The mechanisms that underlie glucose sensing during

advertisement
dme_2376.fm Page 513 Wednesday, April 9, 2008 8:01 PM
DIABETICMedicine
DOI: 10.1111/j.1464-5491.2007.02376.x
Review Article
Blackwell Publishing Ltd
The mechanisms that underlie glucose sensing during
hypoglycaemia in diabetes
R. McCrimmon
Yale University School of Medicine, Department of Internal Medicine, New Haven, CT, USA
Accepted 10 October 2007
Abstract
Hypoglycaemia is a frequent and greatly feared side-effect of insulin therapy, and a major obstacle to achieving nearnormal glucose control. This review will focus on the more recent developments in our understanding of the mechanisms
that underlie the sensing of hypoglycaemia in both non-diabetic and diabetic individuals, and how this mechanism
becomes impaired over time. The research focus of my own laboratory and many others is directed by three principal
questions. Where does the body sense a falling glucose? How does the body detect a falling glucose? And why does this
mechanism fail in Type 1 diabetes? Hypoglycaemia is sensed by specialized neurons found in the brain and periphery, and
of these the ventromedial hypothalamus appears to play a major role. Neurons that react to fluctuations in glucose use
mechanisms very similar to those that operate in pancreatic B- and A-cells, in particular in their use of glucokinase and
the KATP channel as key steps through which the metabolic signal is translated into altered neuronal firing rates. During
hypoglycaemia, glucose-inhibited (GI) neurons may be regulated by the activity of AMP-activated protein kinase. This
sensing mechanism is disturbed by recurrent hypoglycaemia, such that counter-regulatory defence responses are triggered
at a lower glucose level. Why this should occur is not yet known, but it may involve increased metabolism or fuel delivery
to glucose-sensing neurons or alterations in the mechanisms that regulate the stress response.
Diabet. Med. 25, 513–522 (2008)
Keywords hypoglycaemia, glucose-excited neurons, glucose-inhibited neurons, ventromedial hypothalamus, AMPactivated protein kinase
ACTH, adrenocorticotrophic hormone; AgRP, agouti-related peptide; AMP, adenosine monophosphate;
AMPK, AMP-activated protein kinase; APF, action potential frequency; Arc, arcuate nucleus; ATP, adenosine
triphosphate; CRH, corticotrophin-releasing hormone; CSF, cerebrospinal fluid; DMH, dorsomedial hypothalamus; ECF,
extracellular fluid; GABA, gamma-aminobutyric acid; GE, glucose-excited neuron; GH, growth hormone; GI, glucoseinhibited neuron; GLUT, glucose transporter; GK, glucokinase; HAAF, hypoglycaemia-associated autonomic failure;
KATP, ATP-sensitive potassium channel; KO, knock out; MCT2, monocarboxylate transporter 2; MRNA, messanger
ribonucleic acid; NO, nitric oxide; NOS, nitric oxide synthase; POMC, pro-opiomelano cortin; PVN, paraventricular
nucleus; rt-PCR, reverse transcription polymerase chain reaction; T1DM, Type 1 diabetes mellitus; VDCC, voltagedependent calcium channel; VMH, ventromedial hypothalamus; VMN, ventromedial nucleus
Abbreviations
Introduction
Shortly after the introduction of insulin in the management of
T1DM, clinicians became aware of the potential for insulin
therapy to induce iatrogenic hypoglycaemia. Despite the
introduction of insulin analogues and improved delivery
systems, hypoglycaemia remains the major adverse effect of
Correspondence to: Rory J. McCrimmon, Yale University, Department of
Internal Medicine—Section of Endocrinology, PO Box 208020, New Haven,
CT 06520-8020, USA. E-mail: rory.mccrimmon@yale.edu
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
insulin therapy and has emerged as a major limitation to
achieving near-normal glucose control, which is required to
reduce the risk of microvascular complications [1]. An increasing awareness of hypoglycaemia in clinical practice provided
the initial stimulus to a series of excellent and detailed human
physiological studies in the 1980s and 1990s, which greatly
increased our understanding of the counter-regulatory defence
systems that prevent and correct hypoglycaemia and, importantly, how these differed over time in individuals with T1DM
(for a detailed review of this area see [2]). However, in order
to understand the mechanisms that underlie glucose sensing, a
513
dme_2376.fm Page 514 Wednesday, April 9, 2008 8:01 PM
DIABETICMedicine
Glucose sensing during hypoglycaemia • R. McCrimmon
FIGURE 1 Neuroendocrine and neuronal
pathways involved in generating the counterregulatory response to hypoglycaemia. Sensing
of hypoglycaemia by specialized neurons in the
hypothalamus leads to: (i) activation of both
branches of the autonomic nervous system,
culminating (amongst other actions) in the
secretion of epinephrine and noradrenaline
from the adrenal medulla and in the suppression
of insulin release and stimulation of glucagon
release from the pancreas, and (ii) stimulation to
release both anterior and posterior pituitary
hormones such as ACTH and GH.
prerequisite to understanding why this homeostatic system
becomes defective in T1DM, investigators have in recent years
increasingly used more basic in vitro techniques and animal
models. This is an area of research that is developing rapidly,
and focuses on three basic questions. Where does the body
detect hypoglycaemia? How does the body detect hypoglycaemia?
And why does this mechanism become defective over time in
T1DM? This review will focus on the more recent developments in this field, which are beginning to answer these three
important questions.
Where does the body sense a falling
glucose?
Glucose is an integral part of whole-body energy homeostasis
and is tightly regulated by numerous endocrine, neuronal and
behavioural systems, which ensure that glucose levels in the
blood are maintained within a narrow physiological range. It
is perhaps not surprising, given the fundamental importance of
glucose to the organism, that the capacity to sense glucose is
not limited to any single organ system. Apart from the classical
glucose sensor, the pancreatic B-cell, glucose-sensing neurons
in the periphery have, to date, been found in the intestine [3],
hepatoportal vein [4 –9] and carotid body [10]. Within the
brain, glucose-sensing neurons are found in areas such as the
septum [11], amygdala [12], striatum [13], motor cortex [14],
hindbrain [15–19] and hypothalamus [20–25] (Fig. 1). It is
anticipated, but has not as yet been convincingly shown, that
these central and peripheral glucose-sensing neurons form an
integrated neural network that monitors and responds to
changes in the glucose levels to which they are exposed.
514
The reliance of the brain on glucose to meet its energy
demands suggests that, within the context of hypoglycaemia,
brain glucose sensing may predominate. Specialized neurons in
the hypothalamus and hindbrain [26] are recognized as playing
an important role in glucose sensing during hypoglycaemia.
Within the hypothalamus, we now recognize that glucosesensing neurons are found in certain distinct regions, namely
the VMH [27–30] (which includes the arcuate and ventromedial
nuclei), the PVN [31] and the DMH [32]. Perhaps the most
studied of these regions is the VMH. Chemical destruction of
the VMH in a rodent model with ibotenic acid was shown to
cause a ~75% reduction in the hormonal counter-regulatory
response to acute hypoglycaemia [29]. Later, it was shown in
awake rats that local perfusion of the VMH with 2-deoxyglucose
(a non-metabolizable form of glucose that effectively causes
‘local hypoglycaemia’) stimulated a classical systemic counterregulatory response [30], whereas local perfusion of the
VMH with glucose during systemic hypoglycaemia markedly
suppressed the hormonal counter-regulatory response [28].
Taken together, these three studies provided good evidence for
the VMH playing a key role in the sensing of hypoglycaemia.
The glucose-sensing neurons share certain common features.
Within the brain, they localize to regions adjacent to the third
or fourth ventricle or to the circumventricular organs (these
are regions of the brain where the blood–brain barrier is ‘leaky’
or absent). This potentially allows glucose-sensing neurons
direct sampling, and hence monitoring, of glucose levels in the
blood, brain and CSF. This is important because the presence
of the blood–brain barrier ensures that brain glucose levels are
only ~10–30% of the levels seen in the blood [33 –35]. Thus,
glucose-sensing neurons are able to integrate changes in
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
dme_2376.fm Page 515 Wednesday, April 9, 2008 8:01 PM
Review article
glucose within each of these different regions. In addition,
glucose-sensing neurons are generally located in areas involved
in the control of the autonomic nervous system, neuroendocrine
function, nutrient metabolism and energy homeostasis. What
this effectively means is that brain glucose-sensing neurons are
able to receive inputs from blood nutrient levels and a variety of
endocrine hormones, as well as from numerous peripheral and
central sensory systems. This information can then be synthesized
and used to generate the appropriate physiological response,
the ultimate aim of which is to preserve glucose homeostasis.
How does the body detect a falling glucose?
In 1953, Jean Mayer proposed the ‘glucostatic hypothesis’
[36]. Hypothalamic ‘glucoreceptors’, it was proposed, could
sense fluctuations in glucose and translate that signal into a
change in neural activity [36]. The defining feature of these
neurons should be their use of glucose, not simply as a fuel, but
as a signalling molecule that regulates their activity. All neurons
use glucose as their major fuel source and all neurons will
ultimately alter their firing rates when glucose homeostasis is
significantly disrupted, but glucose-sensing neurons alter their
membrane potential, action potential frequency and/or rate of
neurotransmitter release over the more physiological ranges of
glucose to which they are exposed. Such specialized neurons
were first demonstrated by Oomura and colleagues in 1969
[22]. These neurons are ‘glucose’-sensing in so far as glucose is
the major metabolic substrate for the brain, but the fact that
these neurons can use other fuels, such as lactate produced
either by astrocytes [37–39], delivered locally [40,41] or
systemically [42–45] to alter their function, suggests that it is
more likely to be intracellular ATP that determines the activity
of these neurons. This is intriguing because neuronal levels of
ATP are generally thought to be well maintained, thus it is
also likely that subcellular compartmentalization of the glucosesensing apparatus must exist to provide sensing capability.
Such compartmentalization has been shown within pancreatic
β-cells [46].
GE neurons
There are thought to be two predominant subtypes of glucosesensing neurons, namely GE neurons (whose activity increases
as glucose levels rise) and GI neurons (whose activity decreases
as glucose levels rise) [47]. The mechanisms through which
these neurons sense alterations in glucose remain incompletely
understood. Classical glucose sensing in the pancreatic islet
served as a model for the initial research in this field [48]. In the
pancreatic β-cell, glucose is transported into the cell through a
high-capacity glucose transporter (GLUT) to allow rapid equilibration of extracellular and cytosolic glucose. The rate of glucose
metabolism, which is closely coupled to insulin secretion, is
determined by glucokinase (GK), the enzyme in the rate-limiting
step of glycolysis [49]. Metabolism of glucose leads to several
intracellular events, the culmination of which is an increase in
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
DIABETICMedicine
cytosolic calcium. One of the key steps in this signalling pathway is the depolarization of the B-cell that follows closure of
ATP-sensitive potassium (KATP) channels in the plasma membrane [48]. The exciting discovery of glucokinase and KATP
channels in glucose-sensing regions in the brain lead to the
hypothesis that these enzymes might also serve key roles in
glucose sensing, particularly in GE neurons [48,50].
KATP channels have been demonstrated throughout the
brain, including hypothalamic regions thought to be involved
in glucose sensing [27,51–53]. Using single-cell rt-PCR to analyse
glucose-sensing neurons (identified electrophysiologically in
a hypothalamic slice preparation), investigators have shown
that they express mRNA for Kir6.2 and SUR1, the two subunits
that comprise the KATP channel in the pancreatic B-cell [54]. In
addition, electrophysiological studies of the rat [27,55–57]
and mouse [58] have demonstrated that sulphonylureas
(agents that block the KATP channel) can alter the response of
GE neurons to changes in ambient glucose, and Kir6.2 KO
mice show impaired glucose counter-regulation to systemic
hypoglycaemia [58]. Finally, in vivo perfusion of the VMH of
rodents with glibenclamide (a KATP channel blocker) suppresses
[59], whereas diazoxide (a KATP channel opener) amplifies
[60], hormonal counter-regulatory responses to acute hypoglycaemia. However, KATP channels are present on many different
neurons and so while they may be required for glucose sensing
they are unlikely to have a regulatory role.
In the pancreatic B-cell, GK is the critical regulator of
glycolytic production of ATP and KATP channel activity [49].
The pancreatic form of GK is also present in areas of the brain
involved in glucose sensing [61– 63]. GK mRNA is expressed
in ~70% of GE neurons [54], and glucokinase has also been
shown to play a regulatory role in GE neuron-sensing ability
[54,57,61,64]. In addition, the selective down-regulation of
GK in GE neurons in primary VMH neuronal cultures led to
the loss of all demonstrable GE and GI neurons [65]. However,
extracellular levels of glucose are only about 10 –30% of the
levels found in blood. Microdialysis studies in rats [33,34,66]
and human subjects [67] have shown that the ECF glucose
levels to which neurons are exposed are in the range of 1–
2 mM, and fall to a similar degree during acute hypoglycaemia
(~0.5 mM) [33]. Both euglycaemic and hypoglycaemia ECF
glucose levels are beneath the range of glucose levels in which
GK usually acts in a regulatory manner, thus for GK to
perform this role in glucose-sensing neurons it almost certainly
has to operate within a distinct subcellular compartment.
Glucose signalling in the pancreatic B-cell also requires the
GLUT Type 2 (GLUT-2). GLUT-2 mRNA has been found in
brain glucose-sensing regions [54,68]; in transgenic mice,
central GLUT-2 has been shown to be involved in the counterregulatory response to hypoglycaemia. Intriguingly, reintroducing GLUT-2 to glial cells, but not to neurons, restored the
defective counter-regulatory response to hypoglycaemia [69].
Taken together, these studies provide good evidence that the
glucose-sensing mechanism used by the GE neuron has many
similarities with the pancreatic β-cell. In particular, GK and
515
dme_2376.fm Page 516 Wednesday, April 9, 2008 8:01 PM
DIABETICMedicine
Glucose sensing during hypoglycaemia • R. McCrimmon
FIGURE 2 Hypothetical sensing mechanism of
GE neurons. Glucose enters the GE neuron
through GLUT 2 or 3, and is phosphorylated by
GK, acting as the gatekeeper, and regulating the
production of cytosolic ATP in a subcellular
compartment. The ATP closes KATP channels in
the plasma membrane, causing depolarization.
In turn, this leads to Ca2+ influx through
VDCCs, stimulating neurotransmitter release
and/or increased APF. Lactate, produced locally
by astrocytes or arriving systemically, enters the
neuron via MCT2 and can then be metabolized
to form ATP.
FIGURE 3 Hypothetical sensing mechanism of
GI neurons. In GI neurons, GK may once again
act as the gatekeeper. A falling glucose results in
an increase in the AMP : ATP ratio, an effect
that can be mimicked pharmacologically by
AICAR. AMPK, once activated, stimulates the
formation of NOS, which may diffuse out to
adjacent neurons or glial cells. In addition,
AMPK may act on chloride channels leading to
neuronal depolarization and neurotransmitter
release and/or increased APF. This action of
AMPK can be blocked pharmacologically by
compound C.
the KATP channel appear to be key points through which an
increase in the ambient glucose leads to altered firing of GE
neurons (Fig. 2).
GI neurons
In contrast, GI neurons show a decrease in activity as glucose
levels rise [47]. It is perhaps easier to think of GI neurons as
those glucose-sensing neurons that become more active when
glucose levels fall, and as such they may use signalling mechanisms more relevant to the pancreatic A-cell. Unfortunately,
like the A-cell, the signalling pathways used by the GI neuron
are not well understood (Fig. 3). This may reflect the fact that
they are few in number, comprising only 3–14% of neurons in
the ventromedial hypothalamic nucleus [61,70]. However,
recent evidence suggests that GI neurons may be more prevalent:
when improved slice techniques are used in conjunction with
novel methods for identifying GI neurons (e.g. membrane
potential dyes), as many as ~30– 40% of all neurons in the
VMH are reported to be GI neurons [71]. GK mRNA is
expressed in around 40% of GI neurons, and may also serve a
regulatory role in these neurons. [65].
More recently, evidence has also emerged that AMP-activated
protein kinase (AMPK) plays a key role in the sensing pathway
used by GI neurons (Fig. 3). AMPK has been described as an
intracellular ‘fuel gauge’ in that it is activated in response to a
rise in the intracellular ratio of AMP to ATP and acts to switch
off energy-consuming anabolic processes and switch on energyproducing catabolic processes [72]. Within the brain, AMPK
516
expression is thought to be predominantly neuronal in distribution, with very little expression evident in astrocytes [73].
Canabal et al. [71] demonstrated that in VMN GI neurons
exposed to 2.5 mM glucose, AICAR (an activator of AMPK),
mimicked the excitatory effect of low glucose (0.5 mM) on
action potential frequency. Both low glucose and AICAR
were shown to mediate their effects, in part, through an increase
in nitric oxide (NO) production in GI neurons. Conversely,
increased NO production in response to a low glucose was
blocked by compound C, an inhibitor of AMPK [71]. In a
related series of studies, Mountjoy et al. [74] reported that
activation of AMPK with AICAR or inhibition with compound C altered neuronal activity in GI, but not GE, neurons
in an ex vivo hypothalamic cell culture system obtained from
the mediobasal (including VMN and Arc) hypothalamus. In
this study, AMPK had no effect on the KATP channel [74], but
others have suggested that AMPK may instead act on a chloride channel to depolarize the plasma membrane [75]. In
rodent studies, in vivo pharmacological activation of AMPK in
the VMH during acute hypoglycaemia amplifies the glucose
counter-regulatory response in normal Sprague-Dawley rats
[76], and restores the hormonal counter-regulatory response
to hypoglycaemia in rats with defective counter-regulation
[77]. Conversely, down-regulation of AMPK in the VMH using
specific RNA interference suppresses the counter-regulatory
response to acute hypoglycaemia [78]. In addition, mice that
selectively lack the alpha-catalytic subunit of AMPK in
POMC or AgRP neurons (both neuronal types are located in
the medio-basal hypothalamus and are involved in feeding
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
dme_2376.fm Page 517 Wednesday, April 9, 2008 8:01 PM
Review article
behaviour and energy homeostasis) lose the responsiveness
of these neurons to changes in extracellular glucose [79].
What these studies all indicate is that the brain contains
specialized neurons that exist in distinct locations and are able
to monitor and react to alterations in the glucose concentration
to which they are exposed. These neurons, by virtue of specific
sensing systems, translate the rate or quantity of glucose
oxidation into a neural signal that alters neuronal firing rates
and, in the case of hypoglycaemia, leads to the stimulation of
a systemic glucose counter-regulatory defence response. The
glucose-sensing neurons seem to use signalling mechanisms
that are very similar to those used by pancreatic B- and A-cells.
The GE neuron, which seems to operate more under conditions
of eu- or hyperglycaemia, may use glucokinase as its key
regulatory step, and the KATP channel to then translate that
signal into altered neuronal firing rates. In contrast, while the
GI neuron (which operates more under hypoglycaemic
conditions) may still use glucokinase as a regulatory enzyme,
it appears more dependent on alterations in intracellular
AMPK activity that, in turn, may act via a chloride channel to
alter neuronal firing rates. It is highly likely that these two
neuronal populations communicate with each other, perhaps
via the inhibitory neurotransmitter GABA [80], to co-ordinate
their responses to a changing blood glucose. Potentially, the
counterbalance between GI and GE neuronal activity forms
the most sensitive means of regulating and maintaining blood
glucose within a narrow physiological range and ensuring an
adequate supply of glucose to the brain.
Counter-regulation in T1DM
In health, a fall in plasma glucose level is rapidly detected and
a sequence of counter-regulatory responses triggered, which
mainly involve: (1) suppression of insulin secretion; (2) counterregulatory hormone release, which rapidly promotes
endogenous glucose production and limits peripheral glucose
utilization; and (3) subjective awareness of hypoglycaemia. In
T1DM, these compensatory systems are disrupted at every
level. Firstly, for most individuals, insulin delivery from its
subcutaneous depot is unregulated and continues despite the
development of hypoglycaemia. Secondly, most individuals
with T1DM will over time develop defects in the hormonal
counter-regulatory defence against hypoglycaemia. Within
5 years of diagnosis of T1DM, hypoglycaemia fails to stimulate
the release of glucagon, the major counter-regulatory hormone
(Table 1) [81]. As a result, individuals with T1DM are
particularly dependent on the sympathoadrenal (primarily
adrenaline and noradrenaline) response to low blood glucose.
However, within 10 years of diagnosis, the majority of
patients develop additional impairments in sympathoadrenal
and other neurohormonal responses against hypoglycaemia
(Table 1) [81]. In addition, symptom awareness becomes
impaired in individuals with T1DM. The term hypoglycaemiaassociated autonomic failure (HAAF) was introduced by
Cryer to incorporate both defective glucose counter-regulation
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
DIABETICMedicine
Table 1 Percentage of individuals with T1DM showing deficient
responses in each of the major hypoglycaemic counter-regulatory
hormones over time. From Mokan et al. [81]
Duration
of diabetes
Glucagon
(%)
Epinephrine
(%)
Cortisol
(%)
GH
(%)
< 1 year
1–5 years
5–10 years
> 10 years
27
75
100
92
9
25
44
66
0
0
11
25
0
0
11
25
and hypoglycaemia unawareness [1]. The presence of HAAF
greatly increases the risk of suffering severe hypoglycaemia in
patients with T1DM [82].
The major factor leading to the loss of glucagon secretion in
response to acute hypoglycaemia in T1DM appears to be the
loss of insulin secretion from the B-cell. This is because the
glucagon-secretory defect is selective (responses to other stimuli
such as arginine remain intact), and develops concomitantly
with progressive B-cell loss in T1DM [83]. Based on earlier rat
anatomical studies suggesting that blood flow in the islet was
predominantly from B- to A-cell, it was proposed that insulin
under ordinary conditions tonically inhibited glucagon secretion
by the pancreatic A-cell [84 –86]. Hence, the ‘switch-off’
hypothesis posited that during hypoglycaemia when insulin
secretion is suppressed, glucagon is released from the A-cell. In
T1DM, pancreatic B-cells are destroyed in the autoimmune
inflammatory process such that this ‘switch-off’ of insulin no
longer occurs. In fact, intra-islet insulin levels may actually
rise, leading to the suppression of glucagon secretion [84 – 86].
There is good evidence in support of this hypothesis, from both
in vitro [85–89] and in vivo [89] rodent studies. However, the
pancreatic α-cell has an extensive and complex autonomic
innervation (reviewed in detail in Taborsky et al. [90]) with
considerable redundancy, so while insulin may be the primary
factor responsible for this defect, an additional role for
alterations in the autonomic nervous system cannot be excluded.
The almost universal defect in glucagon secretion means that
individuals with T1DM are very reliant on the sympathoadrenal
response to hypoglycaemia to restore blood-glucose levels.
Unfortunately, this response also appears to become defective
over time in T1DM [81]. The major reason for the development
of this defect is thought to be the experience of hypoglycaemia
itself. In the Diabetes Control and Complications Trial, which
compared intensive with standard insulin therapy in T1DM,
the major single factor predicting severe hypoglycaemia in
the study cohort was reporting a previous episode of severe
hypoglycaemia [91]. Intensive insulin therapy, which is
associated with more frequent hypoglycaemia, was shown to
lower the glucose level at which hormonal counter-regulation
was initiated in T1DM [92]. Hypoglycaemia per se is the likely
cause of this and it has been shown in both humans [93 –95]
and rodents [77,96 –97] that single or multiple episodes of
517
dme_2376.fm Page 518 Wednesday, April 9, 2008 8:01 PM
DIABETICMedicine
antecedent hypoglycaemia lead to suppressed adrenaline
responses to a subsequent episode of hypoglycaemia.
Data gleaned from studies in rodents suggest that this defect
arises through changes in key brain glucose-sensing regions
such as the VMH. Recurrent hypoglycaemia has been shown
to markedly suppress the counter-regulatory response to 2deoxyglucose (a non-metabolized form of glucose) perfused
into the VMH of rats [98]. It has also been shown that the
glucose level at which you first see activation of glucoseinhibited neurons in the VMH is lower in rats that have experienced prior recurrent hypoglycaemia [99]. Moreover, in vivo
activation of AMPK in the VMH of rats who have experienced
prior recurrent hypoglycaemia can restore hormonal counterregulatory responses to a subsequent hypoglycaemic challenge
[77]. Similarly, opening of KATP channels in the VMH of recurrently hypoglycaemic rats leads to the restoration of normal
counter-regulatory responses to subsequent hypoglycaemia. If
our model is correct, these studies suggest that the balance
between GE and GI neuronal activity is altered by recurrent
hypoglycaemia. We would anticipate that GE neurons (acting
to suppress counter-regulation) are more likely to be active
and/or that GI neurons (acting to amplify counter-regulation)
are less likely to be active following recurrent hypoglycaemia.
The net effect is that full glucose counter-regulation is initiated at a lower glucose level. This gives individuals less time to
react to, and seek treatment for, hypoglycaemia.
Why does this mechanism fail in Type 1
diabetes?
The question we then need to ask is: why does this happen?
The most obvious explanation is that following recurrent
hypoglycaemia, glucose-sensing neurons are ‘seeing’ higher
glucose and/or ATP levels during a subsequent hypoglycaemic
challenge. This change may arise through an increased supply
of fuel through altered glucose [100], or alternate fuel transport
[101]. However, while these changes suggest an increased
capacity to transport fuels across the blood−brain barrier and
into glucose-sensing neurons, the data from human studies have
been mixed, with some indicating increased whole-brain glucose
[102], or acetate [103], uptake after chronic hypoglycaemia
and others showing no change [104].
Another possibility is that the brain is able to obtain
additional metabolic substrates from more local sources.
Recently, it has emerged that brain glycogen may act as a fuel
reserve during acute hypoglycaemia [105]. Moreover, brain
glycogen levels may actually increase in response to an acute
episode of hypoglycaemia [105,106]. This ‘super-compensation’
of brain glycogen may then provide the additional fuel reserve
that leads to defective glucose sensing during a subsequent
episode of hypoglycaemia. This theory is attractive, but
against it is the fact that brain glycogen levels appear to be very
low and a fraction of the levels seen in other tissues such as
muscle and liver. Also, ‘super-compensation’ of glycogen may
actually have arisen because of the model employed in these
518
Glucose sensing during hypoglycaemia • R. McCrimmon
studies where pre-study infusions of glucose and insulin, rather
than hypoglycaemia per se, may have led to the described
phenomenon. Further research is needed in this area.
It has also emerged that mechanisms for regulating the
magnitude of the neuroendocrine response to acute hypoglycaemia exist within the brain. The CRH family of ligands and
receptors form an ancient and highly conserved means of
regulating the neuroendocrine stress response. Within the
brain, at key sites involved with autonomic activation, CRH
acting through the CRH-R1 receptor appears to lead to an
amplification of the autonomic response to stress whereas
Urocortin (part of the CRH family of peptides), acting through
the CRH-R2 receptor, suppresses the autonomic response to
stress. We were recently able to show that such a mechanism
operated in the VMH, with urocortin locally delivered to the
VMH markedly suppressing the counter-regulatory response
to acute hypoglycaemia [107], whereas CRH amplified the
response [108]. Moreover, unpublished work from our
laboratory would suggest that this counterbalanced system
might change following recurrent hypoglycaemia, with an
upregulation of CRH-R2-mediated suppressive effects in the
VMH. The net effect would be to suppress the counterregulatory response to subsequent hypoglycaemia.
We should not be surprised that there is no single answer
to the phenomenon of HAAF. Hypoglycaemia represents a
profound physiological stress and is likely to activate numerous
physiological defence systems that affect both biological and
behavioural responses. Our understanding of these physiological
defences remains limited, but it is reasonable to conclude that
recurrent hypoglycaemia may lead to an increase in the capacity
of glucose-sensing regions of the brain to use glucose and/or
alternate fuels, as well as to a change in the mechanisms that
fine-tune the hypoglycaemic stress response. The net effect of
these changes is to suppress the glucose counter-regulatory
response to a subsequent episode of hypoglycaemia.
What relevance are studies in rodents to
human disease?
The ultimate aim of all basic research is its translation into the
human model. In hypoglycaemia research, electrophysiological
studies of hypothalamic slice preparations and rodent models
offer much, but one must always be wary of over-interpreting
data. The limitations of cell-based and rodent studies are well
recognized. However, this does not make the work irrelevant
to human disease. Glucose homeostasis, in particular, is
fundamental to the survival of a vast range of species from
single-cell organisms to humans. AMPK, for instance, is a
critical part of the process whereby the yeast cell responds to a
period of fuel deprivation, and likewise in rodents clearly plays
a major role in glucose sensing during hypoglycaemia. Thus, it
appears likely that such ancient and conserved systems may be
important in human disease. Another example is glucokinase,
which, as reviewed earlier, has been shown in rodents to play
a regulatory role in glucose sensing in hypothalamic neurons.
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
dme_2376.fm Page 519 Wednesday, April 9, 2008 8:01 PM
Review article
This finding led to human studies where fructose, a modulator
of glucokinase activity, was delivered systemically during
hypoglycaemia and was shown to amplify the counter-regulatory
response in both non-diabetic [109] and T1DM [110] subjects.
Finally, the recognition that recurrent hypoglycaemia might
lead to adaptive changes that increase the ability of the brain
to use alternate fuels has prompted trials in T1DM subjects
using medium-chain fatty acids that may provide neuroprotection, particularly during the night, while not leading to a
deterioration in overall glucose control.
In summary, our understanding of where and how glucose
is sensed has increased significantly in the last 20 years. We
now recognize that a network of specialized neurons exist that
continually monitor the ambient glucose level and that these
neurons act in such a way as to try to maintain glucose homeostasis. These neurons appear to use sensing mechanisms very
similar to those used within the pancreatic islet, with GK, the
K ATP channel and AMPK all playing key roles in central
glucose sensing. While this information has come largely
from rodent studies using a combination of in vitro and in vivo
techniques, it is anticipated that newer imaging techniques
such as positron emission tomography and magnetic resonance
spectroscopy will allow the translation of these findings into
the human model and determine their relevance to human
disease. The development of novel therapies or treatment
strategies designed to minimize the risk of hypoglycaemia
in insulin-treated individuals with diabetes relies on the
information that these studies will provide.
Competing interests
None to declare.
Acknowledgements
The author acknowledges the post-doctoral fellows and technical
staff who contributed greatly to the research that underpins
this review. The research work of the author is supported by
a research grant from NIDDK (69831), and a Career Development Award from the Juvenile Diabetes Research Foundation.
References
1 Cryer PE. Banting Lecture. Hypoglycemia: the limiting factor in the
management of IDDM. Diabetes 1994; 43: 1378–1389.
2 Frier BM, Fisher BM. Hypoglycaemia and Diabetes: Clinical and
Physiological Aspects. London: Edward Arnold, 1993.
3 Thorens B, Larsen PJ. Gut-derived signaling molecules and vagal
afferents in the control of glucose and energy homeostasis. Curr
Opin Clin Nutr Metab Care 2004; 7: 471–478.
4 Burcelin R, Crivelli V, Perrin C, Da Costa A, Mu J, Kahn BB et al.
GLUT4, AMP kinase, but not the insulin receptor, are required for
hepatoportal glucose sensor-stimulated muscle glucose utilization.
J Clin Invest 2003; 111: 1555–1562.
5 Burcelin R, Da Costa A, Drucker D, Thorens B. Glucose competence
of the hepatoportal vein sensor requires the presence of an activated
glucagon-like peptide-1 receptor. Diabetes 2001; 50: 1720–1728.
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
DIABETICMedicine
6 Burcelin R, Dolci W, Thorens B. Glucose sensing by the hepatoportal
sensor is GLUT2-dependent: in vivo analysis in GLUT2-null mice.
Diabetes 2000; 49: 1643–1648.
7 Donovan CM. Portal vein glucose sensing. Diabetes Nutr Metab
2002; 15: 308–312.
8 Donovan CM, Hamilton-Wessler M, Halter JB, Bergman RN.
Primacy of liver glucosensors in the sympathetic response to
progressive hypoglycemia. Proc Natl Acad Sci USA 1994; 91:
2863–2867.
9 Hevener AL, Bergman RN, Donovan CM. Novel glucosensor for
hypoglycemic detection localized to the portal vein. Diabetes 1997;
46: 1521–1525.
10 Pardal R, Lopez-Barneo J. Low glucose-sensing cells in the carotid
body. Nat Neurosci 2002; 5: 197–198.
11 Shoji S. Glucose regulation of synaptic transmission in the dorsolateral
septal nucleus of the rat. Synapse 1992; 12: 322–332.
12 Nakano Y, Oomura Y, Lenard L, Nishino H, Aou S, Yamamoto T
et al. Feeding-related activity of glucose-and morphine-sensitive
neurons in the monkey amygdala. Brain Res 1986; 399: 167–
172.
13 Lee K, Dixon AK, Freeman TC, Richardson PJ. Identification of an
ATP-sensitive potassium channel current in rat striatal cholinergic
interneurones. J Physiol 1998; 510: 441–453.
14 Lee K, Dixon AK, Rowe IC, Ashford ML, Richardson PJ. The highaffinity sulphonylurea receptor regulates KATP channels in nerve
terminals of the rat motor cortex. J Neurochem 1996; 66: 2562–
2571.
15 Adachi A, Kobashi M, Miyoshi N, Tsukamoto G. Chemosensitive
neurons in the area postrema of the rat and their possible functions.
Brain Res Bull 1991; 26: 137–140.
16 Amoroso S, Schmid-Antomarchi H, Fosset M, Lazdunski M.
Glucose, sulfonylureas, and neurotransmitter release: role of ATPsensitive K+ channels. Science 1990; 247: 852–854.
17 During MJ, Leone P, Davis KE, Kerr D, Sherwin RS. Glucose
modulates rat substantia nigra GABA release in vivo via ATP-sensitive
potassium channels. J Clin Invest 1995; 95: 2403–2408.
18 Finta EP, Harms L, Sevcik J, Fischer HD, Illes P. Effects of potassium
channel openers and their antagonists on rat locus coeruleus
neurones. Br J Pharmacol 1993; 109: 308–315.
19 Karschin A, Brockhaus J, Ballanyi K. KATP channel formation by
the sulphonylurea receptors SUR1 with Kir6.2 subunits in rat dorsal
vagal neurons in situ. J Physiol 1998; 509: 339–346.
20 Funahashi H, Yada T, Muroya S, Takigawa M, Ryushi T, Horie S
et al. The effect of leptin on feeding-regulating neurons in the rat
hypothalamus. Neurosci Lett 1999; 264: 117–120.
21 Nagai K, Niijima A, Nagai N, Hibino H, Chun SJ, Shimizu K et al.
Bilateral lesions of the hypothalamic suprachiasmatic nucleus
eliminated sympathetic response to intracranial injection of 2deoxy-D-glucose and VIP rescued this response. Brain Res Bull
1996; 39: 293–297.
22 Oomura Y, Ono T, Ooyama H, Wayner MJ. Glucose and osmosensitive neurones of the rat hypothalamus. Nature 1969; 222:
282–284.
23 Orsini JC, Himmi T, Wiser AK, Perrin J. Local versus indirect action
of glucose on the lateral hypothalamic neurons sensitive to glycemic
level. Brain Res Bull 1990; 25: 49–53.
24 Orsini JC, Wiser AK, Himmi T, Boyer A, Perrin J. Sensitivity of
lateral hypothalamic neurons to glycemic level: possible involvement
of an indirect adrenergic mechanism. Brain Res Bull 1991; 26: 473–
478.
25 Silver IA, Ereciska M. Glucose-induced intracellular ion changes in
sugar-sensitive hypothalamic neurons. J Neurophysiol 1998; 79:
1733–1745.
26 Ritter S, Dinh TT, Zhang Y. Localization of hindbrain glucoreceptive sites controlling food intake and blood glucose. Brain Res 2000;
856: 37–47.
519
dme_2376.fm Page 520 Wednesday, April 9, 2008 8:01 PM
DIABETICMedicine
27 Ashford ML, Boden PR, Treherne JM. Tolbutamide excites rat
glucoreceptive ventromedial hypothalamic neurones by indirect
inhibition of ATP-K+ channels. Br J Pharmacol 1990; 101: 531–
540.
28 Borg MA, Sherwin RS, Borg WP, Tamborlane WV, Shulman GI.
Local ventromedial hypothalamus glucose perfusion blocks
counterregulation during systemic hypoglycemia in awake rats.
J Clin Invest 1997; 99: 361–365.
29 Borg WP, During MJ, Sherwin RS, Borg MA, Brines ML, Shulman GI.
Ventromedial hypothalamic lesions in rats suppress counterregulatory responses to hypoglycemia. J Clin Invest 1994; 93: 1677–
1682.
30 Borg WP, Sherwin RS, During MJ, Borg MA, Shulman GI. Local
ventromedial hypothalamus glucopenia triggers counterregulatory
hormone release. Diabetes 1995; 44: 180–184.
31 Evans SB, Wilkinson CW, Gronbeck P, Bennett JL, Taborsky GJ Jr,
Figlewicz DP. Inactivation of the PVN during hypoglycemia partially
simulates hypoglycemia-associated autonomic failure. Am J PhysiolRegul Integr Comp Physiol 2003; 284: R57–65.
32 Evans SB, Wilkinson CW, Gronbeck P, Bennett JL, Zavosh A,
Taborsky GJ Jr et al. Inactivation of the DMH selectively inhibits
the ACTH and corticosterone responses to hypoglycemia. Am J
Physiol-Regul Integr Comp Physiol 2004; 286: R123–128.
33 de Vries MG, Arseneau LM, Lawson ME, Beverly JL. Extracellular
glucose in rat ventromedial hypothalamus during acute and recurrent
hypoglycemia. Diabetes 2003; 52: 2767–2773.
34 McNay EC, Gold PE. Extracellular glucose concentrations in the rat
hippocampus measured by zero-net-flux: effects of microdialysis
flow rate, strain, and age. J Neurochem 1999; 72: 785–790.
35 Silver IA, Erecinska M. Extracellular glucose concentration in
mammalian brain: continuous monitoring of changes during
increased neuronal activity and upon limitation in oxygen supply in
normo-, hypo-, and hyperglycemic animals. J Neurosci 1994; 14:
5068–5076.
36 Mayer J. Glucostatic mechanism of regulation of food intake. New
Engl J Med 1953; 249: 13–16.
37 Magistretti PJ, Sorg O, Naichen Y, Pellerin L, de Rham S, Martin JL.
Regulation of astrocyte energy metabolism by neurotransmitters.
Renal Physiol Biochem 1994; 17: 168–171.
38 Magistretti PJ, Sorg O, Yu N, Martin JL, Pellerin L. Neurotransmitters
regulate energy metabolism in astrocytes: implications for the
metabolic trafficking between neural cells. Dev Neurosci 1993; 15:
306–312.
39 Pellerin L, Magistretti PJ. Glutamate uptake into astrocytes
stimulates aerobic glycolysis: a mechanism coupling neuronal activity
to glucose utilization. Proc Natl Acad Sci USA 1994; 91: 10625–
10629.
40 Song Z, Routh VH. Differential effects of glucose and lactate on
glucosensing neurons in the ventromedial hypothalamic nucleus.
Diabetes 2005; 54: 15–22.
41 Borg MA, Tamborlane WV, Shulman GI, Sherwin RS. Local lactate
perfusion of the ventromedial hypothalamus suppresses hypoglycemic
counterregulation. Diabetes 2003; 52: 663–666.
42 King P, Kong MF, Parkin H, MacDonald IA, Barber C, Tattersall RB.
Intravenous lactate prevents cerebral dysfunction during hypoglycaemia in insulin-dependent diabetes mellitus. Clin Sci 1998; 94:
157–163.
43 Maran A, Cranston I, Lomas J, Macdonald I, Amiel SA. Protection
by lactate of cerebral function during hypoglycaemia. Lancet 1994;
343: 16–20.
44 Thurston JH, Hauhart RE. Effect of momentary stress on brain
energy metabolism in weanling mice: apparent use of lactate as
cerebral metabolic fuel concomitant with a decrease in brain glucose
utilization. Metab Brain Dis 1989; 4: 177–186.
45 Thurston JH, Hauhart RE, Schiro JA. Lactate reverses insulininduced hypoglycemic stupor in suckling-weanling mice: biochemical
520
Glucose sensing during hypoglycaemia • R. McCrimmon
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
61
62
63
64
correlates in blood, liver, and brain. J Cereb Blood Flow Metab
1983; 3: 498–506.
Kennedy HJ, Pouli AE, Ainscow EK, Jouaville LS, Rizzuto R,
Rutter GA. Glucose generates sub-plasma membrane ATP microdomains in single islet beta-cells. Potential role for strategically
located mitochondria. J Biol Chem 1999; 274: 13281–13291.
Routh VH. Glucose-sensing neurons: are they physiologically
relevant? Physiol Behav 2002; 76: 403–413.
Schuit FC, Huypens P, Heimberg H, Pipeleers DG. Glucose sensing
in pancreatic beta-cells: a model for the study of other glucoseregulated cells in gut, pancreas, and hypothalamus. Diabetes 2001;
50: 1–11.
Matschinsky FM. Banting Lecture 1995. A lesson in metabolic
regulation inspired by the glucokinase glucose sensor paradigm.
Diabetes 1996; 45: 223–241.
Levin BE, Routh VH, Kang L, Sanders NM, Dunn-Meynell AA.
Neuronal glucosensing: what do we know after 50 years? Diabetes
2004; 53: 2521–2528.
Ashford ML, Boden PR, Treherne JM. Glucose-induced excitation
of hypothalamic neurones is mediated by ATP-sensitive K+ channels.
Pflugers Archiv-Eur J Physiol 1990; 415: 479–483.
Dunn-Meynell AA, Rawson NE, Levin BE. Distribution and phenotype of neurons containing the ATP-sensitive K+ channel in rat
brain. Brain Res 1998; 814: 41–54.
Dunn-Meynell AA, Routh VH, McArdle JJ, Levin BE. Low-affinity
sulfonylurea binding sites reside on neuronal cell bodies in the brain.
Brain Res 1997; 745: 1–9.
Kang L, Routh VH, Kuzhikandathil EV, Gaspers LD, Levin BE.
Physiological and molecular characteristics of rat hypothalamic
ventromedial nucleus glucosensing neurons. Diabetes 2004; 53:
549–559.
Dallaporta M, Perrin J, Orsini JC. Involvement of adenosine
triphosphate-sensitive K+ channels in glucose-sensing in the rat
solitary tract nucleus. Neurosci Lett 2000; 278: 77–80.
Spanswick D, Smith MA, Groppi VE, Logan SD, Ashford ML.
Leptin inhibits hypothalamic neurons by activation of ATP-sensitive
potassium channels. Nature 1997; 390: 521–525.
Yang XJ, Kow LM, Funabashi T, Mobbs CV. Hypothalamic
glucose sensor. Similarities to and differences from pancreatic
beta-cell mechanisms. Diabetes 1999; 48: 1763–1772.
Miki T, Liss B, Minami K, Shiuchi T, Saraya A, Kashima Y et al.
ATP-sensitive K+ channels in the hypothalamus are essential for the
maintenance of glucose homeostasis. Nat Neurosci 2001; 4: 507–512.
Evans ML, McCrimmon RJ, Flanagan DE, Keshavarz T, Fan X,
McNay EC et al. Hypothalamic ATP-sensitive K+ channels play a
key role in sensing hypoglycemia and triggering counterregulatory
epinephrine and glucagon responses. Diabetes 2004; 53: 2542–2551.
McCrimmon RJ, Evans ML, Fan X, McNay EC, Chan O, Ding Y
et al. Activation of KATP channels in the ventromedial hypothalamus amplifies counterregulatory hormone responses to hypoglycemia in normal and recurrently hypoglycemic rats. Diabetes
2005; 54: 3169–3174.
Dunn-Meynell AA, Routh VH, Kang L, Gaspers L, Levin BE.
Glucokinase is the likely mediator of glucosensing in both glucoseexcited and glucose-inhibited central neurons. Diabetes 2002; 51:
2056–2065.
Jetton TL, Liang Y, Pettepher CC, Zimmerman EC, Cox FG,
Horvath K et al. Analysis of upstream glucokinase promoter activity
in transgenic mice and identification of glucokinase in rare neuroendocrine cells in the brain and gut. J Biol Chem 1994; 269: 3641–
3654.
Lynch RM, Tompkins LS, Brooks HL, Dunn-Meynell AA, Levin
BE. Localization of glucokinase gene expression in the rat brain.
Diabetes 2000; 49: 693–700.
Wang R, Liu X, Hentges ST, Dunn-Meynell AA, Levin BE, Wang W
et al. The regulation of glucose-excited neurons in the hypothalamic
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
dme_2376.fm Page 521 Wednesday, April 9, 2008 8:01 PM
Review article
65
66
67
68
69
70
71
72
73
74
75
76
77
78
79
80
arcuate nucleus by glucose and feeding-relevant peptides. Diabetes
2004; 53: 1959–1965.
Kang L, Dunn-Meynell AA, Routh VH, Gaspers LD, Nagata Y,
Nishimura T et al. Glucokinase is a critical regulator of ventromedial
hypothalamic neuronal glucosensing. Diabetes 2006; 55: 412–
420.
McNay EC, Gold PE. Age-related differences in hippocampal extracellular fluid glucose concentration during behavioral testing and
following systemic glucose administration. J Gerontol Series A-Biol
Sci Med Sci 2001; 56: B66–71.
Abi-Saab WM, Maggs DG, Jones T, Jacob R, Srihari V, Thompson
J et al. Striking differences in glucose and lactate levels between
brain extracellular fluid and plasma in conscious human subjects:
effects of hyperglycemia and hypoglycemia. J Cereb Blood Flow
Metab 2002; 22: 271–279.
Leloup C, Arluison M, Lepetit N, Cartier N, Marfaing-Jallat P,
Ferre P et al. Glucose transporter 2 (GLUT 2): expression in specific
brain nuclei. Brain Res 1994; 638: 221–226.
Marty N, Dallaporta M, Foretz M, Emery M, Tarussio D, Bady I
et al. Regulation of glucagon secretion by glucose transporter Type
2 (glut2) and astrocyte-dependent glucose sensors. J Clin Invest
2005; 115: 3545–3553.
Song Z, Levin BE, McArdle JJ, Bakhos N, Routh VH. Convergence
of pre- and postsynaptic influences on glucosensing neurons in
the ventromedial hypothalamic nucleus. Diabetes 2001; 50: 2673–
2681.
Canabal DD, Song Z, Potian JG, Beuve A, McArdle JJ, Routh VH.
Glucose, insulin, and leptin signaling pathways modulate nitric
oxide synthesis in glucose-inhibited neurons in the ventromedial
hypothalamus. Am J Physiol-Regul Integr Comp Physiol 2007;
292: R1418–1428.
Hardie DG, Carling D. The AMP-activated protein kinase. Fuel
gauge of the mammalian cell? Eur J Biochem 1997; 246: 259–273.
Turnley AM, Stapleton D, Mann RJ, Witters LA, Kemp BE,
Bartlett PF. Cellular distribution and developmental expression of
AMP-activated protein kinase isoforms in mouse central nervous
system. J Neurochem 1999; 72: 1707–1716.
Mountjoy PD, Bailey SJ, Rutter GA. Inhibition by glucose or leptin
of hypothalamic neurons expressing neuropeptide Y requires
changes in AMP-activated protein kinase activity. Diabetologia
2007; 50: 168–177.
Fioramonti X, Contie S, Song Z, Routh VH, Lorsignol A, Penicaud
L. Characterization of glucose-sensing neuron subpopulations in
the arcuate nucleus. Diabetes 2007; 56: 1219–1227.
McCrimmon RJ, Fan X, Ding Y, Zhu W, Jacob RJ, Sherwin RS.
Potential role for AMP-activated protein kinase in hypoglycemia
sensing in the ventromedial hypothalamus. Diabetes 2004; 53:
1953–1958.
McCrimmon RJ, Fan X, Cheng H, McNay E, Chan O, Shaw M
et al. Activation of AMP-activated protein kinase within the
ventromedial hypothalamus amplifies counterregulatory hormone
responses in rats with defective counterregulation. Diabetes 2006;
55: 1755–1760.
McCrimmon RJ, Shaw M, Fan X, Cheng H, Ding Y, Wang A
et al. AMP-activated protein kinase (AMPK): a key mediator of
hypoglycemia-sensing in the ventromedial hypothalamus (VMH).
Diabetes 2006; 55 (Suppl. 1): A15.
Claret M, Smith MA, Batterham RL, Selman C, Choudhury AI,
Fryer LGD et al. AMPK is essential for energy homeostasis regulation
and glucose sensing by POMC and AgRP neurons. J Clin Invest
2007; 117: 2325–2336.
Chan O, Zhu W, Ding Y, McCrimmon RJ, Sherwin RS. Blockade
of GABA(A) receptors in the ventromedial hypothalamus further
stimulates glucagon and sympathoadrenal but not the hypothalamopituitary-adrenal response to hypoglycemia. Diabetes 2006; 55:
1080–1087.
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
DIABETICMedicine
81 Mokan M, Mitrakou A, Veneman T, Ryan C, Korytkowski M,
Cryer P et al. Hypoglycemia unawareness in IDDM. Diabetes Care
1994; 17: 1397–1403.
82 White NH, Skor DA, Cryer PE, Levandoski LA, Bier DM, Santiago
JV. Identification of Type 1 diabetic patients at increased risk for
hypoglycemia during intensive insulin therapy. New Engl J Med
1983; 308: 485–491.
83 Fukuda M, Tanaka A, Tahara Y, Ikegami H, Yamamoto Y,
Kumahara Y et al. Correlation between minimal secretory capacity
of pancreatic beta-cells and stability of diabetic control. Diabetes
1988; 37: 81–88.
84 Asplin CM, Paquette TL, Palmer JP. In vivo inhibition of glucagon
secretion by paracrine beta cell activity in man. J Clin Invest 1981;
68: 314–318.
85 Maruyama H, Hisatomi A, Orci L, Grodsky GM, Unger RH.
Insulin within islets is a physiologic glucagon release inhibitor.
J Clin Invest 1984; 74: 2296–2299.
86 Samols E, Stagner JI, Ewart RB, Marks V. The order of islet
microvascular cellular perfusion is B----A----D in the perfused rat
pancreas. J Clin Invest 1988; 82: 350–353.
87 Zhou H, Tran PO, Yang S, Zhang T, LeRoy E, Oseid E et al.
Regulation of alpha-cell function by the beta-cell during hypoglycemia
in Wistar rats: the ‘switch-off’ hypothesis. Diabetes 2004; 53:
1482–1487.
88 Zhou H, Zhang T, Oseid E, Harmon J, Tonooka N, Robertson RP.
Reversal of defective glucagon responses to hypoglycemia in insulindependent autoimmune diabetic BB rats. Endocrinology 2007; 148:
2863–2869.
89 McCrimmon RJ, Evans ML, Jacob RJ, Fan X, Zhu Y, Shulman GI
et al. AICAR and phlorizin reverse the hypoglycemia-specific defect
in glucagon secretion in the diabetic BB rat. Am J Physiol-Endocrinol
Metab 2002; 283: E1076–1083.
90 Taborsky GJ Jr, Ahren B, Havel PJ. Autonomic mediation of
glucagon secretion during hypoglycemia: implications for impaired
alpha-cell responses in Type 1 diabetes. Diabetes 1998; 47: 995–1005.
91 Group TDR. Epidemiology of severe hypoglycemia in the Diabetes
Control and Complications Trial. American Journal of Medicine
1991; 90: 450–459.
92 Amiel SA, Sherwin RS, Simonson DC, Tamborlane WV. Effect of
intensive insulin therapy on glycemic thresholds for counterregulatory
hormone release. Diabetes 1988; 37: 901–907.
93 Davis MR, Shamoon H. Counterregulatory adaptation to recurrent
hypoglycemia in normal humans. J Clin Endocrinol Metab 1991;
73: 995–1001.
94 Davis SN, Galassetti P, Wasserman DH, Tate D. Effects of antecedent
hypoglycemia on subsequent counterregulatory responses to
exercise. Diabetes 2000; 49: 73–81.
95 Heller SR, Cryer PE. Reduced neuroendocrine and symptomatic
responses to subsequent hypoglycemia after 1 episode of hypoglycemia
in nondiabetic humans. Diabetes 1991; 40: 223–226.
96 Flanagan DE, Keshavarz T, Evans ML, Flanagan S, Fan X, Jacob RJ
et al. Role of corticotrophin-releasing hormone in the impairment
of counterregulatory responses to hypoglycemia. Diabetes 2003;
52: 605–613.
97 Powell AM, Sherwin RS, Shulman GI. Impaired hormonal responses
to hypoglycemia in spontaneously diabetic and recurrently hypoglycemic rats. Reversibility and stimulus specificity of the deficits.
J Clin Invest 1993; 92: 2667–2674.
98 Borg MA, Borg WP, Tamborlane WV, Brines ML, Shulman GI,
Sherwin RS. Chronic hypoglycemia and diabetes impair counterregulation induced by localized 2-deoxy-glucose perfusion of the
ventromedial hypothalamus in rats. Diabetes 1999; 48: 584–587.
99 Song Z, Routh VH. Recurrent hypoglycemia reduces the glucose
sensitivity of glucose-inhibited neurons in the ventromedial hypothalamus nucleus. Am J Physiol-Regul Integr Comp Physiol 2006;
291: R1283–1287.
521
dme_2376.fm Page 522 Wednesday, April 9, 2008 8:01 PM
DIABETICMedicine
100 Kumagai AK, Kang YS, Boado RJ, Pardridge WM. Upregulation of
blood−brain barrier GLUT1 glucose transporter protein and
mRNA in experimental chronic hypoglycemia. Diabetes 1995; 44:
1399–1404.
101 Vannucci RC, Nardis EE, Vannucci SJ, Campbell PA. Cerebral
carbohydrate and energy metabolism during hypoglycemia in
newborn dogs. Am J Physiol 1981; 240: R192–199.
102 Boyle PJ, Kempers SF, O’Connor AM, Nagy RJ. Brain glucose
uptake and unawareness of hypoglycemia in patients with
insulin-dependent diabetes mellitus. New Engl J Med 1995; 333:
1726–1731.
103 Mason GF, Petersen KF, Lebon V, Rothman DL, Shulman GI.
Increased brain monocarboxylic acid transport and utilization in
Type 1 diabetes. Diabetes 2006; 55: 929–934.
104 Segel SA, Fanelli CG, Dence CS, Markham J, Videen TO, Paramore DS
et al. Blood-to-brain glucose transport, cerebral glucose metabolism,
and cerebral blood flow are not increased after hypoglycemia.
Diabetes 2001; 50: 1911–1917.
522
Glucose sensing during hypoglycaemia • R. McCrimmon
105 Choi IY, Seaquist ER, Gruetter R. Effect of hypoglycemia on brain
glycogen metabolism in vivo. J Neurosci Res 2003; 72: 25–32.
106 Gruetter R. Glycogen: the forgotten cerebral energy store. J Neurosci
Res 2003; 74: 179–183.
107 McCrimmon RJ, Song Z, Cheng H, McNay EC, Weikart-Yeckel C,
Fan X et al. Corticotrophin-releasing factor receptors within the
ventromedial hypothalamus regulate hypoglycemia-induced
hormonal counterregulation. J Clin Invest 2006; 116: 1723–1730.
108 Cheng H, Zhou L, Zhu W, Wang A, Tang C, Chan O et al. Type 1
corticotrophin releasing factor receptors (CRFR1) in the ventromedial
hypothalamus (VMH) promote hypoglycemia-induced hormonal
counterregulation. Am J Physiol 2007; 293: E705–712.
109 Gabriely I, Hawkins M, Vilcu C, Rossetti L, Shamoon H. Fructose
amplifies counterregulatory responses to hypoglycemia in humans.
Diabetes 2002; 51: 893–900.
110 Gabriely I, Shamoon H. Fructose normalizes specific counterregulatory responses to hypoglycemia in patients with Type 1
diabetes. Diabetes 2005; 54: 609–616.
© 2008 The Author.
Journal compilation © 2008 Blackwell Publishing Ltd. Diabetic Medicine, 25, 513–522
Download