You are on page 1of 13

Ageing Research Reviews 12 (2013) 764–776

Contents lists available at SciVerse ScienceDirect

Ageing Research Reviews


journal homepage: www.elsevier.com/locate/arr

Review

Food for thought: The role of appetitive peptides in age-related


cognitive decline
Jim R. Fadel a , Corinne G. Jolivalt b , Lawrence P. Reagan a,∗
a
Department of Pharmacology, Physiology and Neuroscience, School of Medicine, University of South Carolina, 6439 Garners Ferry Road, Columbia, SC
29208, USA
b
Department of Pathology, Division of Neuropathology, University of California San Diego, 9500 Gilman Drive, San Diego, CA 92093-0612, USA

a r t i c l e i n f o a b s t r a c t

Article history: Through their well described actions in the hypothalamus, appetitive peptides such as insulin, orexin and
Received 1 December 2012 leptin are recognized as important regulators of food intake, body weight and body composition. Beyond
Received in revised form 22 January 2013 these metabolic activities, these peptides also are critically involved in a wide variety of activities ran-
Accepted 25 January 2013
ging from modulation of immune and neuroendocrine function to addictive behaviors and reproduction.
Available online 13 February 2013
The neurological activities of insulin, orexin and leptin also include facilitation of hippocampal synaptic
plasticity and enhancement of cognitive performance. While patients with metabolic disorders such as
Keywords:
obesity and diabetes have greater risk of developing cognitive deficits, dementia and Alzheimer’s dis-
Insulin
Leptin
ease (AD), the underlying mechanisms that are responsible for, or contribute to, age-related cognitive
Orexin decline are poorly understood. In view of the importance of these peptides in metabolic disorders, it is
Hippocampus not surprising that there is a greater focus on their potential role in cognitive deficits associated with
Dementia aging. The goal of this review is to describe the evidence from clinical and pre-clinical studies implicating
Alzheimer’s disease insulin, orexin and leptin in the etiology and progression of age-related cognitive decline. Collectively,
these studies support the hypothesis that leptin and insulin resistance, concepts normally associated
with the hypothalamus, are also applicable to the hippocampus.
© 2013 Elsevier B.V. All rights reserved.

1. Introduction activation of these orexigenic pathways facilitates the performance


of hippocampal-dependent tasks (Stanley et al., 2012). Beyond
Neuroplasticity may be defined as the ability of an organ- these integrative activities, insulin and leptin act directly in the hip-
ism to adapt to changes in the external or internal milieu and pocampus to promote measures of hippocampal synaptic plasticity,
can be assessed using a variety of experimental approaches. In including enhancement of neuronal morphology, increases in neu-
this regard, neuroplasticity can be evaluated through approaches rogenesis/cell proliferation and increases in synaptic transmission
ranging from neuroanatomical analyses to neurochemical and elec- (i.e. long term potentiation; LTP). These neuroanatomical, elec-
trophysiological measures to behavior. An important question is trophysiological and neurochemical enhancements will facilitate
what are the underlying molecular, cellular and neurochemical cognitive/behavioral performance. Conversely, age-related decline
factors that regulate brain plasticity? In addition to neurotrans- in hippocampal neuroplasticity and cognition is associated with
mitters and neurotrophic factors, insulin, leptin and orexin are impairments in the functional activities of insulin, orexin and lep-
examples of neuropeptides that promote hippocampal synaptic tin. The focus of this review is to highlight how these peptides
plasticity (Fig. 1). Insulin derived from the pancreas and leptin that are normally associated with the regulation of metabolism and
derived from adipocytes have well described activities in the body weight play a critical role in the facilitation of hippocampal
hypothalamus, which includes regulation of the orexin system. neuronal plasticity, as well as how the impairments in the neuro-
Orexin neurons from the lateral hypothalamus/perfornical area plasticity activities of these appetitive peptides is a contributing
project directly to the hippocampus and to the medial septum, factor in age-related cognitive decline.
which sends GABAergic, glutamatergic and acetylcholinergic pro-
jections to the hippocampus. Indeed, recent studies indicate that
2. Insulin

∗ Corresponding author at: Department of Pharmacology, Physiology and Neuro-


2.1. Role of insulin in promoting plasticity
science, University of South Carolina School of Medicine, 6439 Garners Ferry Road,
D40, Columbia, SC 29208, USA. Tel.: +1 803 216 3515; fax: +1 803 216 3538. Insulin is a peptidic hormone secreted by the pancreatic islets of
E-mail address: lpreagan@uscmed.sc.edu (L.P. Reagan). Langerhans, regulated by the gut hormone glucagon-like peptide-1

1568-1637/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.arr.2013.01.009
J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776 765

PSD-95 protein (Lee et al., 2005) and promotes internalization of


AMPA receptors causing long term depression (LTD) of excitatory
synaptic transmission (Huang et al., 2003, 2004). By intracere-
broventricular injections, insulin was demonstrated to activate
protein kinase B (PKB or Akt) via the phosphatidylinositol 3-
kinase (PI3-K) pathway in the rat hippocampus (Grillo et al., 2009).
Recently, a GSK3 inhibitor was shown not only to prevent, but also
to reverse, A␤-induced inhibition of LTP in brain slice prepara-
tions, suggesting that over-activation of GSK3␤, that can result from
disrupted insulin signaling but also a number of other pathways
including protein phosphatase (Graff et al., 2010; Dineley et al.,
2010), is responsible for pathological synaptic plasticity (Jo et al.,
2011). More recently, insulin was also shown to increase the fre-
quency of mEPSC (excitatory synapse) and to promote dendritic
spine formation via the PI3 K/Akt/mTOR pathway, playing a cru-
cial role in structural synaptic plasticity (Lee et al., 2011). Another
support of the role of insulin in synaptic plasticity is the recent
demonstration that a reduced insulin receptor expression in mouse
brain impairs hippocampal late-phase LTP and this was associated
with deficits in long term recognition memory (Nistico et al., 2012).

2.2. Role of insulin in age-related cognitive deficits

In this review, we will focus on insulin and age-related cognitive


deficits; however insulin appears to play a role independent of age.
This is underlined in a recent study demonstrating that cognition
is acutely impaired in children with newly diagnosed type 1
diabetes, when insulin regimen is only initiated (Nadebaum et al.,
2012). Similarly, metabolic syndrome in adolescents, that included
insulin resistance for all subjects, results in reductions in cognitive
functions and brain structural integrity, despite a relatively short
duration of poor metabolic control and no vascular disease (Yau
et al., 2012). These results suggest that insulin resistance without
overt hyperglycemia may lead to brain complications. Cognitive
deficits generally associated with aging range from mild cognitive
impairments (MCI) to dementia, including Alzheimer’s disease
(AD). In recent years, the role of insulin in the brain of AD and/or
diabetic patients has emerged as a principal contributor to the
development of cognitive impairments and has become the focus
Fig. 1. The peptides insulin, leptin and orexin work in an integrative fashion to
facilitate hippocampal synaptic plasticity. Insulin released from pancreatic ␤ cells
of detailed studies to understand the mechanisms involved.
and leptin released from adipocytes have well defined metabolic activities in the Diabetes mellitus is sub-classified into type 1 (insulin-deficient)
hypothalamus that include regulation of orexin neuronal populations in the lat- and type 2 (insulin-resistant) diabetes and cognitive deficits occur
eral hypothalamus, perifornical area and dorsomedial hypothalamus (blue arrows). in both of these patient populations (Ryan et al., 1985; Rovet et al.,
Projection sites of these orexin populations include the basal forebrain, where
1987; Reaven et al., 1990; Ryan and Williams, 1993; McCarthy
orexin modulates cholinergic, GABAergic and glutamatergic components of septo-
hippocampal neurotransmission (blue arrows). As such, these orexin-driven basal et al., 2002; Cukierman et al., 2005; Manschot et al., 2006; Biessels
forebrain projections have a significant impact on hippocampal neurochemistry. et al., 2008). Studies have found a relative risk of about 1.9–2.3
Beyond these indirect effects, insulin and leptin can directly modulate hippocampal of developing AD for diabetic patients (Ott et al., 1996; Leibson
synaptic plasticity (red arrows), including facilitation of synaptic transmission (i.e.
et al., 1997; Ronnemaa et al., 2009), and this risk can increase up
LTP), increases in neurogenesis and cell proliferation and promotion of synapto-
genesis and dendritic remodeling (morphology). This facilitation of hippocampal
to 5.5 in the presence of Apo E4, a risk factor for AD (Peila et al.,
synaptic plasticity ultimately leads to the enhancement of cognitive/behavioral 2002). More recently, diabetes was shown to increase not only the
function. See text for details. risk of dementia but also the risk of progression from MCI to AD
(Velayudhan et al., 2010). Uncontrolled diabetes (no insulin treat-
(GLP-1) in response to meal ingestion. Although insulin receptors ment) was associated with development of AD, while patients with
were identified in the brain 3 decades ago (Havrankova et al., 1978, controlled diabetes showed no increased dementia, suggesting a
1979, 1981; Hill et al., 1986; Unger et al., 1989) with functions regu- role of impaired insulin signaling in the development of neurode-
lating glucose metabolism, food intake and body weight (Schwartz generation and AD (Xu et al., 2009). Insulin signaling encompasses
et al., 1992), only within the last decade or so, the role of insulin assorted signaling pathways, including those mediated by phos-
in synaptic plasticity and cognitive functions has become a subject pholipase C␥, mitogen-activated protein kinase (MAPK) and PI3-K
of research. Insulin receptors are localized at the synapse where with the subsequent downstream involvement of GSK3. Recently,
they regulate neurotransmitter release and receptor recruitment we have shown that 9 weeks of peripheral insulin deficiency
(Wan et al., 1997; Jonas et al., 1997; Abbott et al., 1999). Insulin induced by systematic injection of streptozotocin is sufficient to
plays a role in regulating synaptic efficacy in the CNS by rapid cause AD-like features in the brain of type 1 diabetic mice. This
recruitment of GABAA receptors that potentiate inhibitory synap- pathology was associated with learning and memory deficits and
tic transmission (Wan et al., 1997; Taghibiglou et al., 2009). It has decreased insulin receptor expression and Akt phosphorylation
also been shown that insulin enhances NMDA receptor-mediated in the brain leading to increased GSK3 activity, increased tau
synaptic transmission (Liu et al., 1995), stimulates the translation of phosphorylation and increased A␤ protein levels (Jolivalt et al.,
766 J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776

2008). Several other studies in both type 1 and type 2 diabetes fur- correlates with reduced cognitive performance and decreased brain
ther support impaired brain insulin signaling leading to reduced size (Benedict et al., 2012). Brain insulin resistance in AD patients,
PI3-K pathway activity and subsequent increased GSK3 activity. assessed ex vivo by insulin stimulation of the hippocampus and
Recently, we have shown that type 1 diabetes exacerbates brain Western blots of the insulin signaling pathway proteins, was asso-
aging and increase vascular mediated A␤ depositions in the brain ciated with worse performance on tests of working and episodic
of Senescence prone or resistant mice, a model of accelerated aging memory, suggesting that insulin signaling has a direct association
(Currais et al., 2012). Interestingly, intranasal insulin delivery in with cognitive status (Talbot et al., 2012). In a recent prospective
type 1 diabetic mice prevents cognitive decline, cerebral atrophy study following older adults over 9 years, the presence of diabetes
and the decrease of the PI3-K/Akt signaling pathway (Francis et al., mellitus and poor glucose control was associated with worse cog-
2008). In a rat model of type 2 diabetes, intranasal insulin admin- nitive function and greater decline than adults without diabetes,
istration normalized Akt and GSK3␤ activities and reduced tau suggesting that the severity of diabetes may contribute to accel-
phosphorylation while subcutaneous insulin treatment had only erated cognitive aging (Yaffe et al., 2012). A recent autopsy study
minimal effects (Yang et al., 2013). Metformin, a FDA-approved showed decreased activities of the components of the insulin-PI3-
drug used to treat type 2 diabetic patients, administered to high fat K/Akt pathway and increased tau phosphorylation levels in the
diet fed rats for 21 days improved learning and memory deficits as brain of AD and type 2 diabetic patients, alterations that were
well as improved insulin resistance (Pintana et al., 2012). However, more severe in patients with co-morbid AD and type 2 diabetes
in db/db mice, which develop insulin resistance, metformin atten- (Liu et al., 2011). A 4 week diet-induced metabolic syndrome, with
uates AD-like neuropathology, such as increased phosphorylated insulin resistance, in adults with MCI promoted a decrease of A␤
tau but did not improve insulin levels, GSK3 activity or learning protein in cerebrospinal fluid that may correspond to the initiation
behavior (Li et al., 2012a). of A␤ deposition in the brain (Bayer-Carter et al., 2011). In contrast,
AD is characterized by neuronal loss, neurofibrillary tangles insulin infusion in type 2 diabetic patients reduces APP, presenilin-
enriched with highly phosphorylated tau protein and plaques con- 1 and -2 (proteins from the ␤-secretase complex that cleave APP),
taining amyloid ␤ derived from cleavage of Amyloid Precursor and GSK-3␤ mRNA in peripheral blood mononuclear cells (Dandona
Protein (APP). It has been suggested that AD is a neuroendocrine et al., 2011). In a recent study to develop a novel diagnostic marker,
disorder of the brain due to deficiency in brain insulin and impaired plasma A␤ levels were increased in AD patients after a glucose load
insulin signaling (de la Monte and Wands, 2005; Steen et al., despite similar A␤ levels between AD and non-AD patients (Takeda
2005; Revill et al., 2006). The role of the insulin-signaling pathway et al., 2012).
and GSK3 in AD brain was suggested by intracerebroventricular- A number of clinical studies with intranasal administration of
streptozotocin (icv-STZ) rodent models that display AD-type insulin in patients with diabetes, MCI or AD also underscore the role
neurodegeneration, including deficits in learning and memory and mechanism(s) of impaired insulin signaling in the development
(Plaschke and Hoyer, 1993; Duelli et al., 1994; Lannert and Hoyer, of cognitive deficits. In the latest review, Shemesh et al. assessed 8
1998). STZ, when injected directly into the brain, rapidly induced a studies performed in the past 8 years that included healthy, obese,
dramatic reduction of insulin gene expression and subsequent neu- MCI or AD patients and concluded that intranasal insulin has a
rodegeneration and cognitive impairments that were accompanied positive effect on cognitive functions despite limited number of
by an increase of activated GSK3, phosphorylated tau, APP mRNA patients, different doses and tests used in the reviewed studies
and amyloid ␤ (Steen et al., 2005; Lester-Coll et al., 2006; Plaschke (Shemesh et al., 2012). In a recent pilot randomized placebo-
et al., 2010; Santos et al., 2012; Shingo et al., 2012). In addition controlled trial, 4 month treatment with intranasal insulin resulted
to the icv-STZ rat model, transgenic mouse models overexpress- in stabilization or improvement of cognition in MCI and AD patients
ing active GSK3 also displayed cognitive deficits associated with (Craft et al., 2012).
neuronal loss and tau hyperphosphorylation in contrast to mouse
models expressing reduced GSK3 activity [reviewed in Avila et al., 2.4. Potential therapeutic intervention based on insulin action
2010]. These models strengthen the importance of GSK3 in the neu-
rodegeneration. Also, in vitro studies have shown defective insulin Some therapeutic interventions aimed at correcting insulin
signaling as a result of amyloid ␤ oligomer toxicity, suggesting resistance for AD or MCI patients, such as insulin-sensitizers,
that restoration of insulin signaling may slow AD pathogenesis (De insulinotropes, intranasal insulin, antioxidant and anti-inflamma-
Felice et al., 2009). The role of insulin signaling in AD brain is further tory drugs or drugs targeting GSK3 as treatment for type 2 diabetes
supported by animal studies showing exacerbation of tau depo- and AD are described in recent reviews (de la Monte, 2012; Gao
sition into tangles in a mouse model expressing human tau with et al., 2012; Craft et al., 2013). Here, we will summarize only the
concomitant insulin-deficient diabetes (Ke et al., 2009) and studies most recent aspects of intervention based on current results and
showing that induction of type 1 diabetes with STZ in familial AD mechanisms presented above.
transgenic mouse models exaggerated both vascular and AD-like Pioglitazone, an insulin-sensitizer, as well as lithium, a GSK3
pathology (Burdo et al., 2009; Jolivalt et al., 2010). Further support inhibitor, restored memory impairments in icv-STZ rats, behavioral
for the role of impaired insulin signaling as contributor to brain neu- improvements that were associated with a significant attenua-
rodegeneration was demonstrated with insulin-resistance/type 2 tion of GSK3 activity in the hippocampus. Memantine, one of the
diabetes studies showing amyloidosis in a mouse model of AD few approved drugs for the treatment of AD (albeit with mod-
with concomitant diet-induced insulin-resistance (Ho et al., 2004) est effect on moderate to severe AD) improved memory but not
and AD transgenic mice APP23 crossbred with type 2 diabetic GSK3 activity in the icv-STZ rat model (Ponce-Lopez et al., 2011).
ob/ob mice that displayed exacerbated cognitive deficits without Chronic lithium treatment in an aged AD transgenic mouse model
increased A␤ burden in the brain but cerebral inflammation and decreased the cleavage of APP and senile plaque formation, and this
amyloid deposit in blood vessels (Takeda et al., 2010). was accompanied by improvement in spatial learning and mem-
ory abilities (Zhang et al., 2011). In a recent 12 month-long trial
2.3. Translation of animal models with insulin signaling with lithium in MCI patients, cognitive deficits and attention tasks
impairment to the clinical setting were improved along with decreased levels of phosphorylated tau
in the CSF (Forlenza et al., 2011). In our recent work, the role of
In a cohort of 75 year old men and women, impaired insulin GSK3 in learning and memory and synaptic plasticity in type 1 dia-
sensitivity, calculated based on fasting insulin and glucose levels, betic mice was confirmed by treatment with a GSK3 inhibitor or a
J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776 767

neurotrophic factor with GSK3 inhibitory properties (King et al., ventral tegmental area [VTA; for review, see Fiaglewicz and Benoit,
2013). The cognitive improvements were associated with restora- 2009]. For example, lateral hypothalamic (LH) LepR-containing
tion of synaptophysin protein levels in the brain of treated diabetic neurons project to the VTA, where they regulate feeding behavior
mice but not with tau phosphorylation or A␤ protein levels, sug- (Fiaglewicz et al., 2007; Leinninger et al., 2009). These actions of lep-
gesting that, in contrast to synaptic plasticity, tau and A␤ may be tin are mediated through the long form of the LepR (LepRb), which
late contributors to learning and memory processes in diabetic mice is abundantly expressed in several hypothalamic nuclei includ-
brain. These results also support GSK3 inhibition as a treatment for ing the arcuate nucleus and the ventromedial nucleus (Elmquist
type 1-associated cognitive deficits and AD. et al., 1998; Baskin et al., 1999). In the hypothalamus, activation
Glucagon-like peptide-1 (GLP-1) is an incretin hormone regulat- of the LepRb results in a complex and integrated simulation of
ing energy metabolism and insulin secretion with a short half-life several signal transduction cascades. Specifically, the LepRb acti-
due to degradation. GLP-1, but not its degradation metabolites, vates the constitutively associated tyrosine kinase Jak2, resulting
has been shown to improve markers of cognitive functions in in autophosphorylation of Jak2, as well as phosphorylation of sev-
type 2 diabetes models (Porter et al., 2012). A number of long eral tyrosine residues within the intracellular domain of the LepRb.
lasting analogs have been developed, such as 2 drugs on the mar- Phosphorylation of the LepRb at Tyr1138 results in the recruit-
ket to treat type 2 diabetes, exendin-4, commercialized under ment and phosphorylation of the transcriptional regulator STAT3
ByettaTM , and liraglutide. Treatment with liraglutide showed neu- (pSTAT3), thereby leading to nuclear translocation and regulation
roprotective effects in mice with high-fat diet-induced obesity and of a variety of genes including suppressor of cytokine signaling
insulin resistance (Porter et al., 2010) and in an AD mouse model 3 (SOCS3). SOCS3 binds to Tyr985 of the LepRb, as well as to
APP/PS1 in which it rescued memory deficits and impairments in Jak2, to mediate feedback inhibition of the LepRb. Interestingly,
LTP, reduced A␤ levels and amyloid plaque numbers, protected Try985 also provides a binding site for the tyrosine phosphatase
synapses and reduced inflammation in the brain (McClean et al., SHP-2, which ultimately leads to the activation of the ERK signaling
2010, 2011). Similar observations have been made with the long- cascade. Tyr1107 phosphorylation leads to the activation of the
acting GLP-1 analog (Val8 )GLP-1 which protected LTP and reduced transcription factor STAT5. Beyond these Tyr residues on the LepRb,
the dense-core plaque count in aged APP/PS1 mice (Gengler et al., autophosphorylation of Jak2 stimulates the phosphatidylinositol
2012). In recent studies, GLP-1 or analogs were shown to ame- 3-kinase (PI3-K) cascade, leading to the phosphorylation and acti-
liorate neurodegeneration and cognitive performance in icv-STZ vation of Akt [for review of these pathways, see Munzberg and
rats with impaired insulin signaling in the brain. Treatment with Myers, 2005; Myers et al., 2010]. These coordinated signaling
exedin-4 (Ex-4), a long-acting GLP-1 receptor agonist, or (Val8 )GLP- cascades are critical for the regulation of food intake and body
1 improved learning and memory in the Morris water maze and composition. Conversely, obesity and type 2 diabetes are asso-
reversed icv-STZ-induced tau phosphorylation (Li et al., 2012b) via ciated with hypothalamic leptin resistance that results from a
down-regulation of GSK3␤ activity (Chen et al., 2012). Ex-4 was combination of decreased LepRb signaling and decreased BBB
also shown to improve cognitive behavior and neurogenesis in the leptin transport (Banks, 2004). Mechanistically, studies by Banks
hippocampus of adult rodents (Isacson et al., 2011) and in a mouse and Levin demonstrated that decreases in hypothalamic LepRb
model of obesity and insulin-resistance (Gault et al., 2010). Simi- signaling precedes impaired BBB leptin transport in diet-induced
larly, in the STZ mouse model of type 1 diabetes, in which we have obesity models (Levin et al., 2004). As will be described below,
demonstrated AD-like features and insulin signaling impairment hypothalamic leptin resistance also appears to be a hallmark fea-
in the brain (Jolivalt et al., 2008), we have shown that Ex-4 sig- ture of aging. Beyond the hypothalamus, this section will focus on
nificantly prevented memory impairments in the Barnes maze in the role of LepRb in the facilitation of hippocampal synaptic plastic-
diabetic mice after 8 weeks of treatment (Fig. 2), without affecting ity and will discuss the hypothesis that leptin resistance, a concept
plasma glucose or insulin levels (Jolivalt et al., 2011), suggesting normally reserved for the hypothalamus, may be a contributing fac-
that activation of GLP-1 receptor provides neuroprotective effect tor in age-related impairments in hippocampal synaptic plasticity.
in the brain of type 1 diabetic mice independent of direct glucose
or insulin modulation. These latest data showed that activation of 3.2. Leptin facilitation of hippocampal neuroplasticity: preclinical
GLP-1 receptor might have beneficial central effects in both types studies
of diabetes as well as in AD.
The role of insulin and more particularly of impaired insulin Leptin plays a key role in neuronal development (Morrison,
signaling in the brain has been clearly demonstrated in the last 2009) and also facilitates structural and functional activities
decade as a contributor to neurodegeneration and associated cog- in the adult central nervous system (CNS). Indeed, previous
nitive decline. Collectively, these recent data suggest that insulin studies have determined that leptin facilitates hippocampal
delivered directly to the brain and modulators of insulin and/or neuroplasticity in normal control rodents. For example, intrahippo-
insulin signaling pathway may represent potential therapeutic campal administration of leptin elicits dose-dependent effects
interventions against impairments of cognitive performance and on hippocampal-dependent learning (Farr et al., 2006). Similarly,
dementia for patients with diabetes, MCI or AD. intravenous (iv) administration of leptin elicits dose-dependent
effects in the passive avoidance learning and spatial learning in the
water maze (Oomura et al., 2006). Underlying these enhancements
3. Leptin in hippocampal-dependent behaviors, leptin regulates hippocam-
pal plasticity by converting short term potentiation (STP) into
3.1. Leptin signaling in the regulation of metabolic parameters LTP (Shanley et al., 2001), a cellular correlate of learning and
memory processes in the mammalian brain. More recent studies
Leptin is synthesized and secreted by adipocytes and is illustrate that leptin administration enhances LTP in CA1 pyra-
transported across the blood-brain-barrier (BBB) via a saturable midal neurons (Oomura et al., 2006) and dentate gyrus granule
transport system (Banks, 2004). In the hypothalamus, leptin is neurons (Wayner et al., 2004). Interestingly, NMDA receptor sub-
recognized as an important integrator for coordinating the periph- unit composition appears to dictate the age-dependent effects
eral and central signals that regulate food intake, body weight of leptin on synaptic transmission. Specifically, leptin medi-
and body composition [For review see Schwartz et al., 2000]. This ates long term depression (LTD) in neonatal rat hippocampal
includes functional connections with brain regions such as the slices through GluN2B-containing receptors, while eliciting LTP in
768 J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776

Fig. 2. Effect of exendin-4 (Ex-4) on memory retention in the Barnes maze for STZ-diabetic mice. Mice were trained to learn the location of the box for 5 consecutive days
in the Barnes maze, left untested for 3 day and then placed back on the Barnes maze where the box was removed but the cue was present. Panel A: data represent the
time to find the hole where the box was originally. Panel B: percent of errors spent in the quadrant where the box was originally located. Data from control mice in white
bars, Ex-4-treated control mice in gray bars, from untreated diabetic mice in black bars and from Ex-4-treated diabetic mice in hatched bars are mean + SEM, n = 5–6/group.
*p < 0.05 vs diabetic + Ex-4 by one-way ANOVA followed by Bonferroni’s post hoc test.

hippocampal slices from young rats or aged rats through GluN2B hypothalamic–pituitary–adrenal axis dysfunction and alterations
containing receptors (Moult and Harvey, 2011). Beyond these in plasma lipid profiles. As a result, the cause and effect relation-
electrophysiological measures, leptin modulation of glutamater- ship of leptin to neuroplasticity deficits described above must be
gic neurotransmission (Shanley et al., 2001; Durakoglugil et al., interpreted with caution. However, experimental models of AD and
2005; Moult et al., 2009) and glutamate receptor trafficking studies in aged rodents provide additional support for a causative
(Moult and Harvey, 2009; Moult et al., 2010) may also con- role for leptin resistance in age-related cognitive decline. For exam-
tribute to leptin-mediated enhancement of cognitive function. ple, leptin administration to control (six month-old) rats effectively
Confocal immunofluorescence analyses have determined that lep- reduces food intake and hypothalamic NPY mRNA expression,
tin regulates the morphological features of primary hippocampal but is less effective in aged (30 month-old) senescent rats (Shek
cultures, including the motility of dendritic filopodia and hip- and Scarpace, 2000; Scarpace et al., 2000a). These results suggest
pocampal synaptic density (O’Malley et al., 2007). Chronic leptin that aged rats exhibit leptin resistance. Subsequent studies from
administration increases cell proliferation in the dentate gyrus and these same investigators determined that while maximal stimu-
protects hippocampal neurons from excitotoxic damage in both lation of pSTAT3 was similar following peripheral (intravenous)
the in vivo and in vitro settings (Garza et al., 2008). Collectively, administration, the dose response curve for leptin was shifted to
these pre-clinical data support the hypothesis that leptin facili- the right in aged rats (Scarpace et al., 2000b). Interestingly, both
tates morphological, electrophysiology and behavioral measures of decreased pSTAT3 activation and the rightward shift of the lep-
hippocampal synaptic plasticity. tin dose response curve were observed in aged rats following
Neuroplasticity deficits observed in leptin deficient ob/ob mice intracerebroventricular leptin administration (Shek and Scarpace,
and rodents with impaired leptin receptor signaling (db/db mice 2000; Scarpace et al., 2001). Moreover, pSTAT3 transcription factor
and Zucker rats) provide further support for the concept that leptin binding activity was significantly reduced in aged rats (Shek and
is an important regulator and facilitator of hippocampal synaptic Scarpace, 2000; Scarpace et al., 2001). Decreases in leptin receptor
plasticity. This includes deficits in hippocampal long term poten- mRNA expression (Fernandez-Galaz et al., 2001) and protein lev-
tiation (Li et al., 2002; Gerges et al., 2003; Stranahan et al., 2008), els (Scarpace et al., 2001; Fernandez-Galaz et al., 2001), increases
morphological changes such as decreases in spine density in den- in SOCS3 mRNA levels (Wang et al., 2001), as well as age-related
tate gyrus granule neurons (Stranahan et al., 2009), as well as decreases in BBB leptin transport (Fernandez-Galaz et al., 2001)
decreases in cell proliferation and neurogenesis in the dentate may also contribute to decreased leptin responsiveness in aged
gyrus (Stranahan et al., 2008). Ultimately, these electrophysiologi- rats. While these studies focused on hypothalamic measures, the
cal and morphological deficits contribute to the behavioral deficits take home message is that aged rodents exhibit leptin resistance
observed in db/db mice and Zucker rats (Li et al., 2002; Winocur (although see Hosoi et al., 2005). An important future direction
et al., 2005). In this regard, our prior studies revealed that Zucker would be to determine whether age-related leptin resistance is spe-
rats can perform a learning task in the absence of an inter-trial cific for the hypothalamus, or whether leptin resistance extends to
interval (ITI) or when the ITI is very short. However, when the ITI is other brain regions and contributes to age-related neuroplasticity
increased, which more directly assesses hippocampal-dependent deficits in the hippocampus.
function, these leptin receptor deficient rats perform more poorly
when compared to control rats (Winocur et al., 2005). Interestingly, 3.3. Mechanistic basis of leptin-mediated neuroprotection
Zucker rats also exhibit increases in AD-like pathology, including
increases in hyper-phosphorylated tau immunoreactivity in the Studies in experimental models of AD support the notion
dentate gyrus and CA3 region of the hippocampus (Wrighten et al., that leptin provides neuroprotection from the deleterious conse-
2008). Collectively, these data illustrate that hippocampal synaptic quences of aging and have provided insight into the mechanistic
plasticity is impaired in leptin-deficient rodents. basis for this neuroprotection. For example, in the ABPP/PS1 mouse,
One caveat associated with these studies is the identifica- a transgenenic mouse model of AD, hippocampal administration of
tion of the role of leptin receptor deficiency in these models. a lentiviral vector that overexpresses leptin promotes cell prolifer-
Indeed, ob/ob mice, db/db mice and Zucker rats exhibit a wide ation and neurogenesis in the dentate gyrus and also decreases the
array of metabolic and endocrine changes that include, but are not density of Fluro-Jade histochemical labeling in the dentate gryus
limited to glucose intolerance, insulin resistance, leptin resistance, (Perez-Gonzalez et al., 2011). In other transgenic mouse models
J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776 769

of AD, leptin infusion has been shown to decrease amyloid-␤ lev- explanation and also provided analysis in structural parameters in
els in total brain extracts (Fewlass et al., 2004). This same study aged populations. In this regard, elevated plasma leptin levels were
determined that leptin treatment dose-dependently decreased A␤ associated with decreased incidence of dementia and AD. More-
uptake in cloned neuronal cells. Subsequent studies from these over, elevated plasma leptin levels were associated with larger total
same investigators confirmed these in vivo findings and also deter- brain volumes, as well as indirect assessments suggestive of larger
mined that leptin treatment reduced hippocampal amyloid burden hippocampal formation volumes (Lieb et al., 2009). Voxel-based
and that chronic leptin administration reversed the impairment morphometric analyses that provided more direct assessment of
in hippocampal-dependent learning in an AD transgenic mouse hippocampal volumes determined that elevated plasma leptin
model (Greco et al., 2010). Mechanistically, these neuroprotective levels were associated with increased gray matter volumes in
activities of leptin have been proposed to be mediated through right hippocampus and left parahippocampus in a small cohort of
LepRb activation of AMP-activated protein kinase (Greco et al., healthy elderly participants (Narita et al., 2009). Additional support
2009) and STAT5 activation (Marwarha et al., 2011). Recent studies for neurotrophic activities of leptin are provided by studies in
by Doherty et al. support the hypothesis that leptin provides protec- leptin-deficient humans receiving leptin replacement (Matochik
tion of hippocampal neurons from AD-mediated neurotoxicity. For et al., 2005; Paz-Filho et al., 2008), including morphometric
example, amyloid-␤-induced decreases in LTP and enhancement analyses suggesting that leptin replacement in this unique patient
of LTD in hippocampal slices were inhibited by bath application of population increases gray matter volumes in the right hippocam-
leptin. This facilitation of hippocampal synaptic transmission was pal formation (London et al., 2011). Taken together, these studies
inhibited by wortmannin and PI828, implicating leptin-induced suggest that elevated plasma leptin levels may protect structural
activation in the PI3 kinase pathway in these activities (Doherty and functional measures of neuroplasticity from the deleterious
et al., 2013). An obvious question that emerges from these studies in consequences of aging. It is tempting to speculate that the elevated
experimental models of aging and AD is whether these observations plasma leptin levels in these patients are indicative of increased
are translatable to the clinical setting. hippocampal leptin activity, which provides neuroprotection.
Conversely, individuals with reductions in plasma leptin levels or
3.4. Leptin and age-related cognitive decline: clinical those individuals that exhibit hippocampal leptin resistance would
observations not benefit from the neuroprotective activities of leptin and would
thereby be more vulnerable to develop MCI and AD. Clarification
While clinical studies support the concept that cognitive decline of these issues would support the concept that enhancement of
and neuroplasticity deficits in patients with metabolic disorders CNS leptin activity may represent a novel therapeutic option for
like type 2 diabetes is based in part on leptin resistance (Labad et al., the treatment of age-related disorders (Harvey, 2010).
2012), the picture is less clear regarding a causative role for leptin
resistance in age-related cognitive decline. The weaknesses in this
4. The orexins/hypocretins: neuromodulatory peptides at
causal relationship may be related to a variety of factors, includ-
the interface of energy balance and cognition
ing the potential contribution of age-related changes in endocrine
profiles (Rosenbaum et al., 1996; Ostlund et al., 1996; Baumgartner
4.1. Discovery
et al., 1999; Isidori et al., 2000), as well as age-related changes in
body composition and metabolic parameters (Perry et al., 1997;
In 1998, two independent research teams reported the dis-
Isidori et al., 2000), which includes pre-dementia weight loss
covery of a population of neuropeptide-producing cells whose
(Barrett-Connor et al., 1996, 1998). These factors provide a potential
distribution was limited to the lateral hypothalamus, the periforni-
explanation for the disparate findings in studies that have mea-
cal area and contiguous dorsomedial hypothalamus. de Lecea et al.
sured leptin activity in aging. Generally speaking, these studies
(1998) termed these peptides hypocretins (specifically, hypocretin
suggest that plasma leptin levels decrease with age in women, par-
1 and hypocretin 2) due to their restricted hypothalamic expres-
ticularly post-menopausal women and that body mass index (BMI)
sion and their homology to the incretin family of gut hormone
or fat mass are also important determinants in age-related changes
proteins. Meanwhile, Sakurai and colleagues termed these pep-
in plasma leptin levels. Although some studies suggest that leptin
tides orexin A (OxA) and orexin B (OxB), after the Greek word for
levels are decreased in AD patients (Power et al., 2001; Bigalke et al.,
appetite (orexis), because they were distributed most densely in
2011), an important question that these studies cannot address is
the classical ‘feeding center’ of the lateral hypothalamus and ini-
how these changes in plasma levels impact leptin-mediated neu-
tial experiments supported a pro-feeding effect of orexin A upon
roplasticity. More simply, can age-related cognitive decline elicited
intracerebroventricular administration (Sakurai et al., 1998). Two
by hippocampal leptin resistance be accurately assessed by mea-
G protein-coupled receptors have been described for the hypocre-
surement of plasma leptin levels?
tins/orexins (hereafter referred to as orexins for simplicity). The
A number of studies have addressed the relationship of
orexin 1 receptor (Ox1R) binds OxA with 10–100-fold greater affin-
age-related changes in plasma leptin levels to measures of neuro-
ity relative to OxB whereas the orexin 2 receptor (Ox2R) binds
plasticity such as cognitive performance and structural analyses.
OxA and OxB with roughly equal affinity (Smart and Jerman, 2002;
For example, reductions in executive function, which is proposed
Ammoun et al., 2003; Scammell and Winrow, 2011).
to reflect frontal cortical function, was associated with higher
plasma leptin levels in otherwise healthy community-dwelling
elderly participants (Gunstad et al., 2008). Conversely, higher 4.2. Distribution of cell bodies and projections
plasma leptin levels were associated with decreased cognitive
decline in a prospective study involving healthy elderly partici- In rodents and primates, orexins are uniquely localized in a
pants (Holden et al., 2009). Similarly, a more recent prospective posterior hypothalamic region that includes the lateral hypothal-
study reported that higher plasma leptin levels were associated amus, perifornical area and dorsomedial hypothalamus. Despite
with decreased risk of developing dementia/MCI in elderly women their restricted localization pattern and fairly limited number,
(Zeki et al., 2012). The designs of these studies provide a likely however, orexin neurons give rise to a diffuse pattern of projec-
explanation for these dissimilar findings; a single time point mea- tions within and beyond the diencephalon. Ascending fibers, for
sure of cognition vs analysis of cognitive decline over a four-year example, provide orexin innervation of the cerebral cortex (most
period. Results from the Framingham Study support this potential prominently, the prefrontal cortex), as well as subcortical limbic
770 J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776

structures important for learning, memory, motivation and emo- activate behavioral and metabolic pathways that enhance caloric
tion such as the nucleus accumbens, amygdala, basal forebrain expenditure, thus maintaining energy balance. For example, orexin
and hippocampus (Peyron et al., 1998). Among thalamic targets, is required for brown adipose activation (Sellayah et al., 2011;
the paraventricular nucleus of the thalamus receives a particu- Tupone et al., 2011) and food-anticipatory behavioral activation
larly dense orexin innervation (Kirouac et al., 2005). Descending (Mieda et al., 2004). These mechanisms may underlie the appar-
projections innervate numerous brainstem areas and all levels of ently paradoxical association between decreased food intake in
the spinal cord (van den Pol, 1999). Of particular interest, orexins orexin knockout mice and the obese phenotype of these same
provide substantial innervation to nuclei that are the source of dif- animals (Hara et al., 2001; Fujiki et al., 2006). Similarly, human
fuse neuromodulatory transmitters, including dopamine neurons narcolepsy patients (a “natural” human model of orexin deficiency)
of the ventral tegmental area, norepinephrine neurons of the locus have higher rates of obesity (Lammers et al., 1996; Nishino et al.,
coeruleus and cholinergic neurons of the basal forebrain (Fadel and 2001).
Deutch, 2002; Baldo et al., 2003; Fadel et al., 2005; Espana et al.,
2005). Collectively, this pattern of connectivity mediates orexin 4.4. Narcolepsy and sleep
influences over a wide range of cognitive, behavioral, autonomic
and visceral functions, consistent with their description as “physi- Pathological correlates of orexin (dys)function are clearly seen
ological integrators” (de Lecea et al., 2002). in the case of the sleep disorder, narcolepsy. Orexins are deficient (if
Abundant evidence indicates that the orexin neurons are not not completely absent) in most cases of human narcolepsy (Siegel,
functionally homogeneous, but differ along a medial-lateral gra- 1999; Nishino et al., 2000; Thannickal et al., 2000). The narcolep-
dient, with medially located neurons preferentially involved in tic phenotype of orexin knockout mice (Chemelli et al., 1999) and
arousal and stress and the more lateral bank mediating effects the finding of a loss-of-function mutation in Ox2R in a autosomal
on reward-based behaviors, including drug-seeking and feeding recessive form of canine narcolepsy (Lin et al., 1999; Reilly, 1999)
(Harris and Aston-Jones, 2006). Functional descriptions of orexin provide compelling convergent evidence from animal models that
neurons must also take into account that virtually all orexin cells orexin deficiency underlies most cases of narcolepsy. Orexin knock-
colocalize dynorphin (Chou et al., 2001) and at least some are also out mice also exhibit feeding-induced cataplexy, a sudden loss of
glutamatergic (Rosin et al., 2003; Torrealba et al., 2003; Henny et al., muscle tone triggered by strong emotions that is frequently seen
2010). in both canine and human forms of narcolepsy (Clark et al., 2009).

4.3. Orexins and energy balance 4.5. Orexins in cognition and neuroplasticity

The conditions under which the orexin system promotes food Hypothalamic neuropeptides have traditionally been viewed in
intake are not always straightforward, but the data supporting a terms of homeostatic functions mediated largely by modulation
pro-feeding role for these peptides remains compelling. Expression of neuroendocrine and autonomic circuitry. However, homeostatic
of preproorexin and orexin receptors is increased during periods functions such as feeding include cognitive components (interocep-
of food restriction (Cai et al., 1999; Lu et al., 2000; Karteris et al., tive awareness of hunger or energy balance status; remembering
2005). Intracerebroventricular or intra-hypothalamic administra- where to obtain food; attention to food-related cues or threats in
tion of orexin increases food intake in rats (Dube et al., 1999; Haynes the environment; etc.). Thus, it is not surprising that many of these
et al., 1999; Sweet et al., 1999; Baird et al., 2009) and systemic peptide systems, including the orexins, have extensive projections
administration of the Ox1R antagonist SB-334867 attenuates food to brain areas important for cognition and motivated behavior.
intake (Haynes et al., 2000; Rodgers et al., 2002). One such area is the hippocampal formation, which receives a
Like other cellular components of hypothalamic feeding cir- moderate orexin innervation and expresses both Ox1R and Ox2R.
cuitry, orexin neurons respond to several peripherally derived Orexins increase the release of acetylcholine (ACh) in the hip-
cues whose fluctuations are indicative of metabolic status. Con- pocampus (Stanley and Fadel, 2012). Orexin facilitation of synaptic
sistent with a pro-feeding role for orexin peptides these neurons plasticity has been clearly demonstrated in brain regions associated
are inhibited by leptin, with original reports suggesting a direct with drugs of abuse (Borgland et al., 2006). However, orexins also
effect mediated by leptin receptors expressed on orexin cells enhance electrophysiological correlates of hippocampal synaptic
(Hakansson et al., 1999; Yamanaka et al., 2003). However, cur- plasticity (Selbach et al., 2004, 2010). Indeed, direct administra-
rent evidence suggests that leptin inhibition of orexin neurons tion of OxA into the dentate gyrus enhances long-term potentiation
is mediated transynaptically, as leptin activates a population of in an Ox1R-dependent manner (Wayner et al., 2004). Conversely,
LepRb-positive, neurotensin-colocalizing GABAergic LH neurons orexin antagonists impair behavioral measures of hippocampal-
that give rise to local collaterals to inhibit orexin neurons (Louis dependent learning and memory (Akbari et al., 2006, 2008). Orexin
et al., 2010; Leinninger et al., 2011). Conversely, orexin neurons are neurons also give rise to a rather dense projection to the medial
activated by the gut-derived hormone ghrelin and are required for prefrontal cortex where, in rodents, they activate thalamocortical
the effects of this hormone on certain reward-related aspects of projections in a manner that corresponds with attentional func-
feeding (Yamanaka et al., 2003; Perello et al., 2010). Perhaps the tion and cognitive arousal (Lambe et al., 2005; Huang et al., 2006).
clearest response of orexin neurons to a more rapidly fluctuating Orexins increase release of ACh in the prefrontal cortex (Fadel
indicator of energy balance is the activation of these neurons by et al., 2005; Frederick-Duus et al., 2007), and performance in a
hypoglycemia and their striking inhibition by elevated—but physi- cholinergic-dependent attentional task is impaired by systemic
ologically relevant—levels of glucose via activation of tandem-pore or basal forebrain administration of an Ox1R antagonist (Boschen
potassium channels expressed directly on orexin cells (Moriguchi et al., 2009). In non-human primates, both systemic and intranasal
et al., 1999; Cai et al., 2001; Burdakov et al., 2005, 2006). Etholog- delivery of OxA ameliorates the negative effects of sleep depri-
ically, enhanced arousal is necessary during periods of low energy vation on cognitive performance (Deadwyler et al., 2007). Finally,
balance in order to promote cognitive and behavioral aspects of human narcolepsy patients, who presumably lack orexin neurons,
food-seeking. Orexin neurons comprise an ideal neural system to show attentional deficits even during periods of normal wakeful-
integrate these divergent phenomena due to their place at the ness (Rieger et al., 2003). Collectively, these observations clearly
functional intersection of reward, feeding, stress and arousal. Inter- demonstrate a role for the orexins in cognitive function. Specif-
estingly, orexins, while serving a “pro-feeding” role, also appear to ically, under conditions in which the orexin system is normally
J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776 771

activated, such as enhanced arousal or low energy balance, these contribute to age-related deficits in hippocampal plasticity. Dis-
peptides are likely to contribute to attention, learning and memory entangling the causative factors responsible for cognitive decline
processes in a manner that integrates homeostasis and cognition. in dementia and AD, as well as determination of the relative con-
tribution of these various causative factors, represents a major
4.6. Orexins and aging obstacle in the development of effective treatment strategies in
these patient populations. The presence of co-morbid conditions
Convergent studies from the human and animal literature in patients with AD and dementia, such as obesity and type 2
demonstrate that the orexin system is negatively impacted in diabetes, further complicates the identification of the causative
aging. Aged rodents have substantially fewer orexin neurons than factors involved (Wrighten et al., 2008). However, an optimistic
their young counterparts (Sawai et al., 2010; Kessler et al., 2011). viewpoint would contend that these co-morbidities aid in the
While melanin concentrating hormone neurons (which are sim- assessment and identification of potential causative factors in age-
ilarly distributed and also play a role in feeding behavior) show related cognitive decline and implicate appetitive peptides like
some degree of reduction in aged animals, the extent is milder and orexin, insulin and leptin in the pathogenesis of age-related deficits
there is no global age-related neuronal loss in the lateral hypo- in neuroplasticity. An opposing viewpoint would note that while
thalamus (Kessler et al., 2011), suggesting that orexin neurons are elevated plasma leptin levels provide neuroprotection from MCI
preferentially, if not exclusively, susceptible to age-related neu- and AD, elevated plasma leptin levels are associated with cogni-
rodegeneration. Age-related decreases in preproorexin and Ox2R tive decline in obesity and type 2 diabetes (Labad et al., 2012).
gene expression as well as diminished orexin regulation of feeding The key distinction in these studies is that plasma measures do
and neurotransmitter release have also been reported (Terao et al., not provide insight into the activity of appetitive peptides like
2002; Porkka-Heiskanen et al., 2004; Kotz et al., 2005; Stanley et al., leptin and insulin in the CNS. Indeed, it is difficult to predict
2011). Aged rats have reduced orexin innervation of medial septal whether plasma measures of these peripherally derived peptide
GABAergic neurons (Stanley et al., 2011), a functionally significant hormones effectively assess hippocampal insulin and leptin activ-
finding given that orexin receptor-expressing medial septal neu- ity. As described above, the mechanistic basis for leptin and insulin
rons mediate hippocampal theta generation (Gerashchenko et al., resistance includes decreases in BBB transport, as well as decreases
2001). Collectively, these data suggest that not only is the orexin in receptor signaling (Levin et al., 2004). One way to circumvent
system impacted by aging and peripheral metabolic dysfunction, the deficits associated with leptin and insulin resistance would be
but also orexin-based therapies may have therapeutic efficacy in through intranasal administration. The exciting studies related to
treating both the central, cognitive manifestations and some of the the therapeutic potential of intranasal insulin in the treatment of
peripheral correlates of these disorders. AD have been described above. Additionally, studies demonstrat-
In humans, post-mortem studies have also documented ing the metabolic effects of intranasal leptin administration may
decreases in orexin neurons in some age-related neurodegener- also have therapeutic potential in the treatment cognitive decline
ative disorders such as Parkinson’s disease (Thannickal et al., 2007; observed in obesity, type 2 diabetes and AD (Schulz et al., 2004;
Fronczek et al., 2008). Perhaps most relevant to age-related cogni- Waldrop et al., 2010).
tive impairment, a recent study documented a significant (∼40%) As described above, orexin neurons regulate arousal and cog-
loss of orexin neurons, relative to age-matched controls, in post- nitive function according to energy balance (Yamanaka et al.,
mortem brains from AD patients (Fronczek et al., 2012). Because 2003)—a phenomenon mediated to a significant extent via mod-
there was not a young control group included in the Alzheimer’s ulation of orexin activity by circulating factors such as glucose and
study, it is possible that the disease-specific impact is layered onto leptin. Thus, the impact of leptin resistance on behavioral and cog-
an already age-compromised orexin system. When viewed in light nitive correlates of orexin activity is important to our understand-
of the animal data, the clinical findings support the view that the ing of the relationship between age-related metabolic alterations
orexin system is impaired with age and in age-related neurocogni- and cognitive decline. Another question, of particular relevance
tive disorders. to the association between type 2 diabetes and AD, is ‘what is
How might the impact of age-related impairments in orexin the impact of years of hyperglycemia or phasically dysregulated
function be manifest in cognition and homeostasis? Clearly, given glycemic control on a neuronal population that is inhibited by ele-
the role of orexins in arousal and sleep–wake cycles, loss of these vated glucose concentrations?’ In addition, orexin knockout mice
peptides or their receptors could mediate age-related impairments develop insulin resistance in an age-dependent manner (Tsuneki
in sleep architecture. However, while overweight during middle et al., 2008), indicating that central disruptions in orexin function
age is associated with increased risk for AD, recent clinical data sug- can have peripheral metabolic consequences. Collectively, the role
gest that changes in homeostasis and energy balance in the elderly, of orexins in regulating feeding, arousal and cognition, and the asso-
including unexplained weight loss, precede and predict the subse- ciation between aging and orexin hypofunction suggests that this
quent development of AD (Knittweis, 1998; Johnson et al., 2006). An neuropeptide system serves a key integrative function at the inter-
intriguing possibility is that orexin deficiency late in life mediates face between age-related declines in cognitive and homeostatic
changes in sleep and feeding patterns that portend subsequent cog- functions. Orexin dysfunction is likely to contribute to age-related
nitive decline. Via interactions with neurotransmitters and brain cognitive complications of metabolic disorders such as diabetes and
regions classically implicated in senescent dementia, orexin defi- leptin resistance, and direct modulation of the orexin system (e.g.
ciency may also form a mechanistic link between homeostatic by orexin agonists) may have therapeutic efficacy in ameliorating
dysfunction and cognitive impairment. Because homeostatic alter- or reversing age-related cognitive decline in these disorders.
ations in the elderly generally precede the manifestation of frank
dementia by a period of months to several years, orexin-based ther-
apies may also represent a novel target for arresting or slowing this Acknowledgements
progression.
Research in the authors’ labs is supported by: a Career Devel-
5. Conclusions and future perspectives opment Award from the Juvenile Diabetes Research Foundation
and the National Institutes of Health (RO1 AG039736; CG Jolivalt);
As highlighted throughout this review, the clinical and pre- the National Institutes of Health (R01 AG030646) and American
clinical literature clearly indicates that multifactorial mechanisms Federation for Aging Research (JR Fadel); the National Institutes of
772 J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776

Health (RO1 DK017844 and R21 MH086067), the Department of Chemelli, R.M., Willie, J.T., Sinton, C.M., Elmquist, J.K., Scammell, T., Lee, C., Richard-
Veterans Affairs (IO1 BX001804-01) and the University of South son, J.A., Williams, S.C., Xiong, Y., Kisanuki, Y., Fitch, T.E., Nakazato, M., Hammer,
R.E., Saper, C.B., Yanagisawa, M., 1999. Narcolepsy in orexin knockout mice:
Carolina Research Foundation (LP Reagan). The authors would like molecular genetics of sleep regulation. Cell 98, 437–451.
to thank Victoria Macht for assistance in the preparation of the final Chen, S., Liu, A.R., An, F.M., Yao, W.B., Gao, X.D., 2012. Amelioration of neurodegener-
manuscript. ative changes in cellular and rat models of diabetes-related Alzheimer’s disease
by exendin-4. Age (Dordr) 34, 1211–1224.
Chou, T.C., Lee, C.E., Lu, J., Elmquist, J.K., Hara, J., Willie, J.T., Beuckmann, C.T.,
Chemelli, R.M., Sakurai, T., Yanagisawa, M., Saper, C.B., Scammell, T.E., 2001.
References Orexin (hypocretin) neurons contain dynorphin. The Journal of Neuroscience
21, RC168.
Abbott, M.A., Wells, D.G., Fallon, J.R., 1999. The insulin receptor tyrosine kinase sub- Clark, E.L., Baumann, C.R., Cano, G., Scammell, T.E., Mochizuki, T., 2009. Feeding-
strate p58/53 and the insulin receptor are components of CNS synapses. The elicited cataplexy in orexin knockout mice. Neuroscience 161, 970–977.
Journal of Neuroscience 19, 7300–7308. Craft, S., Baker, L.D., Montine, T.J., Minoshima, S., Watson, G.S., Claxton, A., Arbuckle,
Akbari, E., Motamedi, F., Naghdi, N., Noorbakhshnia, M., 2008. The effect of antag- M., Callaghan, M., Tsai, E., Plymate, S.R., Green, P.S., Leverenz, J., Cross, D., Gerton,
onization of orexin 1 receptors in CA1 and dentate gyrus regions on memory B., 2012. Intranasal insulin therapy for Alzheimer disease and amnestic mild
processing in passive avoidance task. Behavioural Brain Research 187, 172–177. cognitive impairment: a pilot clinical trial. Archives in Neurology 69, 29–38.
Akbari, E., Naghdi, N., Motamedi, F., 2006. Functional inactivation of orexin 1 recep- Craft, S., Cholerton, B., Baker, L.D., 2013. Insulin and Alzheimer’s disease: untangling
tors in CA1 region impairs acquisition, consolidation and retrieval in Morris the web. Journal of Alzheimer’s Disease 33 (Supl 1), S263–S275.
water maze task. Behavioural Brain Research 173, 47–52. Cukierman, T., Gerstein, H.C., Williamson, J.D., 2005. Cognitive decline and dementia
Ammoun, S., Holmqvist, T., Shariatmadari, R., Oonk, H.B., Detheux, M., Parmen- in diabetes – systematic overview of prospective observational studies. Dia-
tier, M., Akerman, K.E., Kukkonen, J.P., 2003. Distinct recognition of OX1 and betologia 48, 2460–2469.
OX2 receptors by orexin peptides. Journal of Pharmacology and Experimental Currais, A., Prior, M., Lo, D., Jolivalt, C., Schubert, D., Maher, P., 2012. Diabetes exac-
Therapeutics 305, 507–514. erbates amyloid and neurovascular pathology in aging-accelerated mice. Aging
Avila, J., Wandosell, F., Hernandez, F., 2010. Role of glycogen synthase kinase-3 in Cell 11, 107–1026.
Alzheimer’s disease pathogenesis and glycogen synthase kinase-3 inhibitors. Dandona, P., Mohamed, I., Ghanim, H., Sia, C.L., Dhindsa, S., Dandona, S., Makdissi,
Expert Review of Neurotherapeutics 10, 703–710. A., Chaudhuri, A., 2011. Insulin suppresses the expression of amyloid precursor
Baird, J.P., Choe, A., Loveland, J.L., Beck, J., Mahoney, C.E., Lord, J.S., Grigg, L.A., protein, presenilins, and glycogen synthase kinase-3beta in peripheral blood
2009. Orexin-A hyperphagia: hindbrain participation in consummatory feeding mononuclear cells. Journal of Clinical Endocrinology and Metabolism 96,
responses. Endocrinology 150, 1202–1216. 1783–1788.
Baldo, B.A., Daniel, R.A., Berridge, C.W., Kelley, A.E., 2003. Overlapping distribu- De Felice, F.G., Vieira, M.N., Bomfim, T.R., Decker, H., Velasco, P.T., Lambert, M.P.,
tions of orexin/hypocretin- and dopamine-beta-hydroxylase immunoreactive Viola, K.L., Zhao, W.Q., Ferreira, S.T., Klein, W.L., 2009. Protection of synapses
fibers in rat brain regions mediating arousal, motivation, and stress. Journal of against Alzheimer’s-linked toxins: insulin signaling prevents the pathogenic
Comparative Neurology 464, 220–237. binding of Abeta oligomers. Proceedings of the National Academy of Sciences
Banks, W.A., 2004. The many lives of leptin. Peptides 25, 331–338. of the United States of America 106, 1971–1976.
Barrett-Connor, E., Edelstein, S., Corey-Bloom, J., Wiederholt, W., 1998. Weight loss de la Monte, S.M., 2012. Therapeutic targets of brain insulin resistance in sporadic
precedes dementia in community-dwelling older adults. Journal of Nutrition, Alzheimer’s disease. Frontiers in Bioscience (Elite Edition) 4, 1582–1605.
Health and Aging 2, 113–114. de la Monte, S.M., Wands, J.R., 2005. Review of insulin and insulin-like growth factor
Barrett-Connor, E., Edelstein, S.L., Corey-Bloom, J., Wiederholt, W.C., 1996. Weight expression, signaling, and malfunction in the central nervous system: relevance
loss precedes dementia in community-dwelling older adults. Journal of the to Alzheimer’s disease. Journal of Alzheimer’s Disease 7, 45–61.
American Geriatrics Society 44, 1147–1152. de Lecea, L., Kilduff, T.S., Peyron, C., Gao, X., Foye, P.E., Danielson, P.E., Fukuhara,
Baskin, D.G., Breininger, J.F., Schwartz, M.W., 1999. Leptin receptor mRNA iden- C., Battenberg, E.L., Gautvik, V.T., Bartlett, F.S., Frankel, W.N., van den Pol, A.N.,
tifies a subpopulation of neuropeptide Y neurons activated by fasting in rat Bloom, F.E., Gautvik, K.M., Sutcliffe, J.G., 1998. The hypocretins: hypothalamus-
hypothalamus. Diabetes 48, 828–833. specific peptides with neuroexcitatory activity. Proceedings of the National
Baumgartner, R.N., Waters, D.L., Morley, J.E., Patrick, P., Montoya, G.D., Garry, P.J., Academy of Sciences of the United States of America 95, 322–327.
1999. Age-related changes in sex hormones affect the sex difference in serum de Lecea, L., Sutcliffe, J.G., Fabre, V., 2002. Hypocretins/orexins as integrators of phys-
leptin independently of changes in body fat. Metabolism: Clinical and Experi- iological information: lessons from mutant animals. Neuropeptides 36, 85–95.
mental 48, 378–384. Deadwyler, S.A., Porrino, L., Siegel, J.M., Hampson, R.E., 2007. Systemic and nasal
Bayer-Carter, J.L., Green, P.S., Montine, T.J., VanFossen, B., Baker, L.D., Watson, G.S., delivery of orexin-A (hypocretin-1) reduces the effects of sleep deprivation on
Bonner, L.M., Callaghan, M., Leverenz, J.B., Walter, B.K., Tsai, E., Plymate, S.R., cognitive performance in nonhuman primates. The Journal of Neuroscience 27,
Postupna, N., Wilkinson, C.W., Zhang, J., Lampe, J., Kahn, S.E., Craft, S., 2011. 14239–14247.
Diet intervention and cerebrospinal fluid biomarkers in amnestic mild cognitive Dineley, K.T., Kayed, R., Neugebauer, V., Fu, Y., Zhang, W., Reese, L.C., Taglialatela, G.,
impairment. Archives in Neurology 68, 743–752. 2010. Amyloid-beta oligomers impair fear conditioned memory in a calcineurin-
Benedict, C., Brooks, S.J., Kullberg, J., Burgos, J., Kempton, M.J., Nordenskjold, R., dependent fashion in mice. Journal of Neuroscience Research 88, 2923–2932.
Nylander, R., Kilander, L., Craft, S., Larsson, E.M., Johansson, L., Ahlstrom, H., Doherty, G.H., Beccano-Kelly, D., Yan, S.D., Gunn-Moore, F.J., Harvey, J., 2013. Leptin
Lind, L., Schioth, H.B., 2012. Impaired insulin sensitivity as indexed by the HOMA prevents hippocampal synaptic disruption and neuronal cell death induced by
score is associated with deficits in verbal fluency and temporal lobe gray matter amyloid beta. Neurobiology of Aging 34, 226–237.
volume in the elderly. Diabetes Care 35, 488–494. Dube, M.G., Kalra, S.P., Kalra, P.S., 1999. Food intake elicited by central administra-
Biessels, G.J., Deary, I.J., Ryan, C.M., 2008. Cognition and diabetes: a lifespan perspec- tion of orexins/hypocretins: identification of hypothalamic sites of action. Brain
tive. Lancet Neurology 7, 184–190. Research 842, 473–477.
Bigalke, B., Schreitmuller, B., Sopova, K., Paul, A., Stransky, E., Gawaz, M., Stellos, K., Duelli, R., Schrock, H., Kuschinsky, W., Hoyer, S., 1994. Intracerebroventricular
Laske, C., 2011. Adipocytokines and CD34 progenitor cells in Alzheimer’s disease. injections of streptozotocin induces discrete local changes in cerebral glucose
PLoS ONE 6, e20286. utilization in rats. International Journal of Developmental Neuroscience 12,
Borgland, S.L., Taha, S.A., Sarti, F., Fields, H.L., Bonci, A., 2006. Orexin A in the VTA 737–743.
is critical for the induction of synaptic plasticity and behavioral sensitization to Durakoglugil, M., Irving, A.J., Harvey, J., 2005. Leptin induces a novel form of
cocaine. Neuron 49, 589–601. NMDA receptor-dependent long-term depression. Journal of Neurochemistry
Boschen, K.E., Fadel, J.R., Burk, J.A., 2009. Systemic and intrabasalis administration of 95, 396–405.
the orexin-1 receptor antagonist, SB-334867, disrupts attentional performance Elmquist, J.K., Bjorbaek, C., Ahima, R.S., Flier, J.S., Saper, C.B., 1998. Distributions of
in rats. Psychopharmacology 206, 205–213. leptin receptor mRNA isoforms in the rat brain. Journal of Comparative Neurol-
Burdakov, D., Gerasimenko, O., Verkhratsky, A., 2005. Physiological changes in ogy 395, 535–547.
glucose differentially modulate the excitability of hypothalamic melanin- Espana, R.A., Reis, K.M., Valentino, R.J., Berridge, C.W., 2005. Organization of hypocre-
concentrating hormone and orexin neurons in situ. The Journal of Neuroscience tin/orexin efferents to locus coeruleus and basal forebrain arousal-related
25, 2429–2433. structures. Journal of Comparative Neurology 481, 160–178.
Burdakov, D., Jensen, L.T., Alexopoulos, H., Williams, R.H., Fearon, I.M., O’Kelly, I., Fadel, J., Deutch, A.Y., 2002. Anatomical substrates of orexin-dopamine interactions:
Gerasimenko, O., Fugger, L., Verkhratsky, A., 2006. Tandem-pore K+ channels lateral hypothalamic projections to the ventral tegmental area. Neuroscience
mediate inhibition of orexin neurons by glucose. Neuron 50, 711–722. 111, 379–387.
Burdo, J.R., Chen, Q., Calcutt, N.A., Schubert, D., 2009. The pathological interaction Fadel, J., Pasumarthi, R., Reznikov, L., 2005. Stimulation of cortical acetylcholine
between diabetes and presymptomatic Alzheimer’s disease. Neurobiology of release by orexin A. Neuroscience 130, 541–547.
Aging 30, 1910–1917. Farr, S.A., Banks, W.A., Morley, J.E., 2006. Effects of leptin on memory processing.
Cai, X.J., Evans, M.L., Lister, C.A., Leslie, R.A., Arch, J.R., Wilson, S., Williams, G., 2001. Peptides 27, 1420–1425.
Hypoglycemia activates orexin neurons and selectively increases hypothalamic Fernandez-Galaz, C., Fernandez-Agullo, T., Campoy, F., Arribas, C., Gallardo, N.,
orexin-B levels: responses inhibited by feeding and possibly mediated by the Andres, A., Ros, M., Carrascosa, J.M., 2001. Decreased leptin uptake in hypo-
nucleus of the solitary tract. Diabetes 50, 105–112. thalamic nuclei with ageing in Wistar rats. Journal of Endocrinology 171, 23–32.
Cai, X.J., Widdowson, P.S., Harrold, J., Wilson, S., Buckingham, R.E., Arch, J.R., Fewlass, D.C., Noboa, K., Pi-Sunyer, F.X., Johnston, J.M., Yan, S.D., Tezapsidis, N.,
Tadayyon, M., Clapham, J.C., Wilding, J., Williams, G., 1999. Hypothalamic orexin 2004. Obesity-related leptin regulates Alzheimer’s A beta. FASEB Journal 18,
expression: modulation by blood glucose and feeding. Diabetes 48, 2132–2137. 1870–1878.
J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776 773

Figlewicz, D.P., Benoit, S.C., 2009. Insulin, leptin, and food reward: update 2008. Hill, J.M., Lesniak, M.A., Pert, C.B., Roth, J., 1986. Autoradiographic localization of
American Journal of Physiology. Regulatory, Integrative and Comparative Phys- insulin receptors in rat brain: prominence in olfactory and limbic areas. Neuro-
iology 296, R9–R19. science 17, 1127–1138.
Figlewicz, D.P., MacDonald, N.A., Sipols, A.J., 2007. Modulation of food reward by Ho, L., Qin, W., Pompl, P.N., Xiang, Z., Wang, J., Zhao, Z., Peng, Y., Cambareri, G.,
adiposity signals. Physiology and Behavior 91, 473–478. Rocher, A., Mobbs, C.V., Hof, P.R., Pasinetti, G.M., 2004. Diet-induced insulin
Forlenza, O.V., Diniz, B.S., Radanovic, M., Santos, F.S., Talib, L.L., Gattaz, W.F., 2011. resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s
Disease-modifying properties of long-term lithium treatment for amnestic mild disease. FASEB Journal 18, 902–904.
cognitive impairment: randomised controlled trial. British Journal of Psychiatry Holden, K.F., Lindquist, K., Tylavsky, F.A., Rosano, C., Harris, T.B., Yaffe, K., 2009. Serum
198, 351–356. leptin level and cognition in the elderly: findings from the Health ABC Study.
Francis, G.J., Martinez, J.A., Liu, W.Q., Xu, K., Ayer, A., Fine, J., Tuor, U.I., Glazner, G., Neurobiology of Aging 30, 1483–1489.
Hanson, L.R., Frey, W.H., Toth, C., 2008. Intranasal insulin prevents cognitive Hosoi, T., Okuma, Y., Ono, A., Nomura, Y., 2005. Alteration of leptin-induced STAT3
decline, cerebral atrophy and white matter changes in murine type I diabetic activation in the brain of senescence-accelerated mouse (SAM) P8. Biological
encephalopathy. Brain 131, 3311–3334. and Pharmaceutical Bulletin 28, 1514–1516.
Frederick-Duus, D., Guyton, M.F., Fadel, J., 2007. Food-elicited increases in cortical Huang, C.C., Lee, C.C., Hsu, K.S., 2004. An investigation into signal transduction mech-
acetylcholine release require orexin transmission. Neuroscience 149, 499–507. anisms involved in insulin-induced long-term depression in the CA1 region of
Fronczek, R., Overeem, S., Lee, S.Y., Hegeman, I.M., van Pelt, J., van Duinen, S.G., the hippocampus. Journal of Neurochemistry 89, 217–231.
Lammers, G.J., Swaab, D.F., 2008. Hypocretin (orexin) loss and sleep disturbances Huang, C.C., You, J.L., Lee, C.C., Hsu, K.S., 2003. Insulin induces a novel form of post-
in Parkinson’s disease. Brain 131, e88. synaptic mossy fiber long-term depression in the hippocampus. Molecular and
Fronczek, R., van Geest, S., Frolich, M., Overeem, S., Roelandse, F.W., Lammers, G.J., Cellular Neurosciences 24, 831–841.
Swaab, D.F., 2012. Hypocretin (orexin) loss in Alzheimer’s disease. Neurobiology Huang, H., Ghosh, P., van den Pol, A.N., 2006. Prefrontal cortex-projecting
of Aging 33, 1642–1650. glutamatergic thalamic paraventricular nucleus-excited by hypocretin: a feed-
Fujiki, N., Yoshida, Y., Zhang, S., Sakurai, T., Yanagisawa, M., Nishino, S., 2006. Sex dif- forward circuit that may enhance cognitive arousal. Journal of Neurophysiology
ference in body weight gain and leptin signaling in hypocretin/orexin deficient 95, 1656–1668.
mouse models. Peptides 27, 2326–2331. Isacson, R., Nielsen, E., Dannaeus, K., Bertilsson, G., Patrone, C., Zachrisson, O.,
Gao, C., Holscher, C., Liu, Y., Li, L., 2012. GSK3: a key target for the development of Wikstrom, L., 2011. The glucagon-like peptide 1 receptor agonist exendin-
novel treatments for type 2 diabetes mellitus and Alzheimer disease. Reviews 4 improves reference memory performance and decreases immobility in the
in the Neurosciences 23, 1–11. forced swim test. European Journal of Pharmacology 650, 249–255.
Garza, J.C., Guo, M., Zhang, W., Lu, X.Y., 2008. Leptin promotes adult hippocam- Isidori, A.M., Strollo, F., More, M., Caprio, M., Aversa, A., Moretti, C., Frajese, G.,
pal neurogenesis in vivo and in vitro. Journal of Biological Chemistry 283, Riondino, G., Fabbri, A., 2000. Leptin and aging: correlation with endocrine
18238–18247. changes in male and female healthy adult populations of different body weights.
Gault, V.A., Porter, W.D., Flatt, P.R., Holscher, C., 2010. Actions of exendin-4 therapy Journal of Clinical Endocrinology and Metabolism 85, 1954–1962.
on cognitive function and hippocampal synaptic plasticity in mice fed a high-fat Jo, J., Whitcomb, D.J., Olsen, K.M., Kerrigan, T.L., Lo, S.C., Bru-Mercier, G., Dickinson,
diet. International Journal of Obesity (London) 34, 1341–1344. B., Scullion, S., Sheng, M., Collingridge, G., Cho, K., 2011. Abeta(1-42) inhibition
Gengler, S., McClean, P.L., McCurtin, R., Gault, V.A., Holscher, C., 2012. Val(8)GLP- of LTP is mediated by a signaling pathway involving caspase-3, Akt1 and GSK-
1 rescues synaptic plasticity and reduces dense core plaques in APP/PS1 mice. 3beta. Nature Neuroscience 14, 545–547.
Neurobiology of Aging 33, 265–276. Johnson, D.K., Wilkins, C.H., Morris, J.C., 2006. Accelerated weight loss may precede
Gerashchenko, D., Salin-Pascual, R., Shiromani, P.J., 2001. Effects of hypocretin- diagnosis in Alzheimer disease. Archives in Neurology 63, 1312–1317.
saporin injections into the medial septum on sleep and hippocampal theta. Brain Jolivalt, C.G., Fineman, M., Deacon, C.F., Carr, R.D., Calcutt, N.A., 2011. GLP-1 signals
Research 913, 106–115. via ERK in peripheral nerve and prevents nerve dysfunction in diabetic mice.
Gerges, N.Z., Aleisa, A.M., Alkadhi, K.A., 2003. Impaired long-term potentiation in Diabetes, Obesity and Metabolism 13, 990–1000.
obese zucker rats: possible involvement of presynaptic mechanism. Neuro- Jolivalt, C.G., Hurford, R., Lee, C.A., Dumaop, W., Rockenstein, E., Masliah, E., 2010.
science 120, 535–539. Type 1 diabetes exaggerates features of Alzheimer’s disease in APP transgenic
Graff, J., Koshibu, K., Jouvenceau, A., Dutar, P., Mansuy, I.M., 2010. Protein phos- mice. Experimental Neurology 223, 422–431.
phatase 1-dependent transcriptional programs for long-term memory and Jolivalt, C.G., Lee, C.A., Beiswenger, K.K., Smith, J.L., Orlov, M., Torrance, M.A., Masliah,
plasticity. Learning and Memory 17, 355–363. E., 2008. Defective insulin signaling pathway and increased glycogen synthase
Greco, S.J., Bryan, K.J., Sarkar, S., Zhu, X., Smith, M.A., Ashford, J.W., Johnston, J.M., kinase-3 activity in the brain of diabetic mice: parallels with Alzheimer’s dis-
Tezapsidis, N., Casadesus, G., 2010. Leptin reduces pathology and improves ease and correction by insulin. Journal of Neuroscience Research 86, 3265–
memory in a transgenic mouse model of Alzheimer’s disease. Journal of 3274.
Alzheimer’s Disease 19, 1155–1167. Jonas, E.A., Knox, R.J., Smith, T.C., Wayne, N.L., Connor, J.A., Kaczmarek, L.K., 1997.
Greco, S.J., Sarkar, S., Johnston, J.M., Tezapsidis, N., 2009. Leptin regulates tau phos- Regulation by insulin of a unique neuronal Ca2+ pool and of neuropeptide secre-
phorylation and amyloid through AMPK in neuronal cells. Biochemical and tion. Nature 385, 343–346.
Biophysical Research Communications 380, 98–104. Karteris, E., Machado, R.J., Chen, J., Zervou, S., Hillhouse, E.W., Randeva, H.S.,
Grillo, C.A., Piroli, G.G., Hendry, R.M., Reagan, L.P., 2009. Insulin-stimulated translo- 2005. Food deprivation differentially modulates orexin receptor expression and
cation of GLUT4 to the plasma membrane in rat hippocampus is PI3-kinase signaling in rat hypothalamus and adrenal cortex. American Journal of Physiol-
dependent. Brain Research 1296, 35–45. ogy. Endocrinology and Metabolism 288, E1089–E1100.
Gunstad, J., Spitznagel, M.B., Keary, T.A., Glickman, E., Alexander, T., Karrer, J., Stanek, Ke, Y.D., Delerue, F., Gladbach, A., Gotz, J., Ittner, L.M., 2009. Experimental diabetes
K., Reese, L., Juvancic-Heltzel, J., 2008. Serum leptin levels are associated with mellitus exacerbates tau pathology in a transgenic mouse model of Alzheimer’s
cognitive function in older adults. Brain Research 1230, 233–236. disease. PLoS ONE 4, e7917.
Hakansson, M., de Lecea, L., Sutcliffe, J.G., Yanagisawa, M., Meister, B., 1999. Leptin Kessler, B.A., Stanley, E.M., Frederick-Duus, D., Fadel, J., 2011. Age-related loss of
receptor- and STAT3-immunoreactivities in hypocretin/orexin neurones of the orexin/hypocretin neurons. Neuroscience 178, 82–88.
lateral hypothalamus. Journal of Neuroendocrinology 11, 653–663. King, M.R., Anderson, N.J., Guernsey, L.S., Jolivalt, C.G., 2013. Glycogen synthase
Hara, J., Beuckmann, C.T., Nambu, T., Willie, J.T., Chemelli, R.M., Sinton, C.M., kinase-3 inhibition prevents learning deficits in diabetic mice. Journal of Neu-
Sugiyama, F., Yagami, K., Goto, K., Yanagisawa, M., Sakurai, T., 2001. Genetic abla- roscience Research 91, 506–514.
tion of orexin neurons in mice results in narcolepsy, hypophagia, and obesity. Kirouac, G.J., Parsons, M.P., Li, S., 2005. Orexin (hypocretin) innervation of the para-
Neuron 30, 345–354. ventricular nucleus of the thalamus. Brain Research 1059, 179–188.
Harris, G.C., Aston-Jones, G., 2006. Arousal and reward: a dichotomy in orexin func- Knittweis, J., 1998. Weight loss precedes Alzheimer’s disease symptoms: a case
tion. Trends in the Neurological Sciences 29, 571–577. study. Journal of the American Geriatrics Society 46, 540–541.
Harvey, J., 2010. Leptin: the missing link in Alzheimer disease? Clinical Chemistry Kotz, C.M., Mullett, M.A., Wang, C., 2005. Diminished feeding responsiveness to
56, 696–697. orexin A (hypocretin 1) in aged rats is accompanied by decreased neuronal acti-
Havrankova, J., Brownstein, M., Roth, J., 1981. Insulin and insulin receptors in rodent vation. American Journal of Physiology. Regulatory, Integrative and Comparative
brain. Diabetologia 20 (Suppl.), 268–273. Physiology 289, R359–R366.
Havrankova, J., Roth, J., Brownstein, M., 1978. Insulin receptors are widely dis- Labad, J., Price, J.F., Strachan, M.W., Deary, I.J., Seckl, J.R., Sattar, N., Reynolds, R.M.,
tributed in the central nervous system of the rat. Nature 272, 827–829. 2012. Serum leptin and cognitive function in people with type 2 diabetes. Neu-
Havrankova, J., Roth, J., Brownstein, M.J., 1979. Concentrations of insulin and insulin robiology of Aging 33, 2938–2941.
receptors in the brain are independent of peripheral insulin levels. Studies of Lambe, E.K., Olausson, P., Horst, N.K., Taylor, J.R., Aghajanian, G.K., 2005. Hypocre-
obese and streptozotocin-treated rodents. Journal of Clinical Investigation 64, tin and nicotine excite the same thalamocortical synapses in prefrontal cortex:
636–642. correlation with improved attention in rat. The Journal of Neuroscience 25,
Haynes, A.C., Jackson, B., Chapman, H., Tadayyon, M., Johns, A., Porter, R.A., Arch, 5225–5229.
J.R., 2000. A selective orexin-1 receptor antagonist reduces food consumption Lammers, G.J., Pijl, H., Iestra, J., Langius, J.A., Buunk, G., Meinders, A.E., 1996. Spon-
in male and female rats. Regulatory Peptides 96, 45–51. taneous food choice in narcolepsy. Sleep 19, 75–76.
Haynes, A.C., Jackson, B., Overend, P., Buckingham, R.E., Wilson, S., Tadayyon, M., Lannert, H., Hoyer, S., 1998. Intracerebroventricular administration of streptozotocin
Arch, J.R., 1999. Effects of single and chronic intracerebroventricular adminis- causes long-term diminutions in learning and memory abilities and in cerebral
tration of the orexins on feeding in the rat. Peptides 20, 1099–1105. energy metabolism in adults rats. Behavioral Neuroscience 112, 1199–1208.
Henny, P., Brischoux, F., Mainville, L., Stroh, T., Jones, B.E., 2010. Immunohistochemi- Lee, C.C., Huang, C.C., Hsu, K.S., 2011. Insulin promotes dendritic spine and
cal evidence for synaptic release of glutamate from orexin terminals in the locus synapse formation by the PI3K/Akt/mTOR and Rac1 signaling pathways. Neu-
coeruleus. Neuroscience 169, 1150–1157. ropharmacology 61, 867–879.
774 J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776

Lee, C.C., Huang, C.C., Wu, M.Y., Hsu, K.S., 2005. Insulin stimulates postsynap- Moult, P.R., Cross, A., Santos, S.D., Carvalho, A.L., Lindsay, Y., Connolly, C.N., Irving,
tic density-95 protein translation via the phosphoinositide 3-kinase-Akt- A.J., Leslie, N.R., Harvey, J., 2010. Leptin regulates AMPA receptor trafficking via
mammalian target of rapamycin signaling pathway. Journal of Biological PTEN inhibition. The Journal of Neuroscience 30, 4088–4101.
Chemistry 280, 18543–18550. Moult, P.R., Harvey, J., 2009. Regulation of glutamate receptor trafficking by leptin.
Leibson, C.L., Rocca, W.A., Hanson, V.A., Cha, R., Kokmen, E., O’Brien, P.C., Palumbo, Biochemical Society Transactions 37, 1364–1368.
P.J., 1997. Risk of dementia among persons with diabetes mellitus: a population- Moult, P.R., Harvey, J., 2011. NMDA receptor subunit composition determines the
based cohort study. American Journal of Epidemiology 145, 301–308. polarity of leptin-induced synaptic plasticity. Neuropharmacology 61, 924–936.
Leinninger, G.M., Jo, Y.H., Leshan, R.L., Louis, G.W., Yang, H., Barrera, J.G., Wilson, H., Moult, P.R., Milojkovic, B., Harvey, J., 2009. Leptin reverses long-term poten-
Opland, D.M., Faouzi, M.A., Gong, Y., Jones, J.C., Rhodes, C.J., Chua Jr., S., Diano, S., tiation at hippocampal CA1 synapses. Journal of Neurochemistry 108,
Horvath, T.L., Seeley, R.J., Becker, J.B., Munzberg, H., Myers Jr., M.G., 2009. Leptin 685–696.
acts via leptin receptor-expressing lateral hypothalamic neurons to modulate Munzberg, H., Myers Jr., M.G., 2005. Molecular and anatomical determinants of
the mesolimbic dopamine system and suppress feeding. Cell Metabolism 10, central leptin resistance. Nature Neuroscience 8, 566–570.
89–98. Myers Jr., M.G., Leibel, R.L., Seeley, R.J., Schwartz, M.W., 2010. Obesity and lep-
Leinninger, G.M., Opland, D.M., Jo, Y.H., Faouzi, M., Christensen, L., Cappellucci, L.A., tin resistance: distinguishing cause from effect. Trends in Endocrinology and
Rhodes, C.J., Gnegy, M.E., Becker, J.B., Pothos, E.N., Seasholtz, A.F., Thompson, R.C., Metabolism 21, 643–651.
Myers Jr., M.G., 2011. Leptin action via neurotensin neurons controls orexin, Nadebaum, C., Scratch, S.E., Northam, E.A., Cameron, F.J., 2012. Clinical utility of
the mesolimbic dopamine system and energy balance. Cell Metabolism 14, mental state screening as a predictor of intellectual outcomes 6 months after
313–323. diagnosis of type 1 diabetes. Pediatric Diabetes 13, 632–637.
Lester-Coll, N., Rivera, E.J., Soscia, S.J., Doiron, K., Wands, J.R., de la Monte, S.M., 2006. Narita, K., Kosaka, H., Okazawa, H., Murata, T., Wada, Y., 2009. Relationship between
Intracerebral streptozotocin model of type 3 diabetes: relevance to sporadic plasma leptin level and brain structure in elderly: a voxel-based morphometric
Alzheimer’s disease. Journal of Alzheimer’s Disease 9, 13–33. study. Biological Psychiatry 65, 992–994.
Levin, B.E., Dunn-Meynell, A.A., Banks, W.A., 2004. Obesity-prone rats have nor- Nishino, S., Ripley, B., Overeem, S., Lammers, G.J., Mignot, E., 2000. Hypocretin
mal blood–brain barrier transport but defective central leptin signaling before (orexin) deficiency in human narcolepsy. The Lancet 355, 39–40.
obesity onset. American Journal of Physiology. Regulatory, Integrative and Com- Nishino, S., Ripley, B., Overeem, S., Nevsimalova, S., Lammers, G.J., Vankova, J., Okun,
parative Physiology 286, R143–R150. M., Rogers, W., Brooks, S., Mignot, E., 2001. Low cerebrospinal fluid hypocre-
Li, J., Deng, J., Sheng, W., Zuo, Z., 2012a. Metformin attenuates Alzheimer’s disease- tin (Orexin) and altered energy homeostasis in human narcolepsy. Annals of
like neuropathology in obese, leptin-resistant mice. Pharmacology Biochemistry Neurology 50, 381–388.
and Behavior 101, 564–574. Nistico, R., Cavallucci, V., Piccinin, S., Macri, S., Pignatelli, M., Mehdawy, B., Blan-
Li, L., Zhang, Z.F., Holscher, C., Gao, C., Jiang, Y.H., Liu, Y.Z., 2012b. (Val(8)) dini, F., Laviola, G., Lauro, D., Mercuri, N.B., D’Amelio, M., 2012. Insulin receptor
glucagon-like peptide-1 prevents tau hyperphosphorylation, impairment of beta-subunit haploinsufficiency impairs hippocampal late-phase LTP and recog-
spatial learning and ultra-structural cellular damage induced by streptozotocin nition memory. Neuromolecular Medicine 14, 262–269.
in rat brains. European Journal of Pharmacology 674, 280–286. O’Malley, D., MacDonald, N., Mizielinska, S., Connolly, C.N., Irving, A.J., Harvey, J.,
Li, X.L., Aou, S., Oomura, Y., Hori, N., Fukunaga, K., Hori, T., 2002. Impairment of 2007. Leptin promotes rapid dynamic changes in hippocampal dendritic mor-
long-term potentiation and spatial memory in leptin receptor-deficient rodents. phology. Molecular and Cellular Neurosciences 35, 559–572.
Neuroscience 113, 607–615. Oomura, Y., Hori, N., Shiraishi, T., Fukunaga, K., Takeda, H., Tsuji, M., Matsumiya,
Lieb, W., Beiser, A.S., Vasan, R.S., Tan, Z.S., Au, R., Harris, T.B., Roubenoff, R., Auerbach, T., Ishibashi, M., Aou, S., Li, X.L., Kohno, D., Uramura, K., Sougawa, H., Yada,
S., Decarli, C., Wolf, P.A., Seshadri, S., 2009. Association of plasma leptin levels T., Wayner, M.J., Sasaki, K., 2006. Leptin facilitates learning and memory per-
with incident Alzheimer disease and MRI measures of brain aging. Journal of the formance and enhances hippocampal CA1 long-term potentiation and CaMK II
American Medical Society 302, 2565–2572. phosphorylation in rats. Peptides 27, 2738–2749.
Lin, L., Faraco, J., Li, R., Kadotani, H., Rogers, W., Lin, X., Qiu, X., de Jong, P.J., Nishino, S., Ostlund Jr., R.E., Yang, J.W., Klein, S., Gingerich, R., 1996. Relation between plasma
Mignot, E., 1999. The sleep disorder canine narcolepsy is caused by a mutation leptin concentration and body fat, gender, diet, age, and metabolic covariates.
in the hypocretin (orexin) receptor 2 gene. Cell 98, 365–376. Journal of Clinical Endocrinology and Metabolism 81, 3909–3913.
Liu, L., Brown III, J.C., Webster, W.W., Morrisett, R.A., Monaghan, D.T., 1995. Insulin Ott, A., Stolk, R.P., Hofman, A., van Harskamp, F., Grobbee, D.E., Breteler, M.M., 1996.
potentiates N-methyl-d-aspartate receptor activity in Xenopus oocytes and rat Association of diabetes mellitus and dementia: the Rotterdam Study. Diabetolo-
hippocampus. Neuroscience Letters 192, 5–8. gia 39, 1392–1397.
Liu, Y., Liu, F., Grundke-Iqbal, I., Iqbal, K., Gong, C.X., 2011. Deficient brain insulin Paz-Filho, G.J., Babikian, T., Asarnow, R., Delibasi, T., Esposito, K., Erol, H.K., Wong,
signalling pathway in Alzheimer’s disease and diabetes. Journal of Pathology M.L., Licinio, J., 2008. Leptin replacement improves cognitive development. PLoS
225, 54–62. ONE 3, e3098.
London, E.D., Berman, S.M., Chakrapani, S., Delibasi, T., Monterosso, J., Erol, H.K., Peila, R., Rodriguez, B.L., Launer, L.J., 2002. Type 2 diabetes, APOE gene, and the risk
Paz-Filho, G., Wong, M.L., Licinio, J., 2011. Short-term plasticity of gray mat- for dementia and related pathologies: the Honolulu-Asia Aging Study. Diabetes
ter associated with leptin deficiency and replacement. Journal of Clinical 51, 1256–1262.
Endocrinology and Metabolism 96, E1212–E1220. Perello, M., Sakata, I., Birnbaum, S., Chuang, J.C., Osborne-Lawrence, S., Rovinsky, S.A.,
Louis, G.W., Leinninger, G.M., Rhodes, C.J., Myers Jr., M.G., 2010. Direct innervation Woloszyn, J., Yanagisawa, M., Lutter, M., Zigman, J.M., 2010. Ghrelin increases
and modulation of orexin neurons by lateral hypothalamic LepRb neurons. The the rewarding value of high-fat diet in an orexin-dependent manner. Biological
Journal of Neuroscience 30, 11278–11287. Psychiatry 67, 880–886.
Lu, X.Y., Bagnol, D., Burke, S., Akil, H., Watson, S.J., 2000. Differential distribution and Perez-Gonzalez, R., Antequera, D., Vargas, T., Spuch, C., Bolos, M., Carro, E., 2011.
regulation of OX1 and OX2 orexin/hypocretin receptor messenger RNA in the Leptin induces proliferation of neuronal progenitors and neuroprotection in a
brain upon fasting. Hormones and Behavior 37, 335–344. mouse model of Alzheimer’s disease. Journal of Alzheimer’s Disease 24 (Suppl.
Manschot, S.M., Brands, A.M., van der, G.J., Kessels, R.P., Algra, A., Kappelle, L.J., 2), 17–25.
Biessels, G.J., 2006. Brain magnetic resonance imaging correlates of impaired Perry III, H.M., Morley, J.E., Horowitz, M., Kaiser, F.E., Miller, D.K., Wittert, G., 1997.
cognition in patients with type 2 diabetes. Diabetes 55, 1106–1113. Body composition and age in African-American and Caucasian women: rela-
Marwarha, G., Prasanthi, J.R., Schommer, J., Dasari, B., Ghribi, O., 2011. Molecu- tionship to plasma leptin levels. Metabolism: Clinical and Experimental 46,
lar interplay between leptin, insulin-like growth factor-1, and beta-amyloid in 1399–1405.
organotypic slices from rabbit hippocampus. Molecular Neurodegeneration 6, Peyron, C., Tighe, D.K., van den Pol, A.N., de Lecea, L., Heller, H.C., Sutcliffe, J.G., Kilduff,
41. T.S., 1998. Neurons containing hypocretin (orexin) project to multiple neuronal
Matochik, J.A., London, E.D., Yildiz, B.O., Ozata, M., Caglayan, S., Depaoli, A.M., Wong, systems. The Journal of Neuroscience 18, 9996–10015.
M.L., Licinio, J., 2005. Effect of leptin replacement on brain structure in geneti- Pintana, H., Apaijai, N., Pratchayasakul, W., Chattipakorn, N., Chattipakorn, S.C., 2012.
cally leptin-deficient adults. Journal of Clinical Endocrinology and Metabolism Effects of metformin on learning and memory behaviors and brain mitochon-
90, 2851–2854. drial functions in high fat diet induced insulin resistant rats. Life Sciences 91,
McCarthy, A.M., Lindgren, S., Mengeling, M.A., Tsalikian, E., Engvall, J.C., 2002. Effects 409–414.
of diabetes on learning in children. Pediatrics 109, E9. Plaschke, K., Hoyer, S., 1993. Action of the diabetogenic drug streptozotocin on
McClean, P.L., Gault, V.A., Harriott, P., Holscher, C., 2010. Glucagon-like peptide-1 glycolytic and glycogenolytic metabolism in adult rat brain cortex and hip-
analogues enhance synaptic plasticity in the brain: a link between diabetes and pocampus. International Journal of Developmental Neuroscience 11, 477–
Alzheimer’s disease. European Journal of Pharmacology 630, 158–162. 483.
McClean, P.L., Parthsarathy, V., Faivre, E., Holscher, C., 2011. The diabetes drug Plaschke, K., Kopitz, J., Siegelin, M., Schliebs, R., Salkovic-Petrisic, M., Riederer,
liraglutide prevents degenerative processes in a mouse model of Alzheimer’s P., Hoyer, S., 2010. Insulin-resistant brain state after intracerebroventricu-
disease. The Journal of Neuroscience 31, 6587–6594. lar streptozotocin injection exacerbates Alzheimer-like changes in Tg2576
Mieda, M., Williams, S.C., Sinton, C.M., Richardson, J.A., Sakurai, T., Yanagisawa, M., AbetaPP-overexpressing mice. Journal of Alzheimer’s Disease 19, 691–
2004. Orexin neurons function in an efferent pathway of a food-entrainable 704.
circadian oscillator in eliciting food-anticipatory activity and wakefulness. The Ponce-Lopez, T., Liy-Salmeron, G., Hong, E., Meneses, A., 2011. Lithium, phenserine,
Journal of Neuroscience 24, 10493–10501. memantine and pioglitazone reverse memory deficit and restore phospho-
Moriguchi, T., Sakurai, T., Nambu, T., Yanagisawa, M., Goto, K., 1999. Neurons con- GSK3beta decreased in hippocampus in intracerebroventricular streptozotocin
taining orexin in the lateral hypothalamic area of the adult rat brain are activated induced memory deficit model. Brain Research 1426, 73–85.
by insulin-induced acute hypoglycemia. Neuroscience Letters 264, 101–104. Porkka-Heiskanen, T., Alanko, L., Kalinchuk, A., Heiskanen, S., Stenberg, D., 2004. The
Morrison, C.D., 2009. Leptin signaling in brain: a link between nutrition and cogni- effect of age on prepro-orexin gene expression and contents of orexin A and B
tion? Biochimica et Biophysica Acta 1792, 401–408. in the rat brain. Neurobiology of Aging 25, 231–238.
J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776 775

Porter, D., Faivre, E., Flatt, P.R., Holscher, C., Gault, V.A., 2012. Actions of incretin Shingo, A.S., Kanabayashi, T., Murase, T., Kito, S., 2012. Cognitive decline in STZ-3V
metabolites on locomotor activity, cognitive function and in vivo hippocampal rats is largely due to dysfunctional insulin signalling through the dentate gyrus.
synaptic plasticity in high fat fed mice. Peptides 35, 1–8. Behavioural Brain Research 229, 378–383.
Porter, D.W., Kerr, B.D., Flatt, P.R., Holscher, C., Gault, V.A., 2010. Four weeks admin- Siegel, J.M., 1999. Narcolepsy: a key role for hypocretins (orexins). Cell 98, 409–412.
istration of Liraglutide improves memory and learning as well as glycaemic Smart, D., Jerman, J., 2002. The physiology and pharmacology of the orexins. Phar-
control in mice with high fat dietary-induced obesity and insulin resistance. macology & Therapeutics 94, 51–61.
Diabetes, Obesity and Metabolism 12, 891–899. Stanley, E.M., Fadel, J., 2012. Aging-related deficits in orexin/hypocretin modulation
Power, D.A., Noel, J., Collins, R., O’Neill, D., 2001. Circulating leptin levels and weight of the septohippocampal cholinergic system. Synapse 66, 445–452.
loss in Alzheimer’s disease patients. Dementia and Geriatric Cognitive Disorders Stanley, E.M., Fadel, J.R., Mott, D.D., 2011. Interneuron loss reduces dendritic inhibi-
12, 167–170. tion and GABA release in hippocampus of aged rats. Neurobiology of Aging 33,
Reaven, G.M., Thompson, L.W., Nahum, D., Haskins, E., 1990. Relationship between 431.e1–431.e13.
hyperglycemia and cognitive function in older NIDDM patients. Diabetes Care Stanley, E.M., Wilson, M.A., Fadel, J.R., 2012. Hippocampal neurotransmitter efflux
13, 16–21. during one-trial novel object recognition in rats. Neuroscience Letters 511,
Reilly, C.E., 1999. I. Mutation in the hypocretin (orexin) receptor 2 gene causes canine 38–42.
narcolepsy. Journal of Neurology 246, 985–986. Steen, E., Terry, B.M., Rivera, E.J., Cannon, J.L., Neely, T.R., Tavares, R., Xu, X.J., Wands,
Revill, P., Moral, M.A., Prous, J.R., 2006. Impaired insulin signaling and the pathogen- J.R., de la Monte, S.M., 2005. Impaired insulin and insulin-like growth factor
esis of Alzheimer’s disease. Drugs Today (Barcelona) 42, 785–790. expression and signaling mechanisms in Alzheimer’s disease – is this type 3
Rieger, M., Mayer, G., Gauggel, S., 2003. Attention deficits in patients with narcolepsy. diabetes? Journal of Alzheimer’s Disease 7, 63–80.
Sleep 26, 36–43. Stranahan, A.M., Arumugam, T.V., Cutler, R.G., Lee, K., Egan, J.M., Mattson, M.P.,
Rodgers, R.J., Ishii, Y., Halford, J.C., Blundell, J.E., 2002. Orexins and appetite regula- 2008. Diabetes impairs hippocampal function through glucocorticoid-mediated
tion. Neuropeptides 36, 303–325. effects on new and mature neurons. Nature Neuroscience 11, 309–317.
Ronnemaa, E., Zethelius, B., Sundelof, J., Sundstrom, J., Degerman-Gunnarsson, M., Stranahan, A.M., Lee, K., Martin, B., Maudsley, S., Golden, E., Cutler, R.G., Mattson,
Lannfelt, L., Berne, C., Kilander, L., 2009. Glucose metabolism and the risk of M.P., 2009. Voluntary exercise and caloric restriction enhance hippocampal den-
Alzheimer’s disease and dementia: a population-based 12 year follow-up study dritic spine density and BDNF levels in diabetic mice. Hippocampus 19, 951–961.
in 71-year-old men. Diabetologia 52, 1504–1510. Sweet, D.C., Levine, A.S., Billington, C.J., Kotz, C.M., 1999. Feeding response to central
Rosenbaum, M., Nicolson, M., Hirsch, J., Heymsfield, S.B., Gallagher, D., Chu, F., Leibel, orexins. Brain Research 821, 535–538.
R.L., 1996. Effects of gender, body composition, and menopause on plasma Taghibiglou, C., Bradley, C.A., Gaertner, T., Li, Y., Wang, Y., Wang, Y.T., 2009.
concentrations of leptin. Journal of Clinical Endocrinology and Metabolism 81, Mechanisms involved in cholesterol-induced neuronal insulin resistance. Neu-
3424–3427. ropharmacology 57, 268–276.
Rosin, D.L., Weston, M.C., Sevigny, C.P., Stornetta, R.L., Guyenet, P.G., 2003. Hypo- Takeda, S., Sato, N., Uchio-Yamada, K., Sawada, K., Kunieda, T., Takeuchi, D., Kuri-
thalamic orexin (hypocretin) neurons express vesicular glutamate transporters nami, H., Shinohara, M., Rakugi, H., Morishita, R., 2010. Diabetes-accelerated
VGLUT1 or VGLUT2. Journal of Comparative Neurology 465, 593–603. memory dysfunction via cerebrovascular inflammation and Abeta deposition in
Rovet, J.F., Ehrlich, R.M., Hoppe, M., 1987. Intellectual deficits associated with early an Alzheimer mouse model with diabetes. Proceedings of the National Academy
onset of insulin-dependent diabetes mellitus in children. Diabetes Care 10, of Sciences of the United States of America 107, 7036–7041.
510–515. Takeda, S., Sato, N., Uchio-Yamada, K., Yu, H., Moriguchi, A., Rakugi, H., Morishita, R.,
Ryan, C., Vega, A., Drash, A., 1985. Cognitive deficits in aldolescents who developed 2012. Oral glucose loading modulates plasma beta-amyloid Level in Alzheimer’s
diabetes early in life. Pediatrics 75, 921–927. disease patients: potential diagnostic method for Alzheimer’s disease. Dementia
Ryan, C.M., Williams, T.M., 1993. Effects of insulin-dependent diabetes on learning and Geriatric Cognitive Disorders 34, 25–30.
and memory efficiency in adults. Journal of Clinical and Experimental Neuropsy- Talbot, K., Wang, H.Y., Kazi, H., Han, L.Y., Bakshi, K.P., Stucky, A., Fuino, R.L.,
chology 15, 685–700. Kawaguchi, K.R., Samoyedny, A.J., Wilson, R.S., Arvanitakis, Z., Schneider, J.A.,
Sakurai, T., Amemiya, A., Ishii, M., Matsuzaki, I., Chemelli, R.M., Tanaka, H., Williams, Wolf, B.A., Bennett, D.A., Trojanowski, J.Q., Arnold, S.E., 2012. Demonstrated
S.C., Richardson, J.A., Kozlowski, G.P., Wilson, S., Arch, J.R., Buckingham, R.E., brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1
Haynes, A.C., Carr, S.A., Annan, R.S., McNulty, D.E., Liu, W.S., Terrett, J.A., resistance, IRS-1 dysregulation, and cognitive decline. Journal of Clinical Inves-
Elshourbagy, N.A., Bergsma, D.J., Yanagisawa, M., 1998. Orexins and orexin tigation 122, 1316–1338.
receptors: a family of hypothalamic neuropeptides and G protein-coupled Terao, A., Apte-Deshpande, A., Morairty, S., Freund, Y.R., Kilduff, T.S., 2002. Age-
receptors that regulate feeding behavior. Cell 92, 573–585. related decline in hypocretin (orexin) receptor 2 messenger RNA levels in the
Santos, T.D., Mazucanti, C.H., Xavier, G.F., Torrao, A.D., 2012. Early and late neurode- mouse brain. Neuroscience Letters 332, 190–194.
generation and memory disruption after intracerebroventricular streptozotocin. Thannickal, T.C., Lai, Y.Y., Siegel, J.M., 2007. Hypocretin (orexin) cell loss in Parkin-
Physiology and Behavior 107, 401–413. son’s disease. Brain 130, 1586–1595.
Sawai, N., Ueta, Y., Nakazato, M., Ozawa, H., 2010. Developmental and aging change Thannickal, T.C., Moore, R.Y., Nienhuis, R., Ramanathan, L., Gulyani, S., Aldrich,
of orexin-A and -B immunoreactive neurons in the male rat hypothalamus. M., Cornford, M., Siegel, J.M., 2000. Reduced number of hypocretin neurons in
Neuroscience Letters 468, 51–55. human narcolepsy. Neuron 27, 469–474.
Scammell, T.E., Winrow, C.J., 2011. Orexin receptors: pharmacology and therapeutic Torrealba, F., Yanagisawa, M., Saper, C.B., 2003. Colocalization of orexin a and gluta-
opportunities. Annual Review of Pharmacology and Toxicology 51, 243–266. mate immunoreactivity in axon terminals in the tuberomammillary nucleus in
Scarpace, P.J., Matheny, M., Moore, R.L., Tumer, N., 2000a. Impaired leptin respon- rats. Neuroscience 119, 1033–1044.
siveness in aged rats. Diabetes 49, 431–435. Tsuneki, H., Murata, S., Anzawa, Y., Soeda, Y., Tokai, E., Wada, T., Kimura, I.,
Scarpace, P.J., Matheny, M., Shek, E.W., 2000b. Impaired leptin signal transduction Yanagisawa, M., Sakurai, T., Sasaoka, T., 2008. Age-related insulin resistance in
with age-related obesity. Neuropharmacology 39, 1872–1879. hypothalamus and peripheral tissues of orexin knockout mice. Diabetologia 51,
Scarpace, P.J., Matheny, M., Tumer, N., 2001. Hypothalamic leptin resistance is asso- 657–667.
ciated with impaired leptin signal transduction in aged obese rats. Neuroscience Tupone, D., Madden, C.J., Cano, G., Morrison, S.F., 2011. An orexinergic projection
104, 1111–1117. from perifornical hypothalamus to raphe pallidus increases rat brown adipose
Schulz, C., Paulus, K., Lehnert, H., 2004. Central nervous and metabolic effects of tissue thermogenesis. The Journal of Neuroscience 31, 15944–15955.
intranasally applied leptin. Endocrinology 145, 2696–2701. Unger, J., McNeill, T.H., Moxley III, R.T., White, M., Moss, A., Livingston, J.N., 1989.
Schwartz, M.W., Figlewicz, D.P., Baskin, D.G., Woods, S.C., Porte Jr., D., 1992. Insulin Distribution of insulin receptor-like immunoreactivity in the rat forebrain. Neu-
in the brain: a hormonal regulator of energy balance. Endocrinology Reviews roscience 31, 143–157.
13, 387–414. van den Pol, A.N., 1999. Hypothalamic hypocretin (orexin): robust innervation of
Schwartz, M.W., Woods, S.C., Porte Jr., D., Seeley, R.J., Baskin, D.G., 2000. Central the spinal cord. The Journal of Neuroscience 19, 3171–3182.
nervous system control of food intake. Nature 404, 661–671. Velayudhan, L., Poppe, M., Archer, N., Proitsi, P., Brown, R.G., Lovestone, S., 2010. Risk
Selbach, O., Bohla, C., Barbara, A., Doreulee, N., Eriksson, K.S., Sergeeva, O.A., Haas, of developing dementia in people with diabetes and mild cognitive impairment.
H.L., 2010. Orexins/hypocretins control bistability of hippocampal long-term British Journal of Psychiatry 196, 36–40.
synaptic plasticity through co-activation of multiple kinases. Acta Physiologica Waldrop, M.A., Leinung, M.C., Lee, D.W., Grasso, P., 2010. Intranasal delivery of mouse
(Oxford) 198, 277–285. [d-Leu-4]-OB3, a synthetic peptide amide with leptin-like activity, improves
Selbach, O., Doreulee, N., Bohla, C., Eriksson, K.S., Sergeeva, O.A., Poelchen, W., Brown, energy balance, glycaemic control, insulin sensitivity and bone formation in
R.E., Haas, H.L., 2004. Orexins/hypocretins cause sharp wave- and theta-related leptin-resistant C57BLK/6-m db/db mice. Diabetes, Obesity and Metabolism 12,
synaptic plasticity in the hippocampus via glutamatergic, gabaergic, noradren- 871–875.
ergic, and cholinergic signaling. Neuroscience 127, 519–528. Wan, Q., Xiong, Z.G., Man, H.Y., Ackerley, C.A., Braunton, J., Lu, W.Y., Becker, L.E.,
Sellayah, D., Bharaj, P., Sikder, D., 2011. Orexin is required for brown adipose tissue MacDonald, J.F., Wang, Y.T., 1997. Recruitment of functional GABA(A) receptors
development, differentiation, and function. Cell Metabolism 14, 478–490. to postsynaptic domains by insulin. Nature 388, 686–690.
Shanley, L.J., Irving, A.J., Harvey, J., 2001. Leptin enhances NMDA receptor function Wang, Z.W., Pan, W.T., Lee, Y., Kakuma, T., Zhou, Y.T., Unger, R.H., 2001. The role of
and modulates hippocampal synaptic plasticity. Journal of Neuroscience 21, art- leptin resistance in the lipid abnormalities of aging. FASEB Journal 15, 108–114.
RC186. Wayner, M.J., Armstrong, D.L., Phelix, C.F., Oomura, Y., 2004. Orexin-A (hypocretin-1)
Shek, E.W., Scarpace, P.J., 2000. Resistance to the anorexic and thermogenic effects of and leptin enhance LTP in the dentate gyrus of rats in vivo. Peptides 25, 991–996.
centrally administrated leptin in obese aged rats. Regulatory Peptides 92, 65–71. Winocur, G., Greenwood, C.E., Piroli, G.G., Grillo, C.A., Reznikov, L.R., Reagan, L.P.,
Shemesh, E., Rudich, A., Harman-Boehm, I., Cukierman-Yaffe, T., 2012. Effect of McEwen, B.S., 2005. Memory impairment in obese zucker rats: an investiga-
intranasal insulin on cognitive function: a systematic review. Journal of Clinical tion of cognitive function in an animal model of insulin resistance and obesity.
Endocrinology and Metabolism 97, 366–376. Behavioral Neuroscience 119, 1389–1395.
776 J.R. Fadel et al. / Ageing Research Reviews 12 (2013) 764–776

Wrighten, S.A., Piroli, G.G., Grillo, C.A., Reagan, L.P., 2008. A look inside the diabetic Yang, Y., Ma, D., Wang, Y., Jiang, T., Hu, S., Zhang, M., Yu, X., Gong, C.X., 2013. Intranasal
brain: contributors to diabetes-induced brain aging. Biochimica et Biophysica insulin ameliorates tau hyperphosphorylation in a rat model of type 2 diabetes.
Acta 1792, 44–453. Journal of Alzheimer’s Disease 33, 329–338.
Xu, W.L., von Strauss, E., Qiu, C.X., Winblad, B., Fratiglioni, L., 2009. Uncontrolled Yau, P.L., Castro, M.G., Tagani, A., Tsui, W.H., Convit, A., 2012. Obesity and metabolic
diabetes increases the risk of Alzheimer’s disease: a population-based cohort syndrome and functional and structural brain impairments in adolescence. Pedi-
study. Diabetologia 52, 1031–1039. atrics 130, e856–e864.
Yaffe, K., Falvey, C., Hamilton, N., Schwartz, A.V., Simonsick, E.M., Satterfield, S., Zeki, A.H., Stone, K.L., Haan, M.N., Yaffe, K., 2012. Leptin, mild cognitive impair-
Cauley, J.A., Rosano, C., Launer, L.J., Strotmeyer, E.S., Harris, T.B., 2012. Dia- ment, and dementia among elderly women. Journals of Gerontology. Series A,
betes, glucose control, and 9-year cognitive decline among older adults without Biological Sciences and Medical Sciences 68, 175–180.
dementia. Archives in Neurology 69, 1170–1175. Zhang, X., Heng, X., Li, T., Li, L., Yang, D., Zhang, X., Du, Y., Doody, R.S., Le, W.,
Yamanaka, A., Beuckmann, C.T., Willie, J.T., Hara, J., Tsujino, N., Mieda, M., Tomi- 2011. Long-term treatment with lithium alleviates memory deficits and reduces
naga, M., Yagami, K., Sugiyama, F., Goto, K., Yanagisawa, M., Sakurai, T., 2003. amyloid-beta production in an aged Alzheimer’s disease transgenic mouse
Hypothalamic orexin neurons regulate arousal according to energy balance in model. Journal of Alzheimer’s Disease 24, 739–749.
mice. Neuron 38, 701–713.

You might also like