Cannabis Indica

Page 1
Intranasal insulin prevents cognitive decline,
cerebral atrophy and white matter changes in
murine type I diabetic encephalopathy
George J. Francis,1 Jose A. Martinez,1 Wei Q. Liu,1 Kevin Xu,1 Amit Ayer,1 Jared Fine,2 Ursula I. Tuor,1
Gordon Glazner,3 Leah R. Hanson,2 William H. Frey II2 and Cory Toth1
1Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta,
Canada, 2Alzheimer’s Research Center at Regions Hospital, HealthPartners Research Foundation, St. Paul, MN, USA
and 3Department of Pharmacology and Therapeutics, Division of Neurodegenerative Disorders, University of Manitoba,
St. Boniface Hospital Research Centre, Winnipeg, Manitoba, Canada
Correspondence to: Dr C. Toth, University of Calgary, Department of Clinical Neurosciences, Room 155, 3330 Hospital Drive,
N.W., Calgary, Alberta, Canada T2N 4N1
E-mail: corytoth@shaw.ca
Insulin deficiency in type I diabetes may lead to cognitive impairment, cerebral atrophy and white matter
abnormalities. We studied the impact of a novel delivery system using intranasal insulin (I-I) in a mouse
model of type I diabetes (streptozotocin-induced) for direct targeting of pathological and cognitive deficits
while avoiding potential adverse systemic effects. Daily I-I, subcutaneous insulin (S-I) or placebo in separate
cohorts of diabetic and non-diabetic CD1 mice were delivered over 8 months of life. Radio-labelled insulin
delivery revealed that I-I delivered more rapid and substantial insulin levels within the cerebrum with less
systemic insulin detection when compared with S-I. I-I delivery slowed development of cognitive decline
within weekly cognitive/behavioural testing, ameliorated monthly magnetic resonance imaging abnormalities,
prevented quantitative morphological abnormalities in cerebrum, improved mouse mortality and reversed
diabetes-mediated declines in mRNA and protein for phosphoinositide 3-kinase (PI3K)/Akt and for protein
levels of the transcription factors cyclic AMP response element binding protein (CREB) and glycogen synthase
kinase 3b (GSK-3b) within different cerebral regions. Although the murine diabetic brain was not subject to
cellular loss, a diabetes-mediated loss of protein and mRNA for the synaptic elements synaptophysin and
choline acetyltransferase was prevented with I-I delivery. As a mechanism of delivery, I-I accesses the brain
readily and slows the development of diabetes-induced brain changes as compared to S-I delivery. This therapy
and delivery mode, available in humans, may be of clinical utility for the prevention of pathological changes in
the diabetic human brain.
Keywords: diabetes; insulin; leukoencephalopathy; white matter abnormalities; brain atrophy; cognitive decline
Abbreviations: APP = amyloid precursor protein; CSF = cerebrospinal fluid; DW = diffusion-weighted;
EMSA = electrophoretic mobility shift assay; IGF-1 = insulin-like growth factor; IR = insulin receptor;
NGF = nerve growth factor; SNMTS = spatial non-matching-to-sample; WMAs = white matter abnormalities
Received May 27, 2008. Revised October 3, 2008. Accepted October 8, 2008. Advance Access publication November 16, 2008
Introduction
Diabetes has been associated with cognitive dysfunction, an
elevated risk of dementia, cerebral atrophy and presence of
heightened white matter abnormalities (WMAs). Diabetes-
associated cognitive dysfunction, first described nearly a
century ago (Miles and Root, 1922), occurs in both type of
diabetes. In type I diabetes, impaired learning, memory,
problem solving skills, and intellectual development have
been described (Ryan, 1988; Ryan et al., 1993; Ryan and
Williams, 1993; McCarthy et al., 2002; Schoenle et al.,
2002). For patients with type I diabetes, the greatest impact
of diabetes upon brain structure and function seems to
occur at the extremes of age, with little observable effect
during the middle adult years (Wessels et al., 2007, 2008;
Biessels et al., 2008; Kloppenborg et al., 2008). Also,
cognitive dysfunction does not appear to relate to
doi:10.1093/brain/awn288
Brain (2008),131, 3311^3334
ß The Author (2008). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: journals.permissions@oxfordjournals.org
at Pennsylvania State University on February 23, 2013
Downloaded from
hypoglycaemic episodes in diabetes (Kramer et al., 1998;
Schoenle et al., 2002; Jacobson et al., 2007). Similar
cognitive deficits occur in type II diabetic adult patients,
with impaired performance on abstract reasoning and
complex psychomotor functioning (Reaven et al., 1990;
Strachan et al., 1997; Ryan and Geckle, 2000). Diabetes-
mediated cognitive changes seem to occur at the two
extremes of life: either in childhood (Northam et al., 2001;
Schoenle et al., 2002; Fox et al., 2003) or during later stages
of life (Stewart and Liolitsa, 1999; Awad et al., 2004; van
Harten et al., 2006) when neurodegenerative processes
may initiate (Biessels et al., 2008). There are less clear
indications that diabetes impacts upon cognitive function-
ing in middle-age adults (Stewart and Liolitsa, 1999; Awad
et al., 2004; Jacobson et al., 2007; Weinger et al., 2008).
In animal models of diabetes, cognitive dysfunction has
been demonstrated by impaired performances in the Morris
water maze by streptozotocin (STZ)-induced diabetic rats
(Biessels et al., 1996, 1998). Both pre- and post-synaptic
deficits have been associated with impaired long-term
potentiation in the diabetic hippocampus (Biessels et al.,
1996; Kamal et al., 2006). Short-term replacement of insulin
in STZ-treated rats from the onset of diabetes prevents
cognitive decline and protects against hippocampal poten-
tiation deficits, but cannot reverse these electrophysiological
changes (Biessels et al., 1998). Long-term protection against
development of diabetic encephalopathy via locally deliv-
ered insulin has not yet been attempted.
Structural defects within the diabetes-exposed brain
include cerebral atrophy identified with neuroimaging
techniques (Schmidt et al., 2004; Manschot et al., 2006;
Musen et al., 2006; Ikram et al., 2008; Last et al., 2007).
Cerebral atrophy, possibly acting in concert with WMA, is
associated with cognitive decline (Whitman et al., 2001;
Manschot et al., 2006). Diabetes is a risk factor for WMA
presence in some studies (Pantoni and Garcia, 1997;
Murray et al., 2005; Akisaki et al., 2006; Manschot et al.,
2006; Musen et al., 2006; van Harten et al., 2007), but not
in others (Schmidt et al., 2004). By themselves, WMAs in
humans are a risk factor for stroke (Knopman et al., 2001),
cognitive deficits (Pantoni et al., 2007) and abnormalities in
gait associated with falling (Schwartz et al., 2008). The
presence of cerebral atrophy or WMA has been linked to
cognitive dysfunction in a number of human studies
(Manschot et al., 2006; Verdelho et al., 2007). Rodent
models of diabetes have also demonstrated cerebral atrophy
(Lupien et al., 2006; Toth et al., 2006) and WMAs (Toth
et al., 2006) due to long-term diabetes. Such pathological
changes have been associated with cognitive decline over
time in mouse models of diabetes (Toth et al., 2006) and in
diabetic rats where hippocampal electrophysiological
changes were present (Biessels et al., 1996; Kamal et al.,
2000). One pathogenic factor related to presence of brain
atrophy and WMA in experimental diabetes is the presence
of the receptor for advanced glycation end products
(RAGE) (Toth et al., 2006). However, another important
factor may be relative impaired insulin levels and activity in
the brain exposed to diabetes (Li et al., 2005; Haan, 2006),
as described in the human Alzheimer’s disease brain (Craft
et al., 1998; Hoyer, 2004). Insulin receptors (IRs) are
present at central neurons, synapses, and upon glia (Adamo
et al., 1989; Unger et al., 1989; Wickelgren, 1998; Abbott
et al., 1999; Zhao et al., 2004a); therefore, it is assumed that
insulin and its signalling play an important role in neur-
onal, glial, and overall cognitive and memory functioning.
Insulin replacement within the central nervous system may
prevent or even reverse such diabetes-associated changes,
although its systemic delivery is complicated by hypogly-
caemia. Also, potentially impaired function of the blood–
brain barrier may prevent insulin transport from achieving
sufficient cerebral levels (Cohen, 1993; Mooradian, 1997;
Hattori et al., 2000; Kaiyala et al., 2000). An alternative
method of delivering insulin to the brain could be
instrumental
in
preventing
diabetes-accelerated
neurodegeneration.
Intranasal administration permits insulin or similar
peptides to bypass the periphery and the blood–brain
barrier (Dhanda et al., 2005), reaching the brain and
entering the cerebrospinal fluid (CSF) within minutes.
Proteins with size of up to 20kDa, including insulin,
insulin-like growth factor (IGF)-1 and nerve growth factor
(NGF), have been successfully delivered to the brain using
this method (Chen et al., 1998; Liu et al., 2004; Ross et al.,
2004; Thorne et al., 2004; Reger et al., 2006, 2008; Vig et al.,
2006). Transport of molecules delivered intranasally occurs
through extracellular bulk flow transport along olfactory
and trigeminal perivascular channels, as well as possibly
through axonal transport pathways (Benedict et al., 2004;
Thorne et al., 2004; Reger et al., 2006). This technique
permits targeted delivery to the brain to assess the direct
effect of insulin upon its receptors without significant
changes in plasma insulin or glycaemic levels (Patti et al.,
1995; White and Yenush, 1998; Leinninger et al., 2004;
Reger et al., 2006, 2008; Yu et al., 2006).
We hypothesized that long-term replacement of insulin
within the experimental type I diabetic mouse brain could
slow or prevent such changes. Insulin’s ligation to highly
expressed IR in brain regions including the hippocampus
and upon synapses (Abbott et al., 1999), may promote
learning and memory (Zhao et al., 2004a). We used daily
intranasal delivery of insulin over a life span while per-
forming behavioural and neuroimaging measures to
monitor progression. These studies were also performed
to assist in delineation of insulin’s trophic and anti-
hyperglycaemic effects upon development of diabetic
encephalopathy. Given the absence of neuronal loss in
prior studies of the diabetic murine brain (Toth et al.,
2006), we also examined for evidence of synaptic loss, as
well as the previously detected myelin loss. We postulated
that intranasal insulin delivery would sustain the diabetic
nervous system, potentially limiting cognitive decline, brain
3312
Brain (2008), 131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
atrophy and WMAs, while limiting serious systemic side-
effects that could occur with systemic insulin delivery.
Materials and Methods
Animals
We studied a total of 484 male Swiss Webster wild-type (wt) mice
with initial weight of 20–30g, in strict pathogen-free plastic
sawdust-covered cages with a normal light–dark cycle and free
access to mouse chow and water. All protocols were reviewed and
approved by the University of Calgary Animal Care Committee
using the Canadian Council of Animal Care guidelines. Mice were
anaesthetized with pentobarbital (60mg/kg) prior to all proce-
dures. At the age of 1 month, 304 mice were injected with
streptozotocin (Sigma, St. Louis, MO) intraperitoneally with once
daily doses with each of 60mg/kg, 50mg/kg and then 40mg/kg
over three consecutive days, while the remaining 180 mice were
injected with volume-equated placebo carrier (sodium citrate) for
3 consecutive days. One hundred and forty-four mice injected
with STZ and 80 mice injected with carrier were held aside for
morphological studies to be performed at 1, 3 and 5 months after
injections; the remaining mice were followed for the entire length
of the study (8 months of diabetes or equivalent for carrier-
injected mice) as mortality permitted. Monthly weights were
obtained and monthly whole-blood glucose measurements were
performed using the tail vein and a blood glucometer, (OneTouch
Ultra Meter, LifeScan Canada, Burnaby, BC, Canada) with hyper-
glycaemia verified 1 week after STZ injection; a fasting whole-
blood glucose level of 516mmol/l (normal 5–8mmol/l) was our
criterion for experimental diabetes. In all cases, those mice that
did not develop diabetes as defined above following STZ injections
were excluded from further assessment. Animals were inspected
twice daily, and examined for signs of depressed level of con-
sciousness, ataxia or general malaise. When such signs were
identified, whole-blood glucose testing was performed, with a
measurement of53.5mmol/l defined to represent hypoglycaemia.
No intervention was performed at any time with regards to
additional insulin, glucose or fluid delivery. In situations where
the mouse was obviously ill, euthanasia was performed. In circu-
mstances where severe hyperglycaemia was found (433 mmol/l) in
an ill mouse, euthanasia was again performed.
We studied cohorts with a maximum of eight mice in each
group initially due to resource limitations. After the initial cohorts
containing eight mice each were studied, a second cohort was used
to obtain additional mouse data for mouse cohorts with higher
levels of mortality. For any animal that experienced mortality after
the 20-week point of the cognitive studies, their data were carried
through using the last obtainable data point.
Intranasal insulin or saline delivery
125I-labelled I-I and subcutaneous insulin (S-I) administration
were performed at the University of Minnesota for determination
of targeting of insulin delivery methods. This procedure was
approved by the institutional animal care and use committee at
Regions Hospital. Prior to experimentation, 21 non-diabetic
animals were acclimated to handling for awake intranasal delivery
over $2 weeks. 125I-labelled I-I was provided to 12 Swiss Webster
mice (male, 6–8 weeks, Charles River) and 125I-labelled sub-
cutaneous insulin S-I was provided to nine mice under
pentobarbital anaesthesia (60mg/kg). Insulin (Humulin R, Eli
Lilly, Toronto, Canada) with an initial concentration of 100U/ml
or 4033.98mg/ml was dissolved in PBS and custom-labelled with
125I (GE Healthcare, Piscataway, NJ, USA). Synthesized radio-
labelled insulin solution contained 344.3 mCi/ug. 125I-labelled I-I
delivery was performed in a fume hood behind a lead-
impregnated shield, with anaesthetized mice placed supine. A
mixture of 125I insulin (15.8 mCr) and unlabeled insulin (3.3 mg)
were administered as I-I or S-I. 125I I-I was delivered over
alternating nares as eight 3-ml drops with an Eppendorf pipetter
every 2min, for a total volume of 24 ml. This schedule of delivery
has been modified from a previously used method in rats to
quantify radiolabelled delivery of molecules within the Frey
laboratory (Thorne et al., 2004). For subcutaneous delivery, 125I
S-I was delivered with a single subcutaneous injection of 24 ml in
a fume hood behind a lead-impregnated shield. Each desired dose
contained a calculated radioactive dose of 30 mCi.
At each of 1, 2 and 6h after initiating 125I I-I or S-I delivery,
cardiocentesis was performed for blood extraction, followed by
performance of euthanasia via transcardial perfusion using 120ml
of 4% paraformaldehyde while the mouse was maintained under
anaesthesia. To quantify 125I distribution, blood, urine, lymphatic
and visceral organ structures, as well as portions of the central and
peripheral nervous systems were harvested. Gamma signal was
recorded for each body region with autoradiographic imaging
using a phosphor screen. Concentrations of 125I insulin were
calculated based upon the gamma counting data, tissue weight,
specific activity of the insulin administered and standards
measured. Results were studied for penetration into peripheral
nervous system tissues (reported elsewhere) and central nervous
system tissues.
Daily I-I (Humulin R, Eli Lilly, Toronto, ON) or intranasal
saline (I-S) was administered to both diabetic and non-diabetic
male Swiss Webster mice over 8 months of diabetes. A total of
24 ml containing either a total of 0.87U of insulin or 0.9% saline
only was provided as four 6-ml drops by an Eppendorf pipetter
over alternating nares every minute while each mouse was held in
supine position with neck in extension. Daily S-I (0.87U/d,
Humulin R, Eli Lilly, Toronto, ON) and subcutaneous saline (S-S)
were also administered to either diabetic or non-diabetic male
Swiss Webster mice. All therapies began immediately after
confirmation of presence of diabetes for each cohort. In the first
week, daily glucometer testing was performed for all mice,
followed by once-a-month testing. In this work, mice with
diabetes were indicated with a ‘D’, while mice without diabetes
(control mice) are indicated with a ‘C’. Delivery of subcutaneous
saline is indicated as ‘S-S’, subcutaneous insulin as ‘S-I’, intranasal
saline as ‘I-S’ and intranasal insulin as ‘I-I’.
We attempted to use other control groups, but their utility was
limited in each case. First, we attempted to use subcutaneous
insulin via a sliding scale approach in six diabetic mice in order to
maintain normal or mildly high glycaemia levels. This approach
required daily checks of whole-blood glucose using a tail vein;
over the span of 1 month, all of the six diabetic mice developed
infection over the tail, leading to amputation in three mice (50%)
and was probable cause of death in two mice (33%). During the
course of an 8-month-long study, the morbidity associated with
this procedure would be unacceptable and confounding. We also
attempted to maintain a protected venous catheter in the tail vein
for obtaining whole-blood glucose, but this was associated with
auto-removal of the catheter and failure at re-insertions due to
fibrosis of tail tissues. We also studied six diabetic mice receiving
Intranasal insulin and diabetic encephalopathy
Brain (2008), 131, 3311^3334
3313
at Pennsylvania State University on February 23, 2013
Downloaded from
a half dose of the desired I-I dose (0.43U/d) over 1 month. One
mouse (17%) died of confirmed hypoglycaemia, with the other
five mice surprisingly had no statistical difference in glycaemic
levels when compared to a complementary group of six diabetic
mice receiving subcutaneous placebo injections. Therefore, we
selected the S-I dose to be equivalent to the I-I dose for the
cohorts studied.
Behavioural testing
A minimum of eight mice in each cohort [32 diabetic (D I-I, D I-
S, D S-I and D S-S) and 32 control mice (C I-I, C I-S, C S-I and
C S-S)] had cognitive behavioural testing performed once weekly
for evaluation of procedural, visuospatial and recognition
memory. All behavioural tests were conducted under regular
light between 09:00 and 15:00h after fasting overnight from 19:00
onwards. Every mouse was trained in each test paradigm for
3 weeks before initiation of test recording and prior to diabetes
initiation, beginning after 2 weeks of diabetes at 1.5 months of
age. Behavioural equipment was maintained in the identical
position and climate on each occasion and was recorded by the
same observers in order to ensure stability of distant navigational
cues provided by objects around the testing. Testing always
occurred in the order of Holeboard test, Radial arm test, Object
Recognition test (each performed while fasting) followed by
feeding and then the Morris Water-Maze test 1h later. While the
Holeboard and Radial Arm tests evaluate spatial information
processing and memory, the Morris Water Maze also evaluates
procedural memory and aversive motivation. In contrast, the
Object Recognition test evaluates novelty seeking and exploratory
behaviour. Each mouse performed one trial of each test per week.
Due to the possibility of motor limitations confounding the results
of cognitive testing, concurrent testing of linear swim speed and
linear running speed were performed monthly; once measurable
differences in motor function occurred between interventional
groups in swim or run speed, cognitive testing was discontinued.
The Radial Arm test maze consists of a central platform (35cm
in diameter) and eight arms (each 76cm long and 12cm wide)
constructed of black plastic projecting radially from the platform
with adjacent arms separated by 45 (Schwabe et al., 2006). A food
reward (Cheerio) is placed in the same arm each occasion at 180
from the starting point of the mouse placed in the middle of the
central platform. A mouse was recorded as entering an arm when
it passed the midpoint of the arm in a centrifugal fashion. Each
trial ended when the reward was collected or when 720s expired.
Variables recorded during each test were latency for collecting
reward and the number of errors made, defined as a re-entry into
an arm previously entered, classified as a reference memory error.
The Holeboard test was modified from a previous reported
design (File and Wardill, 1975), and was composed of a
rectangular open field (60 Â 90cm) made of opaque white acrylic
surrounded by opaque walls 60cm high. Eight holes (2.5cm
diameter) were placed in two lines of four, equidistant from each
other and from the walls. Each mouse was started in the same
corner while the food reward (Cheerio) was placed in the same
hole (second hole in the far row, kitty-corner from the starting
point). The latency to collect the reward and the number of times
the mouse placed its head in each individual hole was recorded,
with errors defined as repeat visit of a previously visited hole,
classified as a reference memory error.
The Object Recognition task was performed in an open wooden
box (60 Â 60 Â 60cm) with unique objects to be discriminated
constructed from children’s blocks. On the day of the test, at the
first 2-min sample trial (T1), two identical objects (termed as
sample objects) were presented in two corners of the box. Then, in
the second 2-min choice trial (T2) performed 30min later, one of
the objects presented in T1 was replaced by a new object. Objects
were cleaned between trials and between mouse to prevent the
possibility of scent traces forming an olfactory cue. The time (in
seconds) as well as the number of visits taken by mice in exploring
objects in the two trials were recorded, with exploration
considered as directing the nose to the object at a distance
42m and/or touching it with the nose. This paradigm has been
termed delayed ‘spatial non-matching-to-sample’ (SNMTS) testing
(Rothblat and Kromer, 1991).
A mouse-adapted Morris water-maze task (Morris, 1984;
Crawley, 2000; Whishaw and Kolb, 2005) was performed after
feeding and used a solid-coloured circular pool 88cm in diameter
and 20cm in height filled with water at 25 C. The position of the
10cm radius hidden platform remained fixed for all testing over
the entire study period, and each mouse was placed at an identical
starting position opposite the hidden platform. Animals were left
to swim until either they located the platform, climbing upon it
and staying for at least 2s, or when 300s elapsed. Post-testing,
mice were placed under a heating lamp to warm. Variables
recorded during each test were latency to reach the platform
(escape latency) and the fraction of time spent within the
hemisphere of the pool containing the platform (thigmotaxis).
Magnetic resonance imaging
At each month of diabetes, four mice from each cohort [16
diabetic (D I-I, D I-S, D S-I and D S-S) and 16 control mice
(C I-I, C I-S, C S-I and C S-S)] underwent magnetic resonance
(MR) scanning at the Experimental Imaging Centre at the
University of Calgary. MR images were obtained in animals
anaesthetized by mask with isoflurane using a quadrature volume
coil and a Bruker 9.4Tesla MR imaging system. Respiration and
temperature were monitored and inner core temperature was
maintained to be within 36–37 C with a heated air feedback
system. The head was restrained using ear pins. Three different
sets of MR scans were performed using sequences that acquired
T1-weighted images, diffusion-weighted (DW) images for calculat-
ing an apparent diffusion coefficient of water (ADC) map and T2-
weighted images for determining T2 maps within a total of 24
slices through the cerebrum. T1-weighted images were acquired
using a spin echo sequence with a repetition time (TR) of 500ms
and an echo time (TE) of 8ms. Diffusion-weighted (DW) images
were acquired using a spin echo sequence with TR/TE=1200/
49ms and b values of 46 and 767s/mm2, respectively. T2 maps
were obtained from multi-spin-echo images at TR=1200ms, 12
echoes and TE=12.5ms. The field of view was 2 Â 2cm, with an
acquisition matrix of 256 Â256 and a slice thickness of 0.75mm.
MR images of brain were analysed using locally available software
(Marevisi, IBD) by an observer blinded to the treatment group.
Calculation of T2 values, perfusion values, and ADC values within
brain regions of interest were performed bilaterally for each of the
control and diabetic animals. Apparent visualized abnormalities in
T1- and T2-weighted images were also recorded for each animal.
In addition, MR images were used to calculate brain widths at
pre-determined anatomical landmarks as well as to measure
3314
Brain (2008), 131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
overall brain volume. Volumetric brain measurements were
calculated as a summation of cross-sectional areas for each slice
multiplied by the thickness of the MR slices.
Tissue harvesting
Prior to sacrifice after 1, 3, 5, 7 or 9 months of diabetes, animal
weights and blood glucoses were determined. A total of 0.5ml of
whole intracardiac blood was obtained for glycated haemoglobin
measurements to be performed with affinity chromatography
(Procedure 422; Sigma Diagnostics). Euthanasia for animals was
performed with an overdose of pentobarbital intraperitoneally,
followed by harvesting of brain tissues, placement of half of all
brain tissues in 2% formaldehyde and embedding in paraffin. For
these specimens, 10 mm sections were cut for morphological and
immunohistochemical studies. The other half of mouse brain
tissues were placed in Trizol reagent (Life Technologies Inc.,
Rockville MD, USA) or liquid nitrogen for quantitative real-time
reverse transcriptase polymerase chain reaction (qRT-PCR) and
Western blot studies respectively with storage at –80 C for a
maximum of 1 month.
Brain sectioning and staining
Degrees of myelination were determined by staining paraffin brain
sections for either Luxol Fast Blue (LFB) or myelin basic protein
(MBP) (1:100, Stemcell Technologies Inc., Vancouver). For LFB
staining, slides were de-waxed and stained in LFB overnight at
37 C followed by alcohol washing and subsequent differentiation
of the stain with 0.05% lithium carbonate and then alcohol. Slides
used for MBP detection were incubated in methanol for 20min
and washed in phosphate-buffered solution (PBS), then incubated
in Triton-X for 30min before blocking with 10% normal bovine
serum for 1h. Following PBS washes, slides were incubated with
mouse anti-MBP overnight followed by incubation with the
secondary antibody (bovine anti-mouse IgG Cy3, 1:100 Zymed
Inc., San Francisco) for 1h.
Brain sections stained for LFB were chosen to reflect pre-
defined regions of interest (Appendix 1), representing those
regions known to be abnormal within diabetic human brains, as
well as cortical and subcortical regions important in memory and
cognition (Munoz et al., 1993; de Groot et al., 2000; Tullberg
et al., 2004; Toth et al., 2006). Assessment of MBP labelling was
performed using Image Pro Plus software (Image Pro Plus 5.0,
MediaCybernetics, Silver Spring, MD) in order to measure the
optical density or degree of immunofluorescence within each brain
region of interest. LFB staining was assessed using optical density
measurements with Photoshop software for quantification of blue.
All assessments of brain sections were performed by an
investigator blinded to the experimental conditions.
Additional immunohistochemistry was performed to identify
neurons with microtubule-associated protein (MAP)-2, and to
identify oligodendrocytes with PDGFRa immunohistochemistry.
Background staining was blocked using 1% bovine serum albumin
for 1h, and then slides were stained with monoclonal mouse
anti-MAP-2 (1:100, Abcam, Cambridge, MA) or PDGFRa
(1:100, Abcam, Cambridge, MA) for neuron and oligodendro-
cyte detection, respectively. The secondary antibody used was
anti-mouse IgG Cy3 (1:100, Zymed Inc., San Francisco) in both
cases.
Detailed counts of both neuronal and oligodendroglial cell
numbers within grey matter areas of interest (Appendix 1) were
performed using standard unbiased stereological methods for
those slides stained with the neural marker MAP-2 (Gundersen
et al., 1988; Toth et al., 2006) and the oligodendroglial marker
PDGFRa. Volume and total cell number were performed with the
examiner blinded to the condition and treatment of each mouse
brain. Twelve sampled areas were counted for each brain region in
each brain, with neurons distinguished based upon nuclear size
and appearance.
Quantitative real-time PCR
Total RNA was extracted from brain regions stored in Trizol
reagent (Life Technologies Inc., Rockville, MD). Total RNA (1 mg)
was processed directly to cDNA synthesis using the Taq-
Man\Reverse Transcription Reagents kit (Applied Biosystems).
All PCR primers and TaqMan probes were designed using soft-
ware PrimerExpress (Applied Biosystem) and published sequence
data from the NCBI database. PI3K primer sequences were: for-
ward, 50-AACCCGGCACTGTGCATAAA-30; reverse 50-GCCCATT
GGATTAGCATTGATG-30. Akt primer sequences were: forward,
50-TCTGCCCTGGACTACTTGCACT-30, reverse, 50-GCCCGAAGT
CCGTTATCTTGA-30. NFkBp65 primer sequences were: forward,
50-TGTGCGACAAGGTGCAGAAA-30; and reverse, 50-ACAATG
GCCACTTGCCGAT-30. b-actin primer sequences were: forward,
50-TGTTGTCCCTGTATGCCTCTGGTC-30; reverse, 50-ATGTCA
CGCACGATTTCCCTCTCTC-30. SYP primer sequences were: for-
ward, 50-AAAGGCCTGTCCGATGTGAAG-30; reverse, 50-TCCC
TCAGTTCCTTGCATGTG -30. ChAT primer sequences were:
forward, 50-CTATGAGAGTGCATCCATCCGC-30; reverse, 50-GG
TCAGTCATGGCTTGCACAA-30.
RT-PCR was performed using SYBR Green dye. All reactions
were performed in triplicate in an ABI PRISM 7000 Sequence
Detection System. Data were calculated by the 2–AACT method
and are presented as the fold induction of mRNA for RAGE in
diabetic tissues normalized to 18S for comparison to non-diabetic
tissues (defined as 1.0-fold).
Western blot
Thalami and sensorimotor cortices from brains for each cohort
were homogenized using a RotorStator Homogenizer in ice-cold
lysis buffer (10% glycerol, 2% SDS, 25mM Tris–HCl, pH 7.4,
Roche Mini-Complete Protease Inhibitors). Samples were then
centrifuged at 10000 g for 15min. Supernatant was stored at
–20 C prior to SDS–PAGE and immunoblotting analysis. Equal
amounts (150 mg) of protein were loaded and samples were
separated by SDS–PAGE using 10% polyacrylamide gels with
800V-h of current applied. Separated proteins were transferred
onto nitrocellulose paper (BioRad) over 16 h at 200 mA in Towbin
transfer buffer (25mM Tris, 192mM glycine, 20% v/v methanol,
0.1% v/v SDS). The blot was blocked for 1h in 7.5% (w/v) milk
(Nestle, Carnation) in TBS [50mM Tris, 137mM NaCl, 51mM
KCL, 0.05% (v/v) Tween-20]. The PI3K and Akt pathway were
investigated with PI3K (1:1000), PKB/Akt (1:1000), pAkt (1:1000).
The nuclear signalling transcription factor NFkB p65 and p50
subunits (1:1000 each) were also examined, and additional
immunohistochemistry was performed for CREB (1:200, Abcam
Inc., Cambridge, MA) and glycogen synthase kinase 3b (GSK3b)
(1:200, Abcam Inc., Cambridge, MA). Quantification of synaptic
presence was performed using anti-synaptophysin (SYP) (1:1000;
Santa Cruz, Santa Cruz, CA, USA, polyclonal) and Anti-
Choline Acetyltransferase (ChAT) (1:500, Abcam, Cambridge,
Intranasal insulin and diabetic encephalopathy
Brain (2008),131, 3311^3334
3315
at Pennsylvania State University on February 23, 2013
Downloaded from
UK, polyclonal). Identification of proteins for IRb (C-19) (1:500,
Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) and insulin
(1:500, Abcam, Cambridge, UK, polyclonal) were also performed
using Western blotting. For a housekeeping protein, anti-b-actin
(1:100, Biogenesis Ltd. Poole, UK) was applied to separate blots.
Secondary anti-rabbit, anti-mouse or anti-human IgG HRP Linked
antibody (Cell Signaling) was applied at 1:5000 in each case as
appropriate. Signal detection was performed by exposing of the
blot to enhanced chemi-luminescent reagents ECL (Amersham)
for 2min. The blots were subsequently exposed and captured on
Kodak X-OMAT K film. In each case, three blots were performed,
and analysed with Adobe Photoshop (Adobe Photoshop 9.0,
Adobe, San Jose, CA, 2005) for quantification of relative protein
content.
Analysis of Western blots used a ratio of protein of interest to
b-actin protein for each region of brain tissue and each cohort.
Quantification of the luminosity of each identified protein band
was performed using Adobe Photoshop software (Adobe
Photoshop 7.0, Adobe, San Jose, CA, 2002).
Additional immunohistochemistry
Immunohistochemistry was performed using PI3K (1:200,
Santa Cruz Inc., Santa Cruz, CA), PKB/Akt [1:200, anti-protein
kinase B (Akt), Stressgen, Victoria, Canada], pAkt [1:200, anti-
phospho-Akt (Ser473), Cell Signaling Technologies, Danvers, MA]
and the nuclear signalling transcription factor NFkB p65
subunit (1:200, anti-NFkB p65, Santa Cruz, Santa Cruz, CA)
and p50 subunit (1:200, anti-NFkB p50, Santa Cruz, Santa Cruz,
CA). For synaptic identification, anti-synaptophysin (1:200;
Santa Cruz, Santa Cruz, CA, USA, polyclonal) and Anti-
Choline Acetyltransferase (ChAT) (1:100, Abcam, Cambridge,
UK, polyclonal) were used. Identification of IR and insulin
was performed with immunohistochemistry using antibodies
to IRb (C-19) (1:100, Santa Cruz Biotechnology Inc., Santa
Cruz, CA, USA).
Tissue specimens were examined under fluorescence microscopy
(Zeiss Axioskope, Axiovision and Axiocam, Zeiss Canada,
Toronto, Canada) at 400Â and images obtained were examined
based upon brain regions of interest sectioned at 10 mm (Appendix
1). Calculation of the number of immunofluorescent profiles as
well as the relative luminosity was performed using Adobe
Photoshop (Adobe Photoshop 9.0, Adobe, San Jose, CA, 2005).
In grey matter regions, the total numbers of neurons per trans-
verse section, as well as the numbers of neurons with positive
immunolabelling for the above markers, and their potential
nuclear activation, were recorded. Luminosity was classified as
none-low (luminosity value of 0–150), moderate (150–250) or
high (4250) using Adobe Photoshop software (scale of 0–255 with
arbitrary units). An additional measurement of neuronal nuclear
immunolabelling for pAkt and NFkB was also performed using a
pre-determined luminosity measurement threshold of 150 (no
units), below which negative nuclear reactivity was assigned for
both neurons and glia. All measurements were performed by a
single examiner blinded to the group identity. For synaptic
presence, cortical sections immunostained with synaptophysin and
ChAT were examined with densitometry using Image-Pro Plus
image analysis (Media Cybemetics). A total of 25 randomly chosen
areas of cortex, thalamus, and hippocampus from 10 animals per
cohort group were examined at 400Â.
Electrophoretic mobility shift assays
For evaluation of CREB binding to DNA, brain tissue was obtained
and placed in Totex buffer (20 mM HEPES pH 7.9, 350 mM NaCl,
20% glycerol, 1% igepal, 1mM MgCl2, 0.5mM EDTA, 0.1mM
EGTA, 0.1 mM PMSF, 5 mg/ml aprotinin, 50 mM DTT), followed by
cell lysis on ice for 30 min, centrifugation at 14 000 r.p.m. for 15 min
at 4 C, with supernatant retained. Protein levels were determined by
the Bradford method (Biorad) and samples stored at À80 C. Equal
amounts of protein were incubated in a 20-ml reaction mixture
containing 20 mg of bovine serum albumin; 1 mg of poly (dI–dC);
2 ml of buffer containing 20% glycerol, 100 mM KCl, 0.5 mM EDTA,
0.25% NP-40, 2mM dithiothreitol, 0.1% phenylmethylsulphonyl
fluoride and 20mM HEPES, at pH 7.9; 4 ml of buffer containing
20% Ficoll 400, 300mM KCl, 10mM dithiothreitol, 0.1%
phenylmethylsulphonyl fluoride and 100mM HEPES, pH 7.9; and
20000–50000c.p.m. of 32P-labelled oligonucleotide (S) corre-
sponding to a CREB-binding site (50-CAA TGA CAT GCG GCT
ACG TCA CGG CGC AGT GCC C-30). After 20min at room
temperature, reaction products were separated on a 12% non-
denaturing polyacrylamide gel. Radioactivity of dried gels was
detected by exposure to Kodak X-Omat film, and images on the
developed film were scanned into a computer using a UMAX 1200s
scanner. Densitometry was performed using Scion Image software
(Scion Corp., Frederick, MD).
For evaluation of NFkB binding to DNA, nuclear proteins were
extracted with NE-PERTM Nuclear and Cytoplasmic Extraction
Reagents (Pierce, Rockford, IL). Protein–DNA complexes were
detected using biotin end-labelled double-stranded DNA probes
prepared with the Biotin 30 End DNA Labeling Kit (Pierce). The
binding probe used was 50-TCGACAGA[GGGACTTTCC]GAGA
GGC-30, with the binding site indicated in square brackets, and
bold letters indicating regions of variable nucleotides. Electro-
phoretic mobility shift assay (EMSA) was performed with a
LightShift Chemiluminescent EMSA Kit (Pierce). Briefly, nuclear
extracts (10 mg protein) and the 10 Â binding buffer with 2.5%
glycerol, 5 mM MgCl2, 50ng/ml poly(dI-dC), 0.05% NP-40, 1mM
DTT and 20fmol biotin 30-end labelled double-stranded oligonu-
cleotide were incubated at room temperature for 1h in a volume
of 20 ml. For NFkB supershift analysis, an anti–NFkBp65 poly-
clonal antibody (Santa Cruz; 1 mg per reaction) was incubated
with the nuclear proteins on ice for 1h before labelled oligo-
nucleotide was added. Reaction products were separated by
electrophoresis [5% acrylamide (29:1 acryl/bis)] in 0.5 Â TBE.
After electrophoresis, the protein–DNA complexes were trans-
ferred onto nylon membranes and detected using chemilumines-
cence (LightShift kit; Pierce).
Analysis
All statistical comparisons were intended between the following
groups: D I-I and D S-I; D I-I and D I-S; D I-I and C I-I; D S-I
and D S-S; D S-I and C S-I; C I-I and C S-I; C I-I and C I-S; and
C S-I and C S-S. Comparison testing was not performed between
other grouped cohorts, with Bonferroni corrections applied as
appropriate for the above group comparisons.
Data collected in the groups were expressed as mean Æ standard
error. One-way matched/unmatched ANOVA and Student’s t-tests
were performed to compare means between diabetic and control
groups. The Repeated Measures ANOVA assessment was per-
formed for data obtained during the four cognitive studies, as
individual scoring during 1 week partially depended upon
3316
Brain (2008), 131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
performance of the prior week, with the D I-I group compared to
the D S-I and D I-S groups, and the D S-I group compared to the
D I-S and D S-S groups. Also, Area Under The Curve statistical
testing was performed for cognitive testing other than object
recognition tasks. Again, only the groups intended to have
statistical comparisons were analysed as such. For the purposes
of molecular studies and comparisons, one control (non-diabetic)
group was used as a control value, with subsequent comparisons
to other diabetic groups for the molecular test studied.
Results
Intranasal insulin delivery quantification
Quantification of radiolabelled insulin delivery identified
peak delivery to the brain within 1h after I-I delivery and
around 6h with S-I. Systemic insulin concentrations with
intranasal delivery were limited as compared with sub-
cutaneous delivery (Fig. 1). Mice receiving I-I treatment did
not suffer adverse effects throughout the 1-, 2- and 6-h
monitoring periods before sacrifice. S-I delivery led to much
higher concentrations of whole blood insulin and develop-
ment of hypoglycaemia-induced illness in approximately
one-third of mice. Hepatic and kidney insulin levels were
overall two to three times higher in mice administered S-I.
Diabetes
After STZ injection, mice developed diabetes within 2 weeks
in 262/304 (86%) of animals. Diabetic mice were smaller
than non-diabetic mice throughout life beginning 1 month
after STZ injection (Table 1), with D I-I mice maintaining
weight better than D I-S mice. Hyperglycaemia was
identical in D I-I or D I-S mice, but D S-I mice had less
hyperglycaemia and developed hypoglycaemia associated
with mortality in some instances. Non-diabetic (C) S-I mice
also had increased mortality levels relative to C S-S or C I-I
mice (Table 1). Mouse glycated haemoglobin levels were
increased in all diabetic mice at 9 months of life, and were
identical between I-I and I-S mice, but reduced in S-I mice.
The mortality rate in diabetic mice was significantly higher
than in non-diabetic mice, although mice receiving I-I
had improved mortality relative to I-S, S-S and S-I mice
(Table 1) (Kaplan–Meier survival statistics).
The second cohort groups used to complete data within
each intervention group consisted of four mice in each of
the C S-I, D S-S, D S-I, D I-S and D I-I groups. This led to
a data with a minimum of eight mice in each intervention
group (n =9 D I-I, n =10 D I-S, n =9 D S-I, n =10 D S-S,
n =8C I-I, n = 11 C S-I, n =8C S-S, n = 8 C I-S).
Cognitive behavioural data
Cognitive testing continued until 33 weeks of diabetes. All
cognitive data was based upon a minimum of eight mice in
each cohort group at all time points. Learning processes for
each of the tasks appeared to be similar between diabetic
and non-diabetic mice over the first several weeks (Fig. 2).
Diabetic mice performed better than non-diabetic mice in
the first weeks of the Radial Arm Test, as demonstrated
previously (Toth et al., 2006), hypothesized to be due to
hyperphagia contributing to greater exploratory behaviour.
In general, diabetic mice demonstrated waning perfor-
mances on each of the behavioural tasks after 7–10 weeks of
diabetes, with impaired performances continuing through-
out the remainder of the 33 weeks of testing. Both D I-I
and D S-I mice performed better than diabetic cohorts
(D I-S and D S-S) in the Morris Water Maze, Holeboard
Test and Radial Arm Test tasks (Fig. 2), although D I-I
mice consistently outperformed D S-I mice, particularly in
the Morris Water-Maze task. In the Morris Water Maze,
diabetic mice also spent less time in the hemisphere of the
hidden platform (target zone) than non-diabetic mice,
although D I-I mice continued to spend time in the target
zone similar to that of the non-diabetic mice (Supplemen-
tary Fig. 1). Mistakes made in either of the Holeboard or
Radial Arm Tests were also magnified with diabetes and
increased with duration of diabetes, although D I-I mice
made similar numbers of mistakes as non-diabetic mice
(Supplementary Fig. 1). Mouse performance in the Object
Recognition task demonstrated novelty-seeking behaviour
in control mice and D I-I mice, but not in other diabetic
cohort groups (Fig. 2, Supplementary Fig. 1). Additional
assessments using the Area Under the Curve for each
intervention cohort revealed statistically different perfor-
mances on cognitive testing as described in the figure
legends. Analysis provided by the Repeated Measures
ANOVA testing revealed early and late time points for
differences in intervention groups, as demonstrated in
Fig. 2 and Supplementary Fig. 1.
Taken together, these behavioural experiments demon-
strated better maintenance of visuospatial, procedural and
objection recognition memory functioning in the diabetes-
exposed brain receiving intranasal insulin as compared to
the diabetic mouse brain not receiving intranasal insulin.
MRI and brain weight data
Volumetric measurements of brain demonstrated diffuse
cerebral atrophy after 5 months of diabetes (Fig. 3), with
protection demonstrated in D I-I mice after 8 months of
diabetes. Although not identified within individual brain
regions at earlier time points, diabetes-associated atrophy was
detected in the sensorimotor cortex, caudate/putamen,
corpus callosum, internal capsule, CA3 portion of hippo-
campus and cerebral peduncle (Fig. 3) after 8 months of
diabetes. In each of these brain regions, D I-I mice had
measurable protection from atrophy (Fig. 3). Measurement of
brain mass showed evidence of brain atrophy after 5 months
of diabetes, with protection against loss of brain mass first
detected in D I-I mice at 8 months of diabetes (Fig. 3).
As found previously (Toth et al., 2006), there were no
changes in MR T1 or perfusion-weighted imaging measure-
ments identified. Although initial changes could be seen after
5 months, evidence of diabetes-associated leukoencephalopathy
Intranasal insulin and diabetic encephalopathy
Brain (2008),131, 3311^3334
3317
at Pennsylvania State University on February 23, 2013
Downloaded from
was more easily detected after 8 months of diabetes in all
diabetic mice using quantitative T2 map values. Although
D I-I mice were protected from heightened T2 map values
after 8 months, there were still diabetes-mediated WMA
present in numerous regions of brain described as white
matter regions including corpus callosum and internal
capsule, while similar T2 changes could be identified in
grey matter regions, such as cortex and hippocampus (Fig. 3).
These changes were heterogenous, and were not identified
in other brain regions of interest (Appendix 1).
White matter analysis
Quantitative evaluations of histological sections were
performed to determine myelin presence in brain regions
of interest (Appendix 1). Both LFB preparations and MBP
Fig. 1 Radiolabelled insulin detection. After 1 and 6 h of either I-I (open bars) or S-I (closed bars), I-I led to more rapid and elevated
insulin presence in central nervous system structures, including at cortex and deep brain structures, with much less insulin detected in
blood than with S-I (A). At 6 h after delivery (B), intranasal insulin amounts rose in blood, and subcutaneously delivered insulin had
slowly penetrated nervous system structures, albeit at lesser amounts than with earlier arrival of intranasally delivered insulin.
Significant differences were determined by matched t-tests, with asterisk indicating significant difference (P50.05) between the
intranasal and subcutaneous insulin delivery techniques for each tissue (n = 4 mice in each mouse cohort for each time point).
3318
Brain (2008),131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
Table 1 Murine weights, fasting glycaemia levels, glycated haemoglobin levels and survival numbers at induction of diabetes and at harvesting at months
1, 3, 5 and 8 of diabetes
Timepoint
Prior to injection of
STZ/carrier
Month 1
Month 3
Month 5
Month 8
Murine weight
(gÆ SEM)
(gÆ SEM)
(gÆ SEM)
(gÆ SEM)
(gÆ SEM)
Non-diabetic S-S mice
25.6Æ3.2 (n = 25)
32.7Æ3.8 (n = 25)
39.7Æ4.0 (n = 24)
43.4 Æ 4.3Ã (n =24)
47.2 Æ4.9Ã (n=23)
Non-diabetic S-I mice
25.8Æ 3.9 (n = 25)
30.4Æ 4.1 (n = 20)
34.6Æ 4.7 (n = 17)
36.2 Æ 5.1Ã (n =15)
37.0 Æ 5.4Ã (n=12)
Non-diabetic I-S mice
25.1Æ3.7 (n = 25)
32.1Æ3.9 (n = 25)
40.1Æ4.2 (n = 25)
44.7Æ4.6 (n = 25)
48.1Æ5.2 (n = 24)
Non-diabetic I-I mice
25.4Æ3.3 (n = 25)
31.9Æ3.5 (n = 24)
36.2Æ4.9 (n = 23)
39.1Æ5.3 (n = 22)
43.4Æ 4.8 (n = 21)
Diabetic S-S mice
25.2Æ 3.3 (n = 40)
26.9Æ 4.2 (n = 34) (3 non-diabetic)
28.4Æ 4.3 (n = 30)
30.6Æ 5.7 (n = 20)
31.5Æ5.8 (n = 16)
Diabetic S-I mice
25.4Æ3.4 (n = 40)
26.4Æ 4.8 (n = 31) (2 non-diabetic)
27.2Æ 3.8 (n = 24)
28.2Æ4.1 (n = 16)
28.8Æ 4.9 (n = 12)
Diabetic I-S mice
25.2Æ 3.4 (n = 40)
26.2Æ4.8 (n = 33) (3 non-diabetic)
28.9Æ 4.2 (n = 30)
30.2Æ4.9 (n = 22)
30.4 Æ 5.2& (n=18)
Diabetic I-I mice
25.6Æ3.5 (n = 40)
27.8Æ 4.0 (n = 36) (3 non-diabetic)
30.9Æ 4.5 (n = 35)
34.8Æ 3.6 (n = 33)
35.6 Æ 4.9& (n =30)
Murine glycaemia and
8 month glycated haemoglobin
(mmol/l)
(mmol/l)
(mmol/l)
(mmol/l)
(mmol/l)
(percentage of haemoglobin)
Non-diabetic S-S mice
5.5Æ2.3
5.9Æ 2.6
6.1Æ2.7
6.2Æ 3.0
6.6 Æ3.2Ã (12.4%Æ 4.8%)
Non-diabetic S-I mice
5.4Æ 2.6
3.5Æ2.7
3.9Æ 2.9
4.1Æ3.0
4.0 Æ3.1Ã d (9.2%Æ 4.1%)
Non-diabetic I-S mice
6.0Æ2.8
5.9Æ 2.6
5.9Æ 2.8
6.1Æ3.1
6.3Æ2.7 (12.1% Æ 4.7%)
Non-diabetic I-I mice
5.8Æ 2.6
5.7Æ 2.9
5.6Æ3.0
5.7Æ3.0
5.7Æ3.2 (12.6% Æ 4.9%)
Diabetic S-S mice
5.7Æ 2.7
31.7Æ4.9
32.3Æ 6.1
32.2Æ 6.2
32.4 Æ 6.0 (31.6%Æ 6.2%)&
Diabetic S-I mice
5.6Æ2.6
24.7Æ5.2
25.9Æ5.8
24.3Æ5.6
24.8 Æ 6.1 (24.1%Æ 6.6%)&
Diabetic I-S mice
6.1Æ2.9
32.2Æ4.6
32.1Æ5.3
32.1Æ5.2
32.3Æ5.8 (32.0% Æ 6.0%)
Diabetic I-I mice
5.8Æ 2.8
31.5Æ 4.5
31.6Æ 5.0
31.6Æ 5.2
32.0Æ5.6 (30.2% Æ 6.4%)
Murine survival numbers
Number of mice
Number of mice
Number of mice
Number of mice
Number of mice
Non-diabetic S-S mice
25/25 (100%)
25/25 (100%)
24/25 (960%)
24/25 (96%)
23/25 (92%)Ã
Non-diabetic S-I mice
25/25 (100%)
20/25 (80%)
17/25 (68%)
15/25 (60%)
12/25 (48%)Ã d
Non-diabetic I-S mice
25/25 (100%)
25/25 (100%)
25/25 (100%)
25/25 (100%)
24/25 (96%)
Non-diabetic I-I mice
25/25 (100%)
24/25 (96%)
23/25 (92%)
22/25 (88%)
21/25 (84%)
Diabetic S-S mice
40 (100%)
34/37 (3 non-diabetic, 92%)
30/37 (81%)
20/37 (54%)
16/37 (43%)
Diabetic S-I mice
40 (100%)
31/38 (2 non-diabetic, 82%)
24/38 (63%)
16/38 (42%)
12/38 (32%)
Diabetic I-S mice
40 (100%)
33/36 (3 non-diabetic, 92%)
30/36 (83%)
22/36 (61%)
18/36 (50%)&
Diabetic I-I mice
40 (100%)
36/39 (3 non-diabetic, 92%)
35/39 (90%)
33/39 (85%)
30/39 (77%)& f
Glycated haemoglobin values are presented in italics in the 8 month column for glycemia levels. For murine survival, Kaplan^Meier statistics were performed between cohort groups.
All measures are meanÆ SEM for weights, glycemia levels and glycated haemoglobin levels. ÃIndicates significance at P50.05 with comparison of non-diabetic S-S and S-I mice cohort
groups; &Indicates significance between diabetic cohort groups receiving I-S and I-I; ÈIndicates significance with comparison of diabetic I-I mice to S-S and I-S diabetic cohort groups;
dIndicates significance with comparison of non-diabetic S-I mice to all other non-diabetic mice using multiple ANOVA testing with Bonferroni post hoc t-test comparisons (a = 0.016).
Bold values indicate statistical significance as indicated by the superscripted symbols.
In
tran
asal
in
su
lin
an
d
diabetic
en
cep
h
alo
p
athy
Brain
(20
0
8),131,
3311^333
4
3319
at Pennsylvania State University on February 23, 2013 http://brain.oxfordjournals.org/ Downloaded from
Fig. 2 Cognitivebehaviouraldata.Micewithdiabetesareindicatedwitha‘D’,whilemicewithoutdiabetes(controlmice)areindicatedwitha‘C’.
Delivery of subcutaneous saline is indicated as ‘S-S’, subcutaneous insulin as ‘S-I’, intranasal saline as ‘I-S’ and intranasal insulin as ‘I-I’. No baseline
differences existed between any of the mouse cohorts. Morris Water-Maze testing demonstrated learning ability in each cohort, regardless of
diabetes presence (A).Times to reach the platform continued to improve after 7^9 weeks in non-diabetic mice and D I-I mice, whereas the
performance of other diabetic mice failed to improve with prolongation of their later times to complete testing. D S-I mice performed better
than D S-S or D I-S mice at later time points, while D I-I mice outperformed all other diabetic mice after 9 weeks of time. In the Holeboard test,
both D I-I and D S-I mice outperformed other diabetic mouse cohorts after 7 weeks of testing (B). In the initial stages of the Radial ArmTest (C),
3320
Brain (2008),131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
Fig. 2 Continued
diabetic mice outperformed non-diabetic mice, perhaps due to enhanced search behaviour related to hyperphagia. After 5 weeks, D I-I and
D S-I mice found their targets faster than other diabetic mouse cohorts, whose performance diminished further over time. After 22
weeks, D I-I mouse times were improved relative to D S-I times. The Object RecognitionTask results are presented as the average of each
of 8 consecutive weeks. This task demonstrated less novelty-seeking behaviour in terms of both visits (D) and time spent (E) at novel
objects in theT2 portion of the experiment for D I-S, D S-I and D S-S mouse cohort groups. For A^C, Repeated Measures ANOVA testing
revealed both early and late time points of significance for D S-I and D I-I mouse cohorts, indicated under the graph with red bars (D S-I)
or blue bars (D I-I) for the weeks where significant differences were identified, when compared to D S-S and D I-S mice, or all other
diabetic mouse cohorts respectively. For A^C, additional Area Under The Curve measurements identified significantly improved perfor-
mances for D I-I mice compared to the D S-I and D I-S groups (P50.025 using Bonferroni corrections). For D and E, significant differences
were determined by multiple ANOVA tests, with asterisk indicating significant difference (P50.016 using Bonferroni corrections) between
the D I-I mouse group and other diabetic mouse cohorts (P50.016 using Bonferroni corrections) for the respective time points (n = 8 ^10
mice in each mouse cohort for each time point).
Intranasal insulin and diabetic encephalopathy
Brain (2008),131, 3311^3334
3321
at Pennsylvania State University on February 23, 2013
Downloaded from
Fig. 3 MRI data. The diabetic murine brain demonstrated atrophy and development of white matter changes over time. There were no
differences between cohort groups for volumetric measurements until after 3 months of diabetes, and no difference between cohort
groups for individual brain region volumetric measurements until 5 months of diabetes. Samples of MR T2 images after 8 months of diabetes
are demonstrated using aged-matched non-diabetic mouse brain (top), diabetic mouse brain receiving long-term intranasal insulin (middle)
and diabetic mouse brain receiving intranasal saline (bottom) (A).Volumetric measurements of the entire brain indicated generalized loss of
brain volume over time in diabetes, first demonstrable after 3^5 months of diabetes (B). D I-I mice were protected from cerebral atrophy
when compared to other diabetic mouse cohorts, detectable after 8 months of diabetes. Individual brain regions also showed atrophy after
8 months of diabetes in diabetic mouse cohort groups other than D I-I mice (C). Wet brain mass paralleled MRI volumetric measurements,
again demonstrating cerebral atrophy in diabetic mice after 5 months, with partial protection found in D I-I mice (D). MR T2 map values
were significantly elevated in both white and grey matter regions for diabetic mice, with D I-I mice again protected when compared to
their diabetic cohort groups (E). Significant differences were determined by multiple ANOVA tests, with asterisk (Ã) indicating significant
difference (P50.0125 using Bonferroni corrections) between the indicated mouse group and other non-diabetic mouse cohorts, while o
indicates a significant difference (P50.0125 using Bonferroni corrections) between the D I-I mouse group and the D I-S and D S-S
cohort groups for the respective time points. Finally, g indicates a significant difference (P50.0125 using Bonferroni corrections)
between the D I-I mouse group and all other diabetic mouse cohort groups for the respective time points (n = 4-6 mice in each
mouse cohort for each time point).
3322
Brain (2008),131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
immunohistochemistry detected reductions in myelin quan-
tity within a number of brain regions of interest (Fig. 4).
As determined previously (Toth et al., 2006), none of the
identified WMA had a pattern which could be attributed to
large vessel infarction. Lost MBP expression closely resembled
abnormalities also identified with LFB staining and with T2
measurement changes found with MR imaging (Fig. 4).
As identified previously (Toth et al., 2006), there were
no differences in neuronal density over grey matter regions
of interest after detailed stereological counts. For example,
neuronal densities within the CA1 region of hippocampus
were 1.73Â 106/mm3
in diabetic mice as compared to
1.79Â 106/mm3 in control mice (P= NS). Neuronal densities
within other grey matter brain regions (Appendix 1) were
similar between diabetic and non-diabetic mouse cohort
groups. Oligodendrocyte counts were performed by exam-
ination of immunohistochemistry for PDGFRa within both
regions of white and grey matter (Appendix 1) and revealed a
44% loss of oligodendrocytes within the internal capsule and
corpus callosum regions in D I-S brains as compared to C I-S
brains, with oligodendrocyte loss protected in D I-I brains,
only demonstrating 17% loss (ANOVA, P50.01).
Fig. 3 Continued.
Intranasal insulin and diabetic encephalopathy
Brain (2008),131, 3311^3334
3323
at Pennsylvania State University on February 23, 2013
Downloaded from
mRNA and protein quantification
A relative loss of intraneuronal pAkt and decreased nuclear
pAkt presence (Fig. 5) was associated with diabetic enceph-
alopathy. Insulin provision, however, led to amelioration of
this loss in D S-I mice, and particularly in D I-I mice.
Amongst non-diabetic mouse cohorts, there was no
significant difference in pAkt presence in either cortical or
hippocampal neurons. Quantification of Akt and PI3K
mRNA demonstrated downregulation with diabetes in
general, while a near return to normal for both PI3K and
Akt mRNA levels occurred with I-I delivery in diabetic mice
Fig. 4 Quantification of myelin for both Luxol Fast Blue (LFB) and Myelin Basic Protein (MBP). Data for LFB quantification is demonstrated
as (255 ^ Luminosity), as higher luminosity would indicate greater light passage and therefore, less myelin content. LFB loss occurred in
both white and grey matter regions (A) of the diabetic murine brain as compared to the non-diabetic brain (C S-S) (B), with partial
protection present in the D I-I brain (C) when compared to the D I-S brain (D). (Bars = 20mm) MBP loss also occurred in both white and
grey matter regions (E) of the diabetic murine brain as compared to the non-diabetic murine brain (C S-S) (F), with partial protection
again present in the D I-I brain (G) when compared to the D I-S brain (H). (Bars = 10mm) Significant differences were determined by
multiple ANOVA tests, with asterisk (Ã) indicating significant difference (P50.0125 using Bonferroni corrections) between the indicated
mouse group and other non-diabetic mouse cohorts, while g indicates a significant difference (P50.0125 using Bonferroni corrections)
between the D I-I mouse group and all other diabetic mouse cohort groups for the respective time points (n = 4 mice in each mouse
cohort for each time point).
3324
Brain (2008), 131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
(Fig. 5) in cortex (also seen in hippocampus). Protein
quantification in the hippocampus and cortex of diabetic
mice revealed generalized suppression with diabetes, with
partial protection against loss of PI3K, pAkt, GSK3b,
pGSK3b and pCREB (no significant difference for Akt and
CREB) identified in D I-I mice (Fig. 5). Ratios of
phosphorylated to non-phosphorylated downstream mark-
ers revealed increased activation (phosphorylation) of Akt
Fig. 5 The PI3K/Akt pathway is disturbed in the diabetic murine brain. Levels of pAkt fell within hippocampal neurons exposed to dia-
betes, with partial protection provided in D I-I mice (A). Neuronal activation (presence of pAkt in neuronal nuclei) was also diminished in
diabetic hippocampal neurons (B), with intranasal insulin preventing some of this effect (B). Hippocampal neurons in C S-I mice (C)
demonstrated higher levels of nuclear pAkt presence when compared to D I-I mice (D), which were protected when compared to D S-I
mice (E). (Bars = 25mm) Both PI3K and Akt mRNA fell in diabetic murine brain tissue from both hippocampus (data not shown) and
cortex, (F) although D I-I mice were protected against mRNA loss. Similarly, PI3K and Akt protein levels (representative blots,G) fell
based upon semi-quantitative determination of protein levels in both hippocampus and cortex (H). In addition to changes in PI3K and Akt
protein loss, similar protein loss is seen for PI3K/Akt pathway proteins including pAkt, GSK3b, pGSK3b, CREB and pCREB (G,H). There
was no difference in measurements between non-diabetic cohort mouse groups. Quantitative assessment of electrophoretic mobility shift
assays (EMSA) (example in I) demonstated a loss of CREB DNA binding in diabetic hippocampus, reversed with intranasal insulin provision
(J). Meanwhile, phosphorylation ratios for Akt, CREB, and GSK3b within hippocampus were increased with I-I provision in diabetic mice
(K). Quantitative analysis in each situation was performed using three to five samples for each group with multiple ANOVA tests, appro-
priate Bonferroni corrections, and with asterisk indicating significant difference (P50.016) between groups indicated by horizontal bars.
Intranasal insulin and diabetic encephalopathy
Brain (2008), 131, 3311^3334
3325
at Pennsylvania State University on February 23, 2013
Downloaded from
and CREB, along with increased phosphorylation (inactiva-
tion) of GSK3b in the hippocampus for diabetic mice
receiving I-I (Fig. 5). EMSAs demonstrated maintained
DNA binding for CREB in D I-I mice as compared to D I-S
mice (Fig. 5).
Protein and mRNA for portions of the IR, IRb and IR
subsrate (IRS)-1, were also downregulated in the diabetic
murine brain except when I-I was provided (Fig. 6). Both
IRb and IRS-1 were expressed over the neurolemma of
cortical and hippocampal and cortical neurons, with
decreased levels of expression in diabetes, except in D I-I
mice (Fig. 6). For non-diabetic mice, intranasal insulin
provision also led to elevated IRb levels (Fig. 6).
Important components of the synaptic complex, the
vesicular protein synaptophysin (SYP) and the enzyme
choline acetyltransferase (ChAT), were both downregulated
in diabetic hippocampus and cortex (Fig. 7). Again, D I-I
mice were partially spared from loss of synaptic compo-
nents within the cerebrum based upon immunofluorescence
quantification, protein blotting and qRT-PCR studies
(Fig. 7). There were no differences between non-diabetic
cohort groups despite the provision of I-I or S-I (Fig. 7),
and no loss of synaptic components was demonstrated in
the thalamus in any cohort.
Diabetes led to greater accumulation of NFkB and its
greater nuclear presence (activation) (Fig. 8). Both neurons
Fig. 5 Continued.
3326
Brain (2008),131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
and oligodendrocytes in cortical, hippocampal and sub-
cortical regions demonstrated NFkB activation, with
suppressed levels of activation demonstrated in D I-I
mice. As well, D I-I mice also demonstrated partial sup-
pression of overall NFkB mRNA and protein elevation
occurring in both cortex and hippocampus, which exhibited
age-dependent increases over time for both diabetic and
non-diabetic mice (Fig. 8), but greatest in diabetic mice.
Finally, EMSAs demonstrated depressed levels of DNA
binding for NFkB in D I-I mice as compared to D I-S mice,
but levels remained higher in D I-I mice when compared to
non-diabetic mice (Fig. 8). There were no differences in
NFkB levels between non-diabetic cohorts.
Discussion
Intranasal insulin prevented diabetes-mediated cerebral
neurodegeneration without leading to prominent systemic
effects or modification of glycaemia levels. Replacement of
insulin reversed the downregulated PI3K/Akt pathway to
slow or diminish the development of brain atrophy, WMA
and cognitive decline.
Systemic and neural effects of subcutaneous
and intranasal insulin
Insulin delivered through an intranasal route led to
improvements in behavioural (Fig. 2), morphological (Figs
3 and 4), and molecular abnormalities (Figs 5–8) within the
diabetes-exposed brain. I-I did not confer the risks of
systemic hypoglycaemia identified with S-I delivery in this
long-term experimental model. Not only did S-I delivery in
either of diabetic or non-diabetic mice lead to greater
mortality (Table 1) and higher systemic delivery than I-I
(Fig. 1), but failed to improve a number of morphological
and molecular deficits identified with diabetic encepha-
lopathy. While subcutaneous delivery of insulin led to
improved glycated haemoglobin levels at sacrifice, intranasal
insulin delivery did not, suggesting that the beneficial
effects of I-I in diabetic mice was not due to effects upon
hyperglycaemia, which occurred in the D S-I cohort
(Table 1). Whereas systemic insulin enters the brain via a
receptor-mediated, saturable form of transport (Woods
et al., 2003), I-I directly enters the brain bypassing the
blood–brain barrier and travelling by extra-cellular bulk
flow transport along olfactory and trigeminal perivascular
Fig. 6 Diminution of the insulin pathway in the diabetic murine
brain. Levels of IRb fell within the diabetic murine brain but
replenished with I-I, but not S-I (A). mRNA for both IRb and IRS-1
was also deficient in diabetic murine brain regions (hippocampus
not shown) such as cortex (B). Non-diabetic mice receiving
intranasal insulin demonstrated heightened IRb mRNA, and D I-I
mice received protection against loss of both IRb and IRS-1.
Protein levels (C) for IRb and IRS-1 fell in combination with
diabetes as demonstrated with quantitative measurements using
three protein samples for each group (D), with protection again
offered by I-I in both the diabetic cortex and hippocampus.
There were no differences in measurements between non-diabetic
cohort mouse groups. Multiple ANOVA tests, with asterisk indi-
cating significant difference (P50.016) between groups indicated by
horizontal bars, were performed.
Intranasal insulin and diabetic encephalopathy
Brain (2008),131, 3311^3334
3327
at Pennsylvania State University on February 23, 2013
Downloaded from
Fig. 7 Central synaptic components in the diabetic murine brain are decreased. Choline acetyltransferase (ChAT) and synaptophysin (SYP)
levels are diminished within the cortex and hippocampus (data not shown) of diabetic mouse brains relative to non-diabetic murine brains
(A, D). ChAT and SYP levels within the D I-I brain (B, E) were not different from that of the C I-S brain (A, D), while the D I-S brain
demonstrated loss of ChATand SYP (C, F). Losses in synaptic markers were determined by quantification of density for immunostaining for
both ChAT (G) and SYP (H) within cortex and hippocampus (data not shown). qRT-PCR also demonstrated loss of mRNA for both SYP
and ChAT (I). Finally, quantitative assessment of three protein blots (representative blot, J) also portrays a loss of both SYP and ChAT in
the hippocampus and cortex of the diabetic murine brain, where protection against synaptic loss is again provided by I-I to the diabetic
murine brain (K). Multiple ANOVA tests were performed for each comparison, with asterisk indicating significant difference (P50.05)
between groups indicated by horizontal bars.
3328
Brain (2008), 131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
Fig. 8 Quantification of NFkB expression. Upregulation in diabetic murine brain for NFkB protein and mRNA occurs as compared to non-
diabetic brain. In the hippocampus, neurons identified with NeuN and oligodendrocytes, identified with PDGFRa, co-express NFkBp65
least in the C S-S brain (A) as compared to diabetic murine brain. D I-I mouse brain (B) had less nuclear activation (nuclear presence) than
D I-S mouse brain (C), within both neurons (D) and oligodendroglia (E). mRNA measurements of NFkBp65 expression identified age-
related increases occurring greatest after 3^5 months of diabetes (F), and again ameliorated with I-I. Protein blotting (representative blot,
G) and its quantitative assessment (H) also demonstrated accumulation of NFkBp65 protein over time, accelerated with diabetes, and
partially suppressed with I-I. The amount of NFkB binding to DNA was significantly upregulated with diabetes (I), with protection from its
increase identied in the D I-I cohort, based upon bands obtained from EMSA, an example of which is demonstrated (J). Quantitative
analysis for Western blots was performed using three samples for each group. For comparisons, multiple ANOVA tests, with asterisk
indicating significant difference between groups indicated by horizontal bars, were performed. All diabetic values were significantly less
than non-diabetic values (significance not visually demonstrated).
Intranasal insulin and diabetic encephalopathy
Brain (2008),131, 3311^3334
3329
at Pennsylvania State University on February 23, 2013
Downloaded from
channels, as well as axonal transport pathways (Benedict
et al., 2004; Thorne et al., 2004), leading to faster uptake
and less systemic insulin presence.
The role of insulin as a neuroprotective
trophic factor
Insulin, a highly conserved peptide that is no longer
thought of as solely a promoter of glucose turnover, has
now emerged as a key neurotrophic factor in the nervous
system alongside and interacting with the IGF-I receptor
system, also important in maintaining cognitive function
(Trejo et al., 2008). Insulin is a potent trophic factor which
becomes lost within type I diabetes. The major site of
insulin’s activity, IR, is found in high concentration in the
brain, particularly in the cerebral cortex, olfactory bulb,
hippocampus (Fig. 6), amygdala and septum (Havrankova
et al., 1978a, b; Baskin et al., 1987; Unger et al., 1991).
Reduced levels of insulin and its signalling molecules occur
in the CSF and brain of Alzheimer disease patients (Craft
et al., 1998; Hoyer, 2004). Also, IRs are present at synapses
for both astrocytes and neurons (Abbott et al., 1999). As
demonstrated in our study, diabetic rodents also demon-
strate a loss of insulin transduction machinery (Fig. 6),
which has previously been linked to the increased expres-
sion of amyloid precursor protein (APP), APP’s cleavage
enzyme b-secretase, and other abnormalities typical of the
Alzheimer disease brain (Ho et al., 2004; Salkovic-Petrisic
et al., 2006; Li et al., 2007). It has also been demonstrated
that peripheral hyperinsulinaemia and reduced insulin
signalling increase levels of amyloid (Ab) and tau
hyperphosphorylation, leading to the formation of both
senile plaques and neurofibrillary tangles (Zhao et al.,
2004b; Freude et al., 2005). In addition, insulin promotes
physiologic processes critical for memory, including long-
term potentiation, expression of glutamate receptors, and
modulation of neurotransmitter levels (Craft and Watson,
2004). Finally, insulin diminishes hypothalamic–pituitary–
adrenal-axis activity (Hallschmid et al., 2008), recently
speculated to contribute to diabetes-mediated cognitive
dysfunction (Stranahan et al., 2008).
Potentially preventable changes
in diabetic encephalopathy
Our experiments have demonstrated that long-term intra-
nasal insulin delivery protected against brain atrophy
(Fig. 3). Although other studies have reported evidence of
cerebral neuronal loss in diabetes (Li et al., 2007), we could
not detect any loss of neuronal density in this experimen-
tal diabetic brain model, similar to other recent studies
(Stranahan et al., 2008), but we did determine oligoden-
droglial loss occurs and likely relates to the development
of WMA. One potential cause for our detected diabetes-
associated brain atrophy (Fig. 3) is the presence of large
degree of synaptic loss (Fig. 7), also detected in type II
diabetic rat models (Li et al., 2007). Synaptic loss may
precede other forms of pathology in some models of
Alzheimer disease (Yoshiyama et al., 2007), including neu-
ronal loss. Diabetes also leads to a synergistic potentiation
of synaptic loss in transgenic models of Alzheimer disease
(Burdo et al., 2008). This may certainly be impacted by the
presence of insulin receptor at central synapses (Heidenreich
et al., 1983; Matsumoto and Rhoads, 1990; Wan et al., 1997;
Zhao et al., 1999). Signal transduction by neuronal IRs is
exquisitely sensitive to soluble Ab oligomer-mediated disrup-
tion in vitro, and neuronal response to insulin is also inhib-
ited (Zhao et al., 2008). Such synaptic loss identified in our
experiments of the diabetic brain may herald subsequent
neuronal loss, but other models need to be investigated, and
longer duration studies may be necessary to conclude this.
Insulin’s downstream signalling pathways
Insulin is critical for maintenance of numerous downstream
intracellular signalling pathways. Insulin stimulation upre-
gulates protein–tyrosine phosphorylation (Mahadev et al.,
2004) through downstream activation of IRS-2 (Huang
et al., 2005a). Insulin presence leads to activation of Akt
and phosphorylation of Akt substrates (Fig. 5) (Bruss et al.,
2005). In addition, insulin modulates the inner mitochon-
drial membrane potential through activation of the PI3K
pathway, stimulating phosphorylation of Akt and cAMP
response element-binding protein CREB (Marshall, 1995;
Yao and Cooper, 1995; Fernyhough et al., 2003; Viard et al.,
2004; Huang et al., 2005b), as well as supporting neuritic
extension and branching (Jones et al., 2003). PI3K also
enhances voltage-dependent calcium channel current func-
tioning in diabetic neurons (Viard et al., 2004). In our
studies, downregulation of PI3K/Akt occurred throughout
the diabetic murine brain (Fig. 5), while insulin delivered
intranasally, and to a lesser extent, subcutaneously,
prevented such downregulation while concurrently leading
to improvements in behaviour and morphology.
Insulin’s benefits in the diabetic brain may relate to
inactivation of GSK-3b and activation of CREB. Besides
regulating the transcriptional activities of CREB (Cohen
and Frame, 2001; Grimes and Jope, 2001), GSK-3b is also a
neuron-specific (Leroy and Brion, 1999) apoptosis pro-
moter in the non-phosphorylated, or active state (Hetman
et al., 2000); Akt-mediated phosphorylation of GSK-3b
renders it inactive, giving anti-apoptotic properties (Pap
and Cooper, 1998; Bijur et al., 2000; Hetman et al., 2000).
Similar to insulin in our study, IGF-I also activates the
PI3K/Akt pathway (Dudek et al., 1997; Zheng et al., 2002),
leading to phosphorylation of CREB and GSK-3b
(Leinninger et al., 2004). IGF-I provision also induces oli-
godendrocyte progenitor proliferation via Akt activation
(Cui and Almazan, 2007), with GSK-3b phosphorylation in
oligodendrocyte progenitor cells affecting oligodendrocyte
stability (Frederick et al., 2007). pGSK-3b may also regulate
gene expression and activity of transcriptional factor
3330
Brain (2008),131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
binding to the MAG promoter region (Ogata et al., 2004),
promoting myelination and possibly explaining insulin-
mediated protection against WMA development in the
experimental diabetic murine brain (Figs 3 and 4). While
CREB phosphorylation inhibits apoptosis in neurons
(Walton et al., 1999), the loss of CREB results in impaired
axonal growth (Lonze et al., 2002), suggesting that CREB is
neuroprotective. Intranasal insulin led to heightened
pCREB levels as well as greater CREB DNA binding
indicating transcription (Fig. 5). Overall, we hypothesize
that insulin’s benefits in the diabetic murine brain are due
to maintained phosphorylation of CREB and GSK-3b.
The utility of intranasal delivery in
diabetic encephalopathy
In the case of diabetic encephalopathy, and likely the
Alzheimer disease brain, insulin’s trophic effects are lacking.
I-I delivery without modifying systemic glucose or glycated
haemoglobin (Table 1) (Tomlinson and Gardiner, 2008)
appears to be essential to insulin’s long-term benefits in
brain exposed to diabetes. Human studies have already
demonstrated safe administration of intranasal insulin in
patients with Alzheimer disease, leading to some improve-
ment in cognition and modulating markers of Alzheimer
disease (Reger et al., 2006, 2008). These studies have
concentrated upon the potential benefits of intranasal
insulin delivery upon clinical and pathological markers of
Alzheimer disease. Other clinical studies examining intra-
nasal insulin delivery have also demonstrated effects upon
the CNS, such as improvement in memory (Benedict et al.,
2007, 2008), lowering of food intake (Tomlinson et al.,
2008), and improvement in mood (Hallschmid et al., 2008).
In humans, plasma glucose levels may be minimally
impacted by intranasal insulin delivery (Born et al., 2002;
Reger et al., 2006, 2008; Tomlinson et al., 2008). As of yet,
intranasal insulin delivery to diabetic subjects for the intent
of improving memory or other diabetic complications has
not yet been described, although intranasal insulin delivery
has been examined as a potential method for the treatment
of diabetes itself (Owens et al., 2003; Khafagy et al., 2007).
Our study results must be considered under the
limitations of working in a murine model, and the inability
to achieve a long-term model of murine type I diabetes
with optimal glycaemic management as a suitable control
group. The mouse cohorts were subjected to intensive
testing throughout their lifetime, with the possibility of
stressful impacts upon the results obtained. The use of
multiple cognitive studies, often needed as part of a battery
of tests, may have led to crossover effects affecting results
from one cognitive test based upon the preceding test. It is
also possible that hypoglycaemia may have impacted some
of the results of cognitive testing, and the impact of
hypoglycaemia upon the D I-S and C I-S cohort groups was
anticipated, but difficult to avoid. As well, performance of
behavioural testing within multiple cohorts of mice within
each intervention group may have contributed to perfor-
mance disparities. Based upon our studies, it is difficult to
develop a more appropriate control group of diabetic mice
with long-term glycaemic control based upon the STZ-
induced diabetic model. Despite these difficulties, D S-I
mice performed better than D S-S cohort mice during
portions of cognitive testing. Although cognitive changes in
our mice also seem to occur mainly during the younger and
older age time points (similar to expected changes in type I
diabetic patients), explanations for a relative plateau of
function during the middle-aged years are unknown.
(Wessels et al., 2007, 2008; Biessels et al., 2008;
Kloppenborg et al., 2008;) At this time, our results are
limited to murine models of type I diabetes, and other
forms of diabetes, including models of type II diabetes, will
need study in the future to confirm our results in other
models of diabetes.
Conclusions
Intranasal insulin delivery is a potential therapy to
ameliorate behavioural, morphological and molecular
changes occurring in brain exposed to diabetes over time.
Our results provide strong evidence for benefits of insulin
without impact upon serum glucose levels, indicating that
insulin is an important neurotrophic factor in the manage-
ment of diabetes-mediated brain disease. These data
support the development of clinical studies for the
prevention and slowing of the adverse effects of diabetes
upon the brain using intranasal delivery of insulin.
Supplementary material
Supplementary material is available at Brain online.
Funding
This study was supported by an operating grant from the
Alberta Heritage Foundation for Medical Research and the
Canadian Diabetes Association. C.T. is a Clinical
Investigator of the Alberta Heritage Foundation for
Medical Research and D.W.Z. is a Scientist of the Alberta
Heritage Foundation for Medical Research.
References
Abbott MA, Wells DG, Fallon JR. The insulin receptor tyrosine kinase
substrate p58/53 and the insulin receptor are components of CNS
synapses. J Neurosci 1999; 19: 7300–7308.
Adamo M, Raizada MK, LeRoith D. Insulin and insulin-like growth factor
receptors in the nervous system. Mol Neurobiol 1989; 3: 71–100.
Akisaki T, Sakurai T, Takata T, Umegaki H, Araki A, Mizuno S, et al.
Cognitive dysfunction associates with white matter hyperintensities and
subcortical atrophy on magnetic resonance imaging of the elderly
diabetes mellitus Japanese elderly diabetes intervention trial (J-EDIT).
Diabetes Metab Res Rev 2006; 22: 376–84.
Awad N, Gagnon M, Messier C. The relationship between impaired
glucose tolerance, type 2 diabetes, and cognitive function. J Clin Exp
Neuropsychol 2004; 26: 1044–80.
Intranasal insulin and diabetic encephalopathy
Brain (2008), 131, 3311^3334
3331
at Pennsylvania State University on February 23, 2013
Downloaded from
Baskin DG, Figlewicz DP, Woods SC, Porte D Jr, Dorsa DM. Insulin in the
brain. Annu Rev Physiol 1987; 49: 335–47.
Benedict C, Hallschmid M, Hatke A, Schultes B, Fehm HL, Born J, et al.
Intranasal
insulin
improves
memory
in
humans.
Psychoneuroendocrinology 2004; 29: 1326–34.
Benedict C, Hallschmid M, Schultes B, Born J, Kern W. Intranasal insulin
to improve memory function in humans. Neuroendocrinology 2007; 86:
136–42.
Biessels GJ, Deary IJ, Ryan CM. Cognition and diabetes: a lifespan
perspective. Lancet Neurol 2008; 7: 184–90.
Biessels GJ, Kamal A, Ramakers GM, Urban IJ, Spruijt BM, Erkelens DW,
et al. Place learning and hippocampal synaptic plasticity in streptozo-
tocin-induced diabetic rats. Diabetes 1996; 45: 1259–66.
Biessels GJ, Kamal A, Urban IJ, Spruijt BM, Erkelens DW, Gispen WH.
Water maze learning and hippocampal synaptic plasticity in
streptozotocin-diabetic rats: effects of insulin treatment. Brain Res
1998; 800: 125–35.
Bijur GN, De SP, Jope RS. Glycogen synthase kinase-3beta facilitates
staurosporine- and heat shock-induced apoptosis. Protection by lithium.
J Biol Chem 2000; 275: 7583–90.
Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL. Sniffing
neuropeptides: a transnasal approach to the human brain. Nat Neurosci
2002; 5: 514–6.
Bruss MD, Arias EB, Lienhard GE, Cartee GD. Increased phosphorylation
of Akt substrate of 160 kDa (AS160) in rat skeletal muscle in response to
insulin or contractile activity. Diabetes 2005; 54: 41–50.
Burdo JR, Chen Q, Calcutt NA, Schubert D. The pathological interaction
between diabetes and presymptomatic Alzheimer’s disease. Neurobiol
Aging 2008.
Chen XQ, Fawcett JR, Rahman YE, Ala TA, Frey II WH. Delivery of nerve
growth factor to the brain via the olfactory pathway. J Alzheimers Dis
1998; 1: 35–44.
Cohen RA. Dysfunction of vascular endothelium in diabetes mellitus.
Circulation 1993; 87: V67–76.
Cohen P, Frame S. The renaissance of GSK3. Nat Rev Mol Cell Biol 2001;
2: 769–76.
Craft S, Peskind E, Schwartz MW, Schellenberg GD, Raskind M,
Porte D, Jr. Cerebrospinal fluid and plasma insulin levels in
Alzheimer’s disease: relationship to severity of dementia and apolipo-
protein E genotype. Neurology 1998; 50: 164–8.
Craft S, Watson GS. Insulin and neurodegenerative disease: shared and
specific mechanisms. Lancet Neurol 2004; 3: 169–78.
Crawley JN: Behavioral paradigms. The Morris water task. In: Crawley JN,
editor. What’s wrong with my mouse? Behavioral phenotyping of
transgenic and knockout mice. New York: Wiley-Liss; 2000. p. 87–95.
Cui QL, Almazan G. IGF-I-induced oligodendrocyte progenitor prolifera-
tion requires PI3K/Akt, MEK/ERK, and Src-like tyrosine kinases. J
Neurochem 2007; 100: 1480–93.
de Groot JC, de Leeuw FE, Oudkerk M, van GJ, Hofman A, Jolles J,
Breteler MM. Cerebral white matter lesions and cognitive function: the
Rotterdam Scan Study. Ann Neurol 2000; 47: 145–51.
Dhanda DS, Frey WH, Leopold D, Kompella UB. Nose to brain delivery:
approaches for drug deposition in the human olfactory epithelium.
Drug Deliv Technol 2005; 5: 64–72.
Dudek H, Datta SR, Franke TF, Birnbaum MJ, Yao R, Cooper GM, et al.
Regulation of neuronal survival by the serine-threonine protein kinase
Akt. Science 1997; 275: 661–5.
Fernyhough P, Huang TJ, Verkhratsky A. Mechanism of mitochondrial
dysfunction in diabetic sensory neuropathy. J Peripher Nerv Syst 2003;
8: 227–35.
File SE, Wardill AG. The reliability of the hole-board apparatus.
Psychopharmacologia 1975; 44: 47–51.
Fox MA, Chen RS, Holmes CS. Gender differences in memory and
learning in children with insulin-dependent diabetes mellitus (IDDM)
over a 4-year follow-up interval. J Pediatr Psychol 2003; 28: 569–78.
Frederick TJ, Min J, Altieri SC, Mitchell NE, Wood TL. Synergistic
induction of cyclin D1 in oligodendrocyte progenitor cells by IGF-I and
FGF-2 requires differential stimulation of multiple signaling pathways.
Glia 2007; 55: 1011–22.
Freude S, Plum L, Schnitker J, Leeser U, Udelhoven M, Krone W, et al.
Peripheral hyperinsulinemia promotes tau phosphorylation in vivo.
Diabetes 2005; 54: 3343–8.
Grimes CA, Jope RS. CREB DNA binding activity is inhibited by glycogen
synthase kinase-3 beta and facilitated by lithium. J Neurochem 2001; 78:
1219–32.
Gundersen HJ, Bendtsen TF, Korbo L, Marcussen N, Moller A, Nielsen K,
et al. Some new, simple and efficient stereological methods and their use
in pathological research and diagnosis. APMIS 1988; 96: 379–94.
Haan MN. Therapy Insight: type 2 diabetes mellitus and the risk of late-
onset Alzheimer’s disease. Nat Clin Pract Neurol 2006; 2: 159–66.
Hallschmid M, Benedict C, Schultes B, Born J, Kern W. Obese men
respond to cognitive but not to catabolic brain insulin signaling. Int J
Obes (Lond) 2008; 32: 275–82.
Hattori Y, Hattori S, Sato N, Kasai K. High-glucose-induced nuclear factor
kappaB activation in vascular smooth muscle cells. Cardiovasc Res 2000;
46: 188–97.
Havrankova J, Schmechel D, Roth J, Brownstein M. Identification of
insulin in rat brain. Proc Natl Acad Sci USA 1978a; 75: 5737–41.
Havrankova J, Roth J, Brownstein M. Insulin receptors are widely
distributed in the central nervous system of the rat. Nature 1978b;
272: 827–9.
Heidenreich KA, Zahniser NR, Berhanu P, Brandenburg D, Olefsky JM.
Structural differences between insulin receptors in the brain and
peripheral target tissues. J Biol Chem 1983; 258: 8527–30.
Hetman M, Cavanaugh JE, Kimelman D, Xia Z. Role of glycogen synthase
kinase-3beta in neuronal apoptosis induced by trophic withdrawal.
J Neurosci 2000; 20: 2567–74.
Ho L, Qin W, Pompl PN, Xiang Z, Wang J, Zhao Z, et al. Diet-induced
insulin resistance promotes amyloidosis in a transgenic mouse model of
Alzheimer’s disease. FASEB J 2004; 18: 902–4.
Hoyer S. Glucose metabolism and insulin receptor signal transduction in
Alzheimer disease. Eur J Pharmacol 2004; 490: 115–25.
Huang C, Thirone AC, Huang X, Klip A. Differential contribution of
insulin receptor substrates 1 versus 2 to insulin signaling and glucose
uptake in l6 myotubes. J Biol Chem 2005a; 280: 19426–35.
Huang TJ, Verkhratsky A, Fernyhough P. Insulin enhances mitochondrial
inner membrane potential and increases ATP levels through phosphoi-
nositide 3-kinase in adult sensory neurons. Mol Cell Neurosci 2005b; 28:
42–54.
Ikram MA, Vrooman HA, Vernooij MW, van der LF, Hofman A, van
der LA, et al. Brain tissue volumes in the general elderly population. The
Rotterdam Scan Study. Neurobiol Aging 2008; 29: 882–90.
Jacobson AM, Musen G, Ryan CM, Silvers N, Cleary P, Waberski B, et al.
Long-term effect of diabetes and its treatment on cognitive function.
N Engl J Med 2007; 356: 1842–52.
Jones DM, Tucker BA, Rahimtula M, Mearow KM. The synergistic effects
of NGF and IGF-1 on neurite growth in adult sensory neurons:
convergence on the PI 3-kinase signaling pathway. J Neurochem 2003;
86: 1116–28.
Kaiyala KJ, Prigeon RL, Kahn SE, Woods SC, Schwartz MW. Obesity
induced by a high-fat diet is associated with reduced brain insulin
transport in dogs. Diabetes 2000; 49: 1525–33.
Kamal A, Biessels GJ, Duis SE, Gispen WH. Learning and hippocampal
synaptic plasticity in streptozotocin-diabetic rats: interaction of diabetes
and ageing. Diabetologia 2000; 43: 500–6.
Kamal A, Biessels GJ, Gispen WH, Ramakers GM. Synaptic transmission
changes in the pyramidal cells of the hippocampus in streptozotocin-
induced diabetes mellitus in rats. Brain Res 2006; 1073–1074: 276–80.
Khafagy E, Morishita M, Onuki Y, Takayama K. Current challenges in
non-invasive insulin delivery systems: a comparative review. Adv Drug
Deliv Rev 2007; 59: 1521–46.
Kloppenborg RP, van den Berg E, Kappelle LJ, Biessels GJ. Diabetes and
other vascular risk factors for dementia: which factor matters most? A
systematic review. Eur J Pharmacol 2008; 585: 97–108.
3332
Brain (2008), 131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from
Knopman D, Boland LL, Mosley T, Howard G, Liao D, Szklo M, et al.
Cardiovascular risk factors and cognitive decline in middle-aged adults.
Neurology 2001; 56: 42–8.
Kramer L, Fasching P, Madl C, Schneider B, Damjancic P, Waldhausl W,
et al. Previous episodes of hypoglycemic coma are not associated with
permanent cognitive brain dysfunction in IDDM patients on intensive
insulin treatment. Diabetes 1998; 47: 1909–14.
Last D, Alsop DC, Abduljalil AM, Marquis RP, de BC, Hu K, et al. Global
and regional effects of type 2 diabetes on brain tissue volumes and
cerebral vasoreactivity. Diabetes Care 2007; 30: 1193–9.
Leinninger GM, Backus C, Uhler MD, Lentz SI, Feldman EL. Phosphati-
dylinositol 3-kinase and Akt effectors mediate insulin-like growth factor-I
neuroprotection in dorsal root ganglia neurons. FASEB J 2004; 18: 1544–6.
Leroy K, Brion JP. Developmental expression and localization of
glycogen synthase kinase-3beta in rat brain. J Chem Neuroanat 1999;
16: 279–3.
Li ZG, Zhang W, Sima AA. The role of impaired insulin/IGF action in
primary diabetic encephalopathy. Brain Res 2005; 1037: 12–24.
Li ZG, Zhang W, Sima AA. Alzheimer-like changes in rat models of
spontaneous diabetes. Diabetes 2007; 56: 1817–24.
Liu XF, Fawcett JR, Hanson LR, Frey WH. The window of opportunity for
treatment of focal cerebral ischemic damage with noninvasive intranasal
insulin-like growth factor-I in rats. J Stroke Cerebrovasc Dis 2004; 13:
16–23.
Lonze BE, Riccio A, Cohen S, Ginty DD. Apoptosis, axonal growth defects,
and degeneration of peripheral neurons in mice lacking CREB. Neuron
2002; 34: 371–85.
Lupien SB, Bluhm EJ, Ishii DN. Effect of IGF-I on DNA, RNA, and
protein loss associated with brain atrophy and impaired learning in
diabetic rats. Neurobiol Dis 2006; 21: 487–95.
Mahadev K, Wu X, Motoshima H, Goldstein BJ. Integration of multiple
downstream signals determines the net effect of insulin on MAP kinase
vs. PI 30-kinase activation: potential role of insulin-stimulated H(2)O(2).
Cell Signal 2004; 16: 323–31.
Manschot SM, Brands AM, van der GJ, Kessels RP, Algra A, Kappelle LJ,
et al. Brain magnetic resonance imaging correlates of impaired cognition
in patients with type 2 diabetes. Diabetes 2006; 55: 1106–13.
Marshall CJ. Specificity of receptor tyrosine kinase signaling: transient
versus sustained extracellular signal-regulated kinase activation. Cell
1995; 80: 179–85.
Matsumoto H, Rhoads DE. Specific binding of insulin to membranes from
dendrodendritic synaptosomes of rat olfactory bulb. J Neurochem 1990;
54: 347–50.
McCarthy AM, Lindgren S, Mengeling MA, Tsalikian E, Engvall JC. Effects
of diabetes on learning in children. Pediatrics 2002; 109: E9.
Miles WR, Root HF. Psychologic tests applied to diabetic patients. Arch
Intern Med 1922; 30: 767–77.
Mooradian AD. Central nervous system complications of diabetes
mellitus–a perspective from the blood-brain barrier. Brain Res Brain
Res Rev 1997; 23: 210–8.
Morris R. Developments of a water-maze procedure for studying spatial
learning in the rat. J Neurosci Methods 1984; 11: 47–60.
Munoz DG, Hastak SM, Harper B, Lee D, Hachinski VC. Pathologic
correlates of increased signals of the centrum ovale on magnetic
resonance imaging. Arch Neurol 1993; 50: 492–7.
Musen G, Lyoo IK, Sparks CR, Weinger K, Hwang J, Ryan CM, et al.
Effects of type 1 diabetes on gray matter density as measured by voxel-
based morphometry. Diabetes 2006; 55: 326–33.
Murray AD, Staff RT, Shenkin SD, Deary IJ, Starr JM, Whalley LJ. Brain
white matter hyperintensities: relative importance of vascular risk factors
in nondemented elderly people. Radiology 2005; 237: 251–7.
Northam EA, Anderson PJ, Jacobs R, Hughes M, Warne GL, Werther GA.
Neuropsychological profiles of children with type 1 diabetes 6 years after
disease onset. Diabetes Care 2001; 24: 1541–6.
Ogata T, Iijima S, Hoshikawa S, Miura T, Yamamoto S, Oda H, et al.
Opposing extracellular signal-regulated kinase and Akt pathways control
Schwann cell myelination. J Neurosci 2004; 24: 6724–32.
Owens DR, Zinman B, Bolli G. Alternative routes of insulin delivery.
Diabet Med 2003; 20: 886–98.
Pantoni L, Garcia JH. Pathogenesis of leukoaraiosis: a review. Stroke 1997;
28: 652–9.
Pantoni L, Poggesi A, Inzitari D. The relation between white-matter lesions
and cognition. Curr Opin Neurol 2007; 20: 390–7.
Pap M, Cooper GM. Role of glycogen synthase kinase-3 in the
phosphatidylinositol 3-Kinase/Akt cell survival pathway. J Biol Chem
1998; 273: 19929–32.
Patti ME, Sun XJ, Bruening JC, Araki E, Lipes MA, White MF, et al.
4PS/insulin receptor substrate (IRS)-2 is the alternative substrate of the
insulin receptor in IRS-1-deficient mice. J Biol Chem 1995; 270:
24670–3.
Reaven GM, Thompson LW, Nahum D, Haskins E. Relationship between
hyperglycemia and cognitive function in older NIDDM patients.
Diabetes Care 1990; 13: 16–21.
Reger MA, Watson GS, Frey WH, Baker LD, Cholerton B, Keeling ML,
et al. Effects of intranasal insulin on cognition in memory-impaired
older adults: modulation by APOE genotype. Neurobiol Aging 2006; 27:
451–8.
Reger MA, Watson GS, Green PS, Wilkinson CW, Baker LD, Cholerton B,
et al. Intranasal insulin improves cognition and modulates beta-amyloid
in early AD. Neurology 2008; 70: 440–8.
Ross TM, Martinez PM, Renner JC, Thorne RG, Hanson LR, Frey WH.
Intranasal administration of interferon beta bypasses the blood-brain
barrier to target the central nervous system and cervical lymph nodes: a
non-invasive treatment strategy for multiple sclerosis. J Neuroimmunol
2004; 151: 66–77.
Rothblat LA, Kromer LF. Object recognition memory in the rat: the role of
the hippocampus. Behav Brain Res 1991; 42: 25–32.
Ryan CM. Neurobehavioral complications of type I diabetes. Examination
of possible risk factors. Diabetes Care 1988; 11: 86–93.
Ryan CM, Williams TM. Effects of insulin-dependent diabetes on learning
and memory efficiency in adults. J Clin Exp Neuropsychol 1993; 15:
685–700.
Ryan CM, Williams TM, Finegold DN, Orchard TJ. Cognitive dysfunction
in adults with type 1 (insulin-dependent) diabetes mellitus of long
duration: effects of recurrent hypoglycaemia and other chronic
complications. Diabetologia 1993; 36: 329–34.
Ryan CM, Geckle M. Why is learning and memory dysfunction in Type 2
diabetes limited to older adults? Diabetes Metab Res Rev 2000; 16:
308–15.
Salkovic-Petrisic M, Tribl F, Schmidt M, Hoyer S, Riederer P. Alzheimer-
like changes in protein kinase B and glycogen synthase kinase-3 in rat
frontal cortex and hippocampus after damage to the insulin signalling
pathway. J Neurochem 2006; 96: 1005–15.
Schoenle EJ, Schoenle D, Molinari L, Largo RH. Impaired intellectual
development in children with Type I diabetes: association with HbA(1c),
age at diagnosis and sex. Diabetologia 2002; 45: 108–14.
Schmidt R, Launer LJ, Nilsson LG, Pajak A, Sans S, Berger K, et al. Magnetic
resonance imaging of the brain in diabetes: the Cardiovascular
Determinants of Dementia (CASCADE) Study. Diabetes 2004; 53: 687–92.
Schwabe K, Klein S, Koch M. Behavioural effects of neonatal lesions of the
medial prefrontal cortex and subchronic pubertal treatment with
phencyclidine of adult rats. Behav Brain Res 2006; 168: 150–60.
Schwartz AV, Vittinghoff E, Sellmeyer DE, Feingold KR, de RN,
Strotmeyer ES, et al. Diabetes-related complications, glycemic control,
and falls in older adults. Diabetes Care 2008; 31: 391–6.
Stewart R, Liolitsa D. Type 2 diabetes mellitus, cognitive impairment and
dementia. Diabet Med 1999; 16: 93–112.
Strachan MW, Deary IJ, Ewing FM, Frier BM. Is type II diabetes associated
with an increased risk of cognitive dysfunction? A critical review of
published studies. Diabetes Care 1997; 20: 438–45.
Stranahan AM, Arumugam TV, Cutler RG, Lee K, Egan JM, Mattson MP.
Diabetes impairs hippocampal function through glucocorticoid-
mediated effects on new and mature neurons. Nat Neurosci 2008; 11:
309–17.
Intranasal insulin and diabetic encephalopathy
Brain (2008),131, 3311^3334
3333
at Pennsylvania State University on February 23, 2013
Downloaded from
Thorne RG, Pronk GJ, Padmanabhan V, Frey WH. Delivery of insulin-like
growth factor-I to the rat brain and spinal cord along olfactory and
trigeminal pathways following intranasal administration. Neuroscience
2004; 127: 481–96.
Tomlinson DR, Gardiner NJ. Glucose neurotoxicity. Nat Rev Neurosci
2008; 9: 36–45.
Benedict C, Kern W, Schultes B, Born J, Hallschmid M. Differential
sensitivity of men and women to anorexigenic and memory improving
effects of intranasal insulin. J Clin Endocrinol Metab 2008.
Toth C, Schmidt AM, Tuor UI, Francis G, Foniok T, Brussee V, et al.
Diabetes, leukoencephalopathy and RAGE. Neurobiol Dis 2006; 23:
445–61.
Trejo JL, Llorens-Martın MV, Torres-Alemán I. The effects of exercise on
spatial learning and anxiety-like behavior are mediated by an IGF-I-
dependent mechanism related to hippocampal neurogenesis. Mol Cell
Neurosci. 2008; 37: 402–11.
Tullberg M, Fletcher E, DeCarli C, Mungas D, Reed BR, Harvey DJ, et al.
White matter lesions impair frontal lobe function regardless of their
location. Neurology 2004; 63: 246–53.
Unger J, McNeill TH, Moxley RT, III, White M, Moss A, Livingston JN.
Distribution of insulin receptor-like immunoreactivity in the rat
forebrain. Neuroscience 1989; 31: 143–57.
Unger JW, Livingston JN, Moss AM. Insulin receptors in the central
nervous system: localization, signalling mechanisms and functional
aspects. Prog Neurobiol 1991; 36: 343–62.
van Harten B, de Leeuw FE, Weinstein HC, Scheltens P, Biessels GJ. Brain
imaging in patients with diabetes: a systematic review. Diabetes Care
2006; 29: 2539–48.
van Harten B, Oosterman JM, Potter van Loon BJ, Scheltens P,
Weinstein HC. Brain lesions on MRI in elderly patients with type 2
diabetes mellitus. Eur Neurol 2007; 57: 70–4.
Verdelho A, Madureira S, Ferro JM, Basile AM, Chabriat H, Erkinjuntti T,
et al. Differential impact of cerebral white matter changes, diabetes,
hypertension and stroke on cognitive performance among non-disabled
elderly. The LADIS study. J Neurol Neurosurg Psychiatry 2007; 78:
1325–30.
Viard P, Butcher AJ, Halet G, Davies A, Nurnberg B, Heblich F,
Dolphin AC. PI3K promotes voltage-dependent calcium channel
trafficking to the plasma membrane. Nat Neurosci 2004; 7: 939–46.
Vig PJ, Subramony SH, D’Souza DR, Wei J, Lopez ME. Intranasal
administration of IGF-I improves behavior and Purkinje cell pathology
in SCA1 mice. Brain Res Bull 2006; 69: 573–9.
Walton M, Woodgate AM, Muravlev A, Xu R, During MJ, Dragunow M.
CREB phosphorylation promotes nerve cell survival. J Neurochem 1999;
73: 1836–42.
Wan Q, Xiong ZG, Man HY, Ackerley CA, Braunton J, Lu WY, et al.
Recruitment of functional GABA(A) receptors to postsynaptic domains
by insulin. Nature 1997; 388: 686–90.
Weinger K, Jacobson AM, Musen G, Lyoo IK, Ryan CM, Jimerson DC,
et al. The effects of type 1 diabetes on cerebral white matter.
Diabetologia 2008; 51: 417–25.
Wessels AM, Rombouts SA, Remijnse PL, Boom Y, Scheltens P, Barkhof F,
et al. Cognitive performance in type 1 diabetes patients is associated
with cerebral white matter volume. Diabetologia 2007; 50: 1763–9.
White MF, Yenush L. The IRS-signaling system: a network of docking
proteins that mediate insulin and cytokine action. Curr Top Microbiol
Immunol 1998; 228: 179–208.
Whitman GT, Tang Y, Lin A, Baloh RW. A prospective study of cerebral
white matter abnormalities in older people with gait dysfunction.
Neurology 2001; 57: 990–4.
Wickelgren I. Tracking insulin to the mind. Science 1998; 280: 517–9.
Woods SC, Seeley RJ, Baskin DG, Schwartz MW. Insulin and the blood-
brain barrier. Curr Pharm Des 2003; 9: 795–800.
Yao R, Cooper GM. Requirement for phosphatidylinositol-3 kinase in the
prevention of apoptosis by nerve growth factor. Science 1995; 267: 2003–6.
Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, et al.
Synapse loss and microglial activation precede tangles in a P301S
tauopathy mouse model. Neuron 2007; 53: 337–51.
Yu XR, Jia GR, Gao GD, Wang SH, Han Y, Cao W. Neuroprotection of
insulin against oxidative stress-induced apoptosis in cultured retinal
neurons: involvement of phosphoinositide 3-kinase/Akt signal pathway.
Acta Biochim Biophys Sin (Shanghai) 2006; 38: 241–8.
Zhao W, Chen H, Xu H, Moore E, Meiri N, Quon MJ, et al. Brain insulin
receptors and spatial memory. Correlated changes in gene expression,
tyrosine phosphorylation, and signaling molecules in the hippocampus
of water maze trained rats. J Biol Chem 1999; 274: 34893–902.
Zhao WQ, Chen H, Quon MJ, Alkon DL. Insulin and the insulin receptor
in experimental models of learning and memory. Eur J Pharmacol
2004a; 490: 71–81.
Zhao L, Teter B, Morihara T, Lim GP, Ambegaokar SS, Ubeda OJ, et al.
Insulin-degrading enzyme as a downstream target of insulin receptor
signaling cascade: implications for Alzheimer’s disease intervention.
J Neurosci 2004b; 24: 11120–6.
Zhao WQ, De Felice FG, Fernandez S, Chen H, Lambert MP, Quon MJ,
et al. Amyloid beta oligomers induce impairment of neuronal insulin
receptors. FASEB J 2008; 22: 246–60.
Zheng WH, Kar S, Quirion R. Insulin-like growth factor-1-induced
phosphorylation of transcription factor FKHRL1 is mediated by
phosphatidylinositol 3-kinase/Akt kinase and role of this pathway in
insulin-like growth factor-1-induced survival of cultured hippocampal
neurons. Mol Pharmacol 2002; 62: 225–33.
Appendix 1
The following brain regions were chosen as areas for close
inspection within this study for MRI and myelin quanti-
fication portions of the experiment:
Caudate putamen
Primary motor/sensory cortex
Internal capsule
Cerebral peduncle
CA1 region of hippocampus
CA2/3 region of hippocampus
Ventroposterior region of thalamus
Corpus callosum
Amygdala
Substantia nigra pars reticulata
Subiculum
Lentiform nuclear region
Primary visual cortex
Cerebellum
3334
Brain (2008),131, 3311^3334
G. J. Francis et al.
at Pennsylvania State University on February 23, 2013
Downloaded from

Leave a Reply