Você está na página 1de 8

The Brain-Gut-Islet Connection

Stephen C. Woods, Stephen C. Benoit, and Deborah J. Clegg

Peptide signals from the pancreatic islets and the gastro- secretion. Many of these peptides also serve as critical
intestinal tract influence the regulation of energy ho- satiety signals to the brain that limit meal size. Islet
meostasis by the brain, and the brain in turn influences the hormones influence digestion and control the disposition
secretions of both the islets and the gut. This article of ingested nutrients in addition to providing key signals to
focuses on how insulin interacts with the brain to influence the brain regarding the level of adiposity and circulating
food intake, blood glucose, and cognitive behavior. Insulin energy. This review focuses on one aspect of this complex
is secreted in response to changes of ambient glucose, and network, i.e., the actions of pancreatic insulin within the
the levels achieved are directly proportional to body adi- brain and the pathologies that occur when insulin signal-
posity. Hence, insulin, like leptin, is an adiposity signal. An
increased insulin signal in the mediobasal hypothalamus ing within the brain is compromised. The topic is timely,
indicates that ample or excess energy is available in the with relevant new information appearing almost monthly,
body and elicits responses that limit food intake and and it is also an area of knowledge where the basic tenets
reduce hepatic glucose secretion. Increased insulin (and are being challenged, since insulin was historically
leptin as well) locally within the brain complements other thought not to interact with the brain and subsequently
signals that indicate a surfeit of energy in the body, was thought to mainly influence the control of food intake
including satiety signals generated by the gut during meals, and body weight.
glucose, and some fatty acids. There is compelling evidence
that overlapping intracellular signaling pathways within
the mediobasal hypothalamus mediate the overall catabolic CONTROL OF ENERGY INTAKE
response to these diverse metabolic signals. Insulin recep- The energy equation holds that for body weight to remain
tors are also densely expressed in the hippocampus, and relatively stable over time, food intake must match energy
insulin acts there to facilitate learning and memory. The expenditure, and deviations in either direction will result
function of insulin receptors in other brain areas is poorly in weight gain or loss. Humans and most mammals con-
understood. Obesity and/or the consumption of diets high
in fat render the brain as well as the body insulin resistant.
sume food in discrete episodes or meals. When there are
In the hypothalamus, this is manifest as a reduced ability of no restrictions on when or how much individuals are
insulin to reduce food intake and body weight, and in the allowed to eat (i.e., when they are in a free-feeding or ad
hippocampus, it is manifest as a reduced ability of insulin lib condition), the impetus to begin a meal is rarely if ever
to improve learning and/or memory. Diabetes 55 (Suppl. caused by a biological deficit or need such as insufficient
2):S114 –S121, 2006 glucose. Rather, evidence indicates that the timing of
meals is based on psychological factors such as habit, time
of day, the social situation, convenience, and others (1–3).
Because of this, body weight regulation, the continuous

T
he brain-gut-islet connection refers to the myriad process that ties energy intake to energy expenditure,
ways in which signals arising in the three arms of must logically be manifest as a control over how many
this important axis interact among themselves, calories are consumed once a meal begins, i.e., on meal
and at least one overall function of this inte- size. Consistent with this, during meals, the gut responds
grated control system is the regulation of energy ho- to ingested nutrients by secreting peptide signals propor-
meostasis throughout the body. Many of the interactions tional to the quantity and quality of calories consumed,
of this triad are well known. The brain regulates activity in and some of these secretions function as satiety signals to
both the gut and the pancreatic islets directly via the the brain to limit meal size (4 –7). The prototypical satiety
autonomic nervous system and indirectly via changes in signal is the duodenal peptide cholecystokinin (CCK).
food intake and energy expenditure. Peptide hormones Humans and animals that are administered CCK just
and other signals secreted from the gut, in addition to before eating consume smaller meals, and when adminis-
coordinating the digestion and absorption of nutrients, tered a selective CCK-1 receptor antagonist, they consume
provide the incretin effect that augments prandial insulin larger meals (5,8). Table 1 lists gut and islet signals
secreted during meals that influence meal size and are
considered to be satiety signals.
From the Department of Psychiatry and the Obesity Research Center, Uni- Adiposity signals are hormones secreted in direct pro-
versity of Cincinnati, Cincinnati, Ohio.
Address correspondence and reprint requests to Stephen C. Woods, Depart- portion to body fat. In contrast to satiety signals that are
ment of Psychiatry, University of Cincinnati, 2170 E. Galbraith Rd., Cincinnati, secreted mainly during meals, adiposity signals are toni-
OH 45237. E-mail: steve.woods@psychiatry.uc.edu. cally present, providing a relatively continuous message to
Received for publication 25 March 2006 and accepted in revised form 27
April 2006.
the brain concerning fat stored within the body. Pancre-
This article is based on a presentation at a symposium. The symposium and atic insulin and the adipocyte hormone leptin are the two
the publication of this article were made possible by an unrestricted educa- best-known adiposity signals, and others such as amylin
tional grant from Servier. and adiponectin also circulate in proportion to body fat
ARC, arcuate nucleus; CCK, cholecystokinin.
DOI: 10.2337/db06-S015 and share many of the same properties. Administering
© 2006 by the American Diabetes Association. either insulin or leptin directly into the brain results in
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked “advertisement” in accordance
reduced food intake and body weight, and reductions of
with 18 U.S.C. Section 1734 solely to indicate this fact. either insulin or leptin signaling locally within the brain
S114 DIABETES, VOL. 55, SUPPLEMENT 2, DECEMBER 2006
S.C. WOODS, S.C. BENOIT, AND D.J. CLEGG

TABLE 1
Gastrointestinal hormones that affect satiety
Gastrointestinal peptide Effect on food intake
Cholecystokinin Decrease
Glucagon-like peptide 1 Decrease
Enterostatin Decrease
Bombesin/gastrin-releasing peptide Decrease
Oxyntomodulin Decrease
Gastric leptin Decrease
Glucagon Decrease
Amylin Decrease
Peptide YY (3-36) Decrease
Apolipoprotein A-IV Decrease
Ghrelin Increase*
*Ghrelin is the only gastrointestinal hormone that increases food
intake.

FIG. 1. Insulin is secreted into the blood from the pancreas in direct
results in overeating and weight gain. When an individual’s proportion to the amount of fat stored in white adipose tissue. As it
circulates through brain capillaries, a small amount of insulin is
weight (i.e., body fat) changes, insulin and leptin secretion transported into the brain, where it acts on insulin receptors on
change in parallel, and this is manifest as an altered neurons with either net catabolic or anabolic activity, for example, in
adiposity signal to the brain. There are many reviews of the ARC of the hypothalamus. These neurons in turn influence energy
homeostasis (food intake and energy expenditure) and ultimately the
these phenomena (9 –14). amount of fat stored in the body by exerting a net catabolic or anabolic
The interaction of adiposity signals with satiety signals action.
in the control of food intake is a topic of considerable
interest and beyond the scope of this review. The basic
principle is that adiposity signals modulate the sensitivity body weight is maintained and defended at a level deter-
of the brain to meal-generated satiety signals (10,12,15). As mined by the dose of insulin administered (rev. in 13,31–
an example, the administration of very low doses of either 33). The response to changes of the insulin signal is
insulin or leptin directly into the brain enhances the bidirectional in that the administration of insulin antibod-
efficacy of systemic CCK to reduce food intake (16 –22), ies into or near the mediobasal hypothalamus causes
and a reduced leptin signal in the brain, which also overeating and weight gain (34,35). Likewise, reducing
reduces brain insulin signaling (23), lowers CCK sensitiv- hypothalamic insulin receptor activity either genetically by
ity (24). The implication is that when an individual loses a neuronal-specific knockout of insulin receptors or phar-
weight and insulin and leptin secretion are reduced, brain macologically via antisense oligonucleotides against the
circuits that control meal size are consequently rendered insulin receptor leads to increased food intake and body
less sensitive to meal-generated satiety signals such as fat (36,37). Although insulin has not been administered
CCK. As a result, more calories than normal must be directly into the brains of humans, certain formulations of
consumed before a satiety signal of sufficient magnitude to insulin have been administered intranasally to humans
terminate a meal is generated. Individuals consequently with a consequent increase of cerebrospinal fluid but not
eat larger than normal meals until body weight returns to plasma insulin. Humans receiving insulin in this way eat
normal. Conversely, when an individual gains excess less food and lose body fat (38).
weight, the increased insulin and leptin signal in the brain Insulin enters the brain via receptor-facilitated transport
causes increased sensitivity to satiety signals, and rela- through capillary endothelial cells. The process is satura-
tively small meals are consumed until the excess weight is ble, selective for insulin, and regulated (39 – 42). Insulin
lost. transport into the brain is reduced during fasting (43), by
maintenance on a high-fat diet (44), and in genetic and
INSULIN AS AN ADIPOSITY SIGNAL dietary-induced obesity (45,46). Because brain insulin de-
Basal plasma insulin is low and increases during meals or rives from plasma insulin, it should be the case that
when stimulated by glucose. Basal, prandial, and stimu- experimentally induced increases of plasma insulin enter
lated insulin levels are all direct functions of stored fat, the brain and result in reduced food intake and body
with leaner individuals having lower levels and more weight. Such procedures are confounded by hypoglycemia
obese individuals having higher levels (25–27). Plasma and a consequent increased tendency to eat more food.
insulin is therefore positioned to convey an important However, when insulin is administered systemically at
signal to the brain indicating the degree of adiposity, and doses sufficiently low to preclude hypoglycemia, food
as discussed below, some insulin enters the brain from the intake is reduced (47). Likewise, when sufficient glucose is
circulation and provides a key negative feedback signal in administered in conjunction with systemic insulin to cir-
the regulation of body fat as originally proposed by cumvent hypoglycemia, food intake is also reduced (48).
Kennedy (28) (Fig. 1). When exogenous insulin is admin- Autoradiographic insulin binding, as well as immunohis-
istered near or directly into the mediobasal hypothalamus, tochemical analyses, reveal that insulin receptors are
animals behave as if they have excess fat, i.e., they eat less selectively located in several brain regions (49 –52). The
and lose weight. The response is dose dependent (17,29), areas of highest concentrations are the olfactory bulb,
occurs in all species assessed, and is not secondary to hypothalamus, cerebellum, cortex, and hippocampus (53–
illness or incapacitation (30). When insulin levels in the 55), and most insulin receptor immunoreactivity occurs on
brain are clamped by means of slow steady local infusions, neurons and not on glia.
DIABETES, VOL. 55, SUPPLEMENT 2, DECEMBER 2006 S115
BRAIN-GUT-ISLET AXIS

INSULIN REGULATES ENERGY HOMEOSTASIS IN THE Kemp, A.D. Strader, S.C.B., S.C.W., M. Mangiarachina, N.
HYPOTHALAMUS Geary, unpublished data), and this may be manifest in the
As discussed above, insulin reduces food intake and body selection of foods (75,76) as well as the preferred strategy
weight when delivered into the third cerebral ventricle used by males and females to defend their body weight
(i3vt) or directly into the hypothalamus in or near the (77). The physiological basis of these sex differences is an
arcuate nucleus (ARC). Conversely, reduction of insulin important clinical issue, since males are far more likely to
signaling has the opposite action. Insulin signaling in the develop symptoms of the metabolic syndrome (70),
ARC also initiates a signal via the vagus nerve to the liver whereas females are far more likely to develop eating
to reduce glucose synthesis and secretion into the blood disorders (78).
(56 –58). Hence, insulin’s action in the hypothalamus is Risk factors for developing type 2 diabetes. Although
consistent with its better-known systemic actions, i.e., its insulin and leptin each signal the degree of adiposity to the
net effect is to lower blood glucose (9,11). Reducing the brain, and whereas each lowers food intake as well as
amount of food consumed and neurally reducing hepatic hepatic glucose secretion, they do so by stimulating dif-
glucose output both complement insulin’s ability to facil- ferent ARC neurons and circuits (79) as well as by altering
itate glucose uptake by muscle, liver, and other tissues, as different hepatic enzyme systems (80). Further, each has
well as complement insulin’s direct action in the liver. It important other functions. Insulin is a major controller of
has recently been reported that administering oleic acid or the levels and utilization of glucose throughout most of the
glucose into or near the ARC also reduces food intake body, including the brain (9,11). Leptin also influences
(57,59 – 62). Leptin may also act there to influence glucose glucose parameters via the brain, although via different
homeostasis (63). Hence, ARC neurons respond to diverse neural circuits than stimulated by insulin (80). Low circu-
signals indicative of a surfeit of available energy (i.e., lating leptin and the resultant decrease of leptin signaling
elevated insulin, fatty acids, or glucose) by reducing the have been hypothesized to regulate many vital systems
ingestion of energy and decreasing the secretion of utiliz- when animals are severely hypocaloric and have low body
able energy (especially glucose) into the blood. Rossetti’s fat (81– 83). The secretion of insulin is adjusted in re-
group has postulated that manipulating intracellular met- sponse to every acute change of metabolism (9,84), with
abolic signaling pathways, especially those involved with levels increasing during meals or when glucose is elevated
the oxidation of lipids, comprises the actual stimulus that for some other reason and decreasing during stress and
is important in controlling both food intake and hepatic exercise. The half-life of insulin in the blood (2–3 min) is
glucose production by the hypothalamus (61,64 – 67). consistent with its role as a minute-to-minute indicator of
ongoing metabolism, and all of its fluctuations are directly
proportional to total body fat (26). Leptin is secreted from
INSULIN AND LEPTIN AS ADIPOSITY SIGNALS adipocytes in direct proportion to the amount of stored fat
As discussed above, both leptin and insulin function as (85) (although the actual stimulus is related more to the
adiposity signals, and the two have overlapping actions with metabolic activity of the fat cell than to actual fat storage
regard to the hypothalamic control of metabolism. While the [86] such that dissociations can occur between stored fat
existence of two or more adiposity signals might seem and leptin release, particularly during a fast [86 – 88]).
redundant, insulin and leptin in fact reflect different fat Nonetheless, under normal conditions and with a half-life
stores, sexes, and risk factors for developing type 2 diabetes, of 45 min, plasma leptin levels are a reliable and relatively
cardiovascular problems, and the metabolic syndrome. stable indicator of body fat. Hence, insulin levels reflect
Fat stores. Insulin is secreted in proportion to visceral the interaction of ongoing metabolic processes and body
fat, whereas leptin reflects total fat mass and especially adiposity, whereas leptin levels reflect the activity of
subcutaneous fat (68,69). This is an important distinction adipose cells more directly.
with regard to the message conveyed to the brain, since As described above, both insulin and leptin act in the
visceral fat carries a greater risk factor for the metabolic ARC (and probably other brain areas) to reduce food
complications associated with obesity than does subcuta- intake and body weight, and both also act in the ARC to
neous fat. Elevated visceral fat carries an increased risk reduce hepatic output of glucose. In some instances, the
for insulin resistance, type 2 diabetes, hypertension, car- overlap of function may result from common intracellular
diovascular disease, and certain cancers (68,70,71). Hence, signaling pathways of insulin and leptin, since both acti-
leptin and insulin each convey specific information to the vate a pathway using insulin receptor substrate 1 and 2,
brain regarding the distribution of fat, and the combina- and both cause increased intracellular cAMP degradation
tion of the two additionally conveys information as to the (89 –91). Insulin upregulates the expression of mRNA of the
total fat mass of the body. long form of the leptin receptor (OB-Rb) in neuronal but not
Sex. Women have relatively more subcutaneous fat and other cell types (92), and insulin facilitates leptin’s ability to
higher plasma leptin, whereas men have relatively more activate the JAK-STAT pathway in the hypothalamus (90,93).
visceral fat and higher plasma insulin (68,70,71). Likewise, Both leptin (94,95) and insulin (94,95) exert their catabolic
male rats have relatively more visceral fat and higher action through phosphatidylinositol 3-kinase.
plasma insulin, whereas female rats have more subcuta-
neous fat and higher plasma leptin (72). We have found
that the brain of females is more sensitive to the catabolic INSULIN AND LEPTINⴕS INTERACTION WITH CENTRAL
action of leptin, whereas the brain of males is more NEUROPEPTIDES
sensitive to the catabolic action of insulin (73), and that Insulin and leptin interact with two populations of ARC
estrogen mediates this difference (72). These data are neurons. Activity of those that synthesize proopiomelano-
consistent with a growing literature documenting sex cortin, the precursor molecule of the melanocortins, is
differences in the actions of many compounds that influ- stimulated by insulin and leptin, whereas activity of those
ence energy homeostasis, including CCK (74), insulin that synthesize neuropeptide Y plus Agouti-related protein
(38,73), leptin (73), and ghrelin (D.J.C., L.M. Brown, C.J. is inhibited by insulin and leptin. ␣-Melanocyte–stimulat-
S116 DIABETES, VOL. 55, SUPPLEMENT 2, DECEMBER 2006
S.C. WOODS, S.C. BENOIT, AND D.J. CLEGG

ing hormone is a melanocortin that is derived from ARC not affect peripheral glucose levels. Further, hyperinsu-
proopiomelanocortin and is an agonist at melanocortin linemic states do not appear to increase Alzheimer’s
receptors (MC3R and MC4R) in several hypothalamic patients’ performance on visual-spatial or physical-coordi-
nuclei; Agouti-related protein is an endogenous antagonist nation tasks. This finding adds support to the hypothesis
of these same melanocortin receptors. Central administra- that insulin levels are specific to cognitive process (e.g.,
tion of insulin or leptin, or the administration of ␣-mela- memory) in Alzheimer’s patients. Finally, patients with
nocyte–stimulating hormone or synthetic agonists for type 2 diabetes who are insensitive to the effects of insulin
melanocortin receptors, or the application of treatments (i.e., who are insulin resistant) are also impaired on some
that reduce ARC neuropeptide Y (e.g., antisense oligonu- measures of cognitive ability (114 –117). These and other
cleotides administered directly into the ARC [96]), all findings encourage the hypothesis that insulin plays an
result in reduced food intake and body weight. Conversely, important role in normal cognitive functions and memorial
the administration of neuropeptide Y, Agouti-related pro- processes. Moreover, it encourages the hypothesis that
tein, or synthetic melanocortin antagonists, or the absence alterations in insulin signaling may play an important role
of endogenous insulin or leptin signaling, all result in a net in neurodegenerative disease.
increase of food intake and body weight (rev. in 11,83,97– In addition to the epidemiological and clinical studies, a
101). growing body of evidence from animal experiments also
implicates insulin as an important factor in learning and
memory (118). Experimental removal of insulin leads to
OTHER ACTIONS OF INSULIN WITHIN THE BRAIN disrupted learning and performance. For example, admin-
It is important to consider what a change in the insulin istration of streptozotocin leads to impaired learning on a
signal at its receptor in diverse brain areas actually signi- number of learning tasks in mice (119). In rats, streptozo-
fies. The obvious answer is that acute changes of insulin tocin-induced diabetes leads to disrupted performance on
reflect an increase (or decrease) in energy in the form of avoidance tasks and in Morris water mazes (109,120 –123).
glucose interacting with the pancreas, whereas sustained Central streptozotocin also produces impairment in work-
changes additionally reflect the amount of fat stored in the ing and reference memory in rats (124), and it alters
visceral or abdominal region. Based on this, it is easy to intracellular signaling by N-methyl-D-aspartate (125) and
generate an explanation for why neurons in the medio- ␣-amino-3-hydroxy-5-methyl-4-isoxaziole-propionate (126)
basal hypothalamus express insulin receptors, since these receptors that are implicated in the development of long-
neurons are intimately involved in the regulation of energy term potentiation (127). Thus, disruption of systemic
homeostasis. An intriguing related question, however, con- insulin signaling is associated with impairments of memo-
cerns the rationale for more remote areas of the brain to rial processes in rodents.
express insulin receptors, such as the hippocampus or the Importantly, administration of exogenous insulin im-
olfactory bulb. proves performance on some learning tasks and corrects
In the hippocampus, immunoreactivity for insulin recep- the learning and memory deficits produced by streptozo-
tors is found in the molecular layer of the dentate gyrus tocin or diabetes. For example, administration of exoge-
and on dendrites of CA1 pyramidal cells (51,102). Insulin nous insulin improves rats’ performance in the one-trial
binding in the hippocampus is colocalized with immuno- learning of a passive avoidance task (128,129). It also
labeled phosphotyrosine (54) and insulin receptor sub- ameliorates the diabetic impairments observed on radial-
strate 1 (103), and as is discussed above, these compounds arm maze tasks and water mazes (122,123). A key point is
are important for the insulin receptor’s intracellular sig- that administration of insulin directly into the brain, where
naling pathways (104). Thus, the binding of insulin in the it does not improve peripheral glucose metabolism, also
hippocampus, as it is in the hypothalamus, appears to be improves learning (128,130), although some studies have
closely associated with functional effects of insulin. Glu- reported contradictory findings, i.e., that administration of
cose metabolism in hippocampal cells is sensitive to insulin impairs learning and memory performance (131–
application of exogenous insulin (105), and this sensitivity 134).
depends on the insulin receptor (106,107). Further, insulin Therefore, data from both humans and animals support
receptors in the hippocampus are localized in dendritic the hypothesis that insulin is an important factor in
fields, suggesting a neuromodulatory role (51,54,55). Fur- memorial processes. However, the exact nature of that
thermore, there is a relationship between certain meta- role remains elusive. This is due, in part, to the fact that
bolic disorders and cognitive decline (108). In diabetes, for much of the previous work suffers from one or both of two
example, patients must rely on exogenous insulin admin- serious confounds. First, it is often difficult to disentangle
istration to keep blood glucose levels within a life-sustain- the effects of altered insulin from the effects of altered
ing range. These patients also often exhibit increased metabolism. Large changes in basal insulin levels can
cognitive impairment correlated with the progression of produce profound disruptions in metabolic status that
their diabetes (109). Other disease states are also accom- might account for some of the observed disruptions in
panied by changes in metabolic status. The progression of learning in and of themselves. Second, because most
diverse central nervous system disorders such as Hunting- manipulations of insulin occur systemically, it is difficult to
ton’s disease (110), Parkinson’s disease (111), and schizo- identify the effects of insulin uniquely in regions of the
phrenia is correlated with changes in glucose levels, brain known to be important for learning and memory.
insulin, and metabolic activity. Likewise, the progression Finally, the ability of insulin to improve cognitive behavior
of Alzheimer’s disease is correlated with disturbances in might be task or reinforcement specific. It might be, for
basal insulin levels as well as glucose metabolism (112). example, that increased insulin in the hippocampus, which
More striking is the finding that when Alzheimer’s reflects an increase of available energy in the body,
patients are made hyperinsulinemic, their performance on facilitates learning tasks that help procure food. That is,
several assessments of memorial tasks is improved (113). elevated insulin might well enable remembering where
Importantly, this occurs even at doses of insulin that do food is located, or the best time to access it, or related
DIABETES, VOL. 55, SUPPLEMENT 2, DECEMBER 2006 S117
BRAIN-GUT-ISLET AXIS

factors. Likewise, increased insulin activity in the olfac-


tory bulb could help form associations between specific
odors and food. The point is that any general hypothesis of
insulin action in the brain must account for what changes
of insulin actually reflect.

DIETARY FAT, OBESITY, AND INSULIN RESISTANCE


As previously mentioned, when leptin is administered into
the brains of experimental animals, there is a selective
reduction of body fat, with lean body mass being spared
(135). Likewise, when insulin is administered into the
brain, there is a reduction of the respiratory quotient,
suggesting that the body is oxidizing relatively more fat
(136). These observations suggest that one action of these
adipose signals within the brain is to reduce body fat, and
a corollary of this is that fat ingestion would be expected FIG. 2. Insulin has many actions in the brain. A: Insulin circulates in the
to be reduced as well. Consistent with this, we have blood in direct proportion to stored calories. Within the hypothalamus,
observed that when insulin is administered into the third it regulates food intake and energy expenditure, thus contributing to
cerebral ventricle of rats, fat intake is selectively reduced the maintenance of a relatively stable body weight. B: Insulin levels are
also a function of blood glucose. Elevated glucose increases insulin
(137). Hence, it is reasonable to hypothesize that leptin secretion, and the increased insulin signal in the hypothalamus reduces
and insulin, acting in the brain, reduce body fat by hepatic glucose secretion, thus contributing to the regulation of blood
increasing lipid mobilization and oxidation and simulta- glucose. C: Insulin stimulates receptors in the hippocampus, facilitat-
ing certain kinds of learning and memory. This action, as well as an
neously by reducing the consumption of dietary fat. action in the olfactory bulb, is hypothesized to facilitate foraging
When animals become obese after exposure to high-fat behavior.
diets, they become resistant to leptin and insulin’s ability
to regulate food intake and body weight. The epidemiolog-
ical data that increasing dietary fat accelerates the devel- hippocampal expression of brain-derived neurotrophic
opment of obesity are quite compelling and have been factor mRNA (155). We and others have found that in-
summarized in several reviews (138 –140). Animal studies creased body weight and peripheral metabolic disruptions
provide strong corroborative evidence, i.e., across numer- are also associated with reduced insulin receptor gene
ous experiments, diets, and species, and the conclusion expression in the hippocampus. Several important ques-
that increased consumption of high-fat diets leads to tions remain unanswered, however, including the specific
increased body fat is inescapable (141–148). Importantly, molecular mechanisms by which central insulin resistance
there are strong genetic influences that dictate whether or might lead to decreased expression of such factors as
not a given individual will be prone or resistant to becom- brain-derived neurotrophic factor.
ing obese when exposed to a high-fat diet (141,146,149 –
153). As Bray and Popkin (138) point out, a high-fat diet
can be viewed as the environmental agent that acts on a SUMMARY
susceptible host animal to produce the noninfectious
disease obesity. Importantly, the consequences of obesity, To summarize, increased insulin in the mediobasal hypo-
including dietary-induced obesity, are well documented thalamus provides a signal that ample or excess energy is
and include type 2 diabetes and insulin insensitivity. available in the body, and one consequence is a reduction
Further, some detrimental effects of dietary fat are not of food intake. It has recently been reported that the
limited to obese individuals. For example, we have re- increased hypothalamic insulin signal also elicits a vagal
cently demonstrated that while high-fat diet–induced obe- reflex to the liver that reduces glucose secretion. In-
sity decreases central sensitivity to the anorexic effects of creased insulin (and probably leptin) locally within the
central insulin administration, increased dietary fat, in the hypothalamus therefore can be considered to be analo-
absence of frank obesity, also attenuates the potency of gous to other signals that indicate a surfeit of energy in the
central insulin to reduce food intake and body weight (148; body. In addition to satiety and adiposity signals, this
K. Gotoh, M.D. Wortman, S.C.B., D.J.C., D. D’Alessio, P. includes certain lipids and glucose, and there is compel-
Tso, R.J. Seeley, S.C.W., unpublished data). ling evidence that overlapping intracellular signaling path-
ways within the mediobasal hypothalamus mediate the
HIPPOCAMPAL INSULIN RESISTANCE overall catabolic response to these diverse metabolic
Consistent with the concept that insulin acts in multiple signals. Insulin receptors are also densely expressed in the
regions of the brain to influence multiple processes, di- hippocampus, and there is evidence that insulin acts there
etary-induced insulin resistance has recently been linked to facilitate certain cognitive functions. The function of
to impaired cognitive function in both humans and ani- insulin receptors in other brain areas is poorly understood.
mals. Specifically, high levels of dietary saturated fats and Obesity and/or the consumption of diets high in fat render
simple carbohydrates (which promote insulin resistance the brain as well as the body insulin resistant. In the
and obesity) have been demonstrated to reduce hippocam- hypothalamus, this is manifest as a reduced ability of
pal-dependent memory processes. For example, Gomez- insulin to reduce food intake and body weight, and in the
Pinilla and colleagues (154) have shown that long-term hippocampus, it is manifest as a reduced ability of insulin
access to obesigenic diets impairs rats’ performance in the to improve learning and/or memory. Figure 2 is a model
Morris water maze task, a classic measure of spatial depicting some of the feedback loops involving insulin in
memory. These deficits are associated with reductions in the brain.
S118 DIABETES, VOL. 55, SUPPLEMENT 2, DECEMBER 2006
S.C. WOODS, S.C. BENOIT, AND D.J. CLEGG

REFERENCES tricular infusion of insulin reduces food intake and body weight of
1. Strubbe JH, Woods SC: The timing of meals. Psychol Rev 111:128 –141, baboons. Nature 282:503–505, 1979
2004 30. Chavez M, Seeley RJ, Woods SC: A comparison between the effects of
2. Woods SC: The eating paradox: how we tolerate food. Psychol Rev 98: intraventricular insulin and intraperitoneal LiCl on three measures sen-
488 –505, 1991 sitive to emetic agents. Behav Neurosci 109:547–550, 1995
3. Woods SC, Strubbe JH: The psychobiology of meals. Psychol Bull Rev 31. Schwartz MW, Figlewicz DP, Baskin DG, Woods SC, Porte D Jr: Insulin in
1:141–155, 1994 the brain: a hormonal regulator of energy balance. Endocr Rev 13:387–
414, 1992
4. Kaplan JM, Moran TH: Gastrointestinal signaling in the control of food
32. Woods SC, Chavez M, Park CR, Riedy C, Kaiyala K, Richardson RD,
intake. In Handbook of Behavioral Neurobiology: Neurobiology of Food
Figlewicz DP, Schwartz MW, Porte D, Seeley RJ: The evaluation of insulin
and Fluid Intake. Vol. 4, no. 2. Stricker EM, Woods SC, Eds. New York,
as a metabolic signal controlling behavior via the brain. Neurosci
Kluwer Academic/Plenum, 2004, p. 273–303
Biobehav Rev 20:139 –144, 1995
5. Moran TH, Kinzig KP: Gastrointestinal satiety signals II. Cholecystokinin.
33. Woods SC: Insulin and the brain: a mutual dependency. Prog Psychobiol
Am J Physiol Gastrointest Liver Physiol 286:G183–G188, 2004
Physiol Psychol 16:53– 81, 1996
6. Strader AD, Woods SC: Gastrointestinal hormones and food intake. 34. McGowan MK, Andrews KM, Grossman SP: Chronic intrahypothalamic
Gastroenterology 128:175–191, 2005 infusions of insulin or insulin antibodies alter body weight and food
7. Woods SC: Gastrointestinal satiety signals I. An overview of gastrointes- intake in the rat. Physiol Behav 51:753–766, 1992
tinal signals that influence food intake. Am J Physiol Gastrointest Liver 35. Strubbe JH, Mein CG: Increased feeding in response to bilateral injection
Physiol 286:G7–G13, 2004 of insulin antibodies in the VMH. Physiol Behav 19:309 –313, 1977
8. Smith GP, Gibbs J: The development and proof of the cholecystokinin 36. Brüning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban PC, Klein
hypothesis of satiety. In Multiple Cholecystokinin Receptors in the CNS. R, Krone W, Müller-Wieland D, Kahn CR: Role of brain insulin receptor in
Dourish CT, Cooper SJ, Iversen SD, Iversen LL, Eds. Oxford, U.K., Oxford control of body weight and reproduction. Science 289:2122–2125, 2000
University Press, 1992, p. 166 –182 37. Obici S, Feng Z, Karkanias G, Baskin DG, Rossetti L: Decreasing
9. Porte D Jr, Baskin DG, Schwartz MW: Insulin signaling in the central hypothalamic insulin receptors causes hyperphagia and insulin resistance
nervous system: a critical role in metabolic homeostasis and disease from in rats. Nat Neurosci 5:566 –572, 2002
C. elegans to humans. Diabetes 54:1264 –1276, 2005 38. Hallschmid M, Benedict C, Schultes B, Fehm HL, Born J, Kern W:
10. Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG: Central Intranasal insulin reduces body fat in men but not in women. Diabetes
nervous system control of food intake. Nature 404:661– 671, 2000 53:3024 –3029, 2004
11. Schwartz MW, Porte D Jr: Diabetes, obesity, and the brain. Science 39. Banks WA, Jaspan JB, Huang W, Kastin AJ: Transport of insulin across
307:375–379, 2005 the blood-brain barrier: saturability at euglycemic doses of insulin.
12. Woods SC, Seeley RJ, Porte D Jr, Schwartz MW: Signals that regulate food Peptides 18:1423–1429, 1997
intake and energy homeostasis. Science 280:1378 –1383, 1998 40. Baura G, Foster D, Porte D Jr, Kahn SE, Bergman RN, Cobelli C, Schwartz
13. Woods SC, Seeley RJ: Insulin as an adiposity signal. Int J Obes Relat MW: Saturable transport of insulin from plasma into the central nervous
Metab Disord 25:S35–S38, 2001 system of dogs in vivo: a mechanism for regulated insulin delivery to the
14. Friedman JM: The function of leptin in nutrition, weight, and physiology. brain. J Clin Invest 92:1824 –1830, 1993
Nutr Rev 60:S1–S14, 2002 41. Schwartz MW, Bergman RN, Kahn SE, Taborsky GJ Jr, Fisher LD, Sipols
15. Woods SC, Seeley RJ: Adiposity signals and the control of energy AJ, Woods SC, Steil GM, Porte D Jr: Evidence for uptake of plasma insulin
homeostasis. Nutrition 16:894 –902, 2000 into cerebrospinal fluid through an intermediate compartment in dogs.
16. Figlewicz DP, Sipols AJ, Seeley RJ, Chavez M, Woods SC, Porte DJ: J Clin Invest 88:1272–1281, 1991
Intraventricular insulin enhances the meal-suppressive efficacy of intra- 42. Woods SC, Seeley RJ, Baskin DG, Schwartz MW: Insulin and the blood-
ventricular cholecystokinin octapeptide in the baboon. Behav Neurosci brain barrier. Curr Pharm Des 9:795– 800, 2003
109:567–569, 1995 43. Strubbe JH, Porte DJ, Woods SC: Insulin responses and glucose levels in
17. Riedy CA, Chavez M, Figlewicz DP, Woods SC: Central insulin enhances plasma and cerebrospinal fluid during fasting and refeeding in the rat.
sensitivity to cholecystokinin. Physiol Behav 58:755–760, 1995 Physiol Behav 44:205–208, 1988
44. Kaiyala K, Prigeon RL, Kahn SE, Woods SC, Schwartz MW: Obesity
18. Emond M, Schwartz GJ, Ladenheim EE, Moran TH: Central leptin
induced by a high-fat diet is associated with reduced brain insulin
modulates behavioral and neural responsivity to CCK. Am J Physiol
transport in dogs. Diabetes 49:1525–1533, 2000
Regul Integr Comp Physiol 276:R1545–R1549, 1999
45. Israel PA, Park CR, Schwartz MW, Green PK, Sipols AJ, Woods SC, Porte
19. Matson CA, Wiater MF, Kuijper JL, Weigle DS: Synergy between leptin
D Jr, Figewicz DP: Effect of diet-induced obesity and experimental
and cholecystokinin (CCK) to control daily caloric intake. Peptides 18:
hyperinsulinemia on insulin uptake into CSF of the rat. Brain Res Bull
1275–1278, 1997
30:571–575, 1993
20. Matson CA, Reid DF, Cannon TA, Ritter RC: Cholecystokinin and leptin 46. Stein LJ, Dorsa DM, Baskin DG, Figlewicz DP, Ikeda H, Frankmann SP,
act synergistically to reduce body weight. Am J Physiol Regul Integr Greenwood MR, Porte D Jr, Woods SC: Immunoreactive insulin levels are
Comp Physiol 278:R882–R890, 2000 elevated in the cerebrospinal fluid of genetically obese Zucker rats.
21. Morton GJ, Blevins JE, Williams DL, Niswender KD, Gelling RW, Rhodes Endocrinology 113:2299 –2301, 1983
CJ, Baskin DG, Schwartz MW: Leptin action in the forebrain regulates the 47. Vanderweele DA, Haraczkiewicz E, Van Itallie TB: Elevated insulin and
hindbrain response to satiety signals. J Clin Invest 115:703–710, 2005 satiety in obese and normal weight rats. Appetite 3:99 –109, 1982
22. Barrachina MD, Martinez V, Wang L, Wei JY, Tache Y: Synergistic 48. Nicolaidis S, Rowland N: Metering of intravenous versus oral nutrients
interaction between leptin and cholecystokinin to reduce short-term food and regulation of energy balance. Am J Physiol 231:661– 668, 1976
intake in lean mice. Proc Natl Acad Sci U S A 94:10455–10460, 1997 49. Baskin DG, Marks JL, Schwartz MW, Figewicz DP, Woods SC, Porte D Jr:
23. Ikeda H, West DB, Pustek JJ, Figlewicz DP, Greenwood MRC, Porte D Jr, Insulin and insulin receptors in the brain in relation to food intake and
Woods SC: Intraventricular insulin reduces food intake and body weight body weight. In Endocrine and Nutritional Control of Basic Biological
of lean but not obese Zucker rats. Appetite 7:381–386, 1986 Functions. Lehnert H, Murison R, Weiner H, Hellhammer D, Beyer J, Eds.
24. McMinn JE, Sindelar DK, Havel PJ, Schwartz MW: Leptin deficiency Stuttgart, Hogrefe & Huber, 1990, p. 202–222
induced by fasting impairs the satiety response to cholecystokinin. 50. Baskin DG, Woods SC, West DB, van HM, Posner BI, Dorsa DM, Porte D
Endocrinology 141:4442– 4448, 2000 Jr: Immunocytochemical detection of insulin in rat hypothalamus and its
25. Bagdade JD, Bierman EL, Porte D Jr: The significance of basal insulin possible uptake from cerebrospinal fluid. Endocrinology 113:1818 –1825,
levels in the evaluation of the insulin response to glucose in diabetic and 1983
nondiabetic subjects. J Clin Invest 46:1549 –1557, 1967 51. Corp ES, Woods SC, Porte D Jr, Dorsa DM, Figlewicz DP, Baskin DG:
26. Polonsky KS, Given E, Carter V: Twenty-four-hour profiles and pulsatile Localization of 125I-insulin binding sites in the rat hypothalamus by
patterns of insulin secretion in normal and obese subjects. J Clin Invest quantitative autoradiography. Neurosci Lett 70:17–22, 1986
81:442– 448, 1988 52. Havrankova J, Brownstein M, Roth J: Insulin and insulin receptors in the
27. Woods SC, Decke E, Vasselli JR: Metabolic hormones and regulation of rodent brain. Diabetologia 20:268 –273, 1981
body weight. Psychol Rev 81:26 – 43, 1974 53. Unger J, McNeil TH, Moxley RTI, White M, Moss A, Livingston JN:
28. Kennedy GC: The role of depot fat in the hypothalamic control of food Distribution of insulin receptor-like immunoreactivity in the rat forebrain.
intake in the rat. Proc R Soc Lond (Biol) 140:579 –592, 1953 Neuroscience 31:143–157, 1989
29. Woods SC, Lotter EC, McKay LD, Porte D Jr: Chronic intracerebroven- 54. Unger JW, Moss AM, Livingston JN: Immunohistochemical localization of

DIABETES, VOL. 55, SUPPLEMENT 2, DECEMBER 2006 S119


BRAIN-GUT-ISLET AXIS

insulin receptors and phosphotyrosine in the brainstem of the adult rat. 85. Considine RV, Sinha MK, Heiman ML, Kriaucinas A, Stephens TW, Nyce
Neuroscience 42:853– 861, 1991 MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL, Caro JF: Serum
55. Unger JW, Livingston JN, Moss AM: Insulin receptors in the central immunoreactive-leptin concentrations in normal-weight and obese hu-
nervous system: localization, signalling mechanisms and functional as- mans. N Engl J Med 334:292–295, 1996
pects. Prog Neurobiol 36:343–362, 1991 86. Havel PJ: Update on adipocyte hormones: regulation of energy balance
56. Obici S, Zhang BB, Karkanias G, Rossetti L: Hypothalamic insulin and carbohydrate/lipid metabolism. Diabetes 53 (Suppl. 1):S143–S151,
signaling is required for inhibition of glucose production. Nat Med 2004
8:1376 –1382, 2002 87. Wisse BE, Campfield LA, Marliss EB, Morais JA, Tenenbaum R, Gougeon
57. Lam TK, Pocai A, Gutierrez-Juarez R, Obici S, Bryan J, Aguilar-Bryan L, R: Effect of prolonged moderate and severe energy restriction and
Schwartz GJ, Rossetti L: Hypothalamic sensing of circulating fatty acids refeeding on plasma leptin concentrations in obese women. Am J Clin
is required for glucose homeostasis. Nat Med 11:320 –327, 2005 Nutr 70:321–330, 1999
58. Lin X, Taguchi A, Park S, Kushner JA, Li F, Li Y, White MF: Dysregulation 88. Ahren B, Mansson S, Gingerich RL, Havel PJ: Regulation of plasma leptin
of insulin receptor substrate 2 in beta cells and brain causes obesity and in mice: influence of age, high-fat diet and fasting. Am J Physiol Regul
diabetes. J Clin Invest 114:886 – 888, 2004 Integr Comp Physiol 273:R113–R120, 1997
59. Levin BE, Routh VH, Kang L, Sanders NM, Dunn-Meynell AA: Neuronal 89. Zhao AZ, Shinohara MM, Huang D, Shimizu H, Eldar-Finkelman H, Krebs
glucosensing: what do we know after 50 years? Diabetes 53:2521–2528, EG, Beavo JA, Bornfeldt KE: Leptin induces insulin-like signaling that
2004 antagonizes cAMP elevation by glucagon in hepatocytes. J Biol Chem
60. Obici S, Feng Z, Morgan K, Stein D, Karkanias G, Rossetti L: Central 275:11348 –11354, 2000
administration of oleic acid inhibits glucose production and food intake. 90. Niswender KD, Schwartz MW: Insulin and leptin revisited: adiposity
Diabetes 51:271–275, 2002 signals with overlapping physiological and intracellular signaling capabil-
61. Obici S, Feng Z, Arduini A, Conti R, Rossetti L: Inhibition of hypothalamic ities. Front Neuroendocrinol 24:1–10, 2003
carnitine palmitoyltransferase-1 decreases food intake and glucose pro- 91. Niswender KD, Baskin DG, Schwartz MW: Insulin and its evolving
duction. Nat Med 9:756 –761, 2003 partnership with leptin in the hypothalamic control of energy homeosta-
62. Obici S, Rossetti L: Minireview: nutrient sensing and the regulation of sis. Trends Endocrinol Metab 15:362–369, 2004
insulin action and energy balance. Endocrinology 144:5172–5178, 2003 92. Hikita M, Bujo H, Hirayama S, Takahashi K, Morisaki N, Saito Y:
63. Pocai A, Morgan K, Buettner C, Gutierrez-Juarez R, Obici S, Rossetti L: Differential regulation of leptin receptor expression by insulin and leptin
Central leptin acutely reverses diet-induced hepatic insulin resistance. in neuroblastoma cells. Biochem Biophys Res Commun 271:703–709,
Diabetes 54:3182–3189, 2005 2000
64. He W, Lam TK, Obici S, Rossetti L: Molecular disruption of hypothalamic 93. Carvalheira JB, Siloto RM, Ignacchitti I, Brenelli SL, Carvalho CR, Leite A,
nutrient sensing induces obesity. Nat Neurosci 9:227–233, 2006 Velloso LA, Gontijo JA, Saad MJ: Insulin modulates insulin-induced
65. Pocai A, Obici S, Schwartz GJ, Rossetti L: A brain-liver circuit regulates STAT3 activation in rat hypothalamus. FEBS Lett 500:119 –124, 2001
glucose homeostasis. Cell Metab 1:53– 61, 2005 94. Niswender KD, Morton GJ, Stearns WH, Rhodes CJ, Myers MG Jr,
66. Pocai A, Lam TK, Gutierrez-Juarez R, Obici S, Schwartz GJ, Bryan J, Schwartz MW: Intracellular signaling: key enzyme in leptin-induced
Aguilar-Bryan L, Rossetti L: Hypothalamic K(ATP) channels control anorexia. Nature 413:794 –795, 2001
hepatic glucose production. Nature 434:1026 –1031, 2005 95. Niswender KD, Morrison CD, Clegg DJ, Olson R, Baskin DG, Myers MG Jr,
67. Lam TK, Gutierrez-Juarez R, Pocai A, Rossetti L: Regulation of blood Seeley RJ, Schwartz MW: Insulin activation of phosphatidylinositol
glucose by hypothalamic pyruvate metabolism. Science 309:943–947, 2005 3-kinase in the hypothalamic arcuate nucleus: a key mediator of insulin-
68. Wajchenberg BL, Giannella-Neto D, da Silva ME, Santos RF: Depot- induced anorexia. Diabetes 52:227–231, 2003
specific hormonal characteristics of subcutaneous and visceral adipose 96. Akabayashi A, Wahlestedt C, Alexander JT, Leibowitz SF: Specific inhi-
tissue and their relation to the metabolic syndrome. Horm Metab Res bition of endogenous neuropeptide Y synthesis in arcuate nucleus by
34:616 – 621, 2002 antisense oligonucleotides suppresses feeding behavior and insulin se-
69. Woods SC, Gotoh K, Clegg DJ: Gender differences in the control of energy cretion. Brain Res 21:55– 61, 1994
homeostasis. Exp Biol Med 228:1175–1180, 2003 97. Flier JS: Obesity wars: molecular progress confronts an expanding
70. Bjorntorp P: Body fat distribution, insulin resistance, and metabolic epidemic. Cell 116:337–350, 2004
diseases. Nutrition 13:795– 803, 1997 98. Cowley MA, Smart JL, Rubinstein M, Cerdan MG, Diano S, Horvath TL,
71. Wajchenberg BL: Subcutaneous and visceral adipose tissue: their relation Cone RD, Low MJ: Leptin activates anorexigenic POMC neurons through
to the metabolic syndrome. Endocr Rev 21:697–738, 2000 a neural network in the arcuate nucleus. Nature 411:480 – 484, 2001
72. Clegg DJ, Brown LM, Woods SC, Benoit SC: Gonadal hormones determine 99. Seeley RJ, Drazen DL, Clegg DJ: The critical role of the melanocortin
sensitivity to central leptin and insulin. Diabetes 55:978 –987, 2006 system in the control of energy balance. Ann Rev Nutr 24:133–149, 2004
73. Clegg DJ, Riedy CA, Smith KA, Benoit SC, Woods SC: Differential 100. Benoit SC, Tracy AL, Air EL, Kinzig K, Seeley RJ, Davidson TL: The role
sensitivity to central leptin and insulin in male and female rats. Diabetes of the hypothalamic melanocortin system in behavioral appetitive pro-
52:682– 687, 2003 cesses. Pharmacol Biochem Behav 69:603– 609, 2001
74. Geary N: Estradiol, CCK and satiation. Peptides 22:1251–1263, 2001 101. Baskin DG, Hahn TM, Schwartz MW: Leptin sensitive neurons in the
75. Geary N: Is the control of fat ingestion sexually differentiated? Physiol hypothalamus. Horm Metab Res 31:345–350, 1999
Behav 83:659 – 671, 2004 102. Baskin DG, Porte DJ, Guest K, Dorsa DM: Regional concentrations of
76. Yanovsky S: Sugar and fat: Cravings and aversions. J Nutr 133:835S– 837S, insulin in the rat brain. Endocrinology 112:898 –903, 1983
2003 103. Baskin DG, Sipols AJ, Schwartz MW, White MF: Insulin receptor sub-
77. Valle A, Catala-Niell A, Colom B, Garcia-Palmer FJ, Oliver J, Roca P: Sex strate-1 (IRS-1) expression in rat brain. Endocrinology 134:1952–1955,
related differences in energy balance in response to caloric restriction. 1994
Am J Physiol Endocrinol Metab 289:E15–E22, 2005 104. Baskin DG, Sipols AJ, Schwartz MW, White MF: Immunocytochemical
78. Bulik CM, Reba L, Siega-Riz AM, Reichborn-Kjennerud T: Anorexia detection of insulin receptor substrate-1 (IRS- 1) in rat brain: colocaliza-
nervosa: definition, epidemiology, and cycle of risk. Int J Eat Dis 37 tion with phosphotyrosine. Reg Peptides 48:257–266, 1993
(Suppl. 1):S2–S9, 2005 105. Henneberg N, Hoyer S: Short-term or long-term intracerebroventricular
79. Xu AW, Kaelin CB, Takeda K, Akira S, Schwartz MW, Barsh GS: PI3K (i.c.v.) infusion of insulin exhibits a discrete anabolic effect on cerebral
integrates the action of insulin and leptin on hypothalamic neurons. energy metabolism in the rat. Neurosci Letters 175:153–156, 1994
J Clin Invest 115:951–958, 2005 106. Palovcik R, Phillips M, Kappy M, Raizada M: Insulin inhibits pyramidal
80. Guitterez-Juarez R, Obici S, Rossetti L: Melanocortin-independent effects neurons in hippocampal slices. Brain Res 309:187–191, 1984
of leptin on hepatic glucose fluxes. J Biol Chem 279:49704 – 49715, 2004 107. Phillips M, Palovcik R: Dose-response testing of peptides by hippocampal
81. Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-Flier E, brain slice recording. Meth Enzymol 168:129 –144, 1989
Flier JS: Role of leptin in the neuroendocrine response to fasting. Nature 108. Beal MF: Energetics in the pathogenesis of neurodegenerative diseases.
382:250 –252, 1996 Trends Neurosci 23:298 –304, 2000
82. Ahima RS: Central actions of adipocyte hormones. Trends Endocrinol 109. Biessels G: Cerebral complications of diabetes: clinical findings and
Metab 16:307–313, 2005 pathogenetic mechanisms. Neth J Med 54:35– 45, 1999
83. Kishi T, Elmquist JK: Body weight is regulated by the brain: a link 110. Podolsky S, Leopold N: Abnormal glucose tolerance and arginine toler-
between feeding and emotion. Mol Psychiatry 10:132–146, 2005 ance tests in Huntington’s disease. Gerontology 23:55– 63, 1977
84. Porte DJ, Baskin DG, Schwartz MW: Leptin and insulin action in the 111. Mattson M, Pedersen W, Duan W, Culmsee C, Camandola S: Cellular and
central nervous system. Nutr Rev 60:S20 –S29, 2002 molecular mechanisms underlying perturbed energy metabolism and

S120 DIABETES, VOL. 55, SUPPLEMENT 2, DECEMBER 2006


S.C. WOODS, S.C. BENOIT, AND D.J. CLEGG

neuronal degeneration in Alzheimer’s and Parkinson’s diseases. Ann N Y 133. Kopf S, Baratti C: Effects of posttraining administration of insulin on
Acad Sci 893:154 –175, 1999 retention of a habituation response in mice: participation of a central
112. Nolan JH, Wright CE: Evidence of impaired glucose tolerance and insulin cholinergic mechanism. Neurobiol Learn Mem 71:50 – 61, 1999
resistance in patients with Alzheimer’s disease. Curr Dir Psychol Sci 134. Schwarzberg H, Bernstein H, Reiser M, Gunther O: Intracerebroventric-
10:102–105, 2001 ular administration of insulin attenuates retrieval of a passive avoidance
113. Craft S, Newcomer J, Kanne S, Dagogo-Jack S, Cryer P, Sheline Y, Luby response in rats. Neuropeptides 13:79 – 81, 1989
J, Dagogo-Jack A, Alderson A: Memory improvement following induced 135. Chen G, Koyama K, Yuan X, Lee Y, Zhou YT, O’Doherty R, Newgard CB,
hyperinsulinemia in Alzheimer’s disease. Neurobiol Aging 17:123–130, Unger RH: Disappearance of body fat in normal rats induced by adeno-
1996 virus-mediated leptin gene therapy. Proc Natl Acad Sci U S A 93:14795–
114. Kumari M, Brunner E, Fuhrer R: Minireview: mechanisms by which the 14799, 1996
metabolic syndrome and diabetes impair memory. J Gerontol Series A 136. Park C, Chavez M, Woods SC: IVT insulin decreases respiratory quotient
Biol Sci Med 55:B228 –B232, 2000 in rats. Soc Neurosci Abstr 22:939, 1992
115. Strachan M, Deary I, Ewing F, Frier B: Is type II diabetes associated with 137. Chavez M, Riedy CA, van Dijk G, Woods SC: Central insulin and
an increased risk of cognitive dysfunction? A critical review of published macronutrient intake in the rat. Am J Physiol Regul Integr Comp Physiol
studies. Diabetes Care 20:438 – 445, 1997 271:R727–R731, 1996
116. Stewart R, Liolitsa D: Type 2 diabetes mellitus, cognitive impairment and 138. Bray GA, Popkin BM: Dietary fat does affect obesity. Am J Clin Nutr
dementia. Diabet Med 16:93–112, 1999
68:1157–1173, 1998
117. Richardson J: Cognitive function in diabetes mellitus. Neurosci Biobehav
139. Gibney MJ: Epidemiology of obesity in relation to nutrient intake. Int J
Rev 14:385–388, 1990
Obes 19 (Suppl. 5):S1–S3, 1995
118. Park CR: Cognitive effects of insulin in the central nervous system.
140. Samaras K, Kelly PJ, Chiano MN, Arden N, Spector TD, Campbell LV:
Neurosci Biobehav Rev 25:311–323, 2001
Genes versus environment. Diabetes Care 21:2069 –2076, 1998
119. Flood J, Mooradian A, Morley J: Characteristics of learning and memory
141. Bray GA, Fisler J, York DA: Neuroendocrine control of the development
in streptozocin-induced diabetic mice. Diabetes 39:1391–1398, 1990
120. Kamal A, Biessels G, Urban I, Gispen W: Hippocampal synaptic plasticity of obesity: understanding gained from studies of experimental animal
in streptozotocin-diabetic rats: impairment of long-term potentiation and models. Front Neuroendocrinol 11:128 –181, 1990
facilitation of long-term depression. Neuroscience 90:737–745, 1999 142. Golay A, Bobbioni E: The role of dietary fat in obesity. Int J Obes Rel
121. Kamal A, Biessels G, Duis S, Gispen W: Learning and hippocampal Metab Dis 21 (Suppl. 3):S2–S11, 1997
synaptic plasticity in streptozotocin-diabetic rats: interaction of diabetes 143. Hill JO, Lin D, Yakubu F, Peters JC: Development of dietary obesity in
and ageing. Diabetologia 43:500 –506, 2000 rats: influence of amount and composition of dietary fat. Int J Obes
122. Biessels G, Kamal A, Ramakers G, Urban I, Spruijt B, Erkelens D, Gispen 16:321–333, 1992
W: Place learning and hippocampal synaptic plasticity in streptozotocin- 144. Warwick ZS, Schiffman SS: Role of dietary fat in calorie intake and weight
induced diabetic rats. Diabetes 45:1259 –1266, 1996 gain. Neurosci Biobehav Rev 16:585–596, 1992
123. Biessels G, Kamal A, Urban I, Spruijt B, Erkelens D, Gispen W: Water 145. Warwick ZS: Probing the causes of high-fat diet hyperphagia: a mecha-
maze learning and hippocampal synaptic plasticity in streptozotocin- nistic and behavioral dissection. Neurosci Biobehav Rev 20:155–161, 1996
diabetic rats: effects of insulin treatment. Brain Res 800:125–135, 1998 146. West DB, York B: Dietary fat, genetic predisposition, and obesity: lessons
124. Lannert H, Hoyer S: Intracerebroventricular administration of streptozo- from animal models. Am J Clin Nutr 67 (Suppl. 3):505S–512S, 1998
tocin causes long-term diminutions in learning and memory abilities and 147. Woods SC, Seeley RJ, Rushing PA, D’Alessio DA, Tso P: A controlled
in cerebral energy metabolism in adult rats. Behav Neurosci 112:1199 – high-fat diet induces an obese syndrome in rats. J Nutr 133:1081–1087,
1208, 1998 2003
125. Di Luca M, Ruts L, Gardoni F, Cattabeni F, Biessels G, Gispen W: NMDA 148. Woods SC, D’Alessio DA, Tso P, Rushing PA, Clegg DJ, Benoit SC, Gotoh
receptor subunits are modified transcriptionally and post-translationally K, Liu M, Seeley RJ: Consumption of a high-fat diet alters the homeostatic
in the brain of streptozotocin-diabetic rats. Diabetologia 42:693–701, 1999 regulation of energy balance. Physiol Behav 83:573–578, 2004
126. Chabot C, Massicotte G, Milot M, Trudeau F, Gagne J: Impaired modula- 149. Leibel RL, Chung WK, Chua SC Jr: The molecular genetics of rodent single
tion of AMPA receptors by calcium-dependent processes in streptozoto- gene obesities. J Biol Chem 272:31937–31940, 1997
cin-induced diabetic rats. Brain Res 768:249 –256, 1997 150. Levin BE, Brown KL, Dunn-Meynell AA: Differential effects of diet and
127. Collingridge G: Synaptic plasticity: the role of NMDA receptors in obesity on high and low affinity sulfonylurea binding sites in the rat brain.
learning and memory. Nature 330:604 – 605, 1987 Brain Res 739:293–300, 1996
128. Park C, Seeley R, Craft S, Woods S: Intracerebroventricular insulin 151. Levin BE, Dunn-Meynell AA, Balkan B, Keesey RE: Selective breeding for
enhances memory in a passive-avoidance task. Physiol Behav 68:509 – diet-induced obesity and resistance in Sprague-Dawley rats. Am J Physiol
514, 2000 Regul Integr Comp Physiol 273:R725–R730, 1997
129. Parkes M, White K: Glucose attenuation of memory impairments. Behav 152. Reed DR, Bachmanov AA, Beauchamp GK, Tordoff MG, Price RA:
Neurosci 114:307–319, 2000 Heritable variation in food preferences and their contribution to obesity.
130. Zhao W, Chen H, Xu H, Moore E, Meiri N, Quon M, Alkon D: Brain insulin Behav Genet 27:373–387, 1997
receptors and spatial memory: correlated changes in gene expression, 153. West DB: Genetics of obesity in humans and animal models. Endocrinol
tyrosine phosphorylation, and signaling molecules in the hippocampus of Metab Clin N Am 25:801– 813, 1996
water maze trained rats. J Biol Chem 274:34893–34902, 1999 154. Molteni R, Barnard RJ, Ying Z, Roberts CK, Gomez-Pinilla F: A high-fat,
131. Blanchard J, Duncan P: Effect of combinations of insulin, glucose and refined sugar diet reduces hippocampal brain-derived neurotrophic fac-
scopolamine on radial arm maze performance. Pharmacol Biochem tor, neuronal plasticity, and learning. Neuroscience 112:803– 814, 2002
Behav 58:209 –214, 1997 155. Wu A, Molteni R, Ying Z, Gomez-Pinilla F: A saturated-fat diet aggravates
132. Kopf S, Baratti C: The impairment of retention induced by insulin in mice the outcome of traumatic brain injury on hippocampal plasticity and
may be mediated by a reduction in central cholinergic activity. Neurobiol cognitive function by reducing brain-derived neurotrophic factor. Neuro-
Learn Mem 63:220 –228, 1995 science 119:365–375, 2003

DIABETES, VOL. 55, SUPPLEMENT 2, DECEMBER 2006 S121

Você também pode gostar