neural to intra-
islet acetylcholine delivery by alpha cells indicates that human
islets are better able to respond quickly to dynamic shifts in
blood glucose levels, bypassing neural networks. This may
explain why type 1 diabetic patients can show a reduced
dependency on exogenous insulin application as quickly as
20 days after islet transplantation [61], well before complete
islet innervation is established.
ATP ATP is one of the most important signalling molecules in
the body [62]. Besides its well-established role in glucose-
stimulated insulin secretion [63], ATP may also act as an
extracellular signalling molecule within islets. Although
ATP is found within insulin granules [9], its secretion seems
to precede insulin exocytosis [64,65].
To date, most of the data available on ATP function in islets
is from rodent models. ATP is reported to act via P2Y recep-
tors [66], either stimulating [67] or inhibiting [68] insulin
secretion (Fig. 2). Recent studies in human and NHP islets
show that ATP similarly may act as an autocrine signalling
molecule, taking part in an important positive-feedback loop
in active beta cells (Fig. 2). Although ATP is released at low
glucose concentrations [54], glucose-stimulated ATP has been
suggested to act via ionotropic purinergic P2X
3
receptors in
the membrane of active beta cells, resulting in P2X
3
-mediated
Na
+
Ca
2+
influx and further depolarisation of beta cells and
enhanced insulin secretion [54]. However, another study pro-
duced contradictory data, with P2X isoforms observed to be
less relevant than P2Y
1
receptors in mediating the positive
autocrine role of ATP in human islets [69]. Based on these
intriguing findings, further investigations on the role of intra-
islet ATP signalling are warranted, especially given the
unknown sources of secreted ATP (besides the beta cell) and
the conservation of this autocrine mechanism among different
islet cells.
GABA Since its discovery in the 1950s [70], GABA has been
characterised as a key inhibitory neurotransmitter in the mam-
malian central nervous system (CNS), acting via ionotropic
GABA
A
and metabotropic GABA
B
receptors [71]. In neu-
rons, binding to the various receptor types leads to target cell
hyperpolarisation (i.e. electrical inhibition), either through the
opening of chloride (Cl
) channels and Cl
influx (GABA
A
),
or by the opening of K
+
channels via second-messenger
pathways (GABA
B
)[71]. Again, in islets the majority of data
on the function of GABA originates from rodent models.
Studies have shown that GABA molecules are found in the
cytoplasm of beta cells, both in insulin granules [9] and in
small core vesicles [72]. Acting via GABA
A
receptors,
GABA mediates the glucose-dependent inhibition of gluca-
gon secretion in alpha cells [73] and triggers beta cell prolife-
ration and survival in an autocrine loop [56,74].
Human islet cells contain high levels of the GABA-
synthesising enzyme glutamate decarboxylase 65 (GAD65,
also known as GAD2) [75]. Besides inducing beta cell prolif-
eration [56], GABA seems to work both as an inhibitory and
as an excitatory molecule. In the presence of physiological
glucose concentrations, GABA depolarises beta cells and
stimulates glucose-induced insulin secretion [76], whereas
the role of GABA in glucagon secretion by alpha cells is
controversial [57,76]. In addition, high levels of GABA can
depolarise delta cells, suggesting a role for GABA-mediated
regulation of somatostatin release [76].
As all three main islet endocrine cells contain GABA [76],
an exciting challenge will be to determine whether GABA
secretion from different islet cells is synchronised in vivo,
especially given the apparent antagonistic actions of GABA
on islet cell function.
Glutamate Glutamate is one of the key excitatory neurotrans-
mitters in the CNS. Glutamate acts via ionotropic kainite-type
α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
(AMPA) or N-methyl-D-aspartate (NMDA) receptors [77].
Similar to acetylcholine, GABA, ATP and glutamate,
glutamate-synthesising enzymes and glutamate receptors are
found in the three main endocrine cells in rodent islets [78,
79]. According to one school of thought, in NHPs and humans
[53], intra-islet glutamate is provided by alpha cells, and gluta-
mate is compartmentalised inside glucagon-containing gran-
ules and secreted together with glucagon [80](Fig.2). Once
secreted, glutamate acts mainly in an autocrine fashion, driving
a positive-feedback loop that depolarises the active alpha cell
and upregulates glucagon release (Fig. 2). While this autocrine
function is conserved in rodents and humans, glutamate action
in rodents seems to be mediated by both AMPA and NMDA
receptors [78], while in humans only AMPA receptors are
involved [53]. This scheme has been challenged by the authors
of a recent study using mouse islets who propose a different
model [81], in which glutamate is neither secreted together with
glucagon nor does it modulate glucagon secretion. Rather,
inhibition of glutamate secretion increases cellular pools of
both glutamate and α-ketoglutarate, which are suggested to
enhance somatostatin secretion release from delta cells via a
new AMPA receptor variant [79]. These results highlight the
complexity of intra-islet signalling, and underline the need for
similar studies to be conducted with human islets.
2222 Diabetologia (2015) 58:22182228
Insulin Since the first description of insulin [82], its action
and anti-diabetic potential have been studied for almost a
century. Insulin is critical for proper control of blood glucose
levels and acts in almost all tissues, such as brain, liver, muscle,
adipose tissue and pancreatic islets. Insulin action is mediated
by two different isoforms of the insulin receptor (namely IR-A
and IR-B; Fig. 2). Spatial differences between IR-A and IR-B
on cell membranes may explain their differences in intracellular
signalling networks [83]. Mice lacking IR expression specifi-
cally in beta cells (the βIRKO mouse model) develop a
hyperinsulinaemic state and unlike wild-type mice do not show
acute glucose-induced insulin secretion [84]. Moreover, insulin
released in response to glucose is important for forkhead box
protein O1 (FOXO1) transcriptional activity and K
ATP
channel
opening in beta cells [85,86]. In addition, deletion of IR in
alpha cells (the αIRKO mouse model) leads to hyperglycaemia,
glucose intolerance and hyperglucagonaemia, indicating that
insulin acts also at the alpha cell level to suppress glucagon
secretion [87].
In humans, the results available on intra-islet insulin action
are conflicting. Insulin has been described both as a positive
and a negative regulator of islet cell function. Wu et al [88]
reported that insulin acts via the phosphoinositide 3-kinase
pathway, upregulates pancreatic and duodenal homeobox pro-
tein 1 (PDX-1) transcriptional activity and induces insulin
gene expression. In contrast, Persaud et al noted a negative
effect of insulin on beta cell insulin secretion [89]. Similarly,
human pancreases perfused with low glucose and anti-insulin
antibodies have impaired glucagon secretion [90]. Given the
inconsistency of the experimental data, further studies are
necessary to determine the effects of intra-islet insulin.
Glucagon Glucagon is the key hormone in mammals that
maintains glucose availability during fasting periods. Acting
primarily on the liver, glucagon stimulates hepatic glucose
production and excursion [91]. The systemic role of glucagon
is clearly demonstrated in rodents, where deletion of the glu-
cagon receptor leads to low blood glucose levels,
hyperglucagonaemia and improved glucose tolerance [92].
Glucagon signalling within the islet is not well understood,
mainly due to a potential crosstalk with the pre-proglucagon
incretin pathway [30,93]. Glucagon receptors are found in
both rodent and human islets and in all three major islet cell
types [94,95]. There is consensus that glucagon causes intra-
cellular cAMP levels to rise, which stimulates beta cell
glucose-induced insulin secretion in both rodents and humans
[92,94,96]. This insulinotropic effect of glucagon is also
observed in isolated rodent islets, where deletion or inhibition
of the glucagon receptor blocks insulin secretion and impairs
glucose oxidation [94,97]. In perfused human pancreas, anti-
body neutralisation of glucagon under both low and high
glucose conditions increases insulin secretion [90], contradic-
ting the above data from isolated human islets [94](Fig.2).
Furthermore, glucagon also plays a positive autocrine role by
binding to glucagon receptors on alpha cells to enhance its
own expression [98].
Somatostatin The peptide hormone somatostatin is secreted by
nerve cells in different regions of the brain, by cells in the diges-
tive system and by pancreatic islet delta cells [99](Fig.2).
Somatostatin secreted by gastrointestinal cells is the main source
of circulating somatostatin [99], whereas delta cell-derived
somatostatin plays a key role in intra-islet cell signalling [100]
(Fig. 2). In both rodents and humans, somatostatin inhibits alpha
and beta cell activity, repressing glucagon and insulin secretion
[90,100]. The actions of somatostatin are mediated by five
different isoforms of the somatostatin receptor (SSTR), found
throughout the islet [101,102]. Deletion of the gene encoding
somatostatin in mice leads to a hyperinsulinaemic and
hyperglucagonaemic state, but, surprisingly, without major
whole body metabolic aberrations [100]. Accordingly, somato-
statin neutralisation in perfused human pancreas leads to higher
glucagon and insulin secretion [90].
Recent data indicate that human delta cells are influenced
by alpha and beta cells, thus being an intricate part of a com-
plex paracrine islet cell signalling network [52].
Islet structurefunction and diabetes
Structural changes in pancreatic islets have been observed in
both type 1 and type 2 diabetes (summarised in Table1). Most
of the studies show that besides the loss in beta cell mass in
type 1 diabetes, both rodent and human/NHP islets display an
increase in alpha and delta cell mass and alpha cell prolifera-
tion [6,103105]. Surprisingly, studies have suggested the
presence of glucose-responsive beta cells in patients with long
standing type 1 diabetes [106,107].
In rodent modelsof type 2 diabetes, a large increase in islet
volume/mass is commonly observed, largely as a result of
increased beta cell proliferation and vascular density
[108113]. This massive increase in beta cell mass is attribut-
ed to an intra-islet, rather than systemic, insulin resistance [84,
114]. In type 2 diabetic humans and NHP models, a more
heterogeneous scenario is observed. Studies have described
a loss in beta cell mass coupled to an increase in amyloid
(IAPP) deposition and alpha cell proliferation (Table 1). We
refer to a recent review article describing beta cell pathophys-
iology in type 2 diabetes [115], describing beta cell failure as a
central mechanism in both type 1 and type 2 diabetes.
Interestingly, in non-diabetic humans, insulin resistance has
been reported to contribute to increased beta cell number
and islet size [116]. Recently, in a mouse model, beta cell
dedifferentiation rather than accelerated death has been sug-
gested as a key driver of beta cell failure [117,118]. However,
at this stage, no direct histopathological evidence has been
Diabetologia (2015) 58:22182228 2223
found in human type 2 diabetes to support this observation
besides the well-known imbalance observed in the peripheral
blood between alpha and beta cell hormone levels [12].
Nevertheless,todatewearelimitedtocross-sectional
studies on highly selected patient cohorts (Table 1). In
view of the fact that the data from the UKPDS indicated
a constant loss of beta cell function over time [119], it is
imperative to investigate, longitudinally, the natural history
of human islet dysfunction from pathological onset to pro-
gressive mini-organ failure. Only this approach will allow
the dissection the molecular mechanisms responsible for
the morphological changes of pancreatic islets in the
different metabolic phenotypes [119].
Conclusion
In this review we have summarised key interspecies differ-
ences in the islet cytoarchitecture of rodents, NHPs and
humans (Fig. 1). Such differences might explain disparities
observed in the intra-islet networks regulating islet physiology
and pathophysiology between mice and humans (Fig. 2). In
Tabl e 1 Islet cell phenotype in type 1 and type 2 diabetes in rodents, NHPs and humans
Environment Islet cell changes Species Reference
T1DM Islet endothelial cell density Mouse [122,123]
T1DM Alpha and delta cell mass Mouse [103,104]
T1DM Beta cell mass
Alpha and delta cell mass
Mouse [124]
T1DM Islet alpha cell mass NHP [105]
T1DM Alpha, beta cell proliferation Human [6]
T2DM Beta cell area Mouse [125]
T2DM Pericyte hypertrophy and capillary hypertension db/db mouse [110]
T2DM Total islet blood flow
When normalised to islet content
ob/ob mouse [108]
T2DM Intra-islet vessel diameter
With leptin treatment
ob/ob mouse [109]
T2DM Beta cell mass
Islet organisation with alpha cells in the core
IGF-II TG mouse [126]
T2DM 1st phase insulin secretion,
Alpha:beta cell ratio
Glut2 KO mouse [127]
T2DM Beta cell mass Mouse [113]
T2DM Blood vessel diameter Mouse [111]
T2DM High MW IGF-II
Starfish-shaped islets
GK rat [128]
DIO Beta cell mass Rat [112]
DIOIslet vascularisation Mouse [129]
T2DM Islet amyloid, beta cell mass NHP [5]
T2DM Islet amyloid deposition NHP [130]
T2DM Alpha cell volume NHP [131]
T2DM Large islet alpha:beta cell ratio NHP [13]
T2DM Amyloid deposition Human [132]
T2DM Beta cell mass Human [133]
T2DM Large islet alpha:beta cell ratio
IAPP-positive cells
Human [134]
T2DM Beta cell neogenesis Human [135]
T2DM Beta cell mass Human [4]
T2DM Large islet beta cell mass in head-region of pancreas Human [26]
T2DM Beta cell mass in body and tail region of pancreas Human [136]
T2DM Marginal in beta cells Human [137]
T2DM Intra-islet CD45
+
leucocytes Human [138]
T2DM Intra-islet macrophages and vessel fibrosis Human [37]
DIO, Diet-induced obesity; IAPP, islet amyloid polypeptide; IGF-II, insulin-like growth factor II; MW, molecular weight; T1DM, type 1 diabetes
mellitus; T2DM, type 2 diabetes mellitus
2224 Diabetologia (2015) 58:22182228
view of the evidence provided, we believe that islet and
diabetes research should consider differences in islet
cytoarchitecture when conducting and interpreting results
from one given species, especially rodents. Since human islet
composition is heterogeneous and significantly different from
that of rodents but similar to that of NHPs, the NHP may serve
as a more appropriate animal model for studying human islet
pathophysiology from a translational perspective.
The future of islet research to address existing knowledge
gaps in islet physiology and pathophysiology
Besides understanding the main differences in microanatomy
betweentherodentandthehumanislet,itisessentialtostudy
islet physiology and pathophysiology in an in vivo setting.
Only by using an in vivo platform can researchers truly appre-
ciate an integrated view of islet biology. The islet micro-
environment is highly dynamic, and cross-sectional studies
are limited to snapshots of changes in islet cytoarchitecture
andfunction(Fig.1and Table 1), ignoring the longitudinal
dimension that takes into account the natural history of such
changes. Moreover, isolated islets lack functionally important
nerve fibres and blood vessels, and are thus not under the
influence of crucial peripheral and central regulatory factors.
Therefore, as long as the majority of islet research is conducted
ex vivo, whether in isolated islets or pancreatic slices, our
knowledge of islet pathophysiology will remain incomplete
and lacking the translation from bench to bedside. Over the
years islet researchers have developed various in vivo islet
imaging techniques to provide a glimpse of islet morphology,
function, innervation and vascularisation (reviewed in [22,23]).
The next step in the evolution of these techniques would
involve in vivo islet imaging in the NHP, as these islets rep-
resent a close surrogate of human islet physiology. An increas-
ing number of publications suggest that islets transplanted into
the mouse anterior chamber of the eye (ACE) mirror those of
the endogenous pancreas, both structurally and functionally
[11,37,109,120,121]. Whether or not this holds true during
diabetes pathogenesis (e.g. in states of glucolipotoxicity and
of an immune attack) remains to be elucidated. It is notewor-
thy that studies on the autologous transplantation of NHP
islets following partial pancreatectomy into the ACE are
already under way. If successful, these studies may offer a
quantum leap in understanding human-like islet plasticity,
thus improving translational islet research.
Acknowledgements This work was supported by grants from the Sin-
gapore Ministry of Education, Academic Research Fund Tier 1(2014-T1-
001-149toY.A.)andbyLeeKongChianSchoolofMedicine,Nanyang
Technological University, Singapore (NTU) start-up grants (separately for
Y.A., P.O.B. and B.O.B.). The Lee Kong Chian School of Medicine is a
partnership between Nanyang Technological University, Singapore (NTU)
and Imperial College London. Due to space constraints we have not been
able to provide a comprehensive presentation of the theme and have there-
by disregarded many important contributions to the field of our colleagues.
Duality of interest POB is co-founder and CEO of Biocrine AB, a
diabetes-related biotech company.
Contribution statement All authors were responsible for drafting the
article and revising it critically. All authors approved the version to be
published.
References
1. Sullivan LH (1896) The tall office building artistically considered.
Lippincotts Mag 57:403409
2. Langerhans P (1869) Beitraege zur mikroskopischen Anatomie
der Bauchspeicheldruese. Friedrich-Wilhelm-Universitaet zu
Berlin, Inaugural Dissertation (in German)
3. Laguesse É (1893) Sur la formation des ilots de Langerhans dans
le pancreas. Compt Rend Soc Biol 5:819820 (in French)
4. 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 52:102110
5. de Koning EJ, Bodkin NL, Hansen BC, Clark A (1993) Diabetes
mellitus in Macaca mulatta monkeys is characterised by islet am-
yloidosis and reduction in beta-cell population. Diabetologia 36:
378384
6. Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG
(2010) Evidence of increased islet cell proliferation in patients
with recent-onset type 1 diabetes. Diabetologia 53:20202028
7. 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:E807E814
8. Rodriguez-Diaz R, Abdulreda MH, Formoso AL et al (2011)
Innervation patterns of autonomic axons in the human endocrine
pancreas. Cell Metab 14:4554
9. Suckale J, Solimena M (2010) The insulin secretory granule as a
signaling hub. Trends Endocrinol Metab 21:599609
10. Rutter GA, Hodson DJ (2013) Minireview: intraislet regulation of
insulin secretion in humans. Mol Endocrinol 27:19841995
11. Speier S, Nyqvist D, Cabrera O et al (2008) Noninvasive in vivo
imaging of pancreatic islet cell biology. Nat Med 14:574578
12. Unger RH, Orci L (2010) Paracrinology of islets and the
paracrinopathy of diabetes. Proc Natl Acad Sci U S A 107:
1600916012
13. Fiori JL, Shin YK, Kim Wet al (2013) Resveratrol prevents beta-
cell dedifferentiation in nonhuman primates given a high-fat/high-
sugar diet. Diabetes 62:35003513
14. Cabrera O, Berman DM, Kenyon NS, Ricordi C, Berggren
PO, Caicedo A (2006) The unique cytoarchitecture of hu-
man pancreatic islets has implications for islet cell function.
Proc Natl Acad Sci U S A 103:23342339
15. Stewart AF (2014) Betatrophin versus bitter-trophin and the ele-
phant in the room: time for a new normal in beta-cell regeneration
research. Diabetes 63:11981199
16. Reed JC, Herold KC (2015) Thinking bedside at the bench: the
NOD mouse model of T1DM. Nat Rev Endocrinol 11:308314
17. Rodriguez-Diaz R, Dando R, Jacques-Silva MC et al (2011)
Alpha cells secrete acetylcholine as a non-neuronal paracrine sig-
nal priming beta cell function in humans. Nat Med 17:888892
18. Kim A, Miller K, Jo J, Kilimnik G, Wojcik P, Hara M (2009) Islet
architecture: a comparative study. Islets 1:129136
Diabetologia (2015) 58:22182228 2225
19. Seok J, Warren HS, Cuenca AG et al (2013) Genomic re-
sponses in mouse models poorly mimic human inflammatory
diseases. Proc Natl Acad Sci U S A 110:35073512
20. Westermark P, Andersson A, Westermark GT (2011) Islet amyloid
polypeptide, islet amyloid, and diabetes mellitus. Physiol Rev 91:
795826
21. Mezza T, Kulkarni RN (2014) The regulation of pre- and post-
maturational plasticity of mammalian islet cell mass. Diabetologia
57:12911303
22. Wang P, Medarova Z, Moore A (2011) Molecular imaging: a
promising tool to monitor islet transplantation. J Transl 2011:
202915
23. Leibiger IB, Caicedo A, Berggren PO (2012) Non-invasive
in vivo imaging of pancreatic beta-cell function and survival a
perspective. Acta Physiol 204:178185
24. Brissova M, Fowler MJ, Nicholson WE et al (2005)
Assessment of human pancreatic islet architecture and
composition by laser scanning confocal microscopy.
J Histochem Cytochem: Off J Histochem Soc 53:10871097
25. Andralojc KM, Mercalli A, Nowak KW et al (2009) Ghrelin-
producing epsilon cells in the developing and adult human pan-
creas. Diabetologia 52:486493
26. Wang X, Zielinski MC, Misawa R et al (2013) Quantitative
analysis of pancreatic polypeptide cell distribution in the human
pancreas. PLoS One 8:e55501
27. Rodriguez-Diaz R, Caicedo A (2014) Neural control of the endo-
crine pancreas. Best Pract Res Clin Endocrinol Metab 28:745756
28. Westwell-Roper CY, Ehses JA, Verchere CB (2014) Resident
macrophages mediate islet amyloid polypeptide-induced islet IL-
1βproduction and β-cell dysfunction. Diabetes 63:16981711
29. Bonner-Weir S, Orci L (1982) New perspectives on the microvas-
culature of the islets of Langerhans in the rat. Diabetes 31:883
889
30. Donath MY, Burcelin R (2013) GLP-1 effects on islets: hormonal,
neuronal, or paracrine? Diabetes Care 36(Suppl 2):S145S148
31. Lammert E, Cleaver O, Melton D (2001) Induction of pancreatic
differentiation by signals from blood vessels. Science 294:564
567
32. Lammert E, Gu G, McLaughlin M et al (2003) Role of VEGF-A in
vascularization of pancreatic islets. Curr Biol: CB 13:10701074
33. Gorden DL, Mandriota SJ, Montesano R, Orci L, Pepper MS
(1997) Vascular endothelial growth factor is increased in
devascularized rat islets of Langerhans in vitro. Transplantation
63:436443
34. Samols E, Stagner JI, Ewart RB, Marks V (1988) The order of islet
microvascular cellular perfusion is BAD in the perfused rat
pancreas. J Clin Invest 82:350353
35. Olsson R, Carlsson PO (2011) A low-oxygenated subpopulation
of pancreatic islets constitutes a functional reserve of endocrine
cells. Diabetes 60:20682075
36. Lau J, Svensson J, Grapensparr L, Johansson A, Carlsson PO
(2012) Superior beta cell proliferation, function and gene expres-
sion in a subpopulation of rat islets identified by high blood per-
fusion. Diabetologia 55:13901399
37. Almaca J, Molina J, Arrojo EDR et al (2014) Young capillary ves-
sels rejuvenate aged pancreatic islets. Proc Natl Acad Sci U S A 111:
1761217617
38. Meissner HP (1976) Electrophysiological evidence for coupling
between beta cells of pancreatic islets. Nature 262:502504
39. Ravier MA, Guldenagel M, Charollais A et al (2005) Loss of
connexin36 channels alters beta-cell coupling, islet synchroniza-
tion of glucose-induced Ca
2+
and insulin oscillations, and basal
insulin release. Diabetes 54:17981807
40. Kilimnik G, Jo J, Periwal V, Zielinski MC, Hara M (2012)
Quantification of islet size and architecture. Islets 4:167172
41. Wang X, Misawa R, Zielinski MC et al (2013) Regional differences
in islet distribution in the human pancreas preferential beta-cell
loss in the head region in patients with type 2 diabetes. PLoS One 8:
e67454
42. Bonner-Weir S, Sullivan BA, Weir GC (2015) Human islet
morphology revisited: human and rodent islets are not so
different after all. J Histochem Cytochem. doi:10.1369/
0022155415570969
43. Bosco D, Armanet M, Morel P et al (2010) Unique arrangement of
alpha- and beta-cells in human islets of Langerhans. Diabetes 59:
12021210
44. Meier JJ, Kohler CU, Alkhatib B et al (2010) Beta-cell
development and turnover during prenatal life in humans.
Eur J Endocrinol Eur Fed Endocr Soc 162:559568
45. Gregg BE, Moore PC, Demozay D et al (2012) Formation of a
human beta-cell population within pancreatic islets is set early in
life. J Clin Endocrinol Metab 97:31973206
46. Int Veld P, Lammert E (2015) The dark side of islet vasculature.
Diabetologia 58:46
47. Stagner JI, Samols E (1992) The vascular order of islet cellular
perfusion in the human pancreas. Diabetes 41:9397
48. Stagner JI, Samols E, Koerker DJ, Goodner CJ (1992) Perfusion
with anti-insulin gamma globulin indicates a B to A to D cellular
perfusion sequence in the pancreas of the rhesus monkey, Macaca
mulatta. Pancreas 7:2629
49. Virtanen I, Banerjee M, Palgi J et al (2008) Blood vessels of
human islets of Langerhans are surrounded by a double basement
membrane. Diabetologia 51:11811191
50. Stozer A, Gosak M, Dolensek J et al (2013) Functional connec-
tivity in islets of Langerhans from mouse pancreas tissue slices.
PLoS Comput Biol 9:e1002923
51. Schaeffer M, Hodson DJ, Lafont C, Mollard P (2011) Endocrine
cells and blood vessels work in tandem to generate hormone
pulses. J Mol Endocrinol 47:R59R66
52. Molina J, Rodriguez-Diaz R, Fachado A, Jacques-Silva MC,
Berggren PO, Caicedo A (2014) Control of insulin secretion by
cholinergic signaling in the human pancreatic islet. Diabetes 63:
27142726
53. Cabrera O, Jacques-Silva MC, Speier S et al (2008) Glutamate is a
positive autocrine signal for glucagon release. Cell Metab 7:545
554
54. Jacques-Silva MC, Correa-Medina M, Cabrera O et al (2010)
ATP-g a t e d P 2X
3
receptors constitute a positive autocrine sig-
nal for insulin release in the human pancreatic beta cell.
Proc Natl Acad Sci U S A 107:64656470
55. Fernandez-Alvarez J, Hillaire-Buys D, Loubatières-Mariani MM,
Gomis R, Petit P (2001) P2 receptor agonists stimulate insulin
release from human pancreatic islets. Pancreas 22:6971
56. Purwana I, Zheng J, Li X et al (2014) GABA promotes human
beta-cell proliferation and modulates glucose homeostasis.
Diabetes 63:41974205
57. Taneera J, Jin Z, Jin Y et al (2012) γ-Aminobutyric acid (GABA)
signalling in human pancreatic islets is altered in type 2 diabetes.
Diabetologia 55:19851994
58. Tian J, Dang H, Chen Z et al (2013) γ-Aminobutyric acid regu-
lates both the survival and replication of human beta-cells.
Diabetes 62:37603765
59. Trus MD, Hintz CS, Weinstein JB, Williams AD, Pagliara AS,
Matschinsky FM (1979) A comparison of the effects of glucose
and acetylcholine on insulin release and intermediary metabolism
in rat pancreatic islets. J Biol Chem 254:39213929
60. Renuka TR, Robinson R, Paulose CS (2006) Increased insulin
secretion by muscarinic M1 and M3 receptor function from rat
pancreatic islets in vitro. Neurochem Res 31:313320
2226 Diabetologia (2015) 58:22182228
61. Markmann JF, Deng S, Huang X et al (2003) Insulin indepen-
dence following isolated islet transplantation and single islet infu-
sions. Ann Surg 237:741749, discussion 749-750
62. Burnstock G, Verkhratsky A (2009) Evolutionary origins of the
purinergic signalling system. Acta Physiol 195:415447
63. Cook DL, Hales CN (1984) Intracellular ATP directly blocks K
+
channels in pancreatic B-cells. Nature 311:271273
64. MacDonald PE, Braun M, Galvanovskis J, Rorsman P (2006)
Release of small transmitters through kiss-and-run fusion pores
in rat pancreatic beta cells. Cell Metab 4:283290
65. Obermuller S, Lindqvist A, Karanauskaite J, Galvanovskis J,
Rorsman P, Barg S (2005) Selective nucleotide-release from
dense-core granules in insulin-secreting cells. J Cell Sci 118:
42714282
66. Leon C, Freund M, Latchoumanin O et al (2005) The P2Y
1
re-
ceptor is involved in the maintenance of glucose homeostasis and
in insulin secretion in mice. Purinergic Signal 1:145151
67. Petit P, Manteghetti M, Puech R, Loubatieres-Mariani MM
(1987) ATP and phosphate-modified adenine nucleotide an-
alogues. Effects on insulin secretion and calcium uptake.
Biochem Pharmacol 36:377380
68. Poulsen CR, Bokvist K, Olsen HL et al (1999) Multiple sites of
purinergic control of insulin secretion in mouse pancreatic beta-
cells. Diabetes 48:21712181
69. Khan S, Yan-Do R, Duong E et al (2014) Autocrine activation of
P2Y
1
receptors couples Ca
2+
influx to Ca
2+
release in human
pancreatic beta cells. Diabetologia 57:25352545
70. Roberts E, Frankel S (1950) γ-Aminobutyric acid in brain: its
formation from glutamic acid. J Biol Chem 187:5563
71. Olsen RW, Sieghart W (2009) GABA
A
receptors: subtypes pro-
vide diversity of function and pharmacology. Neuropharmacology
56:141148
72. Braun M, Wendt A, Birnir B et al (2004) Regulated exocytosis of
GABA-containing synaptic-like microvesicles in pancreatic beta-
cells. J Gen Physiol 123:191204
73. Rorsman P, Berggren PO, Bokvist K et al (1989) Glucose-
inhibition of glucagon secretion involves activation of GABA
A
-
receptor chloride channels. Nature 341:233236
74. Soltani N, Qiu H, Aleksic M et al (2011) GABA exerts protective
and regenerative effects on islet beta cells and reverses diabetes.
Proc Natl Acad Sci U S A 108:1169211697
75. Kim J, Richter W, Aanstoot HJ et al (1993) Differential expression
of GAD65 and GAD67 in human, rat, and mouse pancreatic islets.
Diabetes 42:17991808
76. Braun M, Ramracheya R, Bengtsson M et al (2010) γ-
Aminobutyric acid (GABA) is an autocrine excitatory transmitter
in human pancreatic beta-cells. Diabetes 59:16941701
77. Meldrum BS (2000) Glutamate as a neurotransmitter in the brain:
review of physiology and pathology. J Nutr 130:1007S1015S
78. Inagaki N, Kuromi H, Gonoi T et al (1995) Expression and
role of ionotropic glutamate receptors in pancreatic islet cells.
FASEBJ:OffPublFedAmSocExpBiol9:686691
79. MuroyamaA, Uehara S, Yatsushiro S et al (2004) A novel variant
of ionotropic glutamate receptor regulates somatostatin secretion
from delta-cells of islets of Langerhans. Diabetes 53:17431753
80. Hayashi M, Yamada H, Uehara S et al (2003) Secretory granule-
mediated co-secretion of L-glutamate and glucagon triggers gluta-
matergic signal transmission in islets of Langerhans. J Biol Chem
278:19661974
81. Feldmann N, del Rio RM, Gjinovci A, Tamarit-Rodriguez J,
Wollheim CB, Wiederkehr A (2011) Reduction of plasma mem-
brane glutamate transport potentiates insulin but not glucagon
secretion in pancreatic islet cells. Mol Cell Endocrinol 338:4657
82. Banting FG, Best CH, Collip JB, Campbell WR, Fletcher AA
(1922) Pancreatic extracts in the treatment of diabetes mellitus.
Can Med Assoc J 12:141146
83. Uhles S, Moede T, Leibiger B, Berggren PO, Leibiger IB (2003)
Isoform-specific insulin receptor signaling involves different plas-
ma membrane domains. J Cell Biol 163:13271337
84. Kulkarni RN, Bruning JC, Winnay JN, Postic C, Magnuson MA,
Kahn CR (1999) Tissue-specific knockout of the insulin receptor
in pancreatic beta cells creates an insulin secretory defect similar to
that in type 2 diabetes. Cell 96:329339
85. Martinez SC, Cras-Meneur C, Bernal-Mizrachi E, Permutt MA
(2006) Glucose regulates Foxo1 through insulin receptor signaling
in the pancreatic islet beta-cell. Diabetes 55:15811591
86. Khan FA, Goforth PB, Zhang M, Satin LS (2001) Insulin activates
ATP-sensitive K
+
channels in pancreatic beta-cells through a phos-
phatidylinositol 3-kinase-dependent pathway. Diabetes 50:2192
2198
87. Kawamori D, Kurpad AJ, Hu J et al (2009) Insulin signaling in
alpha cells modulates glucagon secretion in vivo. Cell Metab 9:
350361
88. Wu H, MacFarlane WM, Tadayyon M, Arch JR, James RF,
Docherty K (1999) Insulin stimulates pancreatic-duodenal
homoeobox factor-1 (PDX1) DNA-binding activity and insulin
promoter activity in pancreatic beta cells. Biochem J 344:813818
89. Persaud SJ, Asare-Anane H, Jones PM (2002) Insulin receptor
activation inhibits insulin secretion from human islets of
Langerhans. FEBS Lett 510:225228
90. Brunicardi FC, Kleinman R, Moldovan S et al (2001)
Immunoneutralization of somatostatin, insulin, and glucagon
causes alterations in islet cell secretion in the isolated perfused
human pancreas. Pancreas 23:302308
91. Ramnanan CJ, Edgerton DS, Kraft G, Cherrington AD (2011)
Physiologic action of glucagon on liver glucose metabolism.
Diabetes Obes Metab 13(Suppl 1):118125
92. Gelling RW, Du XQ, Dichmann DS et al (2003) Lower blood
glucose, hyperglucagonemia, and pancreatic alpha cell hyperpla-
sia in glucagon receptor knockout mice. Proc Natl Acad Sci U S A
100:14381443
93. Moens K, Flamez D, Van Schravendijk C, Ling Z, Pipeleers D,
Schuit F (1998) Dual glucagon recognition by pancreatic beta-
cells via glucagon and glucagon-like peptide 1 receptors.
Diabetes 47:6672
94. Huypens P, Ling Z, Pipeleers D, Schuit F (2000) Glucagon recep-
tors on human islet cells contribute to glucose competence of
insulin release. Diabetologia 43:10121019
95. Moens K, Heimberg H, Flamez D et al (1996) Expression and
functional activity of glucagon, glucagon-like peptide I, and
glucose-dependent insulinotropic peptide receptors in rat pancre-
atic islet cells. Diabetes 45:257261
96. Kieffer TJ, Heller RS, Unson CG, Weir GC, Habener JF (1996)
Distribution of glucagon receptors on hormone-specific endocrine
cells of rat pancreatic islets. Endocrinology 137:51195125
97. Sorensen H, Winzell MS, Brand CL et al (2006) Glucagon recep-
tor knockout mice display increased insulin sensitivity and im-
paired beta-cell function. Diabetes 55:34633469
98. Leibiger B, Moede T, Muhandiramlage TP et al (2012)
Glucagon regulates its own synthesis by autocrine signaling.
Proc Natl Acad Sci U S A 109:2092520930
99. Gahete MD, Cordoba-Chacon J, Duran-Prado M et al (2010)
Somatostatin and its receptors from fish to mammals.
Ann N Y Acad Sci 1200:4352
100. Hauge-Evans AC, King AJ, Carmignac D et al (2009)
Somatostatin secreted by islet delta-cells fulfills multiple roles
as a paracrine regulator of islet function. Diabetes 58:403411
101. Kumar U, Sasi R, Suresh S et al (1999) Subtype-selective expres-
sion of the five somatostatin receptors (hSSTR1-5) in human pan-
creatic islet cells: a quantitative double-label immunohistochemi-
cal analysis. Diabetes 48:7785
Diabetologia (2015) 58:22182228 2227
102. Wang XP, Yang J, Norman MA, Magnusson J, DeMayo FJ,
Brunicardi FC (2005) SSTR5 ablation in islet results in alterations
in glucose homeostasis in mice. FEBS Lett 579:31073114
103. Zhang Y, Zhang Y, Bone RN et al (2012) Regeneration of pancre-
atic non-beta endocrine cells in adult mice following a single
diabetes-inducing dose of streptozotocin. PLoS One 7:e36675
104. Plesner A, Ten Holder JT, Verchere CB (2014) Islet remodeling in
female mice with spontaneous autoimmune and streptozotocin-
induced diabetes. PLoS One 9:e102843
105. Dufrane D, Maillart JF, Aouassar N, Goebbels RM, Guiot Y,
Gianello P (2009) Native pancreatic alpha-cell adaptation in
streptozotocin-induced diabetic primates: importance for pig islet
xenotransplantation. Xenotransplantation 16:152163
106. Meier JJ, Bhushan A, Butler AE, Rizza RA, Butler PC (2005)
Sustained beta cell apoptosis in patients with long-standing type
1 diabetes: indirect evidence for islet regeneration? Diabetologia
48:22212228
107. Walker JN, Johnson PR, Shigeto M, Hughes SJ, Clark A,
Rorsman P (2011) Glucose-responsive beta cells in islets isolated
from a patient with long-standing type 1 diabetes mellitus.
Diabetologia 54:200202
108. Carlsson PO, Andersson A, Jansson L (1996) Pancreatic islet
blood flow in normal and obese-hyperglycemic (ob/ob) mice.
Am J Physiol 271:E990E995
109. Ilegems E, Dicker A, Speier S et al (2013) Reporter islets in
the eye reveal the plasticity of the endocrine pancreas.
Proc Natl Acad Sci U S A 110:2058120586
110. Nakamura M, Kitamura H, Konishi S et al (1995) The endo-
crine pancreas of spontaneously diabetic db/db mice: micro-
angiopathy as revealed by transmission electron microscopy.
Diabetes Res Clin Pract 30:89100
111. Dai C, Brissova M, Reinert RB et al (2013) Pancreatic islet vas-
culature adapts to insulin resistance through dilation and not an-
giogenesis. Diabetes 62:41444153
112. Paulsen SJ, Jelsing J, Madsen ANet al (2010) Characterization of
beta-cell mass and insulin resistance in diet-induced obese and
diet-resistant rats. Obesity (Silver Spring) 18:266273
113. Ribeiro RA, Santos-Silva JC, Vettorazzi JF et al (2012) Taurine
supplementation prevents morpho-physiological alterations in
high-fat diet mice pancreatic beta-cells. Amino Acids 43:1791
1801
114. Okada T, Liew CW, Hu J et al (2007) Insulin receptors in beta-cells
are critical for islet compensatory growth response to insulin re-
sistance. Proc Natl Acad Sci U S A 104:89778982
115. Halban PA, Polonsky KS, Bowden DWet al (2014) β-Cell failure
in type 2 diabetes: postulated mechanisms and prospects for pre-
vention and treatment. Diabetes Care 37:17511758
116. Mezza T, Muscogiuri G, Sorice GP et al (2014) Insulin resistance
alters islet morphology in nondiabetic humans. Diabetes 63:994
1007
117. Talchai C, Xuan S, Lin HV, Sussel L, Accili D (2012) Pancreatic
beta cell dedifferentiation as a mechanism of diabetic beta cell
failure. Cell 150:12231234
118. Weir GC, Bonner-Weir S (2004) Five stages of evolving beta-cell
dysfunction during progression to diabetes. Diabetes 5(Suppl 3):
S16S21
119. UKPDS (1995) U.K. prospective diabetes study 16. Overview of
6yearstherapy of type II diabetes: a progressive disease. U.K.
Prospective Diabetes Study Group. Diabetes 44:12491258
120. Abdulreda MH, Faleo G, Molano RD et al (2011) High-resolu-
tion, noninvasive longitudinal live imaging of immune responses.
Proc Natl Acad Sci U S A 108:1286312868
121. Rodriguez-Diaz R, Speier S, Molano RD et al (2012) Noninvasive
in vivo model demonstrating the effects of autonomic innervation
on pancreatic islet function. Proc Natl Acad Sci U S A 109:21456
21461
122. Savinov AY, Wong FS, Stonebraker AC, Chervonsky AV
(2003) Presentation of antigen by endothelial cells and
chemoattraction are required for homing of insulin-specific
CD8
+
T cells. J Exp Med 197:643656
123. Akirav EM, Baquero MT, Opare-Addo LW et al (2011) Glucose
and inflammation control islet vascular density and beta-cell func-
tion in NOD mice: control of islet vasculature and vascular endo-
thelial growth factor by glucose. Diabetes 60:876883
124. Reddy S, Pathipati P, Bai Y, Robinson E, Ross JM (2005)
Histopathological changes in insulin, glucagon and somatostatin
cells in the islets of NOD mice during cyclophosphamide-
accelerated diabetes: a combined immunohistochemical and his-
tochemical study. J Mol Histol 36:289300
125. Esterhazy D, Stutzer I, Wang H et al (2011) Bace2 is a beta cell-
enriched protease that regulates pancreatic beta cell function and
mass. Cell Metab 14:365377
126. Devedjian JC, George M, CasellasA et al (2000) Transgenic mice
overexpressing insulin-like growth factor-II in beta cells develop
type 2 diabetes. J Clin Invest 105:731740
127. Guillam MT, Hummler E, Schaerer E et al (1997) Early diabetes
and abnormal postnatal pancreaticislet development in mice lack-
ing Glut-2. Nat Genet 17:327330
128. Höög A, Sandberg-Nordqvist AC, Abdel-Halim SM et al (1996)
Increased amounts of a high molecular weight insulin-like growth
factor II (IGF-II) peptide and IGF-II messenger ribonucleic acid in
pancreatic islets of diabetic Goto-Kakizaki rats. Endocrinology
137:24152423
129. Agudo J, Ayuso E, Jimenez V et al (2012) Vascular endothelial
growth factor-mediated islet hypervascularization and inflamma-
tion contribute to progressive reduction of beta-cell mass. Diabetes
61:28512861
130. Guardado-Mendoza R, Davalli AM, Chavez AO et al (2009)
Pancreatic islet amyloidosis, beta-cell apoptosis, and alpha-cell
proliferation are determinants of islet remodeling in type-2 diabet-
ic baboons. Proc Natl Acad Sci U S A 106:1399213997
131. Guardado-Mendoza R, Jimenez-Ceja L, Majluf-Cruz A et al
(2013) Impact of obesity severity and duration on pancreatic
beta- and alpha-cell dynamics in normoglycemic non-human pri-
mates. Int J Obes (Lond) 37:10711078
132. Jurgens CA, Toukatly MN, Fligner CL et al (2011) β-Cell loss and
β-cell apoptosis in human type 2 diabetes are related to islet am-
yloid deposition. Am J Pathol 178:26322640
133. Henquin JC, Rahier J (2011) Pancreatic alpha cell mass in
European subjects with type 2 diabetes. Diabetologia 54:1720
1725
134. Tomita T (2012) Islet amyloid polypeptide in pancreatic islets
from type 2 diabetic subjects. Islets 4:223232
135. Yoneda S, Uno S, Iwahashi H et al (2013) Predominance of
beta-cell neogenesis rather than replication in humans with an
impaired glucose tolerance and newly diagnosed diabetes.
J Clin Endocrinol Metab 98:20532061
136. Rahier J, Guiot Y, Goebbels RM, Sempoux C, Henquin JC (2008)
Pancreatic beta-cell mass in European subjects with type 2 diabe-
tes. Diabetes Obes Metab 10(Suppl 4):3242
137. Marselli L, Suleiman M, Masini M et al (2014) Are we
overestimating the loss of beta cells in type 2 diabetes?
Diabetologia 57:362365
138. Butcher MJ, Hallinger D, Garcia E et al (2014) Association of
proinflammatory cytokines and islet resident leucocytes with islet
dysfunction in type 2 diabetes. Diabetologia 57:491501
2228 Diabetologia (2015) 58:22182228
... There is some degree of variation in the precise arrangement of the cells between islets, which themselves come in a variety of sizes, and between species [2,8,9]. The precise arrangement of cells may play a role in the signaling between cells and the regulation of hormone secretion [7,[10][11][12][13]. How this arrangement of different cell types in islets [14] relates to innervation [15] of, and blood flow [16,17] through islets is still an area of active study [18,19]. ...
... However, we observe changes in this percentage across developmental stages and between control and diabetic, that correlate with trends in persistence homology of the islets in both cases. These differences in islet cytoarchitecture may affect paracrine signaling between endocrine cells resulting in functional differences [7,[10][11][12][13]. For example, NS-components of β-cells might be indicative of β-cells coupled via gap junction linkages that may play a functional role in coordinated responses to endogenous insulin secretagogues such as glucagon-like peptide-1 (GLP-1), but might not be significant for islet dynamics involved in glucose-stimulated Ca 2+ oscillations [64,65]. ...
Article
Full-text available
The islets of Langerhans are critical endocrine micro-organs that secrete hormones regulating energy metabolism in animals. Insulin and glucagon, secreted by beta and alpha cells, respectively, are responsible for metabolic switching between fat and glucose utilization. Dysfunction in their secretion and/or counter-regulatory influence leads to diabetes. Debate in the field centers on the cytoarchitecture of islets, as the signaling that governs hormonal secretion depends on structural and functional factors, including electrical connectivity, innervation, vascularization, and physical proximity. Much effort has therefore been devoted to elucidating which architectural features are significant for function and how derangements in these features are correlated or causative for dysfunction, especially using quantitative network science or graph theory characterizations. Here, we ask if there are non-local features in islet cytoarchitecture, going beyond standard network statistics, that are relevant to islet function. An example is ring structures, or cycles, of α and δ cells surrounding β cell clusters or the opposite, β cells surrounding α and δ cells. These could appear in two-dimensional islet section images if a sphere consisting of one cell type surrounds a cluster of another cell type. To address these issues, we developed two independent computational approaches, geometric and topological, for such characterizations. For the latter, we introduce an application of topological data analysis to determine locations of topological features that are biologically significant. We show that both approaches, applied to a large collection of islet sections, are in complete agreement in the context both of developmental and diabetes-related changes in islet characteristics. The topological approach can be applied to three-dimensional imaging data for islets as well.
... Images show the fluorescence ratio (F340/ F380) of one representative islet before and after application of 1 mM p116sc, 1 mM p116, and 100 µM carbachol (CCh). b Time course of Ca 2+ responses in five representative regions (1)(2)(3)(4)(5) within the islet is shown and corresponds to c. c Changes in intracellular Ca 2+ concentration induced by p116 (1 mM), p116sc (1 mM), and CCh (100 µM) in pancreatic islets. Relative changes in the Ca 2+ concentration are presented as mean delta ratio (F340/F380) ± SEM from three islets containing five to seven regions of interest (defined as squares of 13 µm side length). ...
Article
Full-text available
Glucose homeostasis is maintained by hormones secreted from different cell types of the pancreatic islets and controlled by manifold input including signals mediated through G protein-coupled receptors (GPCRs). RNA-seq analyses revealed expression of numerous GPCRs in mouse and human pancreatic islets, among them Gpr116/Adgrf5. GPR116 is an adhesion GPCR mainly found in lung and required for surfactant secretion. Here, we demonstrate that GPR116 is involved in the somatostatin release from pancreatic delta cells using a whole-body as well as a cell-specific knock-out mouse model. Interestingly, the whole-body GPR116 deficiency causes further changes such as decreased beta-cell mass, lower number of small islets, and reduced pancreatic insulin content. Glucose homeostasis in global GPR116-deficient mice is maintained by counter-acting mechanisms modulating insulin degradation. Our data highlight an important function of GPR116 in controlling glucose homeostasis.
... Although rat and human islets exhibit similarities, some differences should be considered. For example, human islets have been reported to be more heterogeneous in beta, alpha, and delta cells (Arrojo e Drigo et al., 2015), and the amino acid sequence of angiogenic factors (e.g., VEGF-A) in rats is slightly different from that in humans. The heterogeneity in angiogenic capacities should be evaluated using human islets for clinical application. ...
Article
Full-text available
Pancreatic islet transplantation presents a promising therapy for individuals suffering from type 1 diabetes. To maintain the function of transplanted islets in vivo, it is imperative to induce angiogenesis. However, the mechanisms underlying angiogenesis triggered by islets remain unclear. In this study, we introduced a microphysiological system to study the angiogenic capacity and dynamics of individual islets. The system, which features an open‐top structure, uniquely facilitates the inoculation of islets and the longitudinal observation of vascular formation in in vivo like microenvironment with islet‐endothelial cell communication. By leveraging our system, we discovered notable islet−islet heterogeneity in the angiogenic capacity. Transcriptomic analysis of the vascularized islets revealed that islets with high angiogenic capacity exhibited upregulation of genes related to insulin secretion and downregulation of genes related to angiogenesis and fibroblasts. In conclusion, our microfluidic approach is effective in characterizing the vascular formation of individual islets and holds great promise for elucidating the angiogenic mechanisms that enhance islet transplantation therapy.
... However, understanding human islet and pancreas pathogenesis in detail ultimately requires studies of viable human tissue to enable analysis and direct assessment of islet cell function, regulation, and their cellular interactions. Importantly, animal models, although useful for experimental manipulations and basic mechanistic studies, display fundamental differences and have provided fairly limited translational value (7)(8)(9)(10)(11)(12)(13)(14)(15)(16). This situation has sparked the formation of several initiatives over the last two decades, efforts that aimed to expand access for researchers to human pancreatic specimens (17)(18)(19)(20)(21). ...
Article
Over the last two decades, increased availability of human pancreatic tissues has allowed for major expansions in our understanding of islet biology in health and disease. Indeed, studies of fixed and frozen pancreatic tissues, as well as efforts using viable isolated islets obtained from organ donors, have provided significant insights toward our understanding of diabetes. However, the procedures associated with islet isolation result in distressed cells that have been removed from any surrounding influence. The pancreas tissue slice technology was developed as an in situ approach to overcome certain limitations associated with studies on isolated islets or fixed tissue. In this Perspective, we discuss the value of this novel platform and review how pancreas tissue slices, within a short time, have been integrated in numerous studies of rodent and human islet research. We show that pancreas tissue slices allow for investigations in a less perturbed organ tissue environment, ranging from cellular processes, over peri-islet modulations, to tissue interactions. Finally, we discuss the considerations and limitations of this technology in its future applications. We believe the pancreas tissue slices will help bridge the gap between studies on isolated islets and cells to the systemic conditions by providing new insight into physiological and pathophysiological processes at the organ level. Article Highlights Human pancreas tissue slices represent a novel platform to study human islet biology in close to physiological conditions. Complementary to established technologies, such as isolated islets, single cells, and histological sections, pancreas tissue slices help bridge our understanding of islet physiology and pathophysiology from single cell to intact organ. Diverse sources of viable human pancreas tissue, each with distinct characteristics to be considered, are available to use in tissue slices for the study of diabetes pathogenesis.
... The authors also make claims about the hierarchy of paracrine interactions based on anatomical features of rodent islets, but extensive work by Hara and colleagues has revealed a striking plasticity of islet architecture and cellular composition among various species, with each species having its own islet cytoarchitecture (8,9). In the case of adult human islets, different endocrine cell types are mostly intermingled, and a-cells are located in close proximity to b-cells (10,11). Thus, it is likely that the anatomical arrangement of the human islet further facilitates paracrine interactions between different types of islet cells, including reciprocal paracrine interactions between aand b-cells. ...
Article
Islet transplantation has become an established method for the treatment of insulin-deficient diabetes such as type 1 and type 3C (pancreatogenic). An effective transplantation necessitates a thorough understanding of the islet architecture and related functions to improve engraftment outcomes. However, in chronic pancreatitis (CP), the structural and related functional information is inadequate. Hence, the present study is aimed to understand the cytoarchitecture of endocrine cells and their functional implications in CP with and without diabetes. Herein, a set of human pancreatic tissue specimens (normal, n=5 and CP, n=20) was collected and processed for islet isolation. Furthermore, immunohistochemistry was used to assess the vascular densities, cell mass, organization, and cell–cell interactions. The glucose-stimulated insulin release results revealed that in chronic pancreatitis without diabetes mellitus altered (CPNDA), at basal glucose concentration the insulin secretion was increased by 24.2%, whereas at high glucose concentration the insulin levels were reduced by 77.4%. The impaired insulin secretion may be caused by alterations in the cellular architecture of islets during CP progression, particularly in chronic pancreatitis with diabetes mellitus and CPNDA conditions. Based on the results, a deeper comprehension of islet architecture would be needed to enhance successful transplantation in CP patients:
Article
Full-text available
Type 2 diabetes mellitus (T2D), a major cause of worldwide morbidity and mortality, is characterized by dysfunction of insulin-producing pancreatic islet β cells1,2. T2D genome-wide association studies (GWAS) have identified hundreds of signals in non-coding and β cell regulatory genomic regions, but deciphering their biological mechanisms remains challenging3–5. Here, to identify early disease-driving events, we performed traditional and multiplexed pancreatic tissue imaging, sorted-islet cell transcriptomics and islet functional analysis of early-stage T2D and control donors. By integrating diverse modalities, we show that early-stage T2D is characterized by β cell-intrinsic defects that can be proportioned into gene regulatory modules with enrichment in signals of genetic risk. After identifying the β cell hub gene and transcription factor RFX6 within one such module, we demonstrated multiple layers of genetic risk that converge on an RFX6-mediated network to reduce insulin secretion by β cells. RFX6 perturbation in primary human islet cells alters β cell chromatin architecture at regions enriched for T2D GWAS signals, and population-scale genetic analyses causally link genetically predicted reduced RFX6 expression with increased T2D risk. Understanding the molecular mechanisms of complex, systemic diseases necessitates integration of signals from multiple molecules, cells, organs and individuals, and thus we anticipate that this approach will be a useful template to identify and validate key regulatory networks and master hub genes for other diseases or traits using GWAS data.
Preprint
Full-text available
Pancreatic islets of Langerhans play a pivotal role in regulating blood glucose homeostasis, but critical information regarding their mass, distribution and composition is lacking within a whole organ context. Here, a new 3D imaging pipeline was applied to generate a first complete account of the insulin-producing islets throughout the human pancreas at a microscopic resolution and within a maintained spatial 3D context. These data show that human islets are far more heterogenous than previously accounted for with regards to their size distribution and cellular make up. By deep tissue 3D imaging, this in-depth study demonstrates that 50% of the human insulin-expressing islets are virtually devoid of glucagon-producing a-cells, an observation with significant implications for both experimental and clinical research. One Sentence Summary: New islet heterogeneity identified in the human pancreas
Article
Full-text available
Significance The regulation of blood glucose is a homeostatic process that declines with age, but it is unknown whether this disturbance is a consequence of intrinsic dysfunction of the regulatory organ, the pancreatic islet. In marked contrast to the widely held notion that the insulin-producing pancreatic beta cell loses function with wear and tear, and thus causes age-related disturbances in glucose homeostasis, we show that mouse and human beta cells are fully functional at advanced age. The pancreatic islet as an organ, however, is threatened by vascular senescence. Replacing the islet vasculature in aged islet grafts rejuvenates the islet and fully restores glucose homeostasis, indicating that islet blood vessels should be targeted to mitigate frail glucose homeostasis associated with aging.
Article
Full-text available
According to the classical incretin concept, glucagon-like peptide (GLP)-1 is viewed as a hormone produced in the intestinal L cells and acting via the circulation on satiety in the brain, gut motility, and insulin and glucagon secretion in the pancreatic islet. However, in contrast to typical hormones, plasma levels of GLP-1 are relatively low with a very short half-life. Furthermore, GLP-1 is rapidly inactivated by dipeptidyl peptidase-4 (DPP-4) in the vicinity of L cells within <1 min from the secretion of the gut peptide (1,2). This rapid metabolism of GLP-1 raises questions about how its effects are mediated on target organs such as pancreatic β-cells. In this review, we will discuss possible alternative pathways for the incretin effect on pancreatic islet. These involve L cell–derived GLP-1 via neuronal activation and α-cell–derived GLP-1 via auto/paracrine effects. Of note, GLP-1 is not acting alone, and its effect can be modulated by other factors including GIP. Since no data are available yet in the present context, we have limited this review to GLP-1. ### GLP-1 in the gut-to-brain-to-periphery axis for the control of glucose metabolism Recent rodent data show that GLP-1 can induce its metabolic actions by interacting with its receptors in extrapancreatic locations such as the gut to activate the submucosal and the myenteric nervous plexi (3,4) and the brain, which then transmit the signal to peripheral tissues (5) within minutes from the absorption of glucose and lipids. The final aim of this axis is to anticipate the breakthrough of the nutrients into the blood and their better handling. Indeed, GLP-1 secreted from L cells can influence brain neuronal activities via an alternative neural pathway initiated by sensors in the hepatic portal region (6–8). Thereby, the vagus nerve transmits the metabolic information to the nucleus tractus solitarii in the brain stem, which relays the glucose signal to hypothalamic …
Article
Table 1 Plasma metabolite and hormone levels in 10–15 day old +/+, +/− and −/− mice (13) +/+ +/− −/− Glucose (mM) male 508±0.5 5.49±0.2 11.57±0.9 female 7±0.7 6.08±0.3 12.04±1.0 Insulin (μU nits/ml) 14.6±2.6 11.83±2.0 11.58±1.6 Glucagon (pg/ml) 11 5.08 ±38.7 88.63 ±10.7 399.71 ±78.5 NEFA (μim) 507.4±17.
Article
Our previous experiments on isolated rat pancreas gave evidence for a P 2 purinergic receptor on the insulin-secreting B cell. This work was designed to investigate whether the stimulation of insulin release by phosphorylated adenosine derivatives could also be observed in rat isolated Langerhans islets and whether this stimulation was accompanied by changes in calcium uptake. The results indicate that two structural methylene analogues of ATP and ADP (α,β-methylene ATP and α, β-methylene ADP) display an insulin stimulatory effect comparable to that of ATP, confirming the membrane action of the latter. It was also found that calcium uptake increased concomitantly with insulin release under the effect of α,β-methylene ADP; on the other hand this agent also increased the total exchangeable calcium content of islets at isotopic equilibrium. Verapamil, a blocker of voltage-sensitive calcium channels, counteracted the stimulation of insulin release and also blocked the increase in total exchangeable calcium content. These results demonstrate the involvement of calcium in the stimulus-secretion coupling of insulin release induced by an activator of P2 purinergic receptors and suggest the implication of voltage-sensitive calcium channels.
Article
Studies over the past 35 years in the nonobese diabetic (NOD) mouse have shown that a number of agents can prevent or even reverse type 1 diabetes mellitus (T1DM); however, these successes have not been replicated in human clinical trials. Although some of these interventions have delayed disease onset or progression in subsets of participants, none have resulted in a complete cure. Even in the most robust responders, the treatments do not permanently preserve insulin secretion or stimulate the proliferation of β cells, as has been observed in mice. The shortfalls of translating NOD mouse studies into the clinic questions the value of using this model in preclinical studies. In this Perspectives, we suggest how immunological and genetic differences between NOD mice and humans might contribute to the differential outcomes and suggest ways in which the mouse model might be modified or applied as a tool to develop treatments and improve understanding of clinical trial outcomes.
Article
There has been great interest in understanding how human islets differ from rodent islets. Three major issues about human islet morphology have remained controversial over recent decades: 1) the proportion of the islet made up of β-cells; 2) whether islet cell types have a non-random mantle-core pattern, as seen in rodents, or are randomly scattered throughout the islet; 3) the relation of the different cell types to the blood vessels within the islet, which has implications for intraislet function. We re-examined these issues on immunostained sections of non-diabetic adult human pancreas. The composition of the islets can vary by the analysis method (number vs volume) and by the sampling of islets by size. The majority of adult human islets have clear, non-random clustering of β-cells and blood vessels that penetrate into the β-cell cores. We conclude that although there is far more variability in islet composition both within each human pancreas and among different human pancreas than in rodent pancreas, the islet architecture is not so different between the species. The intrapancreatic variability raises important questions about how islets evolve and function throughout life and how this might relate to the pathogenesis of diabetes.