ArticlePDF AvailableLiterature Review

Amylin at the interface between metabolic and neurodegenerative disorders

Frontiers
Frontiers in Neuroscience
Authors:
  • University of Zurich - Vetsuisse, Switzerland

Abstract and Figures

The pancreatic peptide amylin is best known for its role as a satiation hormone in the control of food intake and as the major component of islet amyloid deposits in the pancreatic islets of patients with type 2 diabetes mellitus (T2DM). Epidemiological studies have established a clear association between metabolic and neurodegenerative disorders in general, and between T2DM and Alzheimer's disease (AD) in particular. Here, we discuss that amylin may be an important player acting at the interface between these metabolic and neurodegenerative disorders. Abnormal amylin production is a hallmark peripheral pathology both in the early (pre-diabetic) and late phases of T2DM, where hyperamylinemic (early phase) and hypoamylinemic (late phase) conditions coincide with hyper- and hypo-insulinemia, respectively. Moreover, there are notable biochemical similarities between amylin and β-amyloids (Aβ), which are both prone to amyloid plaque formation and to cytotoxic effects. Amylin's propensity to form amyloid plaques is not restricted to pancreatic islet cells, but readily extends to the CNS, where it has been found to co-localize with Aβ plaques in at least a subset of AD patients. Hence, amylin may constitute a “second amyloid” in neurodegenerative disorders such as AD. We further argue that hyperamylinemic conditions may be more relevant for the early processes of amyloid formation in the CNS, whereas hypoamylinemic conditions may be more strongly associated with late stages of central amyloid pathologies. Advancing our understanding of these temporal relationships may help to establish amylin-based interventions in the treatment of AD and other neurodegenerative disorders with metabolic comorbidities.
Content may be subject to copyright.
REVIEW
published: 16 June 2015
doi: 10.3389/fnins.2015.00216
Frontiers in Neuroscience | www.frontiersin.org 1June 2015 | Volume 9 | Article 216
Edited by:
Tommaso Cassano,
University of Foggia, Italy
Luca Steardo,
Sapienza University of Rome, Italy
Reviewed by:
Massimo Grilli,
University of Genova, Italy
Fabio Di Domenico,
Sapienza University of Rome, Italy
*Correspondence:
Thomas A. Lutz,
Institute of Veterinary Physiology,
University of Zurich,
Winterthurerstrasse 260, 8057 Zurich,
Switzerland
tomlutz@vetphys.uzh.ch
Specialty section:
This article was submitted to
Neuropharmacology,
a section of the journal
Frontiers in Neuroscience
Received: 30 April 2015
Accepted: 29 May 2015
Published: 16 June 2015
Citation:
Lutz TA and Meyer U (2015) Amylin at
the interface between metabolic and
neurodegenerative disorders.
Front. Neurosci. 9:216.
doi: 10.3389/fnins.2015.00216
Amylin at the interface between
metabolic and neurodegenerative
disorders
Thomas A. Lutz 1, 2*and Urs Meyer 3
1Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland, 2Zurich Center of Integrative Human Physiology,
University of Zurich, Zurich, Switzerland, 3Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich,
Switzerland
The pancreatic peptide amylin is best known for its role as a satiation hormone in
the control of food intake and as the major component of islet amyloid deposits in
the pancreatic islets of patients with type 2 diabetes mellitus (T2DM). Epidemiological
studies have established a clear association between metabolic and neurodegenerative
disorders in general, and between T2DM and Alzheimer’s disease (AD) in particular. Here,
we discuss that amylin may be an important player acting at the interface between these
metabolic and neurodegenerative disorders. Abnormal amylin production is a hallmark
peripheral pathology both in the early (pre-diabetic) and late phases of T2DM, where
hyperamylinemic (early phase) and hypoamylinemic (late phase) conditions coincide
with hyper- and hypo-insulinemia, respectively. Moreover, there are notable biochemical
similarities between amylin and β-amyloids (Aβ), which are both prone to amyloid plaque
formation and to cytotoxic effects. Amylin’s propensity to form amyloid plaques is not
restricted to pancreatic islet cells, but readily extends to the CNS, where it has been found
to co-localize with Aβplaques in at least a subset of AD patients. Hence, amylin may
constitute a “second amyloid” in neurodegenerative disorders such as AD. We further
argue that hyperamylinemic conditions may be more relevant for the early processes of
amyloid formation in the CNS, whereas hypoamylinemic conditions may be more strongly
associated with late stages of central amyloid pathologies. Advancing our understanding
of these temporal relationships may help to establish amylin-based interventions in the
treatment of AD and other neurodegenerative disorders with metabolic comorbidities.
Keywords: amylin, Alzheimer’s disease (AD), β-amyloid, dementia, hyperglycemia, insulin, obesity, type-2 diabetes
Introduction
A growing number of individuals suffer from aging-associated neurological and cognitive
dysfunctions that are characterized by progressive neurodegeneration and dementia. The global
prevalence of dementia is currently estimated to be as high as 30–40 million, and is predicted to
quadruple by the year 2050 (Reitz et al., 2011; Hurd et al., 2013; Reitz and Mayeux, 2014). The
most common form of aging-associated dementia is late-onset or sporadic Alzheimer’s disease
(AD) (Hampel et al., 2011; Reitz et al., 2011), a neurodegenerative disorder affecting multiple
cognitive and behavioral functions. AD patients typically show progressive cognitive decline
along with the concomitant appearance of several neuropathological hallmarks, including amyloid
Lutz and Meyer Amylin in neurodegenerative disorders
plaque deposition, neurofibrillary tangle formation, vascular
malfunction, and synaptic loss (Ferrer, 2012). Several other
pathological processes have been identified in the early stages
of the disease, which in turn may play a major etiopathological
role in sporadic AD. These include increased oxidative stress,
neuroinflammation, and impairments in brain glucose uptake
and metabolism (Eikelenboom et al., 2010; Bassil et al., 2014;
Chen and Zhong, 2014; Morales et al., 2014). Hence, sporadic AD
likely represents a multifactorial disease caused by a multitude
of synergistically interacting pathological changes, resulting in
severe cognitive impairments and extensive brain tissue atrophy
(McDonald, 2002).
In parallel to the global increase in neurodegenerative
disorders, there is a rising prevalence of metabolic diseases, most
notably obesity and type-2 diabetes mellitus (T2DM) (Ginter and
Simko, 2012a,b). Obesity is typically referred to as a medical
condition, in which excess body fat has accumulated to the extent
that the body mass index (BMI) exceeds 30 kg/m2(Chiang et al.,
2011). It is thought to be one of the primary causes of T2DM
(Chiang et al., 2011; Ginter and Simko, 2012a,b). Depending
on its clinical course and disease stage, T2DM involves altered
glycemic regulation and hyperglycemia, resistance to insulin
actions, inadequate insulin secretion from pancreatic β-cells, as
well as progressive loss of pancreatic β-cells which is paralleled by
deposition of islet amyloid polypeptide, or amylin, as pancreatic
amyloid (Kahn et al., 2014).
Besides their negative effects on body homeostasis, peripheral
metabolic diseases also appear to play an important role in
aging-associated cognitive impairments and neurodegenerative
disorders such as AD (de la Monte and Tong, 2014; Ríos
et al., 2014). Epidemiological studies have repeatedly shown a
significant association between (midlife) obesity and late-onset
dementias (Yaffe, 2007; Arnoldussen et al., 2014; Kiliaan et al.,
2014; Emmerzaal et al., 2015). Likewise, patients with T2DM
are 2- to 5-times more likely to develop AD compared to
non-diabetic individuals (Ott et al., 1996; Li and Hölscher,
2007; Butterfield et al., 2014; Desai et al., 2014). Additional
support for these epidemiological associations can be derived
from investigations in animal models showing that diet-
induced obesity and T2DM impair behavioral and cognitive
functions relevant to human dementia (Greenwood and
Winocur, 2005; Winocur and Greenwood, 2005; Morris
and Tangney, 2014) and exacerbate neuropathological and
cognitive deficits in genetic mouse models of AD (Leboucher
et al., 2013; Vandal et al., 2014). Based on these findings, it
has been proposed that AD and related neurodegenerative
disorders may represent a form of “type-3 diabetes.” According
to this hypothesis, impaired brain insulin and insulin-like
growth factor (IGF) signaling, or “brain insulin resistance,
may trigger and promote neurodegenerative processes via
multiple interrelated pathological mechanisms, including
attenuation of brain glucose uptake and subsequent emergence
of central hypometabolism, mitochondrial dysfunction and
oxidative stress, cerebro-vascular disruption, as well as
imbalances in neuroinflammatory reactions (reviewed in
de la Monte and Wands, 2008; de la Monte, 2012; Bassil
et al., 2014; Sebastião et al., 2014). At the same time, some
of the AD-associated neuropathologies themselves, especially
extracellular amyloid plaques, may promote brain insulin
resistance (de la Monte, 2012). Hence, there could be a
positive feedback loop between brain insulin resistance and
amyloid plaque formation, which together may facilitate
progressive neurodegeneration and cognitive decline (de la
Monte, 2012). This hypothesis is also supported by recent
preclinical investigations and clinical trials demonstrating
beneficial effects of compounds with anti-diabetic properties
on attenuating or normalizing cognitive impairments and
AD-related neuropathologies (for review, see Bassil et al., 2014;
Sebastião et al., 2014).
In the present article, we discuss the potential role of the
pancreatic peptide amylin, also called islet amyloid polypeptide
(IAPP), as a link between metabolic and neurodegenerative
disorders in general, and between T2DM and AD in particular.
Amylin is best known for its role as a satiation hormone in
the control of food intake (Lutz, 2005, 2009, 2010) and as
the major component of amyloid deposits in pancreatic islets
of patients with T2DM and of cats (Lutz and Rand, 1993;
Westermark et al., 2000; Osto et al., 2013). Because of its
propensity to form amyloid deposits in susceptible species like
primates and cats, amylin shares several pathological features
with β-amyloids (Aβ), a family of amyloidogenic peptides most
frequently implicated in the neuropathology of AD (Karran
et al., 2011; Seeman and Seeman, 2011). Here, we discuss these
similarities following a concise introduction in amylin’s main
physiological roles. Particular emphasis is then placed on the
potential mechanisms of amylin and Aβamyloidogenesis, and on
the recent findings directly implicating amylin amyloids in the
neuropathology of AD.
Physiological Roles and Actions of Amylin
Amylin Acts as a Satiation Signal
Amylin is a pancreatic β-cell hormone co-released with insulin
in response to food intake (Kahn et al., 1990). It reduces
eating, gastric acid secretion, limits the rate of gastric emptying,
and diminishes pancreatic glucagon secretion (Lutz, 2010). One
of the best-studied functions of amylin relates its role as a
satiation signal: via regulating meal size (Lutz et al., 1995b;
Reidelberger et al., 2002; Mollet et al., 2004), it contributes to
meal termination akin to other typical satiating hormones such
as cholecystokinin (CCK) (Young and Denaro, 1998; Lutz, 2010).
The anorectic actions of amylin represent physiological effects as
eating leads to a rapid increase in circulating amylin levels, and
the administration of exogenous amylin reduces eating within
minutes after application. Further, amylin’s capacity to reduce
meal size is dose dependent and is effectively and specifically
blocked by amylin antagonists like AC187 (Mollet et al., 2004;
Reidelberger et al., 2004). Importantly, amylin and its synthetic
analogs reduce meal size without producing signs of conditioned
taste aversion or visceral illness (Lutz et al., 1995b; Rushing
et al., 2002; Mack et al., 2007). Overall, these data indicate that
amylin acts as a specific and physiological satiating hormone that
mediates its short-term effects on food intake mainly via reducing
meal sizes.
Frontiers in Neuroscience | www.frontiersin.org 2June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
Amylin Acts as Adiposity Signal
The concept of “homeostatic eating controls” classically
distinguishes between adiposity (“tonic”) signals and meal-
associated (“episodic”) signals. Adiposity signals are secreted
by adipose tissue (leptin) or the pancreas (insulin, amylin) in
proportion to body adiposity and are thought to reduce eating
by enhancing the effects of satiation signals. For example, amylin
enhances the satiating effects of cholecystokinin (CCK) (Bhavsar
et al., 1998; Mollet et al., 2003). A number of other findings
support a role for amylin as an adiposity signal. For example,
basal plasma levels of amylin are higher in obese relative to
lean rats (Pieber et al., 1994), and high-fat fed obese rats have
higher baseline amylin levels compared to lean controls (Boyle
and Lutz, 2011; Boyle et al., 2011). Moreover, chronic peripheral
(Arnelo et al., 1996; Lutz et al., 2001) or central (Rushing et al.,
2002) administration of amylin decreases body weight and fat
gain, whereas treatment with amylin antagonists increases body
adiposity (Rushing et al., 2001). Together, these data suggest that
central amylin, like leptin or insulin, may encode a certain level
of body weight, and therefore, may contribute to the relative
constancy of body weight throughout adult life.
Amylin and Energy Expenditure
The physiological roles and actions of amylin have been further
explored using pair-feeding research designs in rats, in which
the amount of food available for vehicle-treated animals was
determined by the amount of food ingested by amylin-treated
animals. These studies showed that the reduction in body fat
is greater in rats chronically treated with exogenous amylin as
compared to pair-fed controls (Isaksson et al., 2005; Roth et al.,
2006; Mack et al., 2007). These findings indicate that amylin not
only reduces food intake but also increases energy expenditure
(Isaksson et al., 2005; Wielinga et al., 2007, 2010). Again, these
dual effects of amylin are similar to those induced by leptin,
which is known to influence energy balance through concomitant
effects on eating and an energy expenditure (Friedman, 1997;
Schwartz et al., 2000, 2003). Increases in energy expenditure can
also been seen in rats after acute central infusion of amylin into
discrete brain areas, including the third cerebral ventricle or area
postrema (AP) (Osaka et al., 2008; Wielinga et al., 2010).
Two recent studies provided novel mechanistic insights into
amylin’s effect on energy metabolism (Zhang et al., 2011;
Fernandes-Santos et al., 2013). Similar to humans, total energy
expenditure in rodents partially depends on the thermogenic
activity of the brown adipose tissue (BAT), which in turn is
controlled by the sympathetic nervous system (Nedergaard and
Cannon, 2014). BAT thermogenic activity is increased by amylin
through actions involving the sympathetic nervous system, and
these effects are enhanced in transgenic mice that overexpress
the receptor activity-modifying protein 1 (RAMP1) of the
amylin receptor complex. More specifically, transgenic mice
with neuronal overexpression of RAMP1 show a potentiated
metabolic response to exogenous amylin treatment and display
a number of basal metabolic changes, including reduced body
weight and fat mass, increased energy expenditure and body
temperature, and hypophagia. It has been further shown that
the effects of RAMP1 overexpression on energy expenditure
are mediated by an increased sympathetic tone in efferents
innervating BAT (Zhang et al., 2011; Fernandes-Santos et al.,
2013).
Central Mechanisms Underlying the
Actions of Amylin
Amylin receptor autoradiography studies demonstrate that
several brain areas have high affinity binding sites for amylin
(Sexton et al., 1994). Amylin binding is particularly strong
in the circumventricular organs such as the subfornical organ
(SFO) and AP. A large number of studies indicate that the AP
plays an important role in mediating the physiological effects
of peripheral amylin. Amylin appears to activate AP neurons
by direct humoral actions, whilst vagal or non-vagal afferents
do not seem to be required in these processes (Edwards et al.,
1998; Lutz et al., 1995a, 1998a,b; Wickbom et al., 2008; Mack
et al., 2010; Braegger et al., 2014). Local amylin administration
into the AP recapitulates the effects of peripheral amylin, and
AP-directed infusion of the amylin antagonist AC187 exerts the
opposite effects. Furthermore, local administration of AC187 into
the AP has been shown to abolish the eating inhibitory effect of
peripheral amylin (Mollet et al., 2004), suggesting that central
actions in the AP are necessary for mediating the anorectic effects
of peripheral amylin.
Amylin-induced activation of AP neurons readily induces a
number of neuronal downstream effects, including stimulation
of dopamine-beta-hydroxylase (DBH) and subsequent increase
in noradrenaline synthesis and release in noradrenergic
output areas (Potes et al., 2010). In addition to its effects
on noradrenergic neurons, amylin may further target other
neurotransmitter systems in the AP, including presynaptic
glutamatergic terminals innervating AP neurons (Fukuda et al.,
2013). The functional contribution of these glutamatergic
terminals, however, remains unknown with regards to amylin’s
effects on food intake, metabolism, and other physiological
processes.
Stimulation of AP neurons by peripheral amylin likely leads
to the activation of a neuroaxis that projects rostrally to the
forebrain and includes the nucleus tractus solitarii (NTS),
lateral parabrachial nuclus (LPB), and central amygdala (CeA)
(Rowland et al., 1997; Riediger et al., 2004). In addition to its
stimulating effects on these brain regions, which most likely
follows primary AP activation (Riediger et al., 2004), amylin
further seems to activate key areas of the mesolimbic dopamine
system, including the ventral tegmental areas (VTA) and nucleus
accumbens (NAc). These two regions display high affinity amylin
binding sites and seem to contain virtually all components
of the amylin receptor complex (Mietlicki-Baase et al., 2013;
Mietlicki-Baase and Hayes, 2014). The mesolimbic dopamine
system has long been recognized to play a crucial role in reward
processing including eating associated rewards (Berridge and
Robinson, 1998; Volkow et al., 2013). With respect to the latter,
it has been suggested that pathological conditions of overeating
may represent a compensation for decreased activity of the
mesolimbic and nigrostriatal dopamine circuits (Wang et al.,
2001). This hypothesis is consistent with the notion that the
Frontiers in Neuroscience | www.frontiersin.org 3June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
rewarding properties of food can stimulate feeding behavior even
when energy requirements have been met, thereby contributing
to excessive weight gain and obesity (Kenny, 2011).
In this context, it is interesting that direct local administration
of the amylin receptor and calcitonin receptor agonist salmon
calcitonin into the VTA of rats has been found to reduce the
intake of standard rodent chow diet and of palatable sucrose.
These anorectic effects appeared to result from reductions in
meal size, suggesting that activation of VTA neurons by salmon
calcitonin may modulate the reward value of food, thereby
reducing the size of a meal. On the contrary, administration
of the amylin antagonist AC187 into the VTA increased
eating, and intra-accumbens injection of AC187 attenuated the
anorectic effects of peripheral treatment with salmon calcitonin
(Mietlicki-Baase et al., 2013); local knockdown of the calcitonin
receptor (CTR)—the core component of functional amylin
receptors (Bailey et al., 2012)—in the VTA also resulted in
hyperphagia (Mietlicki-Baase et al., 2015). Further, salmon
calcitonin administered to the VTA resulted in reduced phasic
dopamine release into the NAc core, and blockade of dopamine
D1 and D2 receptors in the NAc core blunted salmon calcitonin’s
eating inhibitory effect (Mietlicki-Baase et al., 2015). Finally, the
NAc shell may also be directly responsive to amylin receptor
stimulation because low doses of amylin given into the NAc shell
reduced eating in rats; amylin also inhibited the eating response
induced by opioid receptor activation indicating that amylin
signaling may negatively modulate opioid reward driven eating
(Baisley and Baldo, 2014).
Together, these findings indicate that peripheral amylin may,
in addition to its actions on the AP and interconnected areas,
exert a direct influence on VTA or NAc neurons and induce
physiologically relevant effects on satiation and food reward. It
will be important to test in future studies whether the observed
effects via the VTA or NAc do depend on a primary action of
amylin in these areas or perhaps still in the AP. It has been shown
that the rat amylin-1 receptor (Bailey et al., 2012) is activated
equally by amylin but also the neurotransmitter calcitonin gene-
related peptide (CGRP), and, importantly, that the effects of
both peptides at the amylin-1 receptor are blocked by AC187.
Hence, it is possible that primary activation of AP neurons
may trigger CGRP release in various brain areas, including
the VTA or the NAc, to explain the observations discussed
above.
Amylin Amyloid Formation
The aforementioned physiological effects of amylin all require
solubility of the monomeric hormone so that it can effectively
reach the target organs and cells upon release by pancreatic
β-cells. Under certain pathological conditions such as T2DM,
however, amylin can self-aggregate and eventually form insoluble
amylin amyloid plaques. As described in detail in the subsequent
sections, this effect is only relevant in few species, including
humans, other primates and cats when amylin amyloid
loses its common physiological actions, but instead, induces
toxic effects on both peripheral and central organs and
cells.
Mechanisms of Amyloidogenesis
The secreted and biologically active form of amylin contains
37 amino acids. It is initially expressed as a pre-pro-protein
containing 89 amino acids, which includes the actual signal
peptide domain (22 acids) and two short flanking peptides
(Höppener et al., 1994; Marzban et al., 2004, 2005; Wang et al.,
2011). Similar to pro-insulin, pre-pro-amylin is processed to pro-
amylin in the endoplasmic reticulum (ER), during which the
signal peptide and flanking peptides are cleaved from the rest.
The resulting pro-amylin is then further processed by proteolysis
and posttranslational modifications in the Golgi apparatus, where
a disulfide bond between cysteine residues 2 and 7 is formed. The
biologically active amylin is then stored in the halo region of the
secretory granules of the β-cell and is co-secreted with insulin
(Westermark et al., 1996).
The amino acid sequence of amylin is strongly conserved
among mammalian species. Formation of amylin amyloids,
however, can only emerge in the pancreatic islets of humans,
primates, and cats, but not in rodents. The reason for this species
differences is that only the former contain an amyloidogenic
region within their amino acid sequence that is essential for
amyloid formation and toxicity (Betsholtz et al., 1989; Höppener
et al., 1994; Moriarty and Raleigh, 1999; Matveyenko and Butler,
2006). In the case of amylin, amyloidogenesis seems to be
associated with the heterogeneity of the amino acid residues
20–29 of the amylin sequence. In species where pancreatic
amyloid deposition does not occur (e.g., mouse or rat), one or
more proline residues can be found in this region. Hence, it is
believed that the presence of proline prevents the formation of
ordered secondary structures such as the β-sheet conformation,
which is an essential prerequisite for amyloidogenesis (Betsholtz
et al., 1989; Höppener et al., 1994; Moriarty and Raleigh, 1999;
Matveyenko and Butler, 2006).
Under physiological conditions, monomeric amylin is soluble
and natively unfolded. It is believed that amylin is protected
from aggregation under these conditions by interactions with
other components of the secretory vesicles, including insulin,
pro-insulin, or their processing intermediates (Westermark et al.,
1996). It seems that a relative increase in the concentration
of amylin within the halo region of pancreatic islet cells can
initiate their aggregation and formation of fibrils under certain
conditions (Paulsson et al., 2006). When fibril formation begins,
the molecules in β-sheet conformation are bound to each other,
predominantly by hydrogen bounds. The β-sheet conformation
typically forms thin and stable layers, in which the β-strands are
oriented perpendicular to the fibril axis.
Interestingly, amylin’s amyloidogenic properties are not
restricted to mature amylin but further extend to pro-amylin
and its intermediates (Westermark et al., 2000; Paulsson and
Westermark, 2005; Paulsson et al., 2006). In fact, the latter may
be the starting point of oligomerization and amylin aggregation
within the pancreatic islets (Westermark et al., 2000; Jaikaran
and Clark, 2001; Jaikaran et al., 2001; Paulsson et al., 2006).
It should be noted, however, that no precise localization of
the aggregates within the cells has been demonstrated yet.
In studies using baboons, amyloid deposits were observed in
the cytoplasm of β-cells as well as bound to the outer β-cell
Frontiers in Neuroscience | www.frontiersin.org 4June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
membrane (Guardado-Mendoza et al., 2009). In other studies,
where islets from diabetic patients were transplanted into nude
mice or cultured in vitro, amylin-derived amyloid was found
to form an intracellular network, suggesting its presence in the
ER, Golgi apparatus and secretory vesicles (Westermark et al.,
1995a,b; Yagui et al., 1995; Paulsson et al., 2006). Recent evidence
further suggests that the early processes of amyloidogenesis
from amylin depend on pH, which can markedly affect the
electrostatic interactions required for β-sheet formation and
amylin fibrillization (Li et al., 2013; Jha et al., 2014).
Abnormalities in the processing of amylin and its deposition
as amyloid in the islets may contribute to the progressive loss of
pancreatic β-cells, which is a pathological hallmark of T2DM.
Indeed, islet amyloid formation is a striking abnormality of
human T2DM and can be found in more than 90% of T2DM
patients (Clark and Nilsson, 2004). Similarly, islet amyloid is
a very prominent feature in diabetic cats (Lutz and Rand,
1993; Osto et al., 2013). Even though the association between
amylin amyloid deposition and pancreatic β-cell loss in T2DM
seems robust, the underlying etiopathological processes remain
to be determined. According to one hypothesis, prolonged
hyperglycemia may lead to a higher concentration of pro-amylin
within the β-cells (Hou et al., 1999), which in turn may create a
core for amylin aggregation and ultimately cause β-cell loss and
deposition of extracellular amyloid (Paulsson and Westermark,
2005). An alternative (but mutually not exclusive) mechanism for
amylin aggregation may include increased retention of amylin in
β-cell granules as a result of increased sympathetic stimulation
(Watve et al., 2014). In addition, high levels of circulating
free fatty acids, as observed in pre-diabetic patients or patients
with overt T2DM, has been implicated as possible mechanism
facilitating amyloid formation (Li and Hölscher, 2007).
Pathological Effects of Amylin Deposits on
Pancreatic β-Cells
Islet amyloid formation plays a key role in β-cell apoptosis
and dysfunction as seen in T2DM (Westermark et al., 2000,
2011; Paulsson and Westermark, 2005; Höppener and Lips, 2006;
Jurgens et al., 2011). As discussed in the subsequent sections, it is
believed that the toxic effects induced by amylin amyloid deposits
depend on a combination of mechanisms that act both intra- and
extra-cellulary.
One of the prominent features associated with amylin-related
β-cell toxicity is ER stress (Preston et al., 2009). Peripheral insulin
resistance triggers β-cells to produce more insulin in parallel
with an increased synthesis of amylin. As these proteins are
destined for exocytosis, they are transported through the ER
and trans-Golgi complex and stored in secretory granules before
secretion. ER stress is characterized by the unfolded protein
response (UPR), which is induced to cope with an increase
in protein synthesis or obstruction in the ER-Golgi transport
(Preston et al., 2009). UPR involves the upregulation of ER-
located chaperones to assist in the folding of proteins that are
prone to aggregation. Under homeostatic conditions, misfolded
proteins are normally transported to the ubiquitin-proteasome
system for degradation, thus preventing protein aggregation
and amyloidogenesis. Failure of such homestatic responses
typically leads to induction of cell apoptosis (Oyadomari et al.,
2002; Marciniak and Ron, 2006; Davenport et al., 2008). Such
processes may be operational especially during the early phase of
T2DM, during which amylin and insulin syntheses are markedly
increased as a consequence of insulin resistance, leading to non-
homeostatic UPR and eventually β-cell apoptosis.
Abnormally enhanced accumulation of fibrillar material in the
halo region of secretory vesicles may also lead to a disruption
of the vesicles and result in the release of fibrillar material into
the cytosol. Because the proteasome pathway can only degrade
misfolded single proteins but not their aggregates, a cellular
process commonly referred to as “autophagy” dissolves the latter.
It is a well-preserved catabolic process that is activated to degrade
and recycle misfolded proteins and excess or defective organelles
(Stienstra et al., 2014). Under non-pathological conditions,
autophagosomes typically fuse with lysosomes. This causes the
formation of autophagolysosomes, where misfolded proteins or
excess/defective organelles can be degraded efficiently. Under
pathological conditions such as the early phase of T2DM,
however, the formation of autophagolysosomes is attenuated,
which in turn causes the accumulation of material in autophagic
vacuoles without being further processed (Stienstra et al., 2014).
Accumulation of autophagic vacuoles may disturb intracellular
cell transport and induce a cascade of intracellular toxic effects
(Marciniak et al., 2006; Marciniak and Ron, 2006; Davenport
et al., 2008; Stienstra et al., 2014). It has been suggested that
pathological processes involving accumulation of autophagic
vacuoles may also play a role in pancreatic β-cell toxicity (Bursch
et al., 2000; Stienstra et al., 2014).
The overexpression of pro-amylin or amylin during the early
phases of T2DM may further lead to the formation of toxic
oligomers, which enter the cytosol and disrupt the membrane of
organelles such as mitochondria. The disruption of mitochondria
function may then result in increased cellular oxidative stress and
the formation of reactive oxygen species (ROS). Increased ROS
levels may ultimately lead to significant damage to cell structures
and cause cell loss (Parks et al., 2001; Scherz-Shouval and Elazar,
2007; Gurlo et al., 2010). Finally, it appears that increased
amylin production can activate the inflammasome, which is a
multiprotein component of the innate immune system producing
the inflammatory cytokine interleukine-1β(IL-1β). Increased
inflammatory processes in general, and overproduction of IL-1β
in particular, may represent another important mechanism by
which amylin aggregation can cause β-cell death (Masters et al.,
2010; Donath and Shoelson, 2011; Westwell-Roper et al., 2011;
Donath et al., 2013).
Amylin Amyloids: A Comparison with the
Formation and Pathological Consequences
of AβAmyloids
The propensity of amylin to form pancreatic amyloids and their
cytotoxic consequences are remarkably similar to β-amyloids
(Aβ), which are prone to self-aggregate in the CNS following
enzymatic processing of the amyloid precursor protein (APP).
The type-I transmembrane glycoprotein APP is one of the
most abundant proteins in the human CNS and is ubiquitously
Frontiers in Neuroscience | www.frontiersin.org 5June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
expressed in the plasma membrane and in several organelles,
such as the ER, Golgi apparatus, and mitochondria (van der Kant
and Goldstein, 2015).
In general, APP can be processed by two distinct and mutually
exclusive pathways, namely the secretory pathway and the
amyloidogenic pathway (Karran et al., 2011). Whilst the former
leads to soluble fragments, the latter creates proteolytic products
that are prone to self-aggregation. In the secretory pathway, APP
is first cleaved by α-secretase, which results in the release of a
soluble N-terminal fragment (sAPPα) and a C-terminal fragment
(C83). The C-terminal fragment is then further processed by
another cleaving enzyme (γ-secretase) to generate a smaller
and soluble C-terminal fragment of approximately 3 kDa (C3).
Notably, the cleavage of APP by α-secretase occurs within the
sequence of amino acids that would form the Aβpeptide, and
therefore, this proteolytic pathway is crucial and readily prevents
the formation of self-aggregating amyloid peptides (Karran et al.,
2011). In the amyloidogenic pathway, however, APP is cleaved by
β-secretase, which releases a small N-terminal fragment (sAPPβ)
and a long C-terminal fragment (C99). The latter contains the full
amyloidogenic sequence of amino acids and is further processed
by γ-secretase to yield amyloid-β(Aβ) peptides. These are then
released as monomers and eventually aggregate progressively
into oligomers and finally into amyloid plaques.
Aβoligomers are considered the most neurotoxic forms
of all amyloid species. As extensively reviewed elsewhere
(Hardy and Selkoe, 2002; Karran et al., 2011; van der Kant
and Goldstein, 2015), they interact with neuronal and glial
cells to cause mitochondrial dysfunction and oxidative stress,
impairments of intracellular signaling pathways and synaptic
plasticity, and eventually neuronal apoptosis. It is believed
that these mechanisms can give rise to a positive feedback
loop that facilitates the production of Aβpeptides through
stimulation of the amyloidogenic pathway (Hardy and Selkoe,
2002; Karran et al., 2011; van der Kant and Goldstein, 2015).
Hence, the amyloidogenic pathway seems to be more active
under pathological conditions, whereas APP is preferentially
processed via the secretory pathway under physiological
conditions.
The maintenance of physiological conditions is further
dependent on the equilibrium between the production of
(soluble) Aβpeptides and their clearance from the brain (Ueno
et al., 2014; Baranello et al., 2015). Even though the precise
mechanisms by which Aβpeptides are cleared from the CNS
remain to be identified, two proteins seem to be pivotal for
this process, namely apolipoprotein E (APOE) and the insulin-
degrading enzyme (IDE) (Baranello et al., 2015). It has been
proposed that these proteins may bind to the Aβpeptide, thereby
preventing aggregation and promoting clearance from the brain.
Some of the known genetic risk factors of AD, including
polymorphisms in the ε4 allele of APOE, may predisposes
individuals to impaired clearance of Aβpeptides from the
CNS, and thus facilitate the formation of Aβplaques (Baranello
et al., 2015). Aβclearance can also be disrupted by several
metabolic disturbances in general, and by those associated
with T2DM in particular. Indeed, T2DM has been linked to
IDE deficiency, which in turn can facilitate the aggregation of
amyloidogenic proteins (Steneberg et al., 2013), including Aβ
(Farris et al., 2003) and amylin-derived amyloid (Bennett et al.,
2003).
These associations have several important implications. First,
the effects of IDE hypofunction on Aβplaque formation may
represent a crucial pathological link between metabolic disorders
such as T2DM and increased risk of AD. Second, IDE deficiency
may not only be a driving force for amylin oligomerization and
cytotoxicity in pancreatic islet cells, but it may further facilitate
the formation of amylin amyloid in the CNS. In support of the
latter hypothesis and in contrast to the original belief that amylin-
derived amyloid occurs exclusively in pancreatic islets, amylin
oligomers and plaques were recently identified in the temporal
lobe gray matter from patients with T2DM (Jackson et al., 2013).
Moreover, similar amylin deposits were also detected in brain
parenchyma of patients with late onset AD, and these amylin
amyloids co-localized with Aβoligomers and plaques (Jackson
et al., 2013). These observations suggested for the first time that
metabolic disorders such as T2DM and perhaps also aging might
similarly promote the accumulation of amylin amyloid in the
CNS. Based on these findings, it has been hypothesized that
amylin could constitute a “second amyloid” in AD (Jackson et al.,
2013).
More recent studies indicated that amylin and Aβco-localize
in plaques of AD brains, and that amyloid deposits consisting
of both amylin and Aβare also present in blood vessel walls
(Oskarsson et al., 2015). While it is not yet clear whether the
amylin that forms deposits in the brain is produced locally or
is rather secreted by the pancreas, it is of note that intravenous
injection of preformed amylin fibrils did enhance local amyloid
deposition in the pancreas, hence a systemic origin is plausible
(Oskarsson et al., 2015; see also next section).
How Is Altered Amylin Secretion Linked to
Central Amyloid Formation?
Despite the existing evidence for amylin oligomerization in
the brains of T2DM and AD patients (Jackson et al., 2013),
it remains controversial whether this neuropathological effect
can be related to, or even induced by, alterations in peripheral
amylin homeostasis. Some researchers suggest that the primary
mechanism leading to amylin deposition in the brains of patients
with T2DM and AD may be hyperamylinemia, i.e., a pathological
condition of chronic hypersecretion of amylin by pancreatic
β-cells (Srodulski et al., 2014). Hyperamylinemia is frequently
observed in individuals with obesity or pre-diabetic insulin
resistance (Johnson et al., 1989; Enoki et al., 1992) and often
coincides with hyperinsulinemia (Westermark et al., 2011).
Furthermore, genetically enforced overexpression of amylin in
rodents transgenic for the human form of amylin leads to
amylin oligomerization and amyloid formation in pancreatic
β-cells, β-cell apoptosis, and eventual development of T2DM
(Matveyenko and Butler, 2006; Huang et al., 2007). Another
cornerstone of the “hyperamylinemia hypothesis” of central
amylin deposition is that peripherally produced amylin readily
crosses the blood-brain barrier (Banks and Kastin, 1998). Hence,
increased peripheral secretion and central uptake of amylin
Frontiers in Neuroscience | www.frontiersin.org 6June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
may facilitate amylin oligomerization and eventually amyloid
formation in the brain, thereby disrupting the integrity of CNS
structures and functions. Support for this hypothesis has recently
been obtained in a “humanized” rat model of hyperamylinemia
(Srodulski et al., 2014): Rats with pancreatic overexpression of
human amylin showed marked amylin amyloid formation, an
effect that was paralleled by neuroinflammatory processes such
as clustering of activated microglia in areas of amylin infiltration.
These neuropathological changes were further associated with
the development of behavioral and cognitive deficits, including
impaired recognition memory and motor learning (Srodulski
et al., 2014). These findings suggest that hypersecretion of
amyloidogenic forms of amylin might indeed represent a
pathological link between amylin amyloid formation and the
development of neurological deficits.
The “hyperamylinemia hypothesis,” however, may only hold
true for central amylin deposition, but not for the accumulation
and oligomerization of other amyloidogenic peptides such as
Aβ. Indeed, in the Tg2576 genetic mouse model of AD, which
harbors a mutant form of APP that is prone to marked Aβplaque
formation, amylin plasma levels were not significantly elevated
at an age when mice exhibited clear signs of T2DM (Fawver
et al., 2014). These findings argued against the hypothesis that
altered systemic amylin levels might promote cross-seeding of
Aβin the CNS, despite the fact that amylin crosses the blood-
brain barrier and efficiently facilitates Aβoligomerization in vitro
(Fawver et al., 2014). A limitation of these findings, and the
interpretations thereof, is that transgenic Tg2576 mice only
harbor mutations that facilitate Aβplaque formation, but not
amylin plaque formation. Indeed, unlike in humans, primates
and cats, rodent amylin per se is not amyloidogenic (Betsholtz
et al., 1989; Höppener et al., 1994; Moriarty and Raleigh, 1999;
Matveyenko and Butler, 2006). Therefore, the missing association
between systemic amylin and Aβplaque formation in Tg2576
mice may be explained by the native incapacity of mouse amylin
to form amylin plaques, which in turn may limit the capacity of
amylin to cross-seed Aβin the CNS. In this respect, mice clearly
differ from humans.
Adding to these controversies, recent research even suggests
that reduced, rather than increased, systemic levels of amylin
may facilitate the development of AD-like pathology. For
example, Zhu et al. (2015) revealed that the augmentation of
endogenous amylin by chronic systemic administration of non-
amyloidogenic amylin, or its clinical analog pramlintide, has
beneficial effects against neuropathological and cognitive deficits
in a mouse model of AD. Specifically, peripheral treatment
with amylin or pramlintide reduced the number of Aβplaques
and improved the deficits in short- and long-term memory
in PP/PS1 double transgenic mice that harbor five familial
genetic mutations implicated in AD (Zhu et al., 2015). Zhu
et al. (2015) further demonstrated that peripheral amylin or
pramlintide treatment similarly increased the concentrations of
Aβ142 in cerebral spinal fluid (CSF) and serum, suggesting
that the augmentation of non-amyloidogenic amylin might
facilitate the clearance of Aβpeptides from the CNS. Similar
beneficial effects have been found by another research group,
who found that chronic administration of the amylin analog
pramlintide, which lacks amyloidogenic properties, improved
AD-related neuropathological and cognitive deficits in SAMP8
mice, a genetic mouse model of sporadic AD (Adler et al.,
2014). Importantly, there is also initial evidence suggesting that
elders with mild cognitive impairment or established AD have
lower concentrations of plasma amylin compared to age-matched
healthy control subjects (Adler et al., 2014; Qiu and Zhu, 2014).
Taken together, the findings by Adler et al. (2014) and Zhu
et al. (2015) emphasize the possibility that blunted (rather
than increased) levels of peripheral amylin may underlie the
development of neurological and cognitive impairments typically
seen in mild cognitive impairment or established AD. This
pathological link may be explained by a competition between
amylin and Aβto bind to its receptor in the CNS. Amylin
and Aβpeptide both bind to the CTR, which in the CNS is
ubiquitously expressed and complexed with the receptor activity
modifying protein 3 (RAMP3) (Götz et al., 2013). According
to a recent hypothesis (Qiu and Zhu, 2014), increased binding
of Aβto the CTR-RAMP3 complex which corresponds to the
amylin-3 receptor occurs under conditions of hypoamylinemia
when amylin fails to compete with Aβfor binding to this
receptor complex. This in turn may facilitate Aβoligomerization
and plaque formation in the CNS (Qiu and Zhu, 2014).
When hypoamylinemic conditions are restored by exogenous
administration of non-amyloidogenic amylin or its clinical
analogs, non-aggregated amylin may prevent the binding of Aβto
the CTR-RAMP3, thereby reducing the potency of Aβpeptides to
oligomerize (Qiu and Zhu, 2014). In addition, the augmentation
of non-amyloidogenic amylin may facilitate the clearance of
Aβfrom the brain, probably through its effects on cerebral
vasculature (Westfall and Curfman-Falvey, 1995; Edvinsson et al.,
2001; Qiu and Zhu, 2014).
Whatever precise mechanisms involved, it appears that hypo-
and hyper-amylinemic conditions can both be associated with
and promote the pathological effects of amyloid formation in
the CNS. Further investigations are clearly warranted to explore
how alterations in peripheral amylin secretion are linked to
central amyloid formation. In the course of these attempts,
it is of primordial importance to take into account the exact
experimental conditions and in particular the clear species
differences regarding the innate propensity of amylin to from
plaques. Hence, it is absolutely critical to distinguish between
amyloidogenic and non-amyloidogenic forms of amylin; the
former is the case for human, other primates’ and feline amylin,
the latter for rodents (Betsholtz et al., 1989; Höppener et al., 1994;
Moriarty and Raleigh, 1999; Matveyenko and Butler, 2006).
Another important factor that needs careful consideration is
the clinical course of metabolic disturbances across aging, which
in turn may critically determine the nature of peripheral amylin
pathologies. As stated above, individuals with obesity or pre-
diabetic insulin resistance often show signs of hyperamylinemia
that precede the onset of full-blown T2DM (Johnson et al.,
1989; Enoki et al., 1992; Westermark et al., 2011). Chronic
over-production of amylin during these early stages of T2DM
are readily toxic to pancreatic β-cells and eventually lead to β-
cell loss (Höppener and Lips, 2006; Jurgens et al., 2011; Desai
et al., 2014). As a consequence of the latter, the production
Frontiers in Neuroscience | www.frontiersin.org 7June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
and secretion of amylin may be severely impaired, thus leading
to states of hypoamylinemia during late stages of T2DM (Qiu
et al., 2014). Hence, both hyper- and hypo-amylinemia likely
play a role in metabolic diseases such as T2DM depending on
the clinical course and duration of the disease (Zhang et al.,
2014).
The consideration of these temporal changes in amylin
production may be important for attempts to explain why both
hypo- and hyper-amylinemic conditions have been associated
with increased amyloid formation in the CNS. On speculative
ground, hyperamylinemic conditions may be more relevant
for early processes of amyloid formation in the CNS, whereas
hypoamylinemic conditions may be more strongly associated
with late stages of central amyloid pathologies (Figure 1).
The former conditions could indeed offer an explanation why
amylin deposits can be present in brain parenchyma of AD
patients who do not suffer from full-blown T2DM (Jackson
et al., 2013), whereas the latter condition could explain why
augmentation of non-amyloidogenic amylin can exert beneficial
effects on amyloid clearance from the CNS once marked amyloids
had already been established (Adler et al., 2014; Zhu et al.,
2015).
Concluding Remarks
Epidemiological studies have established a clear association
between metabolic and neurodegenerative disorders in general
(Kleinridders et al., 2015), and between T2DM and AD in
particular (Ott et al., 1996; Li and Hölscher, 2007; Butterfield
et al., 2014; Desai et al., 2014). Amylin seems to be an
important player at the interface between these metabolic
and neurodegenerative disorders for several reasons. First,
abnormal amylin production is a hallmark peripheral pathology
both in the early (pre-diabetic) and late phases of T2DM,
where hyperamylinemic (early phase) and hypoamylinemic (late
phase) conditions coincide with hyper- and hypo-insulinemia,
respectively (Figure 1). Second, there are notable biochemical
similarities between amylin and Aβ, which (in some but not all
species) are prone to self-aggregation and amyloid formation.
The propensity of amylin to form amyloid plaques is not
restricted to the peripheral organs such as pancreatic islet cells,
but readily extends to the CNS, where is has been found to co-
localize with Aβplaques in at least a subset of AD patients.
Hence, amylin may constitute a “second amyloid” relevant
for the etiopathogenesis of AD and related neurodegenerative
FIGURE 1 | Model for the temporal association between abnormal
amylin production and peripheral and central pathologies relevant to
neurodegenerative disorders. The graphical illustration shows the putative
relationship between age (x-axis) and peripheral amylin levels (y-axis) under
normal conditions (blue) and conditions of type 2 diabetes mellitus (T2DM;
red). The early phase of T2DM is typically associated with increased
peripheral amylin (and insulin, not shown) secretion. Chronic over-production
of amylin during the early stages of T2DM may facilitate amylin amyloid
formation in pancreatic islet cells and eventually promote islet cell loss. As a
consequence of the latter, the production and secretion of amylin (and
insulin, not shown) may be severely impaired, thus leading to states of
hypoamylinemia during late stages of T2DM. The initial hyperamylinemic
condition occurring during the early phase of T2DM may further lead to
amylin amyloid formation in the brain, which in turn may facilitate central Aβ
amyloid formation and associated neurodegenerative processes during the
late phase of T2DM.
Frontiers in Neuroscience | www.frontiersin.org 8June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
disorders (Jackson et al., 2013). In T2DM, the precise relationship
between altered peripheral amylin production and central amylin
amyloid formation appears complex and may be critically
dependent on the temporal course of progressive pancreatic β-
cell dysfunctions: Hyperamylinemic conditions may be more
relevant for the early processes of amyloid formation in the
CNS, whereas hypoamylinemic conditions may be more strongly
associated with late stages of central amyloid pathologies.
Normalization of hyperamylinemia during the early phase
of T2DM may attenuate of even prevent subsequent amylin
amyloid formation in the CNS, thereby exerting beneficial effects
on progressive neurodegenerative disorders such as AD. On
the other hand, augmentation of non-amyloidogenic amylin
during late phases of T2DM, which are characterized by
hypoamylinemic conditions, may offer a therapeutic strategy
to facilitate amyloid clearance from the CNS and to block
ongoing neurodegenerative processes once marked amyloids are
established. More in-depth knowledge about these temporal
relationships seems highly desirable as it may optimize the
efficacy of amylin-based interventions in the treatment of
AD and related neurodegenerative disorders with metabolic
comorbidities.
Acknowledgments
TL receives financial support granted by the Swiss National
Science Foundation (31003A_156935), the European Union’s
Seventh Framework Programme (FP7/2007–2011) under
grant agreement No. 305707, the National Health and
Medical Research Council (NIDDK). UM receives financial
support granted by the Swiss National Science Foundation
(310030_146217), the European Union’s Seventh Framework
Programme (FP7/2007–2011) under grant agreement No.
259679, the National Health and Medical Research Council
(NHMRC), Australia (APP1057883), and ETH Zurich,
Switzerland (ETH Research Grant 25_13-2).
References
Adler, B. L., Yarchoan, M., Hwang, H. M., Louneva, N., Blair, J. A., Palm, R.,
et al. (2014). Neuroprotective effects of the amylin analogue pramlintide on
Alzheimer’s disease pathogenesis and cognition. Neurobiol. Aging 35, 793–801.
doi: 10.1016/j.neurobiolaging.2013.10.076
Arnelo, U., Permert, J., Adrian, T. E., Larsson, J., Westermark, P., and Reidelberger,
R. D. (1996). Chronic infusion of islet amyloid polypeptide causes anorexia in
rats. Am. J. Physiol. 271, R1654–R1659.
Arnoldussen, I. A., Kiliaan, A. J., and Gustafson, D. R. (2014). Obesity and
dementia: adipokines interact with the brain. Eur. Neuropsychopharmacol. 24,
1982–1999. doi: 10.1016/j.euroneuro.2014.03.002
Bailey, R. J., Walker, C. S., Ferner, A. H., Loomes, K. M., Prijic, G., Halim,
A., et al. (2012). Pharmacological characterization of rat amylin receptors:
implications for the identification of amylin receptor subtypes. Br. J. Pharmacol.
166, 151–167. doi: 10.1111/j.1476-5381.2011.01717.x
Baisley, S. K., and Baldo, B. A. (2014). Amylin receptor signaling in
the nucleus accumbens negatively modulates µ-opioid-driven feeding.
Neuropsychopharmacology 39, 3009–3017. doi: 10.1038/npp.2014.153
Banks, W. A., and Kastin, A. J. (1998). Differential permeability of the blood-brain
barrier to two pancreatic peptides: insulin and amylin. Peptides 19, 883–889.
doi: 10.1016/S0196-9781(98)00018-7
Baranello, R. J., Bharani, K. L., Padmaraju, V., Chopra, N., Lahiri, D. K., Greig,
N. H., et al. (2015). Amyloid-beta protein clearance and degradation (ABCD)
pathways and their role in Alzheimer’s disease. Curr. Alzheimer Res. 12, 32–46.
doi: 10.2174/1567205012666141218140953
Bassil, F., Fernagut, P. O., Bezard, E., and Meissner, W. G. (2014). Insulin, IGF-
1 and GLP-1 signaling in neurodegenerative disorders: targets for disease
modification? Prog. Neurobiol. 118, 1–18. doi: 10.1016/j.pneurobio.2014.02.005
Bennett, R. G., Hamel, F. G., and Duckworth, W. C. (2003). An insulin-degrading
enzyme inhibitor decreases amylin degradation, increases amylin-induced
cytotoxicity, and increases amyloid formation in insulinoma cell cultures.
Diabetes 52, 2315–2320. doi: 10.2337/diabetes.52.9.2315
Berridge, K. C., and Robinson, T. E. (1998). What is the role of dopamine in reward:
hedonic impact, reward learning, or incentive salience? Brain Res. Brain Res.
Rev. 28, 309–369. doi: 10.1016/S0165-0173(98)00019-8
Betsholtz, C., Christmansson, L., Engström, U., Rorsman, F., Svensson, V.,
Johnson, K. H., et al. (1989). Sequence divergence in a specific region of islet
amyloid polypeptide (IAPP) explains differences in islet amyloid formation
between species. FEBS Lett. 251, 261–264. doi: 10.1016/0014-5793(89)81467-X
Bhavsar, S., Watkins, J., and Young, A. (1998). Synergy between amylin and
cholecystokinin for inhibition of food intake in mice. Physiol. Behav. 64,
557–561. doi: 10.1016/S0031-9384(98)00110-3
Boyle, C. N., and Lutz, T. A. (2011). Amylinergic control of food
intake in lean and obese rodents. Physiol. Behav. 105, 129–137. doi:
10.1016/j.physbeh.2011.02.015
Boyle, C. N., Rossier, M. M., and Lutz, T. A. (2011). Influence of high-fat feeding,
diet-induced obesity, and hyperamylinemia on the sensitivity to acute amylin.
Physiol. Behav. 104, 20–28. doi: 10.1016/j.physbeh.2011.04.044
Braegger, F. E., Asarian, L., Dahl, K., Lutz, T. A., and Boyle, C. N. (2014). The role
of the area postrema in the anorectic effects of amylin and salmon calcitonin:
behavioral and neuronal phenotyping. Eur. J. Neurosci. 40, 3055–3066. doi:
10.1111/ejn.12672
Bursch, W., Ellinger, A., Gerner, C., Fröhwein, U., and Schulte-Hermann, R.
(2000). Programmed cell death (PCD). Apoptosis, autophagic PCD, or others?
Ann. N.Y. Acad. Sci. 926, 1–12. doi: 10.1111/j.1749-6632.2000.tb05594.x
Butterfield, D. A., Di Domenico, F., and Barone, E. (2014). Elevated risk
of type 2 diabetes for development of Alzheimer disease: a key role for
oxidative stress in brain. Biochim. Biophys. Acta 1842, 1693–1706. doi:
10.1016/j.bbadis.2014.06.010
Chen, Z., and Zhong, C. (2014). Oxidative stress in Alzheimer’s disease. Neurosci.
Bull. 30, 271–281. doi: 10.1007/s12264-013-1423-y
Chiang, D. J., Pritchard, M. T., and Nagy, L. E. (2011). Obesity, diabetes mellitus,
and liver fibrosis. Am. J. Physiol. Gastrointest. Liver Physiol. 300, G697–G702.
doi: 10.1152/ajpgi.00426.2010
Clark, A., and Nilsson, M. R. (2004). Islet amyloid: a complication of islet
dysfunction or an aetiological factor in Type 2 diabetes? Diabetologia 47,
157–169. doi: 10.1007/s00125-003-1304-4
Davenport, E. L., Morgan, G. J., and Davies, F. E. (2008). Untangling the unfolded
protein response. Cell Cycle 7, 865–869. doi: 10.4161/cc.7.7.5615
de la Monte, S. M. (2012). Contributions of brain insulin resistance and deficiency
in amyloid-related neurodegeneration in Alzheimer’s disease. Drugs 72, 49–66.
doi: 10.2165/11597760-000000000-00000
de la Monte, S. M., and Tong, M. (2014). Brain metabolic dysfunction at
the core of Alzheimer’s disease. Biochem. Pharmacol. 88, 548–559. doi:
10.1016/j.bcp.2013.12.012
de la Monte, S. M., and Wands, J. R. (2008). Alzheimer’s disease is type
3 diabetes-evidence reviewed. J. Diabetes Sci. Technol. 2, 1101–1113. doi:
10.1177/193229680800200619
Desai, G. S., Zheng, C., Geetha, T., Mathews, S. T., White, B. D., Huggins, K.
W., et al. (2014). The pancreas-brain axis: insight into disrupted mechanisms
associating type 2 diabetes and Alzheimer’s disease. J. Alzheimers Dis. 42,
347–356. doi: 10.3233/JAD-140018
Donath, M. Y., Dalmas, É., Sauter, N. S., and Böni-Schnetzler, M. (2013).
Inflammation in obesity and diabetes: islet dysfunction and therapeutic
opportunity. Cell Metab. 17, 860–872. doi: 10.1016/j.cmet.2013.05.001
Frontiers in Neuroscience | www.frontiersin.org 9June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
Donath, M. Y., and Shoelson, S. E. (2011). Type 2 diabetes as an inflammatory
disease. Nat. Rev. Immunol. 11, 98–107. doi: 10.1038/nri2925
Edvinsson, L., Goadsby, P. J., and Uddman, R. (2001). Amylin: localization,
effects on cerebral arteries and on local cerebral blood flow in the cat.
ScientificWorldJournal 1, 168–180. doi: 10.1100/tsw.2001.23
Edwards, G., Bronislava, G., Jodka, C., Dilts, R. P., Miller, C. C., and Young, A.
(1998). Area postrem (AP)-lesions block the regulation of gastric emptying by
amylin. Neurogastroenterol. Motil. 10, 26.
Eikelenboom, P., van Exel, E., Hoozemans, J. J., Veerhuis, R., Rozemuller, A. J.,
and van Gool, W. A. (2010). Neuroinflammation - an early event in both the
history and pathogenesis of Alzheimer’s disease. Neurodegener. Dis. 7, 38–41.
doi: 10.1159/000283480
Emmerzaal, T. L., Kiliaan, A. J., and Gustafson, D. R. (2015). 2003-2013: a decade
of body mass index, Alzheimer’s disease, and dementia. J. Alzheimers Dis. 43,
739–755. doi: 10.3233/JAD-141086
Enoki, S., Mitsukawa, T., Takemura, J., Nakazato, M., Aburaya, J., Toshimori,
H., et al. (1992). Plasma islet amyloid polypeptide levels in obesity, impaired
glucose tolerance and non-insulin-dependent diabetes mellitus. Diabetes Res.
Clin. Pract. 15, 97–102. doi: 10.1016/0168-8227(92)90074-2
Farris, W., Mansourian, S., Chang, Y., Lindsley, L., Eckman, E. A., Frosch,
M. P., et al. (2003). Insulin-degrading enzyme regulates the levels of
insulin, amyloid beta-protein, and the beta-amyloid precursor protein
intracellular domain in vivo.Proc. Natl. Acad. Sci. U.S.A. 100, 4162–4167. doi:
10.1073/pnas.0230450100
Fawver, J. N., Ghiwot, Y., Koola, C., Carrera, W., Rodriguez-Rivera, J.,
Hernandez, C., et al. (2014). Islet amyloid polypeptide (IAPP): a second
amyloid in Alzheimer’s disease. Curr. Alzheimer Res. 11, 928–940. doi:
10.2174/1567205011666141107124538
Fernandes-Santos, C., Zhang, Z., Morgan, D. A., Guo, D. F., Russo, A. F.,
and Rahmouni, K. (2013). Amylin acts in the central nervous system
to increase sympathetic nerve activity. Endocrinology 157, 2481–2488. doi:
10.1210/en.2012-2172
Ferrer, I. (2012). Defining Alzheimer as a common age-related neurodegenerative
process not inevitably leading to dementia. Prog. Neurobiol. 97, 38–51. doi:
10.1016/j.pneurobio.2012.03.005
Friedman, J. M. (1997). Leptin, leptin receptors and the control of body weight.
Eur. J. Med. Res. 2, 7–13.
Fukuda, T., Hirai, Y., Maezawa, H., Kitagawa, Y., and Funahashi, M.
(2013). Electrophysiologically identified presynaptic mechanisms underlying
amylinergic modulation of area postrema neuronal excitability in rat brain
slices. Brain Res. 1494, 9–16. doi: 10.1016/j.brainres.2012.11.051
Ginter, E., and Simko, V. (2012a). Global prevalence and future of diabetes
mellitus. Adv. Exp. Med. Biol. 771, 35–41. doi: 10.1007/978-1-4614-
5441-0_5
Ginter, E., and Simko, V. (2012b). Type 2 diabetes mellitus, pandemic in 21st
century. Adv. Exp. Med. Biol. 771, 42–50. doi: 10.1007/978-1-4614-5441-0_6
Götz, J., Lim, Y. A., and Eckert, A. (2013). Lessons from two prevalent
amyloidoses-what amylin and Aβhave in common. Front. Aging Neurosci. 5:38.
doi: 10.3389/fnagi.2013.00038
Greenwood, C. E., and Winocur, G. (2005). High-fat diets, insulin resistance
and declining cognitive function. Neurobiol. Aging 26(Suppl. 1), 42–45. doi:
10.1016/j.neurobiolaging.2005.08.017
Guardado-Mendoza, R., Davalli, A. M., Chavez, A. O., Hubbard, G. B., Dick,
E. J., Majluf-Cruz, A., et al. (2009). Pancreatic islet amyloidosis, beta-cell
apoptosis, and alpha-cell proliferation are determinants of islet remodeling in
type-2 diabetic baboons. Proc. Natl. Acad. Sci. U.S.A. 106, 13992–13997. doi:
10.1073/pnas.0906471106
Gurlo, T., Ryazantsev, S., Huang, C. J., Yeh, M. W., Reber, H. A., Hines, O. J., et al.
(2010). Evidence for proteotoxicity in beta cells in type 2 diabetes: toxic islet
amyloid polypeptide oligomers form intracellularly in the secretory pathway.
Am. J. Pathol. 176, 861–869. doi: 10.2353/ajpath.2010.090532
Hampel, H., Prvulovic, D., Teipel, S., Jessen, F., Luckhaus, C., Frölich, L., et al.
(2011). German Task Force on Alzheimer’s Disease (GTF-AD). The future
of Alzheimer’s disease: the next 10 years. Prog. Neurobiol. 95, 718–728. doi:
10.1016/j.pneurobio.2011.11.008
Hardy, J., and Selkoe, D. J. (2002). The amyloid hypothesis of Alzheimer’s disease:
progress and problems on the road to therapeutics. Science 297, 353–356. doi:
10.1126/science.1072994
Höppener, J. W., and Lips, C. J. (2006). Role of islet amyloid in type
2 diabetes mellitus. Int. J. Biochem. Cell Biol. 38, 726–736. doi:
10.1016/j.biocel.2005.12.009
Höppener, J. W., Oosterwijk, C., van Hulst, K. L., Verbeek, J. S., Capel, P. J.,
de Koning, E. J., et al. (1994). Molecular physiology of the islet amyloid
polypeptide (IAPP)/amylin gene in man, rat, and transgenic mice. J. Cell.
Biochem. 55(Suppl), 39–53. doi: 10.1002/jcb.240550006
Hou, X., Ling, Z., Quartier, E., Foriers, A., Schuit, F., Pipeleers, D., et al. (1999).
Prolonged exposure of pancreatic beta cells to raised glucose concentrations
results in increased cellular content of islet amyloid polypeptide precursors.
Diabetologia 42, 188–194. doi: 10.1007/s001250051138
Huang, C. J., Lin, C. Y., Haataja, L., Gurlo, T., Butler,A. E., Rizza, R. A., et al. (2007).
High expression rates of human islet amyloid polypeptide induce endoplasmic
reticulum stress mediated beta-cell apoptosis, a characteristic of humans with
type 2 but not type 1 diabetes. Diabetes 56, 2016–2027. doi: 10.2337/db07-0197
Hurd, M. D., Martorell, P., Delavande, A., Mullen, K. J., and Langa, K. M.
(2013). Monetary costs of dementia in the United States. N. Engl. J. Med. 368,
1326–1334. doi: 10.1056/NEJMsa1204629
Isaksson, B., Wang, F., Permert, J., Olsson, M., Fruin, B., Herrington, M. K., et al.
(2005). Chronically administered islet amyloid polypeptide in rats serves as an
adiposity inhibitor and regulates energy homeostasis. Pancreatology 5, 29–36.
doi: 10.1159/000084488
Jackson, K., Barisone, G. A., Diaz, E., Jin, L. W., DeCarli, C., and Despa, F. (2013).
Amylin deposition in the brain: a second amyloid in Alzheimer disease? Ann.
Neurol. 74, 517–526. doi: 10.1002/ana.23956
Jaikaran, E. T., and Clark, A. (2001). Islet amyloid and type 2 diabetes: from
molecular misfolding to islet pathophysiology. Biochim. Biophys. Acta 1537,
179–203. doi: 10.1016/S0925-4439(01)00078-3
Jaikaran, E. T., Higham, C. E., Serpell, L. C., Zurdo, J., Gross, M., Clark, A., et al.
(2001). Identification of a novel human islet amyloid polypeptide beta-sheet
domain and factors influencing fibrillogenesis. J. Mol. Biol. 308, 515–525. doi:
10.1006/jmbi.2001.4593
Jha, S., Snell, J. M., Sheftic, S. R., Patil, S. M., Daniels, S. B., Kolling, F. W., et al.
(2014). pH dependence of amylin fibrillization. Biochemistry 53, 300–310. doi:
10.1021/bi401164k
Johnson, K. H., O’Brien, T. D., Jordan, K., and Westermark, P. (1989). Impaired
glucose tolerance is associated with increased islet amyloid polypeptide (IAPP)
immunoreactivity in pancreatic beta cells. Am. J. Pathol. 135, 245–250.
Jurgens, C. A., Toukatly, M. N., Fligner, C. L., Udayasankar, J., Subramanian, S.
L., Zraika, S., et al. (2011). β-cell loss and β-cell apoptosis in human type 2
diabetes are related to islet amyloid deposition. Am. J. Pathol. 178, 2632–2640.
doi: 10.1016/j.ajpath.2011.02.036
Kahn, S. E., Cooper, M. E., and Del Prato, S. (2014). Pathophysiologyand tre atment
of type 2 diabetes: perspectives on the past, present, and future. Lancet 383,
1068–1083. doi: 10.1016/S0140-6736(13)62154-6
Kahn, S. E., D’Alessio, D. A., Schwartz, M. W., Fujimoto, W. Y., Ensinck,
J. W., Taborsky, G. J. Jr, et al. (1990). Evidence of cosecretion of islet
amyloid polypeptide and insulin by beta-cells. Diabetes 39, 634–638. doi:
10.2337/diab.39.5.634
Karran, E., Mercken, M., and De Strooper, B. (2011). The amyloid cascade
hypothesis for Alzheimer’s disease: an appraisal for the development of
therapeutics. Nat. Rev. Drug Discov. 10, 698–712. doi: 10.1038/nrd3505
Kenny, P. J. (2011). Common cellular and molecular mechanisms in obesity and
drug addiction. Nat. Rev. Neurosci. 12, 638–651. doi: 10.1038/nrn3105
Kiliaan, A. J., Arnoldussen, I. A., and Gustafson, D. R. (2014). Adipokines:
a link between obesity and dementia? Lancet Neurol. 13, 913–923. doi:
10.1016/S1474-4422(14)70085-7
Kleinridders, A., Cai, W., Cappellucci, L., Ghazarian, A., Collins, W. R., Vienberg,
S. G., et al. (2015). Insulin resistance in brain alters dopamine turnover and
causes behavioral disorders. Proc. Natl. Acad. Sci. U.S.A. 112, 3463–3468. doi:
10.1073/pnas.1500877112
Leboucher, A., Laurent, C., Fernandez-Gomez, F. J., Burnouf, S., Troquier, L.,
Eddarkaoui, S., et al. (2013). Detrimental effects of diet-induced obesity on τ
pathology are independent of insulin resistance in τtransgenic mice. Diabetes
62, 1681–1688. doi: 10.2337/db12-0866
Li, L., and Hölscher, C. (2007). Common pathological processes in Alzheimer
disease and type 2 diabetes: a review. Brain Res. Rev. 56, 384–402. doi:
10.1016/j.brainresrev.2007.09.001
Frontiers in Neuroscience | www.frontiersin.org 10 June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
Li, Y., Xu, W., Mu, Y., and Zhang, J. Z. (2013). Acidic pH retards the fibrillization
of human Islet Amyloid Polypeptide due to electrostatic repulsion of histidines.
J. Chem. Phys. 139, 055102. doi: 10.1063/1.4817000
Lutz, T. A. (2005). Pancreatic amylin as a centrally acting satiating hormone. Curr.
Drug Targets 6, 181–189. doi: 10.2174/1389450053174596
Lutz, T. A. (2009). Control of food intake and energy expenditure by amylin—
therapeutic implications. Int. J. Obes. (Lond.), 33(Suppl. 1), S24–S27. doi:
10.1038/ijo.2009.13
Lutz, T. A. (2010). The role of amylin in the control of energy homeostasis.
Am. J. Physiol. Regul. Integr. Comp. Physiol. 298, R1475–R1484. doi:
10.1152/ajpregu.00703.2009
Lutz, T. A., Althaus, J., Rossi, R., and Scharrer, E. (1998a). Anorectic effect of
amylin is not transmitted by capsaicin-sensitive nerve fibers. Am. J. Physiol.
274, R1777–R1782.
Lutz, T. A., Del Prete, E., and Scharrer, E. (1995a). Subdiaphragmatic vagotomy
does not influence the anorectic effect of amylin. Peptides 16, 457–462. doi:
10.1016/0196-9781(94)00203-I
Lutz, T. A., Geary, N., Szabady, M. M., Del Prete, E., and Scharrer, E. (1995b).
Amylin decreases meal size in rats. Physiol. Behav. 58, 1197–1202. doi:
10.1016/0031-9384(95)02067-5
Lutz, T. A., Mollet, A., Rushing, P. A., Riediger, T., and Scharrer, E. (2001). The
anorectic effect of a chronic peripheral infusion of amylin is abolished in area
postrema/nucleus of the solitary tract (AP/NTS) lesioned rats. Int. J. Obes. Relat.
Metab. Disord. 25, 1005–1011. doi: 10.1038/sj.ijo.0801664
Lutz, T. A., and Rand, J. S. (1993). A review of new developments in type 2
diabetes in human beings and cats. Br. Vet. J. 149, 527–536. doi: 10.1016/S0007-
1935(05)80037-5
Lutz, T. A., Senn, M., Althaus, J., Del Prete, E., Ehrensperger, F., and Scharrer, E.
(1998b). Lesion of the area postrema/nucleus of the solitary tract (AP/NTS)
attenuates the anorectic effects of amylin and calcitonin gene-related peptide
(CGRP) in rats. Peptides 19, 309–317. doi: 10.1016/S0196-9781(97)00292-1
Mack, C., Wilson, J., Athanacio, J., Reynolds, J., Laugero, K., Guss, S., et al.
(2007). Pharmacological actions of the peptide hormone amylin in the
long-term regulation of food intake, food preference, and body weight.
Am. J. Physiol. Regul. Integr. Comp. Physiol. 293, R1855–R1863. doi:
10.1152/ajpregu.00297.2007
Mack, C. M., Soares, C. J., Wilson, J. K., Athanacio, J. R., Turek, V. F., Trevaskis,
J. L., et al. (2010). Davalintide (AC2307), a novel amylin-mimetic peptide:
enhanced pharmacological properties over native amylin to reduce food
intake and body weight. Int. J. Obes. (Lond.) 34, 385–395. doi: 10.1038/ijo.
2009.238
Marciniak, S. J., Garcia-Bonilla, L., Hu, J., Harding, H. P., and Ron, D. (2006).
Activation-dependent substrate recruitment by the eukaryotic translation
initiation factor 2 kinase PERK. J. Cell. Biol. 172, 201–209.
Marciniak, S. J., and Ron, D. (2006). Endoplasmic reticulum stress signaling in
disease. Physiol. Rev. 86, 1133–1149. doi: 10.1152/physrev.00015.2006
Marzban, L., Trigo-Gonzalez, G., and Verchere, C. B. (2005). Processing of pro-
islet amyloid polypeptide in the constitutive and regulated secretory pathways
of beta cells. Mol. Endocrinol. 19, 2154–2163. doi: 10.1210/me.2004-0407
Marzban, L., Trigo-Gonzalez, G., Zhu, X., Rhodes, C. J., Halban, P. A., Steiner,
D. F., et al. (2004). Role of beta-cell prohormone convertase (PC)1/3 in
processing of pro-islet amyloid polypeptide. Diabetes 53, 141–148. doi:
10.2337/diabetes.53.1.141
Masters, S. L, Dunne, A., Subramanian, S. L., Hull, R. L., Tannahill, G. M., Sharp,
F. A., et al. (2010). Activation of the NLRP3 inflammasome by islet amyloid
polypeptide provides a mechanism for enhanced IL-1βin type 2 diabetes. Nat.
Immunol. 11, 897–904. doi: 10.1038/ni.1935
Matveyenko, A. V., and Butler, P. C. (2006). Islet amyloid polypeptide (IAPP)
transgenic rodents as models for type 2 diabetes. ILAR J. 47, 225–233. doi:
10.1093/ilar.47.3.225
McDonald, R. J. (2002). Multiple combinations of co-factors produce variants of
age-related cognitive decline: a theory. Can. J. Exp. Psychol. 56, 221–239. doi:
10.1037/h0087399
Mietlicki-Baase, E. G., and Hayes, M. R. (2014). Amylin activates distributed
CNS nuclei to control energy balance. Physiol. Behav. 136, 39–46. doi:
10.1016/j.physbeh.2014.01.013
Mietlicki-Baase, E. G., Reiner, D. J., Cone, J. J., Olivos, D. R., McGrath, L. E.,
Zimmer, D. J., et al. (2015). Amylin modulates the mesolimbic dopamine
system to control energy balance. Neuropsychopharmacology 40, 372–385. doi:
10.1038/npp.2014.180
Mietlicki-Baase, E. G., Rupprecht, L. E., Olivos, D. R., Zimmer, D. J., Alter,
M. D., Pierce, R. C., et al. (2013). Amylin receptor signaling in the ventral
tegmental area is physiologically relevant for the control of food intake.
Neuropsychopharmacology 38, 1685–1697. doi: 10.1038/npp.2013.66
Mollet, A., Gilg, S., Riediger, T., and Lutz, T. A. (2004). Infusion of the amylin
antagonist AC 187 into the area postrema increases food intake in rats. Physiol.
Behav. 81, 149–155. doi: 10.1016/j.physbeh.2004.01.006
Mollet, A., Meier, S., Grabler, V., Gilg, S., Scharrer, E., and Lutz, T. A. (2003).
Endogenous amylin contributes to the anorectic effects of cholecystokinin and
bombesin. Peptides 24, 91–98. doi: 10.1016/S0196-9781(02)00280-2
Morales, I., Guzmán-Martínez, L., Cerda-Troncoso, C., Farías, G. A., and
Maccioni, R. B. (2014). Neuroinflammation in the pathogenesis of Alzheimer’s
disease. A rational framework for the search of novel therapeutic approaches.
Front. Cell. Neurosci. 8:112. doi: 10.3389/fncel.2014.00112
Moriarty, D. F., and Raleigh, D. P. (1999). Effects of sequential proline
substitutions on amyloid formation by human amylin20-29. Biochemistry 38,
1811–1818. doi: 10.1021/bi981658g
Morris, M. C., and Tangney, C. C. (2014). Dietary fat composition
and dementia risk. Neurobiol. Aging 35(Suppl. 2), S59–S64. doi:
10.1016/j.neurobiolaging.2014.03.038
Nedergaard, J., and Cannon, B. (2014). The browning of white adipose tissue:
some burning issues. Cell Metab. 20, 396–407. doi: 10.1016/j.cmet.2014.
07.005
Osaka, T., Tsukamoto,A., Koyama, Y., and Inoue, S. (2008). Central and peripheral
administration of amylin induces energy expenditure in anesthetized rats.
Peptides 29, 1028–1035. doi: 10.1016/j.peptides.2008.02.002
Oskarsson, M. E., Paulsson, J. F., Schultz, S. W., Ingelsson, M., Westermark, P.,
and Westermark, G. T. (2015). In vivo seeding and cross-seeding of localized
amyloidosis: a molecular link between type 2 diabetes and Alzheimer disease.
Am. J. Pathol. 185, 834–846. doi: 10.1016/j.ajpath.2014.11.016
Osto, M., Zini, E., Reusch, C. E., and Lutz, T. A. (2013). Diabetes from
humans to cats. Gen. Comp. Endocrinol. 182, 48–53. doi: 10.1016/j.ygcen.2012.
11.019
Ott, A., Stolk, R. P., Hofman, A., van Harskamp, F., Grobbee, D. E., and Breteler,
M. M. (1996). Association of diabetes mellitus and dementia: the Rotterdam
Study. Diabetologia 39, 1392–1397. doi: 10.1007/s001250050588
Oyadomari, S., Araki, E., and Mori, M. (2002). Endoplasmic reticulum stress-
mediated apoptosis in pancreatic beta-cells. Apoptosis 7, 335–345. doi:
10.1023/A:1016175429877
Parks, J. K., Smith, T. S., Trimmer, P. A., Bennett, J. P. Jr., and Parker, W.
D. Jr. (2001). Neurotoxic Abeta peptides increase oxidative stress in vivo
through NMDA-receptor and nitric-oxide-synthase mechanisms, and inhibit
complex IV activity and induce a mitochondrial permeability transition in vitro.
J. Neurochem. 76, 1050–1056. doi: 10.1046/j.1471-4159.2001.00112.x
Paulsson, J. F., Andersson, A., Westermark, P., and Westermark, G. T. (2006).
Intracellular amyloid-like deposits contain unprocessed pro-islet amyloid
polypeptide (proIAPP) in beta cells of transgenic mice overexpressing the gene
for human IAPP and transplanted human islets. Diabetologia 49, 1237–1246.
doi: 10.1007/s00125-006-0206-7
Paulsson, J. F., and Westermark, G. T. (2005). Aberrant processing of human
proislet amyloid polypeptide results in increased amyloid formation. Diabetes
54, 2117–2125. doi: 10.2337/diabetes.54.7.2117
Pieber, T. R., Roitelman, J., Lee, Y., Luskey, K. L., and Stein, D. T. (1994). Direct
plasma radioimmunoassay for rat amylin-(1-37): concentrations with acquired
and genetic obesity. Am. J. Physiol. 267, E156–E164.
Potes, C. S., Turek, V. F., Cole, R. L., Vu, C., Roland, B. L., Roth, J. D., et al. (2010).
Noradrenergic neurons of the area postrema mediate amylin’s hypophagic
action. Am. J. Physiol. Regul. Integr. Comp. Physiol. 299, R623–R631. doi:
10.1152/ajpregu.00791.2009
Preston, A. M., Gurisik, E., Bartley, C., Laybutt, D. R., and Biden, T. J. (2009).
Reduced endoplasmic reticulum (ER)-to-Golgi protein trafficking contributes
to ER stress in lipotoxic mouse beta cells by promoting protein overload.
Diabetologia 52, 2369–2373. doi: 10.1007/s00125-009-1506-5
Qiu, W. Q., Li, H., Zhu, H., Scott, T., Mwamburi, M., Rosenberg, I., et al. (2014).
Plasma amylin and cognition in diabetes in the absence and the presence of
insulin treatment. J. Diabetes Metab. 5, 458. doi: 10.4172/2155-6156.1000458
Frontiers in Neuroscience | www.frontiersin.org 11 June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
Qiu, W. Q., and Zhu, H. (2014). Amylin and its analogs: a friend or foe
for the treatment of Alzheimer’s disease? Front. Aging Neurosci. 6:186. doi:
10.3389/fnagi.2014.00186
Reidelberger, R. D., Haver, A. C., Arnelo, U., Smith, D. D., Schaffert, C. S.,
and Permert, J. (2004). Amylin receptor blockade stimulates food intake
in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 287, R568–R574. doi:
10.1152/ajpregu.00213.2004
Reidelberger, R. D., Kelsey, L., and Heimann, D. (2002). Effects of amylin-
related peptides on food intake, meal patterns, and gastric emptying in
rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 282, R1395–R1404. doi:
10.1152/ajpregu.00597.2001
Reitz, C., Brayne, C., and Mayeux, R. (2011). Epidemiology of Alzheimer disease.
Nat. Rev. Neurol. 7, 137–152. doi: 10.1038/nrneurol.2011.2
Reitz, C., and Mayeux, R. (2014). Alzheimer disease: epidemiology, diagnostic
criteria, risk factors and biomarkers. Biochem. Pharmacol. 88, 640–651. doi:
10.1016/j.bcp.2013.12.024
Riediger, T., Zuend, D., Becskei, C., and Lutz, T. A. (2004). The anorectic hormone
amylin contributes to feeding-related changes of neuronal activity in key
structures of the gut-brain axis. Am. J. Physiol. Regul. Integr. Comp. Physiol.
286, R114–R122. doi: 10.1152/ajpregu.00333.2003
Ríos, J. A., Cisternas, P., Arrese, M., Barja, S., and Inestrosa, N. C.
(2014). Is Alzheimer’s disease related to metabolic syndrome? A
Wnt signaling conundrum. Prog. Neurobiol. 121, 125–146. doi:
10.1016/j.pneurobio.2014.07.004
Roth, J. D., Hughes, H., Kendall, E., Baron, A. D., and Anderson, C. M. (2006).
Antiobesity effects of the beta-cell hormone amylin in diet-induced obese rats:
effects on food intake, body weight, composition, energy expenditure, and gene
expression. Endocrinology 147, 5855–5864. doi: 10.1210/en.2006-0393
Rowland, N. E., Crews, E. C., and Gentry, R. M. (1997). Comparison of Fos
induced in rat brain by GLP-1 and amylin. Regul. Pept. 71, 171–174. doi:
10.1016/S0167-0115(97)01034-3
Rushing, P. A., Hagan, M. M., Seeley, R. J., Lutz, T. A., D’Alessio, D. A.,
Air, E. L., et al. (2001). Inhibition of central amylin signaling increases
food intake and body adiposity in rats. Endocrinology 142, 5035–5038. doi:
10.1210/endo.142.11.8593
Rushing, P. A., Seeley, R. J., Air, E. L., Lutz, T. A., and Woods, S. C. (2002).
Acute 3rd-ventricular amylin infusion potently reduces food intake but does
not produce aversive consequences. Peptides 23, 985–988. doi: 10.1016/S0196-
9781(02)00022-0
Scherz-Shouval, R., and Elazar, Z. (2007). ROS, mitochondria and the regulation of
autophagy. Trends Cell Biol. 17, 422–427. doi: 10.1016/j.tcb.2007.07.009
Schwartz, M. W., Woods, S. C., Porte, D. Jr., Seeley, R. J., and Baskin, D. G.
(2000). Central nervous system control of food intake. Nature 404, 661–671.
doi: 10.1038/3500753
Schwartz, M. W., Woods, S. C., Seeley, R. J., Barsh, G. S., Baskin, D. G., and Leibel,
R. L. (2003). Is the energy homeostasis system inherently biased toward weight
gain? Diabetes 52, 232–238. doi: 10.2337/diabetes.52.2.232
Sebastião, I., Candeias, E., Santos, M. S., de Oliveira, C. R., Moreira, P. I.,
and Duarte, A. I. (2014). Insulin as a bridge between type 2 diabetes and
Alzheimer disease - How anti-diabetics could be a solution for dementia. Front.
Endocrinol. 5:110. doi: 10.3389/fendo.2014.00110
Seeman, P., and Seeman, N. (2011). Alzheimer’s disease: β-amyloid plaque
formation in human brain. Synapse 65, 1289–1297. doi: 10.1002/syn.20957
Sexton, P. M., Paxinos, G., Kenney, M. A., Wookey, P. J., and Beaumont, K.
(1994). In vitro autoradiographic localization of amylin binding sites in rat
brain. Neuroscience 62, 553–567. doi: 10.1016/0306-4522(94)90388-3
Srodulski, S., Sharma, S., Bachstetter, A. B., Brelsfoard, J. M., Pascual, C., Xie, X.
S., et al. (2014). Neuroinflammation and neurologic deficits in diabetes linked
to brain accumulation of amylin. Mol. Neurodegener. 9, 30. doi: 10.1186/1750-
1326-9-30
Steneberg, P., Bernardo, L., Edfalk, S., Lundberg, L., Backlund, F., Ostenson,
C. G., et al. (2013). The type 2 diabetes-associated gene ide is required for
insulin secretion and suppression of α-synuclein levels in β-cells. Diabetes 62,
2004–2014. doi: 10.2337/db12-1045
Stienstra, R., Haim, Y., Riahi, Y., Netea, M., Rudich, A., and Leibowitz, G.
(2014). Autophagy in adipose tissue and the beta cell: implications for
obesity and diabetes. Diabetologia 57, 1505–1516. doi: 10.1007/s00125-014-
3255-3
Ueno, M., Chiba, Y., Matsumoto, K., Nakagawa, T., and Miyanaka, H. (2014).
Clearance of beta-amyloid in the brain. Curr. Med. Chem. 21, 4085–4090. doi:
10.2174/0929867321666141011194256
Vandal, M., White, P. J., Tremblay, C., St-Amour, I., Chevrier, G., Emond, V.,
et al. (2014). Insulin reverses the high-fat diet-induced increase in brain Aβ
and improves memory in an animal model of Alzheimer disease. Diabetes 63,
4291–4301. doi: 10.2337/db14-0375
van der Kant, R., and Goldstein, L. S. (2015). Cellular functions of the amyloid
precursor protein from development to dementia. Dev. Cell 32, 502–515. doi:
10.1016/j.devcel.2015.01.022
Volkow, N. D., Wang, G. J., Tomasi, D., and Baler, R. D. (2013). The
addictive dimensionality of obesity. Biol. Psychiatry 73, 811–818. doi:
10.1016/j.biopsych.2012.12.020
Wang, G. J., Volkow, N. D., Logan, J., Pappas, N. R., Wong, C. T., Zhu, W., et al.
(2001). Brain dopamine and obesity. Lancet 357, 354–357. doi: 10.1016/S0140-
6736(00)03643-6
Wang, L., Middleton, C. T., Singh, S., Reddy, A. S., Woys, A. M., Strasfeld,
D. B., et al. (2011). 2DIR spectroscopy of human amylin fibrils reflects
stable β-sheet structure. J. Am. Chem. Soc. 133, 16062–16071. doi: 10.1021/
ja204035k
Watve, M., Bodas, A., and Diwekar, M. (2014). Altered autonomic inputs
as a cause of pancreatic β-cell amyloid. Med. Hypotheses 82, 49–53. doi:
10.1016/j.mehy.2013.11.002
Westermark, G., Arora, M. B., Fox, N., Carroll, R., Chan, S. J., Westermark, P.,
et al. (1995a). Amyloid formation in response to beta cell stress occurs in vitro,
but not in vivo, in islets of transgenic mice expressing human islet amyloid
polypeptide. Mol. Med. 1, 542–553.
Westermark, G. T., Steiner, D. F., Gebre-Medhin, S., Engström, U., and
Westermark, P. (2000). Pro islet amyloid polypeptide (ProIAPP)
immunoreactivity in the islets of Langerhans. Ups. J. Med. Sci. 105,
97–106.
Westermark, P., Andersson, A., and Westermark, G. T. (2011). Islet amyloid
polypeptide, islet amyloid, and diabetes mellitus. Physiol. Rev. 91, 795–826. doi:
10.1152/physrev.00042.2009
Westermark, P., Eizirik, D. L., Pipeleers, D. G., Hellerström, C., and Andersson, A.
(1995b). Rapid deposition of amyloid in human islets transplanted into nude
mice. Diabetologia 38, 543–549. doi: 10.1007/BF00400722
Westermark, P., Li, Z. C., Westermark, G. T., Leckström, A., and Steiner, D.
F. (1996). Effects of beta cell granule components on human islet amyloid
polypeptide fibril formation. FEBS Lett. 379, 203–206. doi: 10.1016/0014-
5793(95)01512-4
Westfall, T. C., and Curfman-Falvey, M. (1995). Amylin-induced relaxation of the
perfused mesenteric arterial bed: meditation by calcitonin gene-related peptide
receptors. J. Cardiovasc. Pharmacol. 26, 932–936. doi: 10.1097/00005344-
199512000-00012
Westwell-Roper, C., Dai, D. L., Soukhatcheva, G., Potter, K. J., van Rooijen,
N., Ehses, J. A., et al. (2011). IL-1 blockade attenuates islet amyloid
polypeptide-induced proinflammatory cytokine release and pancreatic islet
graft dysfunction. J. Immunol. 187, 2755–2765. doi: 10.4049/jimmunol.
1002854
Wickbom, J., Herrington, M. K., Permert, J., Jansson, A., and Arnelo, U. (2008).
Gastric emptying in response to IAPP and CCK in rats with subdiaphragmatic
afferent vagotomy. Regul. Pept. 148, 21–25. doi: 10.1016/j.regpep.2008.
03.010
Wielinga, P. Y., Alder, B., and Lutz, T. A. (2007). The acute effect of amylin and
salmon calcitonin on energy expenditure. Physiol. Behav. 91, 212–217. doi:
10.1016/j.physbeh.2007.02.012
Wielinga, P. Y., Lowenstein, C., Muff, S., Munz, M., Woods, S. C., and
Lutz, T. A. (2010). Central amylin acts as an adiposity signal to control
body weight and energy expenditure. Physiol. Behav. 101, 45–52. doi:
10.1016/j.physbeh.2010.04.012
Winocur, G., and Greenwood, C. E. (2005). Studies of the effects of high fat diets
on cognitive function in a rat model. Neurobiol. Aging 26(Suppl. 1), 46–49. doi:
10.1016/j.neurobiolaging.2005.09.003
Yaffe, K. (2007). Metabolic syndrome and cognitive decline. Curr. Alzheimer Res.
4, 123–126. doi: 10.2174/156720507780362191
Yagui, K., Yamaguchi, T., Kanatsuka, A., Shimada, F., Huang, C. I., Tokuyama,
Y., et al. (1995). Formation of islet amyloid fibrils in beta-secretory granules
Frontiers in Neuroscience | www.frontiersin.org 12 June 2015 | Volume 9 | Article 216
Lutz and Meyer Amylin in neurodegenerative disorders
of transgenic mice expressing human islet amyloid polypeptide/amylin. Eur. J.
Endocrinol. 132, 487–496. doi: 10.1530/eje.0.1320487
Young, A., and Denaro, M. (1998). Roles of amylin in diabetes and in regulation of
nutrient load. Nutrition 14, 524–527.
Zhang, S., Liu, H., Chuang, C. L., Li, X., Au, M., Zhang, L., et al. (2014). The
pathogenic mechanism of diabetes varies with the degree of overexpression and
oligomerization of human amylin in the pancreatic islet βcells. FASEB J. 28,
5083–5096. doi: 10.1096/fj.14-251744
Zhang, Z., Liu, X., Morgan, D. A., Kuburas, A., Thedens, D. R., Russo, A. F.,
et al. (2011). Neuronal receptor activity-modifying protein 1 promotes energy
expenditure in mice. Diabetes 60, 1063–1071. doi: 10.2337/db10-0692
Zhu, H., Wang, X., Wallack, M., Li, H., Carreras, I., Dedeoglu, A., et al. (2015).
Intraperitoneal injection of the pancreatic peptide amylin potently reduces
behavioral impairment and brain amyloid pathology in murine models of
Alzheimer’s disease. Mol. Psychiatry 20, 252–262. doi: 10.1038/mp.2014.17
Conflict of Interest Statement: The authors declare that the research was
conducted in the absence of any commercial or financial relationships that could
be construed as a potential conflict of interest.
Copyright © 2015 Lutz and Meyer. This is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). The use,
distribution or reproduction in other forums is permitted, provided the original
author(s) or licensor are credited and that the original publication in this journal
is cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
Frontiers in Neuroscience | www.frontiersin.org 13 June 2015 | Volume 9 | Article 216
... One of the most thoroughly researched functions of amylin is its effect on satiation, caused by the activation of noradrenergic neurons in the glucose-sensitive area postrema (AP) of the medulla oblongata [27]. Nerve fibers containing amylin have also been found in the nucleus tractus solitarius (NTS) and the lateral and parabrachial nucleus (LPBN), which are secondary sites of amylin action. ...
... Leptin is a hormone secreted by the adipose tissue, signalizing the level of triglycerides in the adipose tissue. In rats, simultaneous exogenous administration of leptin and amylin leads to weight loss, although, in more severe obesity, their synergistic effect was less prominent [27,31]. Moreover, amylin administration in animal models has been shown to increase energy expenditure [32,33]. ...
... It has been shown that amyloid can be found in the pancreas of more than 90% of T2DM patients. Several hypotheses have been postulated to explain how these amylin aggregates can cause β-cell loss due to their toxic effects [27]. Notable proposed mechanisms include endoplasmic reticulum correction mechanisms overloading, disturbance of the intracellular cell transport, disruption of the organelles membranes (e.g., mitochondria), and inflammasome activation [27]. ...
Article
Full-text available
Diabetes mellitus is a devastating chronic metabolic disease. Since the majority of type 2 diabetes mellitus patients are overweight or obese, a novel term—diabesity—has emerged. The gut–brain axis plays a critical function in maintaining glucose and energy homeostasis and involves a variety of peptides. Amylin is a neuroendocrine anorexigenic polypeptide hormone, which is co-secreted with insulin from β-cells of the pancreas in response to food consumption. Aside from its effect on glucose homeostasis, amylin inhibits homeostatic and hedonic feeding, induces satiety, and decreases body weight. In this narrative review, we summarized the current evidence and ongoing studies on the mechanism of action, clinical pharmacology, and applications of amylin and its analogs, pramlintide and cagrilintide, in the field of diabetology, endocrinology, and metabolism disorders, such as obesity.
... A nanomolar affinity interaction between early prefibrillar Aβ(1-40) and IAPP species has been shown in vitro to suppress amyloidogenesis [133], whereas seed amounts of Aβ(1-40) fibrils are able to cross-seed IAPP amyloidogenesis in vitro and in animal models in vivo [23,[133][134][135][136]. Furthermore, IAPP has been reported to co-localize with Aβ plaques in human AD-affected brains, suggesting a possible pathophysiological role for the cross-interaction between the two polypeptides [23,137,138]. Andreeto et al. [139] identified hot regions of the Aβ-IAPP interaction interface as high-affinity binding sites in both cross-and self-association. The hydrophobic COOH-terminal part Aβ(29-40) plays a crucial role in the Aβ(1-40)-IAPP interaction [135]. ...
... IAPP IAPP co-localizes with Aβ plaques in human AD-affected brains [23,137,138]. Epidemiological and pathophysiological evidences suggest that the AD and type 2 diabetes are linked to each other [23,138]. ...
... IAPP IAPP co-localizes with Aβ plaques in human AD-affected brains [23,137,138]. Epidemiological and pathophysiological evidences suggest that the AD and type 2 diabetes are linked to each other [23,138]. ...
Article
Full-text available
Amyloid β (Aβ) oligomers have been linked to Alzheimer’s disease (AD) pathogenesis and are the main neurotoxic forms of Aβ. This review focuses on the following: (i) the Aβ(1–42):calmodulin interface as a model for the design of antagonist Aβ peptides and its limitations; (ii) proteolytic degradation as the major source of highly hydrophobic peptides in brain cells; and (iii) brain peptides that have been experimentally demonstrated to bind to Aβ monomers or oligomers, Aβ fibrils, or Aβ plaques. It is highlighted that the hydrophobic amino acid residues of the COOH-terminal segment of Aβ(1–42) play a key role in its interaction with intracellular protein partners linked to its neurotoxicity. The major source of highly hydrophobic endogenous peptides of 8–10 amino acids in neurons is the proteasome activity. Many canonical antigen peptides bound to the major histocompatibility complex class 1 are of this type. These highly hydrophobic peptides bind to Aβ and are likely to be efficient antagonists of the binding of Aβ monomers/oligomers concentrations in the nanomolar range with intracellular proteins. Also, their complexation with Aβ will protect them against endopeptidases, suggesting a putative chaperon-like physiological function for Aβ that has been overlooked until now. Remarkably, the hydrophobic amino acid residues of Aβ responsible for the binding of several neuropeptides partially overlap with those playing a key role in its interaction with intracellular protein partners that mediates its neurotoxicity. Therefore, these latter neuropeptides are also potential candidates to antagonize Aβ peptides binding to target proteins. In conclusion, the analysis performed in this review points out that hydrophobic endogenous brain neuropeptides could be valuable biomarkers to evaluate the risk of the onset of sporadic AD, as well as for the prognosis of AD.
... The formation of hIAPP amyloids is strongly dependent on the region 20-29 (known as the amyloidogenic region). 100 Other studies 101 have shown that hIAPP has the ability to generate hydrogen peroxide (H 2 O 2 ) during amyloid bril formation in vitro. On the contrary, the nontoxic and non-brillogenic analog rat amylin which is called (rIAPP), does not generate H 2 O 2 . ...
Article
Full-text available
Diabetes mellitus, a complex metabolic disorder, presents a growing global health challenge. In 2021, there were 529 million diabetics worldwide. At the super-regional level, Oceania, the Middle East, and North Africa had the highest age-standardized rates. The majority of cases of diabetes in 2021 (>90.0%) were type 2 diabetes, which is largely indicative of the prevalence of diabetes in general, particularly in older adults (K. L. Ong, et al., Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: a systematic analysis for the Global Burden of Disease Study 2021, Lancet, 2023, 402(10397), 203–234). Nowadays, slowing the progression of diabetic complications is the only effective way to manage diabetes with the available therapeutic options. However, novel biomarkers and treatments are urgently needed to control cytokine secretion, advanced glycation end products (AGEs) production, vascular inflammatory effects, and cellular death. Emerging research has highlighted the intricate interplay between reactive oxygen species (ROS) and protein aggregation in the pathogenesis of diabetes. In this scenario, the main aim of this paper is to provide a comprehensive review of the current understanding of the molecular mechanisms underlying ROS-induced cellular damage and protein aggregation, specifically focusing on their contribution to diabetes development. The role of ROS as key mediators of oxidative stress in diabetes is discussed, emphasizing their impact on cellular components and signaling. Additionally, the involvement of protein aggregation in impairing cellular function and insulin signaling is explored. The synergistic effects of ROS and protein aggregation in promoting β-cell dysfunction and insulin resistance are examined, shedding light on potential targets for therapeutic intervention.
... Since this hormone competes with pathological Aβ proteins for receptor binding sites and even helps eliminate amyloid from the CNS, it has a direct implication in inflammatory regulation and neuronal death (Lutz and Meyer, 2015;Bharadwaj et al., 2017;Reiner et al., 2017). ...
... A noteworthy correlation between obesity, which is proposed to be one of the primary causes of T2DM and late-onset dementias has been established by various studies (Atti et al., 2019;Feinkohl et al., 2019;Kouvari et al., 2022;Tahmi et al., 2021). T2DM individuals are also two to five times more likely to develop Alzheimer's disease (Lutz & Meyer, 2015;Mumme et al., 2019;Wang et al., 2021). According to some researchers, the mechanism behind amylin deposition in the brains of patients carrying T2DM and AD may be hyperamylasemia, frequently noted among individuals with obesity or pre-diabetic insulin resistance. ...
Article
Aim Intra-pancreatic fat deposition (IPFD) while hypothesised to impair beta-cell function, its impact on alpha-cells remains unclear. We evaluated the association between IPFD and markers of pancreatic cells function using whey protein. Methods Twenty overweight women with impaired fasting glucose (IFG) and low or high IPFD (<4.66% vs ≥4.66%) consumed 3 beverage treatments: 0 g (water control), 12.5 g (low-dose) and 50.0 g (high-dose) whey protein, after an overnight fast, in randomised order. Blood glucose, insulin, C-peptide, glucagon, gastric-inhibitory polypeptide (GIP), glucagon-like peptide-1 (GLP-1) and amylin were analysed postprandially over 4 h. Incremental area-under-the-curve (iAUC), incremental maximum concentration (iCmax), and time to maximum concentration (Tmax) for these were compared between IPFD groups using repeated measures linear mixed models, also controlled for age (pcov). Results iAUC and iCmax glucose and insulin while similar between the two IPFD groups, high IPFD and ageing contributed to higher postprandial glucagon (iAUC: p = 0.012; pcov = 0.004; iCmax: p = 0.069; pcov = 0.021) and GLP-1 (iAUC: p = 0.006; pcov = 0.064; iCmax: p = 0.011; pcov = 0.122) concentrations. Conclusion In our cohort, there was no evidence that IPFD impaired protein-induced insulin secretion. Conversely, IPFD may be associated with increased protein-induced glucagon secretion, a novel observation which warrants further investigation into its relevance in the pathogenesis of dysglycaemia and type-2 diabetes.
Article
Full-text available
Proteinopathy is characterized by the accumulation of aggregates of a specific protein in a target organ, tissue, or cell. The aggregation of the same protein can cause different pathologies as single protein can adopt various amyloidogenic, disease-specific conformations. The conformation governs the interaction of amyloid aggregates with other proteins that are prone to misfolding and, thus, determines disease-specific spectrum of concomitant pathologies. In this regard, a detailed description of amyloid protein conformation as well as spectrum of its interaction with other proteins become a key point for drafting of precise description of the disease. The majority of clinical cases of neuronal proteinopathies is caused by the aggregation of rather limited range of amyloidogenic proteins. Here, we provided the characterization of pathologies, related to the aggregation of amyloid β peptide, tau protein, α-synuclein, TDP-43, and amylin, giving a short description of pathologies themselves, recent advances in elucidation of misfolded protein conformation, with emphasis on those protein aggregates extracted from biological samples, what is known about the interaction of this proteins, and the influence of this interaction on the progression of underlying disease and comorbidities.
Article
Full-text available
Significance Both types 1 and 2 diabetes are associated with increased risks of age-related decay in cognitive function and mood disorders, especially depression. Insulin action has been shown to regulate neuronal signaling and plasticity. Here we investigate whether brain-specific knockout of insulin receptor (NIRKO) in mice causes behavioral changes and how these are mechanistically linked. We find that NIRKO mice exhibit age-related anxiety and depressive-like behavior. This is due to altered mitochondrial function, aberrant monoamine oxidase (MAO) expression, and increased dopamine turnover in the mesolimbic system, and can be reversed by treatment with Mao inhibitors. Thus, brain insulin resistance alters dopamine turnover and induces anxiety and depressive-like behaviors. These findings demonstrate a potential molecular link between central insulin resistance and behavioral disorders.
Article
Full-text available
Background: Plasma amylin is positively associated with cognitive function in humans. Amylin treatment improves memory in Alzheimer's mouse models. However, the relationship between plasma amylin, diabetes and cognition is not clear. Objectives: In this study we examined the concentration of plasma amylin, its relationship with diabetes and cognition. Material and method: A cross-sectional, homebound elderly population with data of plasma amylin under fasting condition and cognitive measurements was used. Results: We found that subjects with a long and chronic duration of diabetes were more likely to take insulin treatment and have reduced secretion of amylin. Compared to non-diabetics, diabetic subjects without insulin treatment had a higher concentration, but those with insulin treatment had a lower concentration, of plasma amylin [median (Q1, Q3): 20 (11.0, 36.2) vs. 25.2 (13.2, 50.6) vs. 15.0 (4.9, 33.8), p<0.0001]. In the whole sample vs. in the absence of diabetes, plasma amylin was positively associated with logical memory delayed recall (β= +0.61, SE=0.25, p=0.02 vs. β=+0.80, SE=0.33, p=0.02) and block design (β=+0.62, SE=0.24, p=0.009 vs. β=+0.93, SE=0.31, p=0.003), and negatively associated with Trailmaking A scores (β= -6.21, SE=1.55, p<0.0001 vs. β=-7.51, SE=1.95, p=0.0001) and Trailmaking B (β= -4.32, SE=2.13, p=0.04 vs. β= -5.86, SE=2.73, p=0.04). All these relationships disappeared in the presence of diabetes regardless the treatment. Conclusion: This study suggests that secretion of amylin by pancreas compensates and then deteriorates depending on the duration of diabetes. Amylin's activities for cognition are impaired in the presence of diabetes.
Article
Full-text available
Several proteins have been identified as amyloid forming in humans, and independent of protein origin, the fibrils are morphologically similar. Therefore, there is a potential for structures with amyloid seeding ability to induce both homologous and heterologous fibril growth; thus, molecular interaction can constitute a link between different amyloid forms. Intravenous injection with preformed fibrils from islet amyloid polypeptide (IAPP), proIAPP, or amyloid-beta (Aβ) into human IAPP transgenic mice triggered IAPP amyloid formation in pancreas in 5 of 7 mice in each group, demonstrating that IAPP amyloid could be enhanced through homologous and heterologous seeding with higher efficiency for the former mechanism. Proximity ligation assay was used for colocalization studies of IAPP and Aβ in islet amyloid in type 2 diabetic patients and Aβ deposits in brains of patients with Alzheimer disease. Aβ reactivity was not detected in islet amyloid although islet β cells express AβPP and convertases necessary for Aβ production. By contrast, IAPP and proIAPP were detected in cerebral and vascular Aβ deposits, and presence of proximity ligation signal at both locations showed that the peptides were <40 nm apart. It is not clear whether IAPP present in brain originates from pancreas or is locally produced. Heterologous seeding between IAPP and Aβ shown here may represent a molecular link between type 2 diabetes and Alzheimer disease. Copyright © 2015 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.
Article
Full-text available
Amyloid-β proteins(Aβ) of 42 (Aβ42)and 40 aa (Aβ40) accumulate as senile plaques (SP) and cerebrovascular amyloid protein deposits that are defining diagnostic features of Alzheimer's disease (AD). A number of rare mutations linked to familial AD (FAD) on the Aβ precursor protein (APP),Presenilin-1 (PS1), Presenilin-2 (PS2),Adamalysin10,and other genetic risk factors for sporadic AD such as the γ4 allele of Apolipoprotein E (ApoE-γ4) foster the accumulation of Aβ and also induce the entire spectrum of pathology associated with the disease. Aβ accumulation is therefore a key pathological event and a prime target for the prevention and treatment of AD. APP is sequentially processed by β -site APP cleaving enzyme (BACE1)and β-secretase, a multisubunit PS1/PS2-containing integral membrane protease, to generate Aβ. Although Aβ accumulates in all forms of AD, the only pathways known to be affected in FAD increase Aβ production by APP gene duplication or via base substitutions on APP and γ-secretase subunits PS1 and PS2 that either specifically increase the yield of the longer Aβ42 or both Aβ40 and Aβ42.However, the vast majority of AD patients accumulate Aβ without these known mutations. This led to proposals that impairment of Aβdegradation or clearance may play a key rolein AD pathogenesis. Several candidate enzymes, including Insulin-degrading enzyme (IDE), Neprilysin (NEP), Endothelin-converting enzyme (ECE), Angiotensin converting enzyme (ACE), Plasmin, and Matrix metalloproteinases (MMPs) have been identified and some have even been successfully evaluated in animal models. Several studies also have demonstrated the capacity of γ-secretase inhibitors to paradoxically increase the yield of Aβ and we have recently established that the mechanism is by skirting Aβ degradation. This review outlines major cellular pathways of Aβ degradation to provide a basis for future efforts to fully characterize the panel of pathways responsible for Aβturnover.
Article
Full-text available
Amyloid formation is the pathological hallmark of type 2 diabetes (T2D) and Alzheimer's disease (AD). These diseases are marked by extracellular amyloid deposits of islet amyloid polypeptide (IAPP) in the pancreas and amyloid β (Aβ) in the brain. Since IAPP may enter the brain and disparate amyloids can cross-seed each other to augment amyloid formation, we hypothesized that pancreatic derived IAPP may enter the brain to augment misfolding of Aβ in AD. The corollaries for validity of this hypothesis are that IAPP enters the brain, augments Aβ misfolding, [3] associates with Aβ plaques, and most importantly plasma levels correlate with AD diagnosis. We demonstrate the first 3 corollaries that: (1) IAPP is present in the brain in human cerebrospinal fluid (CSF), (2) synthetic IAPP promoted oligomerization of Aβ in vitro, and (3) endogenous IAPP localized to Aβ oligomers and plaques. For the 4th corollary, we did not observe correlation of peripheral IAPP levels with AD pathology in either an African American cohort or AD transgenic mice. In the African American cohort, with increased risk for both T2D and AD, peripheral IAPP levels were not significantly different in samples with no disease, T2D, AD, or both T2D and AD. In the Tg2576 AD mouse model, IAPP plasma levels were not significantly elevated at an age where the mice exhibit the glucose intolerance of pre-diabetes. Based on this negative data, it appears unlikely that peripheral IAPP cross-seeds or "infects" Aβ pathology in AD brain. However, we provide novel and additional data which demonstrate that IAPP protein is present in astrocytes in murine brain and secreted from primary cultured astrocytes. This preliminary report suggests a potential and novel association between brain derived IAPP and AD, however whether astrocytic derived IAPP cross-seeds Aβ in the brain requires further research.
Article
What roles do mesolimbic and neostriatal dopamine systems play in reward? Do they mediate the hedonic impact of rewarding stimuli? Do they mediate hedonic reward learning and associative prediction? Our review of the literature, together with results of a new study of residual reward capacity after dopamine depletion, indicates the answer to both questions is 'no'. Rather, dopamine systems may mediate the incentive salience of rewards, modulating their motivational value in a manner separable from hedonia and reward learning. In a study of the consequences of dopamine loss, rats were depleted of dopamine in the nucleus accumbens and neostriatum by up to 99% using 6-hydroxydopamine. In a series of experiments, we applied the 'taste reactivity' measure of affective reactions (gapes, etc.) to assess the capacity of dopamine-depleted rats for: 1) normal affect (hedonic and aversive reactions), 2) modulation of hedonic affect by associative learning (taste aversion conditioning), and 3) hedonic enhancement of affect by non-dopaminergic pharmacological manipulation of palatability (benzodiazepine administration). We found normal hedonic reaction patterns to sucrose vs. quinine, normal learning of new hedonic stimulus values (a change in palatability based on predictive relations), and normal pharmacological hedonic enhancement of palatability. We discuss these results in the context of hypotheses and data concerning the role of dopamine in reward. We review neurochemical, electrophysiological, and other behavioral evidence. We conclude that dopamine systems are not needed either to mediate the hedonic pleasure of reinforcers or to mediate predictive associations involved in hedonic reward learning. We conclude instead that dopamine may be more important to incentive salience attributions to the neural representations of reward-related stimuli. Incentive salience, we suggest, is a distinct component of motivation and reward. In other words, dopamine systems are necessary for 'wanting' incentives, but not for 'liking' them or for learning new 'likes' and 'dislikes'.
Article
Amylin is a 37-amino acid peptide that shares considerable homology with calcitonin gene-related peptide (CGRP). Both peptides exert glycoregulatory actions and produce vasodilation of the cardiovascular system. We wished to determine if amylin exerts vasodilatory action in the perfused mesenteric arterial bed in a manner similar to that of CGRP and if so, to determine if amylin and CGRP share a common mechanism of action. Amylin 10 -8 , 10 -7 , and 10 -6 M produced significant decreases in perfusion pressure by 18, 34, and 45 mm Hg, respectively, of the perfused mesenteric arterial bed pretreated with guanethidine (7 x 10 -6 M) and precontracted with methoxamine (10 -6 -10 -5 M). Amylin was ∼10 times less potent than CGRP. This vasodilatory effect was not antagonized by atropine in a concentration (10 -6 M) that blocked the vasodilatory action of acetylcholine (ACh) or of nadolol in a concentration that blocked the response to isoproterenol (ISO 10 -6 M). In contrast, the CGRP receptor antagonist [8-37]hCGRP blocked the response of both amylin and CGRP while failing to block the effect of ISO. The depressor effects of CGRP (10 -8 M), amylin (10 -6 M), and ISO (10 -5 M) were 38, 43, and 42 mm Hg without and 5, 12, and 44 mm Hg with [8-37]hCGRP (10 -7 M), respectively. Simultaneous administration of CGRP and amylin failed to produce an additive effect. The depressor effects of CGRP (10 -8 M), amylin (10 -7 M), and CGRP (10 -8 M) plus amylin (10 -7 M) were 50, 32, and 45 mm Hg, respectively. We conclude that amylin exerts a vasodilator action in the perfused mesenteric arterial bed by acting on CGRP 1 receptors and suggest that this glycoregulatory hormone may also exert regulatory actions in the vasculature in a manner similar to that of CGRP.
Article
Amylin is a 37-amino acid peptide hormone that is co-secreted with insulin by pancreatic β cells in response to feeding. We recently reported that amylin potently reduces food intake, body weight, and adiposity when delivered into the 3rd cerebral ventricle (i3vt) of rats. We have now infused i3vt a specific antagonist (AC187) to ascertain the physiological relevance of central amylin in the control of energy balance. After establishing the ability of i3vt AC187 to block the anorexic effect of i3vt amylin, we performed an experiment to examine the impact of acute inhibition of central amylin signaling on feeding. Separate groups (n = 7/group) of ad lib-fed male Long Evans rats were given one bolus i3vt infusion of synthetic cerebrospinal fluid vehicle (CSF) or AC187 (250 or 1000 pmol). Acute infusion of AC187 tended to increase 1-h food intake and significantly elevated 4-h intake. Both the 250 and 1000 pmol doses produced significant increases as compared to CSF. In another experiment designed to tonically inhibit central amylin signaling over an extended period, two other groups of rats (n = 6/group) received continuous i3vt infusion of CSF or 100 pmol/h AC187 over 14 days via implantable osmotic pumps. Rats receiving AC187 ate significantly more food over the 14-day infusion period relative to controls (CSF = 322 ± 6 g, AC187 = 360 ± 12 g). Although body weight was not significantly affected, body fat was increased by about 30% in the AC187 rats, with no difference in lean tissue between the groups. Additionally, although fasting plasma glucose did not differ between the CSF and AC187 groups after 14 days of infusion, plasma insulin was significantly elevated in the AC187 rats. In summary, the present results document significant increases of food intake and body adiposity resulting from inhibition of central amylin signaling. They are consistent with our hypothesis that CNS actions of endogenous amylin contribute to the long-term regulation of energy balance.
Article
Amyloid precursor protein (APP) is a key player in Alzheimer's disease (AD). The Aβ fragments of APP are the major constituent of AD-associated amyloid plaques, and mutations or duplications of the gene coding for APP can cause familial AD. Here we review the roles of APP in neuronal development, signaling, intracellular transport, and other aspects of neuronal homeostasis. We suggest that APP acts as a signaling nexus that transduces information about a range of extracellular conditions, including neuronal damage, to induction of intracellular signaling events. Subtle disruptions of APP signaling functions may be major contributors to AD-causing neuronal dysfunction. Copyright © 2015 Elsevier Inc. All rights reserved.