SlideShare uma empresa Scribd logo
1 de 22
Baixar para ler offline
Intranasal administration of insulin to the brain impacts cognitive function

                                    and peripheral metabolism




                            Volker Ott1*, Christian Benedict2, Bernd Schultes3,

                                      Jan Born4, Manfred Hallschmid1



1
Department of Neuroendocrinology, University of Luebeck, Germany; 2Department of Neuroscience,

Functional Pharmacology, Uppsala University, Uppsala, Sweden; 3Interdisciplinary Obesity Center,

Cantonal Hospital St. Gallen, Switzerland; 4Department of Medical Psychology and Behavioral

Neurobiology, University of Tübingen, Tübingen, Germany



Key Terms:       Insulin, intranasal, central nervous system, glucose homeostasis, energy homeostasis,

                 thermogenesis, cognitive function, insulin resistance, hepatic glucose production

Word Count:      3487



* To whom correspondence and reprint requests should be addressed:

Department of Neuroendocrinology, Hs. 50.1

University of Luebeck

Ratzeburger Allee 160

23538 Luebeck, Germany

Phone: ++49-451-500-5375

Fax:     ++49-451-500-3640

E-mail: ott@kfg.uni-luebeck.de




This is an Accepted Article that has been peer-reviewed and approved for publication in the Diabetes,
Obesity and Metabolism, but has yet to undergo copy-editing and proof correction. Please cite this
article as an "Accepted Article"; doi: 10.1111/j.1463-1326.2011.01490.x

                                                                                                         1
Abstract

In recent years, the central nervous system (CNS) has emerged as a principle site of insulin action.

This notion is supported by studies in animals relying on intracerebroventricular insulin infusion and

by experiments in humans that make use of the intranasal pathway of insulin administration to the

brain. Employing neurobehavioral and metabolic measurements as well as functional imaging

techniques, these studies have provided insight into a broad range of central and peripheral effects of

brain insulin. The present review focuses on CNS effects of insulin administered via the intranasal

route on cognition, in particular memory function, and whole-body energy homeostasis including

glucose metabolism. Furthermore, evidence is reviewed that suggests a pathophysiological role of

impaired brain insulin signaling in obesity and type 2 diabetes, which are hallmarked by peripheral

and possibly central nervous insulin resistance, as well as in conditions such as Alzheimer´s disease

where      CNS     insulin     resistance    might     contribute     to    cognitive     dysfunction.




                                                                                                     2
Introduction

In the mid-eighteen-hundreds, Claude Bernard demonstrated that puncture of the fourth cerebral

ventricle induces glucosuria in mice [1], giving rise to the assumption that the central nervous system

(CNS) is involved in glucose homeostasis. However, interest in the role the brain might play in the

regulation of glucose metabolism abated and was only sparked again more than a century later.

Havrankova and coworkers demonstrated in 1978 that insulin receptors are present throughout the rat

CNS [2], followed closely by the demonstration that insulin receptors are also expressed in the human

brain [3,4]. The insulin receptor, a tyrosine kinase receptor, is found in particularly high densities in

brain regions like the olfactory bulb, the cerebellum, the dentate gyrus, the pyriform cortex, the

hippocampus, the choroid plexus and the arcuate nucleus of the hypothalamus [5]. Some animal

studies suggest that insulin gene expression takes place within the CNS [6,7]. However, although

indicators of insulin transcription in human brain tissue have been presented [8], solid evidence for

local insulin production in the human CNS is still lacking [9]. It is rather assumed that peripheral

insulin crosses the blood-brain barrier (BBB) by a saturable, receptor-mediated transport mechanism

[9-11] and by binding to brain insulin receptors affects functions as diverse as energy and glucose

homeostasis [12-14], reproduction [13], growth [15] and neuronal plasticity [16]. Woods and co-

workers were the first to perform seminal studies indicating that insulin, circulating within the blood

stream in proportion to body fat stores, acts as an adiposity signal within in the CNS. In conjunction

with the adipokine leptin it provides the brain with negative feedback on the amount of peripheral

energy (i.e., fat) depots [17; for review see reference 18]. In line with this notion, CNS administration

of insulin reduces body adiposity by down-regulating food intake [12,19,20]. This catabolic effect has

been observed mainly in males, indicating that the role of insulin in central nervous body weight

regulation may have sex-specific properties [21-23]. Moreover, as will be outlined in this review,

insulin’s impact on the brain exceeds its involvement in energy homeostasis and pertains to cognitive

functions (Figure 1). Brain insulin signaling might even constitute a neuroendocrine link between both

domains and is therefore emerging as a potential target in the treatment of metabolic and cognitive

disorders [24].


                                                                                                       3
Enhancing central nervous insulin signaling by intranasal insulin administration in humans

Whereas in animals effective modes of insulin administration to the CNS, e.g., direct

intracerebroventricular (ICV) [25] or hypothalamic infusion [26] are routinely employed, insulin

administration to the human brain is more complicated. The conventional way of increasing CNS

concentrations of insulin to investigate effects of brain insulin relies on the intravenous (IV) infusion

of the hormone which has been shown to result in an increase in cerebrospinal fluid (CSF) insulin

concentrations [27]. This parenteral route, however, faces several serious drawbacks. The fall in blood

glucose levels resulting from systemic insulin infusion triggers the graded activation of endocrine axes

that can affect brain function [28], and below certain threshold levels inevitably impairs cognition

[29]. Insulin-induced hypoglycemia and its potentially harmful effects can be prevented by

simultaneous continuous glucose infusion that per se may exert a biasing impact on (cognitive) brain

functioning. Moreover, the euglycemic-hyperinsulinemic clamp procedure implies considerable time

and labor investments and, generally, systemic insulin administration does not permit the dissection of

insulin’s effects on the CNS from its direct peripheral actions e.g. in liver [30] and adipose tissue [31].

These methodological limitations are avoided by the intranasal (IN) route of administration that has

been shown in humans to bypass the BBB and effectively deliver insulin as well as other peptide

hormones to the CNS within one hour after administration in the absence of relevant systemic

absorption [32]. Accordingly, findings in animals demonstrate that intranasally administered

neuropeptides reach brain structures involved in the regulation of metabolism and cognition [33,34].

Intra-neuronal transport of neuropeptides from the nasal cavity to the olfactory bulb takes several

hours [35]. Thus, extra-neuronal passage through intercellular clefts of the olfactory epithelium,

situated on the superior turbinate and opposite the nasal septum [36], is assumed to be the preferential

path of peptide transport into the CNS compartment [32,37], with additional transport along trigeminal

nerve branches to brainstem regions [38].

        IN administration of insulin preparations for the purpose of systemic insulin substitution, i.e.,

as an alternative approach to subcutaneous insulin injection, is not within the scope of this review.




                                                                                                         4
Information on this aspect of nasal insulin administration can be found elsewhere [e.g. references

39,40].



Insulin modulates neurobehavioral measures of brain activity and cognition in humans

Insulin effects on human brain activity have been revealed in a number of studies relying on different

methodological approaches. CNS responses to IN insulin were observed in the form of distinct

alterations in auditory evoked electroencephalographic brain potential responses during an oddball-

paradigm in healthy men while peripheral blood glucose levels remained unchanged [41] . In a related

study, the IN administration of 60 international units of insulin induced a negative shift in direct

current brain potentials that was also found after IV bolus injection of the hormone [42]. Both the IN

and the intravenous effects emerged within 20 min after insulin administration, indicating that

increases in systemic insulin concentrations are rapidly reported to the brain and that IN delivery of

the compound bypassing the body periphery can have a comparable impact on brain activity. The

impact of systemic insulin on cerebrocortical activity was likewise measured in euglycemic-

hyperinsulinemic clamp studies that utilized magnetoencephalographic (MEG) recordings and

demonstrated that obesity [43,44] and the fat-mass and obesity associated (FTO) allele variant

rs8050136 [45] modify insulin’s effects on cerebrocortical beta- and theta-wave activity .

          Experiments employing functional magnetic resonance imaging (fMRI) have shown a positive

relationship between plasma insulin levels and activation of the right hippocampus in response to

viewing photographs of high-caloric food items [46]. In some contrast to these results, in another

fMRI study, food picture-related activity of this and other brain regions was found to be reduced after

IN insulin in comparison to placebo administration [47], raising the question if insulin acting on the

hippocampus is relevant for the regulation of ingestive behavior. On the other hand, the hippocampus

is highly relevant for the formation and maintenance of declarative memory, i.e. memory for facts and

episodes that is accessible to conscious recollection [for review see reference 48]. The ability to

acquire and retain memories depends on synaptic plasticity. Thus, long-term potentiation (LTP) and

long-term depression (LTD) of synaptic transmission, i.e. the augmentation or reduction of synaptic

efficacy are assumed to be important modulators of the strength of a memory representation [49,50].


                                                                                                     5
Several studies indicate that insulin contributes to changes in hippocampal synaptic plasticity by

potentiating LTD and LTP, respectively, at different synapses [for review see reference 51]. Moreover,

insulin receptors have been found to increase synapse density and dendritic plasticity in structures that

process visual input [52]. In addition to these mechanims, insulin may promote glucose utilization of

neuronal networks [53]. Although globally glucose transport to the CNS is assumed to be insulin-

independent [54-56], hyperinsulinemia has been shown          in rodents to exert effects on glucose

metabolism in regions like the anterior hypothalamus and the basolateral amygdale [57].

        In accordance with insulin’s effects on synaptic plasticity [52] and regional glucose uptake

[52], central nervous administration of the hormone via the IN route has been shown to improve

memory functions in studies in healthy humans [23,58,59]. In experiments performed in our lab, a

declarative memory test was conducted at the beginning and end of eight weeks of IN insulin

treatment (160 international units/d). In brief, lists of 30 words were presented and in addition to an

immediate recall 3 min after presentation, in a delayed recall one week later subjects wrote down all

words they still remembered. The delayed recall of words was significantly improved after eight

weeks of IN insulin administration whereas immediate word recall and non-declarative memory

functions were not affected [58]. In line with the strong accumulation of insulin receptors in

hippocampal and cortical brain structures [60], this finding indicates that insulin signaling contributes

to the formation of declarative, hippocampus-dependent memory contents. Noteworthy, beneficial

effects of IN insulin on declarative memory are not restricted to healthy subjects but have also been

shown in memory-impaired subjects [e.g. references 61,62]. Suzanne Craft and co-workers performed

a study in adults with mild cognitive impairments including amnestic symptoms (e.g., due to

Alzheimer´s disease) who were treated with IN insulin over a period of three weeks (2x20

international units/d) [61]. The primary outcome measure was the recall of a story containing 44

informational bits to which subjects listened and that they were asked to recall immediately and after a

20-minute delay. Patients treated with insulin showed significantly increased memory savings over the

21-day period compared to placebo. Considering reports of impaired brain glucose metabolism in

Alzheimer’s disease [63-65], it might be speculated that these effects and, in particular, acute insulin-

induced enhancements of cognitive function in memory-impaired patients occurring within minutes


                                                                                                       6
[66] at least in part derive from increases in cerebral glucose metabolism. In related animal studies, IN

administration of the peptide slowed the development of diabetes-induced brain changes in a murine

model of type 1 diabetes [67].

        CNS insulin signaling has been linked not only to cognitive but also to emotional functions of

the brain. Most recently, lentivirus-mediated downregulation of hypothalamic insulin receptor

expression in rats has been shown to elicit depressive and anxiety-like behaviors [68]. Vice versa, the

8-week IN insulin treatment described above induced an improvement in rated mood in our human

subjects [58]. In mice, IN insulin enhanced object-memory and induced anxiolytic behavioral effects

[69]. However, in mice with impaired glucose tolerance due to diet-induced obesity receiving the same

dose of IN insulin both effects were abrogated [69]. These findings suggest that disturbed CNS insulin

signaling/CNS insulin resistance might link metabolic disorders like obesity with cognitive

impairments and also depressive symptoms. Further evidence for this assumption is discussed below.



CNS insulin signaling and peripheral metabolism

In animal experiments, brain insulin signaling has emerged as an important regulator of energy

balance [70-72]. Insulin’s net effect on energy homeostasis depends on several factors. Whereas

intravenously administered insulin exerts direct peripheral and, after BBB transport, central nervous

effects, intransally administered insulin selectively targets the CNS. In this context it is interesting to

note that the central nervous action of insulin on energy homeostasis partly opposes its peripheral

effects. Whereas after peripheral (IV or subcutaneous) administration insulin acts as an anabolic

hormone by promoting weight gain in form of muscle and fat mass [73,74], IN and ICV insulin

administration in humans and animals, respectively, induces catabolic effects by reducing food intake

[19,23] and as a consequence body fat content [20,21] particularly in the male organism [21-23]. In

parallel, central insulin exerts an anabolic impact on adipose tissue: in addition to the inhibition of

lipolysis by peripheral insulin [75,76], CNS insulin has been found to likewise inhibit lipolysis and

also to enhance lipogenesis [75,77,78]. Recent murine data also suggest that hypothalamic insulin

signaling potentiates brown adipose tissue thermogenesis through inhibition of warm sensitive neurons

[79]. Our group corroborated these findings in humans by demonstrating that IN insulin enhances


                                                                                                         7
postprandial thermogenesis [80]. Thus, the catabolic effect of IN insulin appears to stem from reduced

energy intake [21,23] and increased energy expenditure [79,80] alike.

        Within the last decade, evidence has amounted that the impact of brain insulin signaling on

energy balance extends to glucose homeostasis. Hepatic glucose metabolism is an important

determinant of euglycemia [81]. By glycogenesis on the one hand and glycogenolysis and

gluconeogenesis on the other hand the liver stabilizes plasma glucose concentrations during

(postprandial) glucose abundance and (fasting) glucose depletion, respectively [82]. These processes

have long been known to be mediated by direct insulin action on hepatic insulin receptors and indirect

insulin effects on liver functions, including the downregulation of glucagon secretion and circulating

plasma nonesterified fatty acid concentrations [for      review see reference 83]. However, hepatic

glucose metabolism also seems to be under the control of a brain-liver axis. Obici and co-workers

have shown in rodents that genetic downregulation of hypothalamic insulin receptor expression

disinhibits hepatic glucose production [84]. This finding clearly hints at a reduction in hepatic insulin

sensitivity as a consequence of impaired hypothalamic insulin signaling. Fittingly, insulin has been

found to open ATP-sensitive potassium channels on glucose-responsive hypothalamic neurons and

the resulting neuronal hyperpolarization seems to be responsible for the vagal transmission of a signal

that downregulates hepatic glucose production [14,84]. However, in an experiment in dogs,

quadrupling the concentration of circulating insulin selectively in brain afferent arteries did not

enhance the inhibition of hepatic glucose production [85] which leaves open the question whether the

contribution of hypothalamic insulin signaling to insulin’s hepatic effects has a species-dependent

component.

        In humans it has recently been shown that IV pretreatment with insulin potentiates glucose-

induced pancreatic insulin secretion by 40%, suggesting that circulating insulin exerts a direct positive

feedback on its own secretion [86]. Interestingly, a similar effect was found for brain insulin over 30

years ago in dogs, where ICV insulin administration increased pancreatic insulin secretion via a feed-

forward mechanism [87,88]. This brain-pancreatic crosstalk involving the vagal nerve has been

hypothesized to be another regulator of blood glucose. While effects of CNS insulin on peripheral

insulin sensitivity and pancreatic insulin secretion in humans are largely unexplored, two recent


                                                                                                       8
studies have gathered evidence that insulin delivery to the brain does affect peripheral glucose

metabolism. IN insulin administration before intake of a liquid meal reduced postprandial circulating

insulin levels in healthy subjects while plasma glucose levels were unchanged in comparison to

placebo [80]. This finding suggests that brain insulin administration can enhance postprandial

peripheral insulin sensitivity, adding to the feed-forward effect of brain insulin on pancreatic insulin

secretion observed in animals. Another recent set of experiments performed by Stockhorst and

colleagues indicates that such brain-pancreatic cross-talk is accessible to classical conditioning [89].

On day 1, the investigators administered IN insulin vs. placebo that both have the same specific odor

due to the formulation with meta-cresol (a stabilizing agent in insulin solutions). They found an

increase in serum insulin concentrations and a reduction in blood glucose levels (within the

euglycemic range) after insulin compared with placebo, suggesting that activation of the brain-liver

axis enhanced pancreatic insulin secretion. On day 2, the procedure was repeated but placebo was

administered in both groups with the smell of meta-cresol functioning as a conditioned stimulus. Here,

the presentation of the conditioned stimulus alone after pretreatment with IN insulin on day 1 was

sufficient to cause an even enhanced increasing effect on serum insulin concentrations, which points to

a significant contribution of neurocognitive learning mechanisms to the regulation of peripheral

glucose homeostasis by brain insulin.


Central nervous system insulin resistance

Peripheral insulin resistance is a well-known feature of type 2 diabetes and obesity. Insulin resistance

in central nervous structures might likewise contribute to the development not only of these metabolic

disorders but also of cognitive impairments. Raising systemic insulin levels by IV infusion results in

increased CSF insulin concentrations in healthy, normal-weight subjects [27]. Obese subjects and

Alzheimer patients seem to display relatively decreased CSF insulin concentrations suggesting

reduced insulin transport across the BBB [90,91]. Likewise, in comparison to normal-weight subjects,

overweight humans show a decrease in MEG-recorded cortical activity during hyperinsulinemic-

euglycemic clamp experiments that is directly related to the amount of body fat and the degree of

peripheral insulin resistance [43]. These findings support the notion that in obesity both BBB insulin

transport and the central nervous sensitivity to insulin are reduced. Related studies relying on acute

                                                                                                      9
and prolonged IN administration of the hormone have refined this picture. In further MEG-based

experiments, IN insulin acutely increased cerebrocortical activity in response to food vs. non-food

pictures in lean but not in obese subjects [92]. Long-term (8 weeks) administration of IN insulin in

obese subjects failed to affect body weight and fat mass [93] but still enhanced declarative memory

and dampened HPA-axis activity to a degree comparable with that observed in normal-weight subjects

[58,94]. This differential insulin response implies that brain regions involved in energy and glucose

homeostasis might be particularly prone to develop insulin resistance in obesity. In accordance with

these findings, in rats diet-induced obesity abolishes the catabolic actions of ICV insulin

administration, and a reduction in insulin receptor density in the hypothalamic arcuate nucleus causes

hyperphagia (and, as mentioned above, also disinhibits hepatic glucose production) [84]. On a

molecular level, activation of the PI-3 signaling cascade subsequent to the binding of insulin to its

receptor mediates the majority of central nervous insulin effects on energy homeostasis [for review see

reference 95], while disturbances of this pathway are regarded as a likely cause of neuronal insulin

resistance [for review see 96].

        Reduced central nervous sensitivity might represent a pathophysiological link between

obesity, peripheral insulin resistance and cognitive disorders that have been found to be significantly

related in epidemiological studies [97,98]. Central insulin resistance seems to impair neuronal

plasticity via detrimental effects on glutamatergic and cholinergic pathways [99,100]. Such processes

are assumed to be influenced by genetic predisposition. One indicator for this assumption is that IN

insulin administration to patients with memory impairments improved memory functions

predominantly in non-carriers of the APOE*E4 allele, a risk factor for the development of Alzheimer’s

disease [62,66; for review see reference 101]. In this context, several further genetic polymorphisms

associated with reduced central nervous responsiveness to insulin have been characterized. Subjects

with the FTO gene polymorphism rs8050136 as well as carriers of the Gly972Arg polymorphism of

the Insulin Receptor Substrate 1 (IRS1) exhibit a decreased cerebrocortical response to IV insulin

[43,45]. Interestingly, these data support the suggested connection between the FTO variant and a

hyperphagic phenotype [102] characterised by a predilection for energy-dense foods [103], which

might involve decreased insulin sensitivity of food reward-related brain pathways [104].


                                                                                                    10
Potential therapies aimed at overcoming CNS insulin resistance might include the IN

administration of insulin but could also rely on the insulin-sensitizing properties of the peroxisome

proliferator-activated receptor-ƴ (PPAR-ƴ) agonist rosiglitazone [105-108] and of metformin [109].

Regarding glucose homeostasis, enhancing central nervous/hypothalamic insulin signaling by insulin

administration to the brain might reinforce a vagally transmitted inhibitory signal on hepatic

gluconeogenesis [110], whose disinhibition represents a hallmark of type 2 diabetes and peripheral

insulin resistance [84]. The latter, moreover, appears to be highly associated with central nervous

insulin resistance [111]. IN insulin has been found to reduce HPA axis activity [58,93,94], thus

potentially opposing visceral adiposity and cognitive impairments due to stress-induced chronic HPA-

axis overactivation [112-114]. Moreover, in light of IN insulin’s acute memory-improving effects in

patients with mild cognitive impairments [61,62], the compound might ameliorate the harmful effects

on cognition that obesity and diabetes are suspected to engender [97].

        Notwithstanding these encouraging results, some caveats need to be addressed. In light of the

hyperinsulinemia that accompanies peripheral insulin resistance, it might be argued that the reduction

of CSF insulin levels observed in obese subjects [91] could represent a protective mechanism that

limits central nervous hyperinsulinemia and the potentially detrimental sequelae of cellular insulin

resistance inside the brain. Although speculative, this assumption is in line with findings that hint at a

dose-dependent directionality of central insulin’s impact on memory function. For example, acute IN

insulin administration to Alzheimer patients improved verbal memory recall only at lower doses (20

international units), whereas higher doses (up to 60 international units) were not effective and, in

carriers of the APOE*E4 allele, even induced a decline in memory performance [62]. In healthy

subjects, the induction of acute moderate euglycemic hyperinsulinemia has been found to trigger

central nervous system inflammation and beta-amyloid formation [115], both of which are known risk

factors for the development of cognitive impairments. The notion that brain hyperinsulinemia might

promote central nervous insulin resistance is supported by a recent in vitro-study showing that

prolonged (4-24 h) exposure of hypothalamic cells to high concentrations of insulin led to inactivation

and degradation of the insulin receptor and IRS-1 [116]. Against this background and considering that

long-term data on therapeutic and side effects of IN insulin in humans are so far lacking, obviously

                                                                                                       11
much work is still necessary to sound the potential of brain insulin administration in the treatment of

cognitive and metabolic disorders.



Conclusion

Insulin binding to its receptors in the brain impacts a number of pivotal physiological functions,

including energy uptake and expenditure, glucose metabolism, adipocyte function and cognition. The

experimental data briefly summarized here clearly implicate CNS insulin resistance as a potentially

important factor in the pathophysiology of obesity and systemic insulin resistance as well as of

cognitive impairments like Alzheimer’s disease. Moreover, the association between these disorders

found in epidemiological investigations may at least partly rely on dysregulated central nervous

insulin signaling. Although further studies are needed to substantiate the promising results of proof-of-

concept experiments on central nervous insulin administration, overcoming insulin resistance in the

brain may prove a viable therapeutic option in the treatment of these increasingly prevalent afflictions.




Acknowledgments

This work was supported by Deutsche Forschungsgemeinschaft (KFO126/B5), Germany. The funding

source   had    no   input   in   the   preparation,   review,   or   approval    of   the   manuscript.




                                                                                                      12
Reference List

 1. Bernard C. Leçons de physiologie expérimentale appliquée à la médecine faites au
    Collège de France. Baillère et Fils Paris, France 1855;296-313.

 2. Havrankova J., Roth J., Brownstein M. Insulin receptors are widely distributed in the
    central nervous system of the rat. Nature 1978; 272:827-829.

 3. Sara V.R., Hall K., Von H.H., Humbel R., Sjogren B., Wetterberg L. Evidence for the
    presence of specific receptors for insulin-like growth factors 1 (IGE-1) and 2 (IGF-2)
    and insulin throughout the adult human brain. Neurosci Lett 1982; 34:39-44.

 4. Potau N., Escofet M.A., Martinez M.C. Ontogenesis of insulin receptors in human
    cerebral cortex. J Endocrinol Invest 1991; 14:53-58.

 5. Marks J.L., Porte D., Jr., Stahl W.L., Baskin D.G. Localization of insulin receptor
    mRNA in rat brain by in situ hybridization. Endocrinology 1990; 127:3234-3236.

 6. Devaskar S.U., Giddings S.J., Rajakumar P.A., Carnaghi L.R., Menon R.K., Zahm
    D.S. Insulin gene expression and insulin synthesis in mammalian neuronal cells. J Biol
    Chem 1994; 269:8445-8454.

 7. Hrytsenko O., Wright J.R., Jr., Morrison C.M., Pohajdak B. Insulin expression in the
    brain and pituitary cells of tilapia (Oreochromis niloticus). Brain Res 2007; 1135:31-
    40.

 8. Steen E., Terry B.M., Rivera E.J. et al. Impaired insulin and insulin-like growth factor
    expression and signaling mechanisms in Alzheimer's disease--is this type 3 diabetes? J
    Alzheimers Dis 2005; 7:63-80.

 9. Woods S.C., Seeley R.J., Baskin D.G., Schwartz M.W. Insulin and the blood-brain
    barrier. Curr Pharm Des 2003; 9:795-800.

10. Banks W.A., Jaspan J.B., Kastin A.J. Selective, physiological transport of insulin
    across the blood-brain barrier: novel demonstration by species-specific
    radioimmunoassays. Peptides 1997; 18:1257-1262.

11. Schwartz M.W., Bergman R.N., Kahn S.E. et al. Evidence for entry of plasma insulin
    into cerebrospinal fluid through an intermediate compartment in dogs. Quantitative
    aspects and implications for transport. J Clin Invest 1991; 88:1272-1281.

12. Air E.L., Benoit S.C., Clegg D.J., Seeley R.J., Woods S.C. Insulin and leptin combine
    additively to reduce food intake and body weight in rats. Endocrinology 2002;
    143:2449-2452.

13. Bruning J.C., Gautam D., Burks D.J. et al. Role of brain insulin receptor in control of
    body weight and reproduction. Science 2000; 289:2122-2125.

14. Pocai A., Lam T.K., Gutierrez-Juarez R. et al. Hypothalamic K(ATP) channels control
    hepatic glucose production. Nature 2005; 434:1026-1031.


                                                                                         13
15. Rulifson E.J., Kim S.K., Nusse R. Ablation of insulin-producing neurons in flies:
    growth and diabetic phenotypes. Science 2002; 296:1118-1120.

16. Chiu S.L., Cline H.T. Insulin receptor signaling in the development of neuronal
    structure and function. Neural Dev 2010; 5:7.

17. Seeley R.J., Woods S.C. Monitoring of stored and available fuel by the CNS:
    implications for obesity. Nat Rev Neurosci 2003; 4:901-909.

18. Woods S.C., D'Alessio D.A. Central control of body weight and appetite. J Clin
    Endocrinol Metab 2008; 93:S37-S50.

19. Honda K., Kamisoyama H., Saneyasu T., Sugahara K., Hasegawa S. Central
    administration of insulin suppresses food intake in chicks. Neurosci Lett 2007;
    423:153-157.

20. Brown L.M., Clegg D.J., Benoit S.C., Woods S.C. Intraventricular insulin and leptin
    reduce food intake and body weight in C57BL/6J mice. Physiol Behav 2006; 89:687-
    691.

21. Hallschmid M., Benedict C., Schultes B., Fehm H.L., Born J., Kern W. Intranasal
    insulin reduces body fat in men but not in women. Diabetes 2004; 53:3024-3029.

22. Clegg D.J., Riedy C.A., Smith K.A., Benoit S.C., Woods S.C. Differential sensitivity
    to central leptin and insulin in male and female rats. Diabetes 2003; 52:682-687.

23. Benedict C., Kern W., Schultes B., Born J., Hallschmid M. Differential sensitivity of
    men and women to anorexigenic and memory-improving effects of intranasal insulin.
    J Clin Endocrinol Metab 2008; 93:1339-1344.

24. Hallschmid M., Schultes B. Central nervous insulin resistance: a promising target in
    the treatment of metabolic and cognitive disorders? Diabetologia 2009; 52:2264-2269.

25. Woods S.C., Lotter E.C., McKay L.D., Porte D., Jr. Chronic intracerebroventricular
    infusion of insulin reduces food intake and body weight of baboons. Nature 1979;
    282:503-505.

26. van Dijk G., de Groote C., Chavez M., van der Werf Y., Steffens A.B., Strubbe J.H.
    Insulin in the arcuate nucleus of the hypothalamus reduces fat consumption in rats.
    Brain Res 1997; 777:147-152.

27. Wallum B.J., Taborsky G.J., Jr., Porte D., Jr. et al. Cerebrospinal fluid insulin levels
    increase during intravenous insulin infusions in man. J Clin Endocrinol Metab 1987;
    64:190-194.

28. Tesfaye N., Seaquist E.R. Neuroendocrine responses to hypoglycemia. Ann N Y Acad
    Sci 2010; 1212:12-28.

29. Mitrakou A., Ryan C., Veneman T. et al. Hierarchy of glycemic thresholds for
    counterregulatory hormone secretion, symptoms, and cerebral dysfunction. Am J
    Physiol 1991; 260:E67-E74.



                                                                                         14
30. Cherrington A.D., Edgerton D., Sindelar D.K. The direct and indirect effects of insulin
    on hepatic glucose production in vivo. Diabetologia 1998; 41:987-996.

31. Urbanet R., Pilon C., Calcagno A. et al. Analysis of insulin sensitivity in adipose
    tissue of patients with primary aldosteronism. J Clin Endocrinol Metab 2010;
    95:4037-4042.

32. Born J., Lange T., Kern W., McGregor G.P., Bickel U., Fehm H.L. Sniffing
    neuropeptides: a transnasal approach to the human brain. Nat Neurosci 2002; 5:514-
    516.

33. Alcala-Barraza S.R., Lee M.S., Hanson L.R., McDonald A.A., Frey W.H., McLoon
    L.K. Intranasal delivery of neurotrophic factors BDNF, CNTF, EPO, and NT-4 to the
    CNS. J Drug Target 2010; 18:179-190.

34. Fliedner S., Schulz C., Lehnert H. Brain uptake of intranasally applied radioiodinated
    leptin in Wistar rats. Endocrinology 2006; 147:2088-2094.

35. Thorne R.G., Emory C.R., Ala T.A., Frey W.H. Quantitative analysis of the olfactory
    pathway for drug delivery to the brain. Brain Res 1995; 692:278-282.

36. Illum L. Transport of drugs from the nasal cavity to the central nervous system. Eur J
    Pharm Sci 2000; 11:1-18.

37. Dhuria S.V., Hanson L.R., Frey W.H. Intranasal delivery to the central nervous
    system: mechanisms and experimental considerations. J Pharm Sci 2010; 99:1654-
    1673.

38. Thorne R.G., Pronk G.J., Padmanabhan V., Frey W.H. Delivery of insulin-like growth
    factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways
    following intranasal administration. Neuroscience 2004; 127:481-496.

39. Henkin R.I. Inhaled insulin-intrapulmonary, intranasal, and other routes of
    administration: mechanisms of action. Nutrition 2010; 26:33-39.

40. Duan X., Mao S. New strategies to improve the intranasal absorption of insulin. Drug
    Discov Today 2010; 15:416-427.

41. Kern W., Born J., Schreiber H., Fehm H.L. Central nervous system effects of
    intranasally administered insulin during euglycemia in men. Diabetes 1999; 48:557-
    563.

42. Hallschmid M., Schultes B., Marshall L. et al. Transcortical direct current potential
    shift reflects immediate signaling of systemic insulin to the human brain. Diabetes
    2004; 53:2202-2208.

43. Tschritter O., Preissl H., Hennige A.M. et al. The cerebrocortical response to
    hyperinsulinemia is reduced in overweight humans: a magnetoencephalographic study.
    Proc Natl Acad Sci U S A 2006; 103:12103-12108.

44. Tschritter O., Preissl H., Hennige A.M. et al. The insulin effect on cerebrocortical
    theta activity is associated with serum concentrations of saturated nonesterified Fatty
    acids. J Clin Endocrinol Metab 2009; 94:4600-4607.

                                                                                        15
45. Tschritter O., Preissl H., Yokoyama Y., Machicao F., Haring H.U., Fritsche A.
    Variation in the FTO gene locus is associated with cerebrocortical insulin resistance in
    humans. Diabetologia 2007; 50:2602-2603.

46. Wallner-Liebmann S., Koschutnig K., Reishofer G. et al. Insulin and hippocampus
    activation in response to images of high-calorie food in normal weight and obese
    adolescents. Obesity (Silver Spring) 2010; 18:1552-1557.

47. Guthoff M., Grichisch Y., Canova C. et al. Insulin modulates food-related activity in
    the central nervous system. J Clin Endocrinol Metab 2010; 95:748-755.

48. Diekelmann S., Born J. The memory function of sleep. Nat Rev Neurosci 2010;
    11:114-126.

49. Feldman D.E. Synaptic mechanisms for plasticity in neocortex. Annu Rev Neurosci
    2009; 32:33-55.

50. Huang C.C., Lee C.C., Hsu K.S. The role of insulin receptor signaling in synaptic
    plasticity and cognitive function. Chang Gung Med J 2010; 33:115-125.

51. Moult P.R., Harvey J. Hormonal regulation of hippocampal dendritic morphology and
    synaptic plasticity. Cell Adh Migr 2008; 2:269-275.

52. Chiu S.L., Chen C.M., Cline H.T. Insulin receptor signaling regulates synapse
    number, dendritic plasticity, and circuit function in vivo. Neuron 2008; 58:708-719.

53. Craft S., Watson G.S. Insulin and neurodegenerative disease: shared and specific
    mechanisms. Lancet Neurol 2004; 3:169-178.

54. Seaquist E.R., Damberg G.S., Tkac I., Gruetter R. The effect of insulin on in vivo
    cerebral glucose concentrations and rates of glucose transport/metabolism in humans.
    Diabetes 2001; 50:2203-2209.

55. Fanelli C.G., Dence C.S., Markham J. et al. Blood-to-brain glucose transport and
    cerebral glucose metabolism are not reduced in poorly controlled type 1 diabetes.
    Diabetes 1998; 47:1444-1450.

56. Hasselbalch S.G., Knudsen G.M., Videbaek C. et al. No effect of insulin on glucose
    blood-brain barrier transport and cerebral metabolism in humans. Diabetes 1999;
    48:1915-1921.

57. Doyle P., Cusin I., Rohner-Jeanrenaud F., Jeanrenaud B. Four-day hyperinsulinemia in
    euglycemic conditions alters local cerebral glucose utilization in specific brain nuclei
    of freely moving rats. Brain Res 1995; 684:47-55.

58. Benedict C., Hallschmid M., Hatke A. et al. Intranasal insulin improves memory in
    humans. Psychoneuroendocrinology 2004; 29:1326-1334.

59. Benedict C., Hallschmid M., Schmitz K. et al. Intranasal insulin improves memory in
    humans: superiority of insulin aspart. Neuropsychopharmacology 2007; 32:239-243.




                                                                                         16
60. Unger J.W., Livingston J.N., Moss A.M. Insulin receptors in the central nervous
    system: localization, signalling mechanisms and functional aspects. Prog Neurobiol
    1991; 36:343-362.

61. Reger M.A., Watson G.S., Green P.S. et al. Intranasal insulin improves cognition and
    modulates beta-amyloid in early AD. Neurology 2008; 70:440-448.

62. Reger M.A., Watson G.S., Green P.S. et al. Intranasal insulin administration dose-
    dependently modulates verbal memory and plasma amyloid-beta in memory-impaired
    older adults. J Alzheimers Dis 2008; 13:323-331.

63. Mosconi L., Pupi A., De Leon M.J. Brain glucose hypometabolism and oxidative
    stress in preclinical Alzheimer's disease. Ann N Y Acad Sci 2008; 1147:180-195.

64. Mosconi L., Mistur R., Switalski R. et al. Declining brain glucose metabolism in
    normal individuals with a maternal history of Alzheimer disease. Neurology 2009;
    72:513-520.

65. Yasuno F., Imamura T., Hirono N. et al. Age at onset and regional cerebral glucose
    metabolism in Alzheimer's disease. Dement Geriatr Cogn Disord 1998; 9:63-67.

66. Reger M.A., Watson G.S., Frey W.H. et al. Effects of intranasal insulin on cognition
    in memory-impaired older adults: modulation by APOE genotype. Neurobiol Aging
    2006; 27:451-458.

67. Francis G.J., Martinez J.A., Liu W.Q. et al. Intranasal insulin prevents cognitive
    decline, cerebral atrophy and white matter changes in murine type I diabetic
    encephalopathy. Brain 2008; 131:3311-3334.

68. Grillo C.A., Piroli G.G., Kaigler K.F., Wilson S.P., Wilson M.A., Reagan L.P.
    Downregulation of hypothalamic insulin receptor expression elicits depressive-like
    behaviors in rats. Behav Brain Res 2011; 222:230-235.

69. Marks D.R., Tucker K., Cavallin M.A., Mast T.G., Fadool D.A. Awake intranasal
    insulin delivery modifies protein complexes and alters memory, anxiety, and olfactory
    behaviors. J Neurosci 2009; 29:6734-6751.

70. Lin H.V., Plum L., Ono H. et al. Divergent regulation of energy expenditure and
    hepatic glucose production by insulin receptor in agouti-related protein and POMC
    neurons. Diabetes 2010; 59:337-346.

71. Morton G.J., Cummings D.E., Baskin D.G., Barsh G.S., Schwartz M.W. Central
    nervous system control of food intake and body weight. Nature 2006; 443:289-295.

72. Porte D., Jr., Baskin D.G., Schwartz M.W. Insulin signaling in the central nervous
    system: a critical role in metabolic homeostasis and disease from C. elegans to
    humans. Diabetes 2005; 54:1264-1276.

73. McFarlane S.I. Insulin therapy and type 2 diabetes: management of weight gain. J Clin
    Hypertens (Greenwich ) 2009; 11:601-607.

74. Russell-Jones D., Khan R. Insulin-associated weight gain in diabetes--causes, effects
    and coping strategies. Diabetes Obes Metab 2007; 9:799-812.

                                                                                      17
75. Enoksson S., Degerman E., Hagstrom-Toft E., Large V., Arner P. Various
    phosphodiesterase subtypes mediate the in vivo antilipolytic effect of insulin on
    adipose tissue and skeletal muscle in man. Diabetologia 1998; 41:560-568.

76. Stralfors P., Bjorgell P., Belfrage P. Hormonal regulation of hormone-sensitive lipase
    in intact adipocytes: identification of phosphorylated sites and effects on the
    phosphorylation by lipolytic hormones and insulin. Proc Natl Acad Sci U S A 1984;
    81:3317-3321.

77. Koch L., Wunderlich F.T., Seibler J. et al. Central insulin action regulates peripheral
    glucose and fat metabolism in mice. J Clin Invest 2008; 118:2132-2147.

78. Scherer T., O'Hare J., Diggs-Andrews K. et al. Brain insulin controls adipose tissue
    lipolysis and lipogenesis. Cell Metab 2011; 13:183-194.

79. Sanchez-Alavez M., Tabarean I.V., Osborn O. et al. Insulin causes hyperthermia by
    direct inhibition of warm-sensitive neurons. Diabetes 2010; 59:43-50.

80. Benedict C., Brede S., Schioth H.B. et al. Intranasal insulin enhances postprandial
    thermogenesis and lowers postprandial serum insulin levels in healthy men. Diabetes
    2011; 60:114-118.

81. Bergman R.N. Orchestration of glucose homeostasis: from a small acorn to the
    California oak. Diabetes 2007; 56:1489-1501.

82. Cherrington A.D. Banting Lecture 1997. Control of glucose uptake and release by the
    liver in vivo. Diabetes 1999; 48:1198-1214.

83. Cherrington A.D. The role of hepatic insulin receptors in the regulation of glucose
    production. J Clin Invest 2005; 115:1136-1139.

84. Obici S., Feng Z., Karkanias G., Baskin D.G., Rossetti L. Decreasing hypothalamic
    insulin receptors causes hyperphagia and insulin resistance in rats. Nat Neurosci 2002;
    5:566-572.

85. Edgerton D.S., Lautz M., Scott M. et al. Insulin's direct effects on the liver dominate
    the control of hepatic glucose production. J Clin Invest 2006; 116:521-527.

86. Bouche C., Lopez X., Fleischman A. et al. Insulin enhances glucose-stimulated insulin
    secretion in healthy humans. Proc Natl Acad Sci U S A 2010; 107:4770-4775.

87. Chen M., Woods S.C., Porte D., Jr. Effect of cerebral intraventricular insulin on
    pancreatic insulin secretion in the dog. Diabetes 1975; 24:910-914.

88. Woods S.C., Porte D., Jr. Effect of intracisternal insulin on plasma glucose and insulin
    in the dog. Diabetes 1975; 24:905-909.

89. Stockhorst U., de Fries D., Steingrueber H.J., Scherbaum W.A. Unconditioned and
    conditioned effects of intranasally administered insulin vs placebo in healthy men: a
    randomised controlled trial. Diabetologia 2011; 54:1502-1506.




                                                                                         18
90. Craft S., Peskind E., Schwartz M.W., Schellenberg G.D., Raskind M., Porte D., Jr.
     Cerebrospinal fluid and plasma insulin levels in Alzheimer's disease: relationship to
     severity of dementia and apolipoprotein E genotype. Neurology 1998; 50:164-168.

 91. Kern W., Benedict C., Schultes B. et al. Low cerebrospinal fluid insulin levels in
     obese humans. Diabetologia 2006; 49:2790-2792.

 92. Guthoff M., Stingl K.T., Tschritter O. et al. The insulin-mediated modulation of
     visually evoked magnetic fields is reduced in obese subjects. PLoS One 2011;
     6:e19482.

 93. Hallschmid M., Benedict C., Schultes B., Born J., Kern W. Obese men respond to
     cognitive but not to catabolic brain insulin signaling. Int J Obes (Lond) 2008; 32:275-
     282.

 94. Bohringer A., Schwabe L., Richter S., Schachinger H. Intranasal insulin attenuates the
     hypothalamic-pituitary-adrenal    axis    response     to    psychosocial      stress.
     Psychoneuroendocrinology 2008; 33:1394-1400.

 95. Marino J.S., Xu Y., Hill J.W. Central insulin and leptin-mediated autonomic control of
     glucose homeostasis. Trends Endocrinol Metab 2011.

 96. Schwartz M.W., Porte D., Jr. Diabetes, obesity, and the brain. Science 2005; 307:375-
     379.

 97. Whitmer R.A., Gustafson D.R., Barrett-Connor E., Haan M.N., Gunderson E.P., Yaffe
     K. Central obesity and increased risk of dementia more than three decades later.
     Neurology 2008; 71:1057-1064.

 98. Raffaitin C., Feart C., Le G.M. et al. Metabolic syndrome and cognitive decline in
     French elders: the Three-City Study. Neurology 2011; 76:518-525.

 99. Trudeau F., Gagnon S., Massicotte G. Hippocampal synaptic plasticity and glutamate
     receptor regulation: influences of diabetes mellitus. Eur J Pharmacol 2004; 490:177-
     186.

100. Kodl C.T., Seaquist E.R. Cognitive dysfunction and diabetes mellitus. Endocr Rev
     2008; 29:494-511.

101. Raber J., Huang Y., Ashford J.W. ApoE genotype accounts for the vast majority of
     AD risk and AD pathology. Neurobiol Aging 2004; 25:641-650.

102. Speakman J.R., Rance K.A., Johnstone A.M. Polymorphisms of the FTO gene are
     associated with variation in energy intake, but not energy expenditure. Obesity (Silver
     Spring) 2008; 16:1961-1965.

103. Cecil J.E., Tavendale R., Watt P., Hetherington M.M., Palmer C.N. An obesity-
     associated FTO gene variant and increased energy intake in children. N Engl J Med
     2008; 359:2558-2566.

104. Konner A.C., Klockener T., Bruning J.C. Control of energy homeostasis by insulin
     and leptin: targeting the arcuate nucleus and beyond. Physiol Behav 2009; 97:632-638.


                                                                                         19
105. Landreth G., Jiang Q., Mandrekar S., Heneka M. PPARgamma agonists as
     therapeutics for the treatment of Alzheimer's disease. Neurotherapeutics 2008; 5:481-
     489.

106. Pathan A.R., Gaikwad A.B., Viswanad B., Ramarao P. Rosiglitazone attenuates the
     cognitive deficits induced by high fat diet feeding in rats. Eur J Pharmacol 2008;
     589:176-179.

107. Risner M.E., Saunders A.M., Altman J.F. et al. Efficacy of rosiglitazone in a
     genetically defined population with mild-to-moderate Alzheimer's disease.
     Pharmacogenomics J 2006; 6:246-254.

108. Watson G.S., Cholerton B.A., Reger M.A. et al. Preserved cognition in patients with
     early Alzheimer disease and amnestic mild cognitive impairment during treatment
     with rosiglitazone: a preliminary study. Am J Geriatr Psychiatry 2005; 13:950-958.

109. Gupta A., Bisht B., Dey C.S. Peripheral insulin-sensitizer drug metformin ameliorates
     neuronal insulin resistance and Alzheimer's-like changes. Neuropharmacology 2011;
     60:910-920.

110. Obici S., Zhang B.B., Karkanias G., Rossetti L. Hypothalamic insulin signaling is
     required for inhibition of glucose production. Nat Med 2002; 8:1376-1382.

111. Anthony K., Reed L.J., Dunn J.T. et al. Attenuation of insulin-evoked responses in
     brain networks controlling appetite and reward in insulin resistance: the cerebral basis
     for impaired control of food intake in metabolic syndrome? Diabetes 2006; 55:2986-
     2992.

112. Dallman M.F., Pecoraro N.C., la Fleur S.E. et al. Glucocorticoids, chronic stress, and
     obesity. Prog Brain Res 2006; 153:75-105.

113. Hibberd C., Yau J.L., Seckl J.R. Glucocorticoids and the ageing hippocampus. J Anat
     2000; 197 Pt 4:553-562.

114. Strachan M.W., Reynolds R.M., Frier B.M., Mitchell R.J., Price J.F. The role of
     metabolic derangements and glucocorticoid excess in the aetiology of cognitive
     impairment in type 2 diabetes. Implications for future therapeutic strategies. Diabetes
     Obes Metab 2009; 11:407-414.

115. Fishel M.A., Watson G.S., Montine T.J. et al. Hyperinsulinemia provokes
     synchronous increases in central inflammation and beta-amyloid in normal adults.
     Arch Neurol 2005; 62:1539-1544.

116. Mayer C.M., Belsham D.D. Central insulin signaling is attenuated by long-term
     insulin exposure via insulin receptor substrate-1 serine phosphorylation, proteasomal
     degradation, and lysosomal insulin receptor degradation. Endocrinology 2010; 151:75-
     84.




                                                                                          20
Figure 1. Insulin-mediated crosstalk between brain and body periphery

Intranasal/CNS and systemic insulin affect hepatic glucose production, pancreatic insulin secretion,

adipocyte function, energy homeostasis, and cognitive function. Respective citations in text boxes

refer to the reference list of the main text.




                                                                                                 21
2-4-1-9-5-8-3
                                                                                                                          Moon-Night


                                                                                                                 Intranasal/CNS insulin enhances
    Intranasal insulin                                                                                           declarative (e.g. word pairs) and
 bypasses the BBB and                                                                                              working memory (e.g. number
 achieves maximal CSF                                                                                             sequences) learning [23;58;59].
concentrations within 40
      minutes [32].




  Intranasal                              Insulin reaches the brain via
                                          receptor-mediated saturable
    insulin                                transport across the blood-                                         Intranasal/CNS insulin decreases
                                               brain-barrier [9;10].                    CNS insulin, like          food intake and enhances
           CNS insulin inhibits                                                         systemic insulin,         postprandial thermogenesis
         hepatic gluconeogenesis                                                      inhibits lipolysis and               [21;23;80].
                                                   CNS insulin increases
           via vagal efferences                 pancreatic insulin secretion         stimulates lipogenesis
                 [14;110].                      (feed-forward loop [87;88].)                 [77;78].




        Liver                                           Pancreas
                            Systemic insulin
                                                                                                        Adipocytes
                             inhibits gluco-                                  Systemic insulin
                            neogenesis via                                 inhibits lipolysis and
                             hepatic insulin                                    stimulates
                           receptors [82;83].                              lipogenesis [75;76].



                                                                                                                                                     22

Mais conteúdo relacionado

Mais procurados

Newer insulins in clinical practice
Newer insulins in clinical practiceNewer insulins in clinical practice
Newer insulins in clinical practiceDr. Arun Sharma, MD
 
NOVEL APPROACHES TO INSULIN DELIVERY.
NOVEL APPROACHES TO INSULIN DELIVERY.NOVEL APPROACHES TO INSULIN DELIVERY.
NOVEL APPROACHES TO INSULIN DELIVERY.manish kumar
 
Why we need new analog insulin
Why we need new analog insulinWhy we need new analog insulin
Why we need new analog insulinko ko
 
Rusham presentation on human insulin
Rusham presentation on human insulinRusham presentation on human insulin
Rusham presentation on human insulinRusham Das
 
Dr sherif - Entero insular axis
Dr sherif -       Entero insular axis Dr sherif -       Entero insular axis
Dr sherif - Entero insular axis drsherif36
 
Industrial Production of Insulin
Industrial Production of Insulin Industrial Production of Insulin
Industrial Production of Insulin Zohaib HUSSAIN
 
NOVEL APPROACHES TO DELIVER INSULIN
NOVEL APPROACHES TO DELIVER INSULINNOVEL APPROACHES TO DELIVER INSULIN
NOVEL APPROACHES TO DELIVER INSULINSHASHANK SONI
 
Mechanism of action of insulin
Mechanism of action of insulinMechanism of action of insulin
Mechanism of action of insulinakash mahadev
 
Insulin 301 abbotsford
Insulin 301 abbotsfordInsulin 301 abbotsford
Insulin 301 abbotsfordIhsaan Peer
 

Mais procurados (20)

Newer insulins in clinical practice
Newer insulins in clinical practiceNewer insulins in clinical practice
Newer insulins in clinical practice
 
Insulins And Insulin Delivery
Insulins And Insulin DeliveryInsulins And Insulin Delivery
Insulins And Insulin Delivery
 
Insulin22
Insulin22Insulin22
Insulin22
 
NOVEL APPROACHES TO INSULIN DELIVERY.
NOVEL APPROACHES TO INSULIN DELIVERY.NOVEL APPROACHES TO INSULIN DELIVERY.
NOVEL APPROACHES TO INSULIN DELIVERY.
 
Why we need new analog insulin
Why we need new analog insulinWhy we need new analog insulin
Why we need new analog insulin
 
Insulin+and+Oral
Insulin+and+OralInsulin+and+Oral
Insulin+and+Oral
 
Insulin & Oh Gs(10 13)
Insulin & Oh Gs(10 13)Insulin & Oh Gs(10 13)
Insulin & Oh Gs(10 13)
 
Sleep article
Sleep articleSleep article
Sleep article
 
Insulin old & new
Insulin old & newInsulin old & new
Insulin old & new
 
Rusham presentation on human insulin
Rusham presentation on human insulinRusham presentation on human insulin
Rusham presentation on human insulin
 
Insulin
InsulinInsulin
Insulin
 
C peptide
C  peptideC  peptide
C peptide
 
Insulin
InsulinInsulin
Insulin
 
Basics of Insulin
Basics of InsulinBasics of Insulin
Basics of Insulin
 
Insulin
InsulinInsulin
Insulin
 
Dr sherif - Entero insular axis
Dr sherif -       Entero insular axis Dr sherif -       Entero insular axis
Dr sherif - Entero insular axis
 
Industrial Production of Insulin
Industrial Production of Insulin Industrial Production of Insulin
Industrial Production of Insulin
 
NOVEL APPROACHES TO DELIVER INSULIN
NOVEL APPROACHES TO DELIVER INSULINNOVEL APPROACHES TO DELIVER INSULIN
NOVEL APPROACHES TO DELIVER INSULIN
 
Mechanism of action of insulin
Mechanism of action of insulinMechanism of action of insulin
Mechanism of action of insulin
 
Insulin 301 abbotsford
Insulin 301 abbotsfordInsulin 301 abbotsford
Insulin 301 abbotsford
 

Semelhante a Insulina intranasal e cognição

Insulina e cérebro
Insulina  e  cérebroInsulina  e  cérebro
Insulina e cérebroRuy Pantoja
 
Current Paradigms to Explore the Gut Microbiota Linkage to Neurological Disor...
Current Paradigms to Explore the Gut Microbiota Linkage to Neurological Disor...Current Paradigms to Explore the Gut Microbiota Linkage to Neurological Disor...
Current Paradigms to Explore the Gut Microbiota Linkage to Neurological Disor...Dr Varruchi Sharma
 
Significance of sprouted broccoli
Significance of sprouted broccoliSignificance of sprouted broccoli
Significance of sprouted broccoliDr. sreeremya S
 
Brain as an endocrine organ
Brain as an endocrine organBrain as an endocrine organ
Brain as an endocrine organalaa wafa
 
Brain as an endocrine organ
Brain as an endocrine organBrain as an endocrine organ
Brain as an endocrine organalaa wafa
 
Modern neurosciapproachcs ppaper13
Modern neurosciapproachcs ppaper13Modern neurosciapproachcs ppaper13
Modern neurosciapproachcs ppaper13Nathanael Amparo
 
Regeneration of mind and brain
Regeneration of mind and brainRegeneration of mind and brain
Regeneration of mind and brainDevashish Konar
 
ENKEPHALIN PPT (final).. (2)-1.pdf
ENKEPHALIN PPT (final).. (2)-1.pdfENKEPHALIN PPT (final).. (2)-1.pdf
ENKEPHALIN PPT (final).. (2)-1.pdfsaleeemmalik259
 
Patrik Brundin - Are Synucleinopathies Prion Diseases?
Patrik Brundin - Are Synucleinopathies Prion Diseases?Patrik Brundin - Are Synucleinopathies Prion Diseases?
Patrik Brundin - Are Synucleinopathies Prion Diseases?Alzforum
 
parkinson's disease-1.pptx
parkinson's disease-1.pptxparkinson's disease-1.pptx
parkinson's disease-1.pptxfawwazumar3
 
Efficacy of Souvenaid in Mild Alzheimer’s Disease: Results from a Randomized,...
Efficacy of Souvenaid in Mild Alzheimer’s Disease: Results from a Randomized,...Efficacy of Souvenaid in Mild Alzheimer’s Disease: Results from a Randomized,...
Efficacy of Souvenaid in Mild Alzheimer’s Disease: Results from a Randomized,...Nutricia
 
Pengantar sistem saraf brain stem
Pengantar sistem saraf brain  stemPengantar sistem saraf brain  stem
Pengantar sistem saraf brain stemNurfitriBustamam
 
Cognition and Behavioral Effects in Epilepsy: A Review
Cognition and Behavioral Effects in Epilepsy: A ReviewCognition and Behavioral Effects in Epilepsy: A Review
Cognition and Behavioral Effects in Epilepsy: A ReviewBRNSS Publication Hub
 

Semelhante a Insulina intranasal e cognição (20)

Insulina e cérebro
Insulina  e  cérebroInsulina  e  cérebro
Insulina e cérebro
 
Current Paradigms to Explore the Gut Microbiota Linkage to Neurological Disor...
Current Paradigms to Explore the Gut Microbiota Linkage to Neurological Disor...Current Paradigms to Explore the Gut Microbiota Linkage to Neurological Disor...
Current Paradigms to Explore the Gut Microbiota Linkage to Neurological Disor...
 
International Journal of Sports Science & Medicine
International Journal of Sports Science & MedicineInternational Journal of Sports Science & Medicine
International Journal of Sports Science & Medicine
 
C-Fos Protein Immunoreactivity in Raphe Nuclei of Trained and Sedentary Rats
C-Fos Protein Immunoreactivity in Raphe Nuclei of Trained and Sedentary Rats C-Fos Protein Immunoreactivity in Raphe Nuclei of Trained and Sedentary Rats
C-Fos Protein Immunoreactivity in Raphe Nuclei of Trained and Sedentary Rats
 
Significance of sprouted broccoli
Significance of sprouted broccoliSignificance of sprouted broccoli
Significance of sprouted broccoli
 
Brain as an endocrine organ
Brain as an endocrine organBrain as an endocrine organ
Brain as an endocrine organ
 
Brain as an endocrine organ
Brain as an endocrine organBrain as an endocrine organ
Brain as an endocrine organ
 
Modern neurosciapproachcs ppaper13
Modern neurosciapproachcs ppaper13Modern neurosciapproachcs ppaper13
Modern neurosciapproachcs ppaper13
 
Regeneration of mind and brain
Regeneration of mind and brainRegeneration of mind and brain
Regeneration of mind and brain
 
ENKEPHALIN PPT (final).. (2)-1.pdf
ENKEPHALIN PPT (final).. (2)-1.pdfENKEPHALIN PPT (final).. (2)-1.pdf
ENKEPHALIN PPT (final).. (2)-1.pdf
 
1.pptx
1.pptx1.pptx
1.pptx
 
Patrik Brundin - Are Synucleinopathies Prion Diseases?
Patrik Brundin - Are Synucleinopathies Prion Diseases?Patrik Brundin - Are Synucleinopathies Prion Diseases?
Patrik Brundin - Are Synucleinopathies Prion Diseases?
 
parkinson's disease-1.pptx
parkinson's disease-1.pptxparkinson's disease-1.pptx
parkinson's disease-1.pptx
 
Yang{JMCC_2009]
Yang{JMCC_2009]Yang{JMCC_2009]
Yang{JMCC_2009]
 
Efficacy of Souvenaid in Mild Alzheimer’s Disease: Results from a Randomized,...
Efficacy of Souvenaid in Mild Alzheimer’s Disease: Results from a Randomized,...Efficacy of Souvenaid in Mild Alzheimer’s Disease: Results from a Randomized,...
Efficacy of Souvenaid in Mild Alzheimer’s Disease: Results from a Randomized,...
 
Pengantar sistem saraf brain stem
Pengantar sistem saraf brain  stemPengantar sistem saraf brain  stem
Pengantar sistem saraf brain stem
 
01_IJPBA_1884_20.pdf
01_IJPBA_1884_20.pdf01_IJPBA_1884_20.pdf
01_IJPBA_1884_20.pdf
 
01_IJPBA_1884_20.pdf
01_IJPBA_1884_20.pdf01_IJPBA_1884_20.pdf
01_IJPBA_1884_20.pdf
 
01_IJPBA_1884_20.pdf
01_IJPBA_1884_20.pdf01_IJPBA_1884_20.pdf
01_IJPBA_1884_20.pdf
 
Cognition and Behavioral Effects in Epilepsy: A Review
Cognition and Behavioral Effects in Epilepsy: A ReviewCognition and Behavioral Effects in Epilepsy: A Review
Cognition and Behavioral Effects in Epilepsy: A Review
 

Mais de Ruy Pantoja

Dm e efeitos da canela
Dm e efeitos da canelaDm e efeitos da canela
Dm e efeitos da canelaRuy Pantoja
 
Dieta hiperproteica para obeso com resistência à insulina
Dieta hiperproteica para obeso com resistência à insulinaDieta hiperproteica para obeso com resistência à insulina
Dieta hiperproteica para obeso com resistência à insulinaRuy Pantoja
 
Ressecção parcial de pâncreas x regeneração x terapia incretínica humanos
Ressecção parcial de pâncreas x regeneração x terapia incretínica humanosRessecção parcial de pâncreas x regeneração x terapia incretínica humanos
Ressecção parcial de pâncreas x regeneração x terapia incretínica humanosRuy Pantoja
 
Brown adipose tissue
Brown adipose tissueBrown adipose tissue
Brown adipose tissueRuy Pantoja
 
Não somos quentes o suficiente ?
Não somos quentes o suficiente ?Não somos quentes o suficiente ?
Não somos quentes o suficiente ?Ruy Pantoja
 
MACROPHAGE AND BROWN FAT
MACROPHAGE AND BROWN FATMACROPHAGE AND BROWN FAT
MACROPHAGE AND BROWN FATRuy Pantoja
 
Beta cell stress en dm2 b
Beta cell stress en dm2 bBeta cell stress en dm2 b
Beta cell stress en dm2 bRuy Pantoja
 
Beta cell stres in dm2 a
Beta cell stres in dm2 aBeta cell stres in dm2 a
Beta cell stres in dm2 aRuy Pantoja
 
Beta cell failure, stress and dm2
Beta cell failure, stress and dm2Beta cell failure, stress and dm2
Beta cell failure, stress and dm2Ruy Pantoja
 
Dpp4 e angiogênese
Dpp4 e angiogêneseDpp4 e angiogênese
Dpp4 e angiogêneseRuy Pantoja
 
DIABETES AND CANCER
DIABETES AND CANCERDIABETES AND CANCER
DIABETES AND CANCERRuy Pantoja
 
Sitagliptina e proteção em ca diferenciado da tireóide.x
Sitagliptina e proteção em ca diferenciado da tireóide.xSitagliptina e proteção em ca diferenciado da tireóide.x
Sitagliptina e proteção em ca diferenciado da tireóide.xRuy Pantoja
 
Exetimibe melhora dm diminuindo nafld.x
Exetimibe melhora dm diminuindo nafld.xExetimibe melhora dm diminuindo nafld.x
Exetimibe melhora dm diminuindo nafld.xRuy Pantoja
 
Dpp4 i for the treatment of ACNE
Dpp4 i for the treatment of ACNEDpp4 i for the treatment of ACNE
Dpp4 i for the treatment of ACNERuy Pantoja
 
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇASGLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇASRuy Pantoja
 
To my colleagues from many parts of the world
To my colleagues from many parts of the worldTo my colleagues from many parts of the world
To my colleagues from many parts of the worldRuy Pantoja
 
FENTERMIN DOENSNT INCREASE HYPERTENSION
FENTERMIN DOENSNT INCREASE HYPERTENSIONFENTERMIN DOENSNT INCREASE HYPERTENSION
FENTERMIN DOENSNT INCREASE HYPERTENSIONRuy Pantoja
 
Doença cardiovascular
Doença cardiovascularDoença cardiovascular
Doença cardiovascularRuy Pantoja
 
Cancer cell metabolism
Cancer cell metabolismCancer cell metabolism
Cancer cell metabolismRuy Pantoja
 

Mais de Ruy Pantoja (20)

Dm e efeitos da canela
Dm e efeitos da canelaDm e efeitos da canela
Dm e efeitos da canela
 
Drug discovery
Drug discoveryDrug discovery
Drug discovery
 
Dieta hiperproteica para obeso com resistência à insulina
Dieta hiperproteica para obeso com resistência à insulinaDieta hiperproteica para obeso com resistência à insulina
Dieta hiperproteica para obeso com resistência à insulina
 
Ressecção parcial de pâncreas x regeneração x terapia incretínica humanos
Ressecção parcial de pâncreas x regeneração x terapia incretínica humanosRessecção parcial de pâncreas x regeneração x terapia incretínica humanos
Ressecção parcial de pâncreas x regeneração x terapia incretínica humanos
 
Brown adipose tissue
Brown adipose tissueBrown adipose tissue
Brown adipose tissue
 
Não somos quentes o suficiente ?
Não somos quentes o suficiente ?Não somos quentes o suficiente ?
Não somos quentes o suficiente ?
 
MACROPHAGE AND BROWN FAT
MACROPHAGE AND BROWN FATMACROPHAGE AND BROWN FAT
MACROPHAGE AND BROWN FAT
 
Beta cell stress en dm2 b
Beta cell stress en dm2 bBeta cell stress en dm2 b
Beta cell stress en dm2 b
 
Beta cell stres in dm2 a
Beta cell stres in dm2 aBeta cell stres in dm2 a
Beta cell stres in dm2 a
 
Beta cell failure, stress and dm2
Beta cell failure, stress and dm2Beta cell failure, stress and dm2
Beta cell failure, stress and dm2
 
Dpp4 e angiogênese
Dpp4 e angiogêneseDpp4 e angiogênese
Dpp4 e angiogênese
 
DIABETES AND CANCER
DIABETES AND CANCERDIABETES AND CANCER
DIABETES AND CANCER
 
Sitagliptina e proteção em ca diferenciado da tireóide.x
Sitagliptina e proteção em ca diferenciado da tireóide.xSitagliptina e proteção em ca diferenciado da tireóide.x
Sitagliptina e proteção em ca diferenciado da tireóide.x
 
Exetimibe melhora dm diminuindo nafld.x
Exetimibe melhora dm diminuindo nafld.xExetimibe melhora dm diminuindo nafld.x
Exetimibe melhora dm diminuindo nafld.x
 
Dpp4 i for the treatment of ACNE
Dpp4 i for the treatment of ACNEDpp4 i for the treatment of ACNE
Dpp4 i for the treatment of ACNE
 
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇASGLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
GLP 1 E GIP: DIFERENÇAS E SEMELHANÇAS
 
To my colleagues from many parts of the world
To my colleagues from many parts of the worldTo my colleagues from many parts of the world
To my colleagues from many parts of the world
 
FENTERMIN DOENSNT INCREASE HYPERTENSION
FENTERMIN DOENSNT INCREASE HYPERTENSIONFENTERMIN DOENSNT INCREASE HYPERTENSION
FENTERMIN DOENSNT INCREASE HYPERTENSION
 
Doença cardiovascular
Doença cardiovascularDoença cardiovascular
Doença cardiovascular
 
Cancer cell metabolism
Cancer cell metabolismCancer cell metabolism
Cancer cell metabolism
 

Último

Dyslexia AI Workshop for Slideshare.pptx
Dyslexia AI Workshop for Slideshare.pptxDyslexia AI Workshop for Slideshare.pptx
Dyslexia AI Workshop for Slideshare.pptxcallscotland1987
 
Activity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdfActivity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdfciinovamais
 
Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...
Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...
Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...christianmathematics
 
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdfUGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdfNirmal Dwivedi
 
How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17Celine George
 
How to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POSHow to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POSCeline George
 
psychiatric nursing HISTORY COLLECTION .docx
psychiatric  nursing HISTORY  COLLECTION  .docxpsychiatric  nursing HISTORY  COLLECTION  .docx
psychiatric nursing HISTORY COLLECTION .docxPoojaSen20
 
Third Battle of Panipat detailed notes.pptx
Third Battle of Panipat detailed notes.pptxThird Battle of Panipat detailed notes.pptx
Third Battle of Panipat detailed notes.pptxAmita Gupta
 
ICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxAreebaZafar22
 
ICT role in 21st century education and it's challenges.
ICT role in 21st century education and it's challenges.ICT role in 21st century education and it's challenges.
ICT role in 21st century education and it's challenges.MaryamAhmad92
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introductionMaksud Ahmed
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdfQucHHunhnh
 
Key note speaker Neum_Admir Softic_ENG.pdf
Key note speaker Neum_Admir Softic_ENG.pdfKey note speaker Neum_Admir Softic_ENG.pdf
Key note speaker Neum_Admir Softic_ENG.pdfAdmir Softic
 
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...ZurliaSoop
 
This PowerPoint helps students to consider the concept of infinity.
This PowerPoint helps students to consider the concept of infinity.This PowerPoint helps students to consider the concept of infinity.
This PowerPoint helps students to consider the concept of infinity.christianmathematics
 
Understanding Accommodations and Modifications
Understanding  Accommodations and ModificationsUnderstanding  Accommodations and Modifications
Understanding Accommodations and ModificationsMJDuyan
 
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxBasic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxDenish Jangid
 
Food safety_Challenges food safety laboratories_.pdf
Food safety_Challenges food safety laboratories_.pdfFood safety_Challenges food safety laboratories_.pdf
Food safety_Challenges food safety laboratories_.pdfSherif Taha
 

Último (20)

Dyslexia AI Workshop for Slideshare.pptx
Dyslexia AI Workshop for Slideshare.pptxDyslexia AI Workshop for Slideshare.pptx
Dyslexia AI Workshop for Slideshare.pptx
 
Activity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdfActivity 01 - Artificial Culture (1).pdf
Activity 01 - Artificial Culture (1).pdf
 
Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...
Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...
Explore beautiful and ugly buildings. Mathematics helps us create beautiful d...
 
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdfUGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
UGC NET Paper 1 Mathematical Reasoning & Aptitude.pdf
 
How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17
 
How to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POSHow to Manage Global Discount in Odoo 17 POS
How to Manage Global Discount in Odoo 17 POS
 
psychiatric nursing HISTORY COLLECTION .docx
psychiatric  nursing HISTORY  COLLECTION  .docxpsychiatric  nursing HISTORY  COLLECTION  .docx
psychiatric nursing HISTORY COLLECTION .docx
 
Third Battle of Panipat detailed notes.pptx
Third Battle of Panipat detailed notes.pptxThird Battle of Panipat detailed notes.pptx
Third Battle of Panipat detailed notes.pptx
 
Spatium Project Simulation student brief
Spatium Project Simulation student briefSpatium Project Simulation student brief
Spatium Project Simulation student brief
 
ICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptx
 
ICT role in 21st century education and it's challenges.
ICT role in 21st century education and it's challenges.ICT role in 21st century education and it's challenges.
ICT role in 21st century education and it's challenges.
 
microwave assisted reaction. General introduction
microwave assisted reaction. General introductionmicrowave assisted reaction. General introduction
microwave assisted reaction. General introduction
 
1029-Danh muc Sach Giao Khoa khoi 6.pdf
1029-Danh muc Sach Giao Khoa khoi  6.pdf1029-Danh muc Sach Giao Khoa khoi  6.pdf
1029-Danh muc Sach Giao Khoa khoi 6.pdf
 
Key note speaker Neum_Admir Softic_ENG.pdf
Key note speaker Neum_Admir Softic_ENG.pdfKey note speaker Neum_Admir Softic_ENG.pdf
Key note speaker Neum_Admir Softic_ENG.pdf
 
Mehran University Newsletter Vol-X, Issue-I, 2024
Mehran University Newsletter Vol-X, Issue-I, 2024Mehran University Newsletter Vol-X, Issue-I, 2024
Mehran University Newsletter Vol-X, Issue-I, 2024
 
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
Jual Obat Aborsi Hongkong ( Asli No.1 ) 085657271886 Obat Penggugur Kandungan...
 
This PowerPoint helps students to consider the concept of infinity.
This PowerPoint helps students to consider the concept of infinity.This PowerPoint helps students to consider the concept of infinity.
This PowerPoint helps students to consider the concept of infinity.
 
Understanding Accommodations and Modifications
Understanding  Accommodations and ModificationsUnderstanding  Accommodations and Modifications
Understanding Accommodations and Modifications
 
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxBasic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
 
Food safety_Challenges food safety laboratories_.pdf
Food safety_Challenges food safety laboratories_.pdfFood safety_Challenges food safety laboratories_.pdf
Food safety_Challenges food safety laboratories_.pdf
 

Insulina intranasal e cognição

  • 1. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism Volker Ott1*, Christian Benedict2, Bernd Schultes3, Jan Born4, Manfred Hallschmid1 1 Department of Neuroendocrinology, University of Luebeck, Germany; 2Department of Neuroscience, Functional Pharmacology, Uppsala University, Uppsala, Sweden; 3Interdisciplinary Obesity Center, Cantonal Hospital St. Gallen, Switzerland; 4Department of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany Key Terms: Insulin, intranasal, central nervous system, glucose homeostasis, energy homeostasis, thermogenesis, cognitive function, insulin resistance, hepatic glucose production Word Count: 3487 * To whom correspondence and reprint requests should be addressed: Department of Neuroendocrinology, Hs. 50.1 University of Luebeck Ratzeburger Allee 160 23538 Luebeck, Germany Phone: ++49-451-500-5375 Fax: ++49-451-500-3640 E-mail: ott@kfg.uni-luebeck.de This is an Accepted Article that has been peer-reviewed and approved for publication in the Diabetes, Obesity and Metabolism, but has yet to undergo copy-editing and proof correction. Please cite this article as an "Accepted Article"; doi: 10.1111/j.1463-1326.2011.01490.x 1
  • 2. Abstract In recent years, the central nervous system (CNS) has emerged as a principle site of insulin action. This notion is supported by studies in animals relying on intracerebroventricular insulin infusion and by experiments in humans that make use of the intranasal pathway of insulin administration to the brain. Employing neurobehavioral and metabolic measurements as well as functional imaging techniques, these studies have provided insight into a broad range of central and peripheral effects of brain insulin. The present review focuses on CNS effects of insulin administered via the intranasal route on cognition, in particular memory function, and whole-body energy homeostasis including glucose metabolism. Furthermore, evidence is reviewed that suggests a pathophysiological role of impaired brain insulin signaling in obesity and type 2 diabetes, which are hallmarked by peripheral and possibly central nervous insulin resistance, as well as in conditions such as Alzheimer´s disease where CNS insulin resistance might contribute to cognitive dysfunction. 2
  • 3. Introduction In the mid-eighteen-hundreds, Claude Bernard demonstrated that puncture of the fourth cerebral ventricle induces glucosuria in mice [1], giving rise to the assumption that the central nervous system (CNS) is involved in glucose homeostasis. However, interest in the role the brain might play in the regulation of glucose metabolism abated and was only sparked again more than a century later. Havrankova and coworkers demonstrated in 1978 that insulin receptors are present throughout the rat CNS [2], followed closely by the demonstration that insulin receptors are also expressed in the human brain [3,4]. The insulin receptor, a tyrosine kinase receptor, is found in particularly high densities in brain regions like the olfactory bulb, the cerebellum, the dentate gyrus, the pyriform cortex, the hippocampus, the choroid plexus and the arcuate nucleus of the hypothalamus [5]. Some animal studies suggest that insulin gene expression takes place within the CNS [6,7]. However, although indicators of insulin transcription in human brain tissue have been presented [8], solid evidence for local insulin production in the human CNS is still lacking [9]. It is rather assumed that peripheral insulin crosses the blood-brain barrier (BBB) by a saturable, receptor-mediated transport mechanism [9-11] and by binding to brain insulin receptors affects functions as diverse as energy and glucose homeostasis [12-14], reproduction [13], growth [15] and neuronal plasticity [16]. Woods and co- workers were the first to perform seminal studies indicating that insulin, circulating within the blood stream in proportion to body fat stores, acts as an adiposity signal within in the CNS. In conjunction with the adipokine leptin it provides the brain with negative feedback on the amount of peripheral energy (i.e., fat) depots [17; for review see reference 18]. In line with this notion, CNS administration of insulin reduces body adiposity by down-regulating food intake [12,19,20]. This catabolic effect has been observed mainly in males, indicating that the role of insulin in central nervous body weight regulation may have sex-specific properties [21-23]. Moreover, as will be outlined in this review, insulin’s impact on the brain exceeds its involvement in energy homeostasis and pertains to cognitive functions (Figure 1). Brain insulin signaling might even constitute a neuroendocrine link between both domains and is therefore emerging as a potential target in the treatment of metabolic and cognitive disorders [24]. 3
  • 4. Enhancing central nervous insulin signaling by intranasal insulin administration in humans Whereas in animals effective modes of insulin administration to the CNS, e.g., direct intracerebroventricular (ICV) [25] or hypothalamic infusion [26] are routinely employed, insulin administration to the human brain is more complicated. The conventional way of increasing CNS concentrations of insulin to investigate effects of brain insulin relies on the intravenous (IV) infusion of the hormone which has been shown to result in an increase in cerebrospinal fluid (CSF) insulin concentrations [27]. This parenteral route, however, faces several serious drawbacks. The fall in blood glucose levels resulting from systemic insulin infusion triggers the graded activation of endocrine axes that can affect brain function [28], and below certain threshold levels inevitably impairs cognition [29]. Insulin-induced hypoglycemia and its potentially harmful effects can be prevented by simultaneous continuous glucose infusion that per se may exert a biasing impact on (cognitive) brain functioning. Moreover, the euglycemic-hyperinsulinemic clamp procedure implies considerable time and labor investments and, generally, systemic insulin administration does not permit the dissection of insulin’s effects on the CNS from its direct peripheral actions e.g. in liver [30] and adipose tissue [31]. These methodological limitations are avoided by the intranasal (IN) route of administration that has been shown in humans to bypass the BBB and effectively deliver insulin as well as other peptide hormones to the CNS within one hour after administration in the absence of relevant systemic absorption [32]. Accordingly, findings in animals demonstrate that intranasally administered neuropeptides reach brain structures involved in the regulation of metabolism and cognition [33,34]. Intra-neuronal transport of neuropeptides from the nasal cavity to the olfactory bulb takes several hours [35]. Thus, extra-neuronal passage through intercellular clefts of the olfactory epithelium, situated on the superior turbinate and opposite the nasal septum [36], is assumed to be the preferential path of peptide transport into the CNS compartment [32,37], with additional transport along trigeminal nerve branches to brainstem regions [38]. IN administration of insulin preparations for the purpose of systemic insulin substitution, i.e., as an alternative approach to subcutaneous insulin injection, is not within the scope of this review. 4
  • 5. Information on this aspect of nasal insulin administration can be found elsewhere [e.g. references 39,40]. Insulin modulates neurobehavioral measures of brain activity and cognition in humans Insulin effects on human brain activity have been revealed in a number of studies relying on different methodological approaches. CNS responses to IN insulin were observed in the form of distinct alterations in auditory evoked electroencephalographic brain potential responses during an oddball- paradigm in healthy men while peripheral blood glucose levels remained unchanged [41] . In a related study, the IN administration of 60 international units of insulin induced a negative shift in direct current brain potentials that was also found after IV bolus injection of the hormone [42]. Both the IN and the intravenous effects emerged within 20 min after insulin administration, indicating that increases in systemic insulin concentrations are rapidly reported to the brain and that IN delivery of the compound bypassing the body periphery can have a comparable impact on brain activity. The impact of systemic insulin on cerebrocortical activity was likewise measured in euglycemic- hyperinsulinemic clamp studies that utilized magnetoencephalographic (MEG) recordings and demonstrated that obesity [43,44] and the fat-mass and obesity associated (FTO) allele variant rs8050136 [45] modify insulin’s effects on cerebrocortical beta- and theta-wave activity . Experiments employing functional magnetic resonance imaging (fMRI) have shown a positive relationship between plasma insulin levels and activation of the right hippocampus in response to viewing photographs of high-caloric food items [46]. In some contrast to these results, in another fMRI study, food picture-related activity of this and other brain regions was found to be reduced after IN insulin in comparison to placebo administration [47], raising the question if insulin acting on the hippocampus is relevant for the regulation of ingestive behavior. On the other hand, the hippocampus is highly relevant for the formation and maintenance of declarative memory, i.e. memory for facts and episodes that is accessible to conscious recollection [for review see reference 48]. The ability to acquire and retain memories depends on synaptic plasticity. Thus, long-term potentiation (LTP) and long-term depression (LTD) of synaptic transmission, i.e. the augmentation or reduction of synaptic efficacy are assumed to be important modulators of the strength of a memory representation [49,50]. 5
  • 6. Several studies indicate that insulin contributes to changes in hippocampal synaptic plasticity by potentiating LTD and LTP, respectively, at different synapses [for review see reference 51]. Moreover, insulin receptors have been found to increase synapse density and dendritic plasticity in structures that process visual input [52]. In addition to these mechanims, insulin may promote glucose utilization of neuronal networks [53]. Although globally glucose transport to the CNS is assumed to be insulin- independent [54-56], hyperinsulinemia has been shown in rodents to exert effects on glucose metabolism in regions like the anterior hypothalamus and the basolateral amygdale [57]. In accordance with insulin’s effects on synaptic plasticity [52] and regional glucose uptake [52], central nervous administration of the hormone via the IN route has been shown to improve memory functions in studies in healthy humans [23,58,59]. In experiments performed in our lab, a declarative memory test was conducted at the beginning and end of eight weeks of IN insulin treatment (160 international units/d). In brief, lists of 30 words were presented and in addition to an immediate recall 3 min after presentation, in a delayed recall one week later subjects wrote down all words they still remembered. The delayed recall of words was significantly improved after eight weeks of IN insulin administration whereas immediate word recall and non-declarative memory functions were not affected [58]. In line with the strong accumulation of insulin receptors in hippocampal and cortical brain structures [60], this finding indicates that insulin signaling contributes to the formation of declarative, hippocampus-dependent memory contents. Noteworthy, beneficial effects of IN insulin on declarative memory are not restricted to healthy subjects but have also been shown in memory-impaired subjects [e.g. references 61,62]. Suzanne Craft and co-workers performed a study in adults with mild cognitive impairments including amnestic symptoms (e.g., due to Alzheimer´s disease) who were treated with IN insulin over a period of three weeks (2x20 international units/d) [61]. The primary outcome measure was the recall of a story containing 44 informational bits to which subjects listened and that they were asked to recall immediately and after a 20-minute delay. Patients treated with insulin showed significantly increased memory savings over the 21-day period compared to placebo. Considering reports of impaired brain glucose metabolism in Alzheimer’s disease [63-65], it might be speculated that these effects and, in particular, acute insulin- induced enhancements of cognitive function in memory-impaired patients occurring within minutes 6
  • 7. [66] at least in part derive from increases in cerebral glucose metabolism. In related animal studies, IN administration of the peptide slowed the development of diabetes-induced brain changes in a murine model of type 1 diabetes [67]. CNS insulin signaling has been linked not only to cognitive but also to emotional functions of the brain. Most recently, lentivirus-mediated downregulation of hypothalamic insulin receptor expression in rats has been shown to elicit depressive and anxiety-like behaviors [68]. Vice versa, the 8-week IN insulin treatment described above induced an improvement in rated mood in our human subjects [58]. In mice, IN insulin enhanced object-memory and induced anxiolytic behavioral effects [69]. However, in mice with impaired glucose tolerance due to diet-induced obesity receiving the same dose of IN insulin both effects were abrogated [69]. These findings suggest that disturbed CNS insulin signaling/CNS insulin resistance might link metabolic disorders like obesity with cognitive impairments and also depressive symptoms. Further evidence for this assumption is discussed below. CNS insulin signaling and peripheral metabolism In animal experiments, brain insulin signaling has emerged as an important regulator of energy balance [70-72]. Insulin’s net effect on energy homeostasis depends on several factors. Whereas intravenously administered insulin exerts direct peripheral and, after BBB transport, central nervous effects, intransally administered insulin selectively targets the CNS. In this context it is interesting to note that the central nervous action of insulin on energy homeostasis partly opposes its peripheral effects. Whereas after peripheral (IV or subcutaneous) administration insulin acts as an anabolic hormone by promoting weight gain in form of muscle and fat mass [73,74], IN and ICV insulin administration in humans and animals, respectively, induces catabolic effects by reducing food intake [19,23] and as a consequence body fat content [20,21] particularly in the male organism [21-23]. In parallel, central insulin exerts an anabolic impact on adipose tissue: in addition to the inhibition of lipolysis by peripheral insulin [75,76], CNS insulin has been found to likewise inhibit lipolysis and also to enhance lipogenesis [75,77,78]. Recent murine data also suggest that hypothalamic insulin signaling potentiates brown adipose tissue thermogenesis through inhibition of warm sensitive neurons [79]. Our group corroborated these findings in humans by demonstrating that IN insulin enhances 7
  • 8. postprandial thermogenesis [80]. Thus, the catabolic effect of IN insulin appears to stem from reduced energy intake [21,23] and increased energy expenditure [79,80] alike. Within the last decade, evidence has amounted that the impact of brain insulin signaling on energy balance extends to glucose homeostasis. Hepatic glucose metabolism is an important determinant of euglycemia [81]. By glycogenesis on the one hand and glycogenolysis and gluconeogenesis on the other hand the liver stabilizes plasma glucose concentrations during (postprandial) glucose abundance and (fasting) glucose depletion, respectively [82]. These processes have long been known to be mediated by direct insulin action on hepatic insulin receptors and indirect insulin effects on liver functions, including the downregulation of glucagon secretion and circulating plasma nonesterified fatty acid concentrations [for review see reference 83]. However, hepatic glucose metabolism also seems to be under the control of a brain-liver axis. Obici and co-workers have shown in rodents that genetic downregulation of hypothalamic insulin receptor expression disinhibits hepatic glucose production [84]. This finding clearly hints at a reduction in hepatic insulin sensitivity as a consequence of impaired hypothalamic insulin signaling. Fittingly, insulin has been found to open ATP-sensitive potassium channels on glucose-responsive hypothalamic neurons and the resulting neuronal hyperpolarization seems to be responsible for the vagal transmission of a signal that downregulates hepatic glucose production [14,84]. However, in an experiment in dogs, quadrupling the concentration of circulating insulin selectively in brain afferent arteries did not enhance the inhibition of hepatic glucose production [85] which leaves open the question whether the contribution of hypothalamic insulin signaling to insulin’s hepatic effects has a species-dependent component. In humans it has recently been shown that IV pretreatment with insulin potentiates glucose- induced pancreatic insulin secretion by 40%, suggesting that circulating insulin exerts a direct positive feedback on its own secretion [86]. Interestingly, a similar effect was found for brain insulin over 30 years ago in dogs, where ICV insulin administration increased pancreatic insulin secretion via a feed- forward mechanism [87,88]. This brain-pancreatic crosstalk involving the vagal nerve has been hypothesized to be another regulator of blood glucose. While effects of CNS insulin on peripheral insulin sensitivity and pancreatic insulin secretion in humans are largely unexplored, two recent 8
  • 9. studies have gathered evidence that insulin delivery to the brain does affect peripheral glucose metabolism. IN insulin administration before intake of a liquid meal reduced postprandial circulating insulin levels in healthy subjects while plasma glucose levels were unchanged in comparison to placebo [80]. This finding suggests that brain insulin administration can enhance postprandial peripheral insulin sensitivity, adding to the feed-forward effect of brain insulin on pancreatic insulin secretion observed in animals. Another recent set of experiments performed by Stockhorst and colleagues indicates that such brain-pancreatic cross-talk is accessible to classical conditioning [89]. On day 1, the investigators administered IN insulin vs. placebo that both have the same specific odor due to the formulation with meta-cresol (a stabilizing agent in insulin solutions). They found an increase in serum insulin concentrations and a reduction in blood glucose levels (within the euglycemic range) after insulin compared with placebo, suggesting that activation of the brain-liver axis enhanced pancreatic insulin secretion. On day 2, the procedure was repeated but placebo was administered in both groups with the smell of meta-cresol functioning as a conditioned stimulus. Here, the presentation of the conditioned stimulus alone after pretreatment with IN insulin on day 1 was sufficient to cause an even enhanced increasing effect on serum insulin concentrations, which points to a significant contribution of neurocognitive learning mechanisms to the regulation of peripheral glucose homeostasis by brain insulin. Central nervous system insulin resistance Peripheral insulin resistance is a well-known feature of type 2 diabetes and obesity. Insulin resistance in central nervous structures might likewise contribute to the development not only of these metabolic disorders but also of cognitive impairments. Raising systemic insulin levels by IV infusion results in increased CSF insulin concentrations in healthy, normal-weight subjects [27]. Obese subjects and Alzheimer patients seem to display relatively decreased CSF insulin concentrations suggesting reduced insulin transport across the BBB [90,91]. Likewise, in comparison to normal-weight subjects, overweight humans show a decrease in MEG-recorded cortical activity during hyperinsulinemic- euglycemic clamp experiments that is directly related to the amount of body fat and the degree of peripheral insulin resistance [43]. These findings support the notion that in obesity both BBB insulin transport and the central nervous sensitivity to insulin are reduced. Related studies relying on acute 9
  • 10. and prolonged IN administration of the hormone have refined this picture. In further MEG-based experiments, IN insulin acutely increased cerebrocortical activity in response to food vs. non-food pictures in lean but not in obese subjects [92]. Long-term (8 weeks) administration of IN insulin in obese subjects failed to affect body weight and fat mass [93] but still enhanced declarative memory and dampened HPA-axis activity to a degree comparable with that observed in normal-weight subjects [58,94]. This differential insulin response implies that brain regions involved in energy and glucose homeostasis might be particularly prone to develop insulin resistance in obesity. In accordance with these findings, in rats diet-induced obesity abolishes the catabolic actions of ICV insulin administration, and a reduction in insulin receptor density in the hypothalamic arcuate nucleus causes hyperphagia (and, as mentioned above, also disinhibits hepatic glucose production) [84]. On a molecular level, activation of the PI-3 signaling cascade subsequent to the binding of insulin to its receptor mediates the majority of central nervous insulin effects on energy homeostasis [for review see reference 95], while disturbances of this pathway are regarded as a likely cause of neuronal insulin resistance [for review see 96]. Reduced central nervous sensitivity might represent a pathophysiological link between obesity, peripheral insulin resistance and cognitive disorders that have been found to be significantly related in epidemiological studies [97,98]. Central insulin resistance seems to impair neuronal plasticity via detrimental effects on glutamatergic and cholinergic pathways [99,100]. Such processes are assumed to be influenced by genetic predisposition. One indicator for this assumption is that IN insulin administration to patients with memory impairments improved memory functions predominantly in non-carriers of the APOE*E4 allele, a risk factor for the development of Alzheimer’s disease [62,66; for review see reference 101]. In this context, several further genetic polymorphisms associated with reduced central nervous responsiveness to insulin have been characterized. Subjects with the FTO gene polymorphism rs8050136 as well as carriers of the Gly972Arg polymorphism of the Insulin Receptor Substrate 1 (IRS1) exhibit a decreased cerebrocortical response to IV insulin [43,45]. Interestingly, these data support the suggested connection between the FTO variant and a hyperphagic phenotype [102] characterised by a predilection for energy-dense foods [103], which might involve decreased insulin sensitivity of food reward-related brain pathways [104]. 10
  • 11. Potential therapies aimed at overcoming CNS insulin resistance might include the IN administration of insulin but could also rely on the insulin-sensitizing properties of the peroxisome proliferator-activated receptor-ƴ (PPAR-ƴ) agonist rosiglitazone [105-108] and of metformin [109]. Regarding glucose homeostasis, enhancing central nervous/hypothalamic insulin signaling by insulin administration to the brain might reinforce a vagally transmitted inhibitory signal on hepatic gluconeogenesis [110], whose disinhibition represents a hallmark of type 2 diabetes and peripheral insulin resistance [84]. The latter, moreover, appears to be highly associated with central nervous insulin resistance [111]. IN insulin has been found to reduce HPA axis activity [58,93,94], thus potentially opposing visceral adiposity and cognitive impairments due to stress-induced chronic HPA- axis overactivation [112-114]. Moreover, in light of IN insulin’s acute memory-improving effects in patients with mild cognitive impairments [61,62], the compound might ameliorate the harmful effects on cognition that obesity and diabetes are suspected to engender [97]. Notwithstanding these encouraging results, some caveats need to be addressed. In light of the hyperinsulinemia that accompanies peripheral insulin resistance, it might be argued that the reduction of CSF insulin levels observed in obese subjects [91] could represent a protective mechanism that limits central nervous hyperinsulinemia and the potentially detrimental sequelae of cellular insulin resistance inside the brain. Although speculative, this assumption is in line with findings that hint at a dose-dependent directionality of central insulin’s impact on memory function. For example, acute IN insulin administration to Alzheimer patients improved verbal memory recall only at lower doses (20 international units), whereas higher doses (up to 60 international units) were not effective and, in carriers of the APOE*E4 allele, even induced a decline in memory performance [62]. In healthy subjects, the induction of acute moderate euglycemic hyperinsulinemia has been found to trigger central nervous system inflammation and beta-amyloid formation [115], both of which are known risk factors for the development of cognitive impairments. The notion that brain hyperinsulinemia might promote central nervous insulin resistance is supported by a recent in vitro-study showing that prolonged (4-24 h) exposure of hypothalamic cells to high concentrations of insulin led to inactivation and degradation of the insulin receptor and IRS-1 [116]. Against this background and considering that long-term data on therapeutic and side effects of IN insulin in humans are so far lacking, obviously 11
  • 12. much work is still necessary to sound the potential of brain insulin administration in the treatment of cognitive and metabolic disorders. Conclusion Insulin binding to its receptors in the brain impacts a number of pivotal physiological functions, including energy uptake and expenditure, glucose metabolism, adipocyte function and cognition. The experimental data briefly summarized here clearly implicate CNS insulin resistance as a potentially important factor in the pathophysiology of obesity and systemic insulin resistance as well as of cognitive impairments like Alzheimer’s disease. Moreover, the association between these disorders found in epidemiological investigations may at least partly rely on dysregulated central nervous insulin signaling. Although further studies are needed to substantiate the promising results of proof-of- concept experiments on central nervous insulin administration, overcoming insulin resistance in the brain may prove a viable therapeutic option in the treatment of these increasingly prevalent afflictions. Acknowledgments This work was supported by Deutsche Forschungsgemeinschaft (KFO126/B5), Germany. The funding source had no input in the preparation, review, or approval of the manuscript. 12
  • 13. Reference List 1. Bernard C. Leçons de physiologie expérimentale appliquée à la médecine faites au Collège de France. Baillère et Fils Paris, France 1855;296-313. 2. Havrankova J., Roth J., Brownstein M. Insulin receptors are widely distributed in the central nervous system of the rat. Nature 1978; 272:827-829. 3. Sara V.R., Hall K., Von H.H., Humbel R., Sjogren B., Wetterberg L. Evidence for the presence of specific receptors for insulin-like growth factors 1 (IGE-1) and 2 (IGF-2) and insulin throughout the adult human brain. Neurosci Lett 1982; 34:39-44. 4. Potau N., Escofet M.A., Martinez M.C. Ontogenesis of insulin receptors in human cerebral cortex. J Endocrinol Invest 1991; 14:53-58. 5. Marks J.L., Porte D., Jr., Stahl W.L., Baskin D.G. Localization of insulin receptor mRNA in rat brain by in situ hybridization. Endocrinology 1990; 127:3234-3236. 6. Devaskar S.U., Giddings S.J., Rajakumar P.A., Carnaghi L.R., Menon R.K., Zahm D.S. Insulin gene expression and insulin synthesis in mammalian neuronal cells. J Biol Chem 1994; 269:8445-8454. 7. Hrytsenko O., Wright J.R., Jr., Morrison C.M., Pohajdak B. Insulin expression in the brain and pituitary cells of tilapia (Oreochromis niloticus). Brain Res 2007; 1135:31- 40. 8. Steen E., Terry B.M., Rivera E.J. et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer's disease--is this type 3 diabetes? J Alzheimers Dis 2005; 7:63-80. 9. Woods S.C., Seeley R.J., Baskin D.G., Schwartz M.W. Insulin and the blood-brain barrier. Curr Pharm Des 2003; 9:795-800. 10. Banks W.A., Jaspan J.B., Kastin A.J. Selective, physiological transport of insulin across the blood-brain barrier: novel demonstration by species-specific radioimmunoassays. Peptides 1997; 18:1257-1262. 11. Schwartz M.W., Bergman R.N., Kahn S.E. et al. Evidence for entry of plasma insulin into cerebrospinal fluid through an intermediate compartment in dogs. Quantitative aspects and implications for transport. J Clin Invest 1991; 88:1272-1281. 12. Air E.L., Benoit S.C., Clegg D.J., Seeley R.J., Woods S.C. Insulin and leptin combine additively to reduce food intake and body weight in rats. Endocrinology 2002; 143:2449-2452. 13. Bruning J.C., Gautam D., Burks D.J. et al. Role of brain insulin receptor in control of body weight and reproduction. Science 2000; 289:2122-2125. 14. Pocai A., Lam T.K., Gutierrez-Juarez R. et al. Hypothalamic K(ATP) channels control hepatic glucose production. Nature 2005; 434:1026-1031. 13
  • 14. 15. Rulifson E.J., Kim S.K., Nusse R. Ablation of insulin-producing neurons in flies: growth and diabetic phenotypes. Science 2002; 296:1118-1120. 16. Chiu S.L., Cline H.T. Insulin receptor signaling in the development of neuronal structure and function. Neural Dev 2010; 5:7. 17. Seeley R.J., Woods S.C. Monitoring of stored and available fuel by the CNS: implications for obesity. Nat Rev Neurosci 2003; 4:901-909. 18. Woods S.C., D'Alessio D.A. Central control of body weight and appetite. J Clin Endocrinol Metab 2008; 93:S37-S50. 19. Honda K., Kamisoyama H., Saneyasu T., Sugahara K., Hasegawa S. Central administration of insulin suppresses food intake in chicks. Neurosci Lett 2007; 423:153-157. 20. Brown L.M., Clegg D.J., Benoit S.C., Woods S.C. Intraventricular insulin and leptin reduce food intake and body weight in C57BL/6J mice. Physiol Behav 2006; 89:687- 691. 21. Hallschmid M., Benedict C., Schultes B., Fehm H.L., Born J., Kern W. Intranasal insulin reduces body fat in men but not in women. Diabetes 2004; 53:3024-3029. 22. Clegg D.J., Riedy C.A., Smith K.A., Benoit S.C., Woods S.C. Differential sensitivity to central leptin and insulin in male and female rats. Diabetes 2003; 52:682-687. 23. Benedict C., Kern W., Schultes B., Born J., Hallschmid M. Differential sensitivity of men and women to anorexigenic and memory-improving effects of intranasal insulin. J Clin Endocrinol Metab 2008; 93:1339-1344. 24. Hallschmid M., Schultes B. Central nervous insulin resistance: a promising target in the treatment of metabolic and cognitive disorders? Diabetologia 2009; 52:2264-2269. 25. Woods S.C., Lotter E.C., McKay L.D., Porte D., Jr. Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature 1979; 282:503-505. 26. van Dijk G., de Groote C., Chavez M., van der Werf Y., Steffens A.B., Strubbe J.H. Insulin in the arcuate nucleus of the hypothalamus reduces fat consumption in rats. Brain Res 1997; 777:147-152. 27. Wallum B.J., Taborsky G.J., Jr., Porte D., Jr. et al. Cerebrospinal fluid insulin levels increase during intravenous insulin infusions in man. J Clin Endocrinol Metab 1987; 64:190-194. 28. Tesfaye N., Seaquist E.R. Neuroendocrine responses to hypoglycemia. Ann N Y Acad Sci 2010; 1212:12-28. 29. Mitrakou A., Ryan C., Veneman T. et al. Hierarchy of glycemic thresholds for counterregulatory hormone secretion, symptoms, and cerebral dysfunction. Am J Physiol 1991; 260:E67-E74. 14
  • 15. 30. Cherrington A.D., Edgerton D., Sindelar D.K. The direct and indirect effects of insulin on hepatic glucose production in vivo. Diabetologia 1998; 41:987-996. 31. Urbanet R., Pilon C., Calcagno A. et al. Analysis of insulin sensitivity in adipose tissue of patients with primary aldosteronism. J Clin Endocrinol Metab 2010; 95:4037-4042. 32. Born J., Lange T., Kern W., McGregor G.P., Bickel U., Fehm H.L. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci 2002; 5:514- 516. 33. Alcala-Barraza S.R., Lee M.S., Hanson L.R., McDonald A.A., Frey W.H., McLoon L.K. Intranasal delivery of neurotrophic factors BDNF, CNTF, EPO, and NT-4 to the CNS. J Drug Target 2010; 18:179-190. 34. Fliedner S., Schulz C., Lehnert H. Brain uptake of intranasally applied radioiodinated leptin in Wistar rats. Endocrinology 2006; 147:2088-2094. 35. Thorne R.G., Emory C.R., Ala T.A., Frey W.H. Quantitative analysis of the olfactory pathway for drug delivery to the brain. Brain Res 1995; 692:278-282. 36. Illum L. Transport of drugs from the nasal cavity to the central nervous system. Eur J Pharm Sci 2000; 11:1-18. 37. Dhuria S.V., Hanson L.R., Frey W.H. Intranasal delivery to the central nervous system: mechanisms and experimental considerations. J Pharm Sci 2010; 99:1654- 1673. 38. Thorne R.G., Pronk G.J., Padmanabhan V., Frey W.H. Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration. Neuroscience 2004; 127:481-496. 39. Henkin R.I. Inhaled insulin-intrapulmonary, intranasal, and other routes of administration: mechanisms of action. Nutrition 2010; 26:33-39. 40. Duan X., Mao S. New strategies to improve the intranasal absorption of insulin. Drug Discov Today 2010; 15:416-427. 41. Kern W., Born J., Schreiber H., Fehm H.L. Central nervous system effects of intranasally administered insulin during euglycemia in men. Diabetes 1999; 48:557- 563. 42. Hallschmid M., Schultes B., Marshall L. et al. Transcortical direct current potential shift reflects immediate signaling of systemic insulin to the human brain. Diabetes 2004; 53:2202-2208. 43. Tschritter O., Preissl H., Hennige A.M. et al. The cerebrocortical response to hyperinsulinemia is reduced in overweight humans: a magnetoencephalographic study. Proc Natl Acad Sci U S A 2006; 103:12103-12108. 44. Tschritter O., Preissl H., Hennige A.M. et al. The insulin effect on cerebrocortical theta activity is associated with serum concentrations of saturated nonesterified Fatty acids. J Clin Endocrinol Metab 2009; 94:4600-4607. 15
  • 16. 45. Tschritter O., Preissl H., Yokoyama Y., Machicao F., Haring H.U., Fritsche A. Variation in the FTO gene locus is associated with cerebrocortical insulin resistance in humans. Diabetologia 2007; 50:2602-2603. 46. Wallner-Liebmann S., Koschutnig K., Reishofer G. et al. Insulin and hippocampus activation in response to images of high-calorie food in normal weight and obese adolescents. Obesity (Silver Spring) 2010; 18:1552-1557. 47. Guthoff M., Grichisch Y., Canova C. et al. Insulin modulates food-related activity in the central nervous system. J Clin Endocrinol Metab 2010; 95:748-755. 48. Diekelmann S., Born J. The memory function of sleep. Nat Rev Neurosci 2010; 11:114-126. 49. Feldman D.E. Synaptic mechanisms for plasticity in neocortex. Annu Rev Neurosci 2009; 32:33-55. 50. Huang C.C., Lee C.C., Hsu K.S. The role of insulin receptor signaling in synaptic plasticity and cognitive function. Chang Gung Med J 2010; 33:115-125. 51. Moult P.R., Harvey J. Hormonal regulation of hippocampal dendritic morphology and synaptic plasticity. Cell Adh Migr 2008; 2:269-275. 52. Chiu S.L., Chen C.M., Cline H.T. Insulin receptor signaling regulates synapse number, dendritic plasticity, and circuit function in vivo. Neuron 2008; 58:708-719. 53. Craft S., Watson G.S. Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol 2004; 3:169-178. 54. Seaquist E.R., Damberg G.S., Tkac I., Gruetter R. The effect of insulin on in vivo cerebral glucose concentrations and rates of glucose transport/metabolism in humans. Diabetes 2001; 50:2203-2209. 55. Fanelli C.G., Dence C.S., Markham J. et al. Blood-to-brain glucose transport and cerebral glucose metabolism are not reduced in poorly controlled type 1 diabetes. Diabetes 1998; 47:1444-1450. 56. Hasselbalch S.G., Knudsen G.M., Videbaek C. et al. No effect of insulin on glucose blood-brain barrier transport and cerebral metabolism in humans. Diabetes 1999; 48:1915-1921. 57. Doyle P., Cusin I., Rohner-Jeanrenaud F., Jeanrenaud B. Four-day hyperinsulinemia in euglycemic conditions alters local cerebral glucose utilization in specific brain nuclei of freely moving rats. Brain Res 1995; 684:47-55. 58. Benedict C., Hallschmid M., Hatke A. et al. Intranasal insulin improves memory in humans. Psychoneuroendocrinology 2004; 29:1326-1334. 59. Benedict C., Hallschmid M., Schmitz K. et al. Intranasal insulin improves memory in humans: superiority of insulin aspart. Neuropsychopharmacology 2007; 32:239-243. 16
  • 17. 60. Unger J.W., Livingston J.N., Moss A.M. Insulin receptors in the central nervous system: localization, signalling mechanisms and functional aspects. Prog Neurobiol 1991; 36:343-362. 61. Reger M.A., Watson G.S., Green P.S. et al. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology 2008; 70:440-448. 62. Reger M.A., Watson G.S., Green P.S. et al. Intranasal insulin administration dose- dependently modulates verbal memory and plasma amyloid-beta in memory-impaired older adults. J Alzheimers Dis 2008; 13:323-331. 63. Mosconi L., Pupi A., De Leon M.J. Brain glucose hypometabolism and oxidative stress in preclinical Alzheimer's disease. Ann N Y Acad Sci 2008; 1147:180-195. 64. Mosconi L., Mistur R., Switalski R. et al. Declining brain glucose metabolism in normal individuals with a maternal history of Alzheimer disease. Neurology 2009; 72:513-520. 65. Yasuno F., Imamura T., Hirono N. et al. Age at onset and regional cerebral glucose metabolism in Alzheimer's disease. Dement Geriatr Cogn Disord 1998; 9:63-67. 66. Reger M.A., Watson G.S., Frey W.H. et al. Effects of intranasal insulin on cognition in memory-impaired older adults: modulation by APOE genotype. Neurobiol Aging 2006; 27:451-458. 67. Francis G.J., Martinez J.A., Liu W.Q. et al. Intranasal insulin prevents cognitive decline, cerebral atrophy and white matter changes in murine type I diabetic encephalopathy. Brain 2008; 131:3311-3334. 68. Grillo C.A., Piroli G.G., Kaigler K.F., Wilson S.P., Wilson M.A., Reagan L.P. Downregulation of hypothalamic insulin receptor expression elicits depressive-like behaviors in rats. Behav Brain Res 2011; 222:230-235. 69. Marks D.R., Tucker K., Cavallin M.A., Mast T.G., Fadool D.A. Awake intranasal insulin delivery modifies protein complexes and alters memory, anxiety, and olfactory behaviors. J Neurosci 2009; 29:6734-6751. 70. Lin H.V., Plum L., Ono H. et al. Divergent regulation of energy expenditure and hepatic glucose production by insulin receptor in agouti-related protein and POMC neurons. Diabetes 2010; 59:337-346. 71. Morton G.J., Cummings D.E., Baskin D.G., Barsh G.S., Schwartz M.W. Central nervous system control of food intake and body weight. Nature 2006; 443:289-295. 72. Porte D., Jr., Baskin D.G., Schwartz M.W. Insulin signaling in the central nervous system: a critical role in metabolic homeostasis and disease from C. elegans to humans. Diabetes 2005; 54:1264-1276. 73. McFarlane S.I. Insulin therapy and type 2 diabetes: management of weight gain. J Clin Hypertens (Greenwich ) 2009; 11:601-607. 74. Russell-Jones D., Khan R. Insulin-associated weight gain in diabetes--causes, effects and coping strategies. Diabetes Obes Metab 2007; 9:799-812. 17
  • 18. 75. Enoksson S., Degerman E., Hagstrom-Toft E., Large V., Arner P. Various phosphodiesterase subtypes mediate the in vivo antilipolytic effect of insulin on adipose tissue and skeletal muscle in man. Diabetologia 1998; 41:560-568. 76. Stralfors P., Bjorgell P., Belfrage P. Hormonal regulation of hormone-sensitive lipase in intact adipocytes: identification of phosphorylated sites and effects on the phosphorylation by lipolytic hormones and insulin. Proc Natl Acad Sci U S A 1984; 81:3317-3321. 77. Koch L., Wunderlich F.T., Seibler J. et al. Central insulin action regulates peripheral glucose and fat metabolism in mice. J Clin Invest 2008; 118:2132-2147. 78. Scherer T., O'Hare J., Diggs-Andrews K. et al. Brain insulin controls adipose tissue lipolysis and lipogenesis. Cell Metab 2011; 13:183-194. 79. Sanchez-Alavez M., Tabarean I.V., Osborn O. et al. Insulin causes hyperthermia by direct inhibition of warm-sensitive neurons. Diabetes 2010; 59:43-50. 80. Benedict C., Brede S., Schioth H.B. et al. Intranasal insulin enhances postprandial thermogenesis and lowers postprandial serum insulin levels in healthy men. Diabetes 2011; 60:114-118. 81. Bergman R.N. Orchestration of glucose homeostasis: from a small acorn to the California oak. Diabetes 2007; 56:1489-1501. 82. Cherrington A.D. Banting Lecture 1997. Control of glucose uptake and release by the liver in vivo. Diabetes 1999; 48:1198-1214. 83. Cherrington A.D. The role of hepatic insulin receptors in the regulation of glucose production. J Clin Invest 2005; 115:1136-1139. 84. Obici S., Feng Z., Karkanias G., Baskin D.G., Rossetti L. Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats. Nat Neurosci 2002; 5:566-572. 85. Edgerton D.S., Lautz M., Scott M. et al. Insulin's direct effects on the liver dominate the control of hepatic glucose production. J Clin Invest 2006; 116:521-527. 86. Bouche C., Lopez X., Fleischman A. et al. Insulin enhances glucose-stimulated insulin secretion in healthy humans. Proc Natl Acad Sci U S A 2010; 107:4770-4775. 87. Chen M., Woods S.C., Porte D., Jr. Effect of cerebral intraventricular insulin on pancreatic insulin secretion in the dog. Diabetes 1975; 24:910-914. 88. Woods S.C., Porte D., Jr. Effect of intracisternal insulin on plasma glucose and insulin in the dog. Diabetes 1975; 24:905-909. 89. Stockhorst U., de Fries D., Steingrueber H.J., Scherbaum W.A. Unconditioned and conditioned effects of intranasally administered insulin vs placebo in healthy men: a randomised controlled trial. Diabetologia 2011; 54:1502-1506. 18
  • 19. 90. Craft S., Peskind E., Schwartz M.W., Schellenberg G.D., Raskind M., Porte D., Jr. Cerebrospinal fluid and plasma insulin levels in Alzheimer's disease: relationship to severity of dementia and apolipoprotein E genotype. Neurology 1998; 50:164-168. 91. Kern W., Benedict C., Schultes B. et al. Low cerebrospinal fluid insulin levels in obese humans. Diabetologia 2006; 49:2790-2792. 92. Guthoff M., Stingl K.T., Tschritter O. et al. The insulin-mediated modulation of visually evoked magnetic fields is reduced in obese subjects. PLoS One 2011; 6:e19482. 93. Hallschmid M., Benedict C., Schultes B., Born J., Kern W. Obese men respond to cognitive but not to catabolic brain insulin signaling. Int J Obes (Lond) 2008; 32:275- 282. 94. Bohringer A., Schwabe L., Richter S., Schachinger H. Intranasal insulin attenuates the hypothalamic-pituitary-adrenal axis response to psychosocial stress. Psychoneuroendocrinology 2008; 33:1394-1400. 95. Marino J.S., Xu Y., Hill J.W. Central insulin and leptin-mediated autonomic control of glucose homeostasis. Trends Endocrinol Metab 2011. 96. Schwartz M.W., Porte D., Jr. Diabetes, obesity, and the brain. Science 2005; 307:375- 379. 97. Whitmer R.A., Gustafson D.R., Barrett-Connor E., Haan M.N., Gunderson E.P., Yaffe K. Central obesity and increased risk of dementia more than three decades later. Neurology 2008; 71:1057-1064. 98. Raffaitin C., Feart C., Le G.M. et al. Metabolic syndrome and cognitive decline in French elders: the Three-City Study. Neurology 2011; 76:518-525. 99. Trudeau F., Gagnon S., Massicotte G. Hippocampal synaptic plasticity and glutamate receptor regulation: influences of diabetes mellitus. Eur J Pharmacol 2004; 490:177- 186. 100. Kodl C.T., Seaquist E.R. Cognitive dysfunction and diabetes mellitus. Endocr Rev 2008; 29:494-511. 101. Raber J., Huang Y., Ashford J.W. ApoE genotype accounts for the vast majority of AD risk and AD pathology. Neurobiol Aging 2004; 25:641-650. 102. Speakman J.R., Rance K.A., Johnstone A.M. Polymorphisms of the FTO gene are associated with variation in energy intake, but not energy expenditure. Obesity (Silver Spring) 2008; 16:1961-1965. 103. Cecil J.E., Tavendale R., Watt P., Hetherington M.M., Palmer C.N. An obesity- associated FTO gene variant and increased energy intake in children. N Engl J Med 2008; 359:2558-2566. 104. Konner A.C., Klockener T., Bruning J.C. Control of energy homeostasis by insulin and leptin: targeting the arcuate nucleus and beyond. Physiol Behav 2009; 97:632-638. 19
  • 20. 105. Landreth G., Jiang Q., Mandrekar S., Heneka M. PPARgamma agonists as therapeutics for the treatment of Alzheimer's disease. Neurotherapeutics 2008; 5:481- 489. 106. Pathan A.R., Gaikwad A.B., Viswanad B., Ramarao P. Rosiglitazone attenuates the cognitive deficits induced by high fat diet feeding in rats. Eur J Pharmacol 2008; 589:176-179. 107. Risner M.E., Saunders A.M., Altman J.F. et al. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer's disease. Pharmacogenomics J 2006; 6:246-254. 108. Watson G.S., Cholerton B.A., Reger M.A. et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. Am J Geriatr Psychiatry 2005; 13:950-958. 109. Gupta A., Bisht B., Dey C.S. Peripheral insulin-sensitizer drug metformin ameliorates neuronal insulin resistance and Alzheimer's-like changes. Neuropharmacology 2011; 60:910-920. 110. Obici S., Zhang B.B., Karkanias G., Rossetti L. Hypothalamic insulin signaling is required for inhibition of glucose production. Nat Med 2002; 8:1376-1382. 111. Anthony K., Reed L.J., Dunn J.T. et al. Attenuation of insulin-evoked responses in brain networks controlling appetite and reward in insulin resistance: the cerebral basis for impaired control of food intake in metabolic syndrome? Diabetes 2006; 55:2986- 2992. 112. Dallman M.F., Pecoraro N.C., la Fleur S.E. et al. Glucocorticoids, chronic stress, and obesity. Prog Brain Res 2006; 153:75-105. 113. Hibberd C., Yau J.L., Seckl J.R. Glucocorticoids and the ageing hippocampus. J Anat 2000; 197 Pt 4:553-562. 114. Strachan M.W., Reynolds R.M., Frier B.M., Mitchell R.J., Price J.F. The role of metabolic derangements and glucocorticoid excess in the aetiology of cognitive impairment in type 2 diabetes. Implications for future therapeutic strategies. Diabetes Obes Metab 2009; 11:407-414. 115. Fishel M.A., Watson G.S., Montine T.J. et al. Hyperinsulinemia provokes synchronous increases in central inflammation and beta-amyloid in normal adults. Arch Neurol 2005; 62:1539-1544. 116. Mayer C.M., Belsham D.D. Central insulin signaling is attenuated by long-term insulin exposure via insulin receptor substrate-1 serine phosphorylation, proteasomal degradation, and lysosomal insulin receptor degradation. Endocrinology 2010; 151:75- 84. 20
  • 21. Figure 1. Insulin-mediated crosstalk between brain and body periphery Intranasal/CNS and systemic insulin affect hepatic glucose production, pancreatic insulin secretion, adipocyte function, energy homeostasis, and cognitive function. Respective citations in text boxes refer to the reference list of the main text. 21
  • 22. 2-4-1-9-5-8-3 Moon-Night Intranasal/CNS insulin enhances Intranasal insulin declarative (e.g. word pairs) and bypasses the BBB and working memory (e.g. number achieves maximal CSF sequences) learning [23;58;59]. concentrations within 40 minutes [32]. Intranasal Insulin reaches the brain via receptor-mediated saturable insulin transport across the blood- Intranasal/CNS insulin decreases brain-barrier [9;10]. CNS insulin, like food intake and enhances CNS insulin inhibits systemic insulin, postprandial thermogenesis hepatic gluconeogenesis inhibits lipolysis and [21;23;80]. CNS insulin increases via vagal efferences pancreatic insulin secretion stimulates lipogenesis [14;110]. (feed-forward loop [87;88].) [77;78]. Liver Pancreas Systemic insulin Adipocytes inhibits gluco- Systemic insulin neogenesis via inhibits lipolysis and hepatic insulin stimulates receptors [82;83]. lipogenesis [75;76]. 22