mini-review: insulin resistant brain state and its ... · web viewregulation of brain insulin, in...

54
Mini-review INSULIN RESISTANT BRAIN STATE AND ITS LINK TO DIABETES MELLITUS

Upload: others

Post on 31-Jan-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Mini-review: INSULIN RESISTANT BRAIN STATE AND ITS LINK TO DIABETES MELLITUS

M Šalković-Petrišić, Z. Lacković

Mini-review

INSULIN RESISTANT BRAIN STATE AND ITS LINK TO DIABETES MELLITUS

Running title:

brain insulin resistance

Melita Šalkovic-Petrišić*1, Zdravko Lacković1

*1Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Medical School & Croation Institute for Brain Research, University of Zagreb, Šalata 11, HR-10 000 Zagreb, Croatia.

telephone: +385-1-45 90 219

telefax: +385-1-45 66 843

e-mail:

[email protected]

* To whom the correspondence should be addressed

Abbreviations:

CNS, central nervous system; BBB, blood-brain barrier; IR, insulin receptor; IRS, insulin receptor substrate; PKB/Act, protein kinase B /Act; PI3, phosphatydilinositol–3 kinase; GSK-3, glycogen synthase kinase-3; APP, amyloid precursor protein; Aβs, amyloid-β peptides; GLUT, glucose transporter; MAPK, mitogen activated protein kinase; IGF-1, insulin-like growth factor-1

ABSTRACT

Insulin resistant brain state is a dysfunction of the neuronal insulin receptor (IR) signalling, demonstrated mostly in phosphatydilinositol-3 kinase pathway that resembles IR signaling dysfunction in the peripheral insulin resistance in type 2 diabetes mellitus. The resulted brain insulin dysfunction may be manifested as altered feeding behaviour and cognitive deficits in learning and memory in relation to aging, obesity, diabetes mellitus type 2, and late-onset sporadic Alzehimer’s disease. These conditions represent both, primarily central (aging and Alzheimer’s disease) and primarily peripheral (obesity and type diabetes) insulin resistance state. Current knowledge suggests a bi-directional communication between these two insulin resistant states, one increasing the risk for development of another and vice versa. In spite of reported genetic, metabolic, nutritional and environmental contributing factors, it is still not clear in which conditions insulin resistant brain state and the peripheral insulin resistance co-exist with manifested clinical symptoms of both disorders. Research is focused on discovering the trigger determining which target tissue, neuronal or non-neural, becomes first resistant to insulin and what initiates the afterwards development of insulin resistance in the left over target tissue.

INTRODUCTION

With a progression of brain research in diabetes mellitus, new perceptions have emerged revealing that within the brain, a condition of “cerebral” diabetes could develop. To set the stage for defining the “cerebral” diabetes in this review, it is necessary to turn first to the main features of diabetes mellitus.

Diabetes mellitus is a heterogeneous metabolic disorder characterized by an inappropriate glucose metabolism that leads to impaired removal of glucose from the circulation. The clearance of glucose from blood is mediated by insulin which following binding to insulin receptor (IR) activates cellular glucose uptake. Therefore, absolute or relative lack of insulin and/or impaired insulin action via IR causes delayed or almost absent metabolism of circulating glucose. Type 1 diabetes mellitus, in general, is caused by autoimmune destruction of insulin producing/secreting β cells in pancreas resulting in lack of insulin, while type 2 diabetes mellitus is associated with dysfunction of insulin intracellular signal transduction due to the cell does not couple insulin binding to IR with activation of the particular cell function, i.e. cell becomes resistant to the action of insulin (86). Insulin signaling within the cell is mediated in general by two functional cascades, one acting through phosphatidylinositol-3 (PI3) kinase pathway and mediating insulin effects on intermediary metabolism and glucose uptake, and one acting through mitogen activated protein kinase (MAPK) pathway, mediating cell growth and mitosis (50). Clinical insulin resistance seen in type 2 diabetes has been reported to involve inhibition of insulin-activated PI3 kinase pathway, that may be induced by products of fatty acid metabolism, cytokines and inflammatory cascade products (e.g. tumor necrosis factor-α /TNF-α/), etc. (25,27,56).

In line with these key features of diabetes mellitus, “cerebral” diabetes could be roughly define as a state of dysfunction of brain glucose metabolism and brain insulin system. Term “insulin resistant brain state” has recently been introduced instead of “cerebral diabetes”. Research of the brain insulin pathophysiology has been intensified in the last decade and dysregulation of insulin secretion and transport in the CNS along with insulin deficiency or resistance in the brain has been reported in relation to aging, obesity, diabetes mellitus and late-onset sporadic Alzheimer’s disease (32,47). This mini-review summarizes current knowledge about insulin resistant brain state and its relationship to diabetes mellitus, particularly type 2 diabetes.

BRAIN INSULIN

Insulin and IR are found throughout the central nervous system (CNS) (83). Since the main feature of the insulin resistant brain state is disorder of the IR signaling cascade (47), the review is focused primarily to this aspect and therefore, brain insulin and IR distribution, regulation, function and dissimilarities to the peripheral insulin system will be briefly summarized.

Insulin origin. The source of insulin in the brain might be either peripheral or local in origin, or both (3,83). Insulin from the blood crosses the blood brain barrier (BBB) by a saturable transport mechanism, but all regions of the BBB are not equally permeable to insulin. Namely, BBB transports insulin into the pons-medulla and hypothalamus twice time (4), and into the olfactory bulb two to eight times (5) faster than into the whole brain. This is consistent with finding of regionally specific distribution of insulin in the CNS with the olfactory bulb and hypothalamus containing the highest concentration of insulin (6). There are also several lines of evidence consistent with a local synthesis of insulin in the brain. Insulin mRNA is distributed in a highly specific pattern with the highest density in pyramidal cells of the hippocampus and a high density in the medial prefrontal cortex, the enthorinal cortex, perirhinal cortex, thalamus, granule cell layer of the olfactory bulb, and hypothalamus (24,104). Synthesized insulin was shown to be secreted into the extracellular space specifically by neurones (35).

Insulin receptor and its signalling. Two different types of IR have been found in adult mammalian brain; a peripheral type, detected in lower density on glia cells, and a neuron-specific type of IR that has been widely distributed in the CNS on the regionally specific basis (6). High concentrations of this neuronal IR mRNA (the following text refers to this type of IR) were found in the chorioid plexus, olfactory bulb, piryform cortex, amygdaloid nucleus, hippocampus, hypothalamic nucleus and cerebellar cortex (60). Expression of IR protein levels is in accordance with its mRNA levels in most areas, but significant discrepancy is found in the hippocampus and cerebellar cortex, suggesting a higher stability of IR in the hippocampus and probably a rapid turnover of the receptor in the cerebellar cortex neurons (106). The mayor molecular structure and most of the biochemical properties of the neuronal IR are indistinguishable from those found in the periphery, but some structural and functional differences between the CNS and the peripheral IR have been suggested; the molecular mass of alpha and beta subunits of the brain IR is slightly lower than that in the peripheral tissue (42), and high concentrations of insulin downregulate the peripheral IR, but have no effect on the brain IR (12). It has to be mentioned that two insulin receptor isoforms (IR-A and IR-B) generated by alternative mRNA splicing have been identified at the periphery, and their expression found to be highly regulated in tissue specific fashion (7). Namely, IR exists in two isoforms differing by the absence (Ex11-) or presence (Ex11+) of 12 amino acids in the C-terminus of the alpha-subunit due to alternative splicing of exon 11.Ex11- binds insulin with two-fold higher affinity than Ex11+. This difference is paralleled by a decreased sensitivity for metabolic actions of insulin. However, data reported so far suggest that heterogeneity of IR in different tissues including brain is unrelated to alternative splicing of IR gene (13,51). Also, inconsistent data have been reported regarding the abundance of the two isoforms in relation to insulin resistance in type 2 diabetes in humans (84).

Insulin signal transduction in the brain is similar to that at the periphery. Insulin receptor belongs to the receptor tyrosine kinase superfamily. Briefly, activated IR recruits insulin receptor substrate (IRS-1/2) adapter protein (also regionally specifically distributed in the CNS and co-expressed with IR in certain population of neurones /29/) to its phosphorylated docking site, which then becomes phosphorylated on tyrosine residues and capable to recruit various SH2 domain-containing signaling molecules, among which are PI3 kinase and adapter proteins for MAPK pathway (Fig. 1) (50). Activation of MAPK pathway leads to insulin-induced mitogenic effects. Activation of PI3 kinase pathway transduces the signal to protein kinase B (PKB/Act), that in the peripheral insulin-sensitive tissue and as also shown in some brain regions, triggers glucose transporter GLUT4 translocation and consequently cellular glucose uptake (50,98). In addition, PKB/Act can modulate the glycogen synthase kinase (GSK-3) pathway that in the peripheral insulin-sensitive tissue leads to glycogen synthesis (50). In the brain, beside other roles, GSK-3 has been related to the regulation of amyloid-β peptides (Aβs) and tau-protein phosphorylation (47). Therefore, metabolism of amyloid precursor protein (APP), the intracellular formation of secreted APPs and Aβs, and the release of APPs and Aβs into the extracellular space, as well as the balanced phosphorylation of tau-protein, are under control of insulin/IR signal transduction (47). Factors that affect phosphorylation/dephosphorylation homeostasis of elements in insulin signal transduction are capable of modifying this cascade causing its dysfunction, e.g., phosphorylation of particular serine and threonine residues of IRS protein reduces insulin-stimulated IRS-1 associated PI3 kinase activity that at the periphery has been causative of insulin resistance and type 2 diabetes (27).

Role of insulin in the brain. The biological effects of brain insulin depend on the availability of the hormone in the brain, its binding and activating specific brain IR with related signaling pathways, and to a minor extent also on regulation of the brain insulin that is synthesized within the CNS. Regional and state-dependent differences in transport across the BBB and in its distribution within the CNS have been suggested as possible factors that may explain how brain insulin can have so diverse effects (3). Namely, while the insulin/IR associated with the hypothalamus plays important role in regulation of the body energy homeostasis and feeding, the hippocampus- and cerebral cortex-distributed insulin/IR has been shown to be involved in brain cognitive function, including learning and memory (75,105).

Glucose is the primary fuel for the brain and cerebral metabolism of glucose requires its transport across the BBB by insulin-insensitive glucose transport GLUT1 (64), that has been found downregulated in uncontrolled diabetes (69), and upregulated in chronic hypoglycaemia (63). Majority of glucose utilization within the CNS appears to be mediated through glucose transporters GLUT1 and GLUT3, both insulin-insensitive (28). However, rapid and transient changes in cerebral glucose utilization may occur via insulin-sensitive glucose transporters GLUT4 and GLUT8, found in the brain (76,77,98). Glucose transporter GLUT4 is the main transporter type responsible for glucose uptake in the peripheral insulin target cell, and in the brain neuronal localization of GLUT4 mRNA and protein in hypothalamus, cerebellum, cortex and hippocampus exhibits overlapping distribution with IR, as well (57,65). Activation of brain IR leads to stimulation of the glycolytic key enzymes and pyruvate oxidation in the brain (48,78).

Growing evidence suggests that insulin interacts with both orexigenic and anorexigenic peptides in the brain in the control of feeding behaviour, maintenance of body weight and energy homeostasis, but most of the research has been focused on insulin “cross-talk” with leptin in the hypothalamus, as reviewed elsewhere (36,72). Both insulin and leptin acutely regulate the membrane potential and firing rates of a specific subset of hypothalamic neurones, the effect being dependent on signaling through PI3 kinase (89,90). Genetic studies demonstrated that brain-specific knockouts of the IR and animals lacking IRS-2 show a phenotype of obesity and reproductive dysfunction (14,16), and knockdown of IR expression locally in the hypothalamus resulted in cumulative food intake of 152% and fat mass of 186% relative to controls (73). These findings support the conclusion that insulin action in the hypothalamus is essential in the regulation of energy homeostasis. It has been suggested that central insulin has fundamentally catabolic (i.e. reducing food intake and body weight), whereas peripheral insulin has anabolic (i.e. increasing energy storage and potentially increasing body weight if an individual has too much insulin) activity (72). Like most physiological systems, the peripheral and central actions of insulin regarding this issue are balanced. Therefore, the lack of catabolic insulin action in the brain (coupled with disturbances like the decrease in serum leptin) leads to marked increases in food intake and obesity that is often associated with insulin resistance and hyperinsulinemia (72).

Insulin receptor signaling plays a role in synaptic plasticity in hippocampus by modulating activity of excitatory and inhibitory receptors and consequently affecting cognitive functions like learning and memory (105). During learning insulin binds to its receptors, and activated IR may be involved in memory formation via potentiation of glutamatergic NMDA receptor channel activity leading to increase in Ca 2+ influx and long-term potentiation (85). Via PI3 kinase it may be involved in long-term depression by internalisation of glutamatergic AMPA receptors (59), or via recruiting of functional GABA receptors to the postsynaptic membrane (99). Additionally, IR induced activation of MAPK pathway after learning may lead to regulation of gene expression that is required for long-term memory storage, while IR interaction with G-protein coupled receptor may activate protein kinase C leading to facilitation of short-term memory encoding (105). Also, IR-IRS-PI3 kinase pathway may trigger endothelial nitric oxide synthase (eNOS) activity and generation of nitric oxide that acts as a retrograde messenger for release of neurotransmitters involved in learning and synaptic plasticity (26,68).

Regulation of brain insulin. It is still a controversial issue if the effects of insulin on the brain should mainly be regarded as an extension of its peripheral action, or insulin effects on the brain that are clearly independent from its peripheral metabolic effects. If the latter is the case, how can these peripheral and central effects of insulin be regulated independent from each other, assuming that circulating insulin is the major source of insulin in the brain. This suggests not only the complexity of brain insulin regulation, but also the potential significance of the centrally originated portion of brain insulin.

Brain insulin is subject to a multifactorial control exerted at various levels. It can be regulated both peripherally and centrally; biosynthesis and secretion in the pancreas along with transport in the brain, internalisation, storage, stability, as well as its local synthesis and release within the CNS may be affected by numerous metabolites, circulating hormones, regulatory peptides and neurotransmitters (36). The molecular mechanisms involved in the production and release of insulin in the CNS seem to share similarities to that in the periphery: structures specific for glucose sensing and metabolizing at the level of beta cells, such as glucose transporter GLUT2 and glucokinase, have been demonstrated co-localized in particular hypothalamic cells (49); both beta cells and hypothalamic neurons contain ATP-sensitive-K+ channels, a key protein in the glucose response mechanism and insulin secretion (2,97). Furthermore, insulin released from adult rat brain synaptosomes under depolarising conditions depends on calcium influx, suggesting that insulin is stored in the adult rat brain in synaptic vesicles within nerve endings from which it can be mobilized by exocytosis in association to neural activity (101).

Regulation of brain insulin, in particular its release, has been investigated at the level of hypothalamus. Increased insulin release in hypothalamus has been found in relation to carbohydrate meals (37), and local glucose (38,82) and serotonin (74) increament, respectively. This regulation showed regional specificity, since the modifications induced in the hypothalamus were not observed in the cerebellum, and neither insulinemia nor glycemia were affected. Leptin may interact with insulin directly or via other neuromodulators (serotonin, melanocortin, etc.) and there is also a cross-talk of insulin and leptin receptors in the CNS (72). The potential effects of glucocorticoids on brain insulin could be a result of both peripheral and central action, where local regulation has been suggested to involve effects via glucose and serotonin (72). Participation of other hormones and regulatory peptides in regulation of brain insulin is also being investigated.

INSULIN RESISTANT BRAIN STATE AND AGING

It has been clinically demonstrated that with physiological aging a resistance to the action of insulin develops at the periphery, leading to slightly increased glycemia and insulin levels, as reviewed elsewhere (32). Several causes have been postulated for this insulin resistance with aging, among them also the alteration of peripheral IR signal transduction, such as decrease in IRS-1 protein level and its tyrosine residua phosphorylation (18), decrease in IRS-1 association with PI3 kinase and decrease in insulin-stimulated MAPK activity (33,67). The regulation of neuronal glucose metabolism during aging is diminished in the brain, as a result of complex age-associated alterations, but mainly because of the decreased neuronal insulin signal transduction (31). Namely, insulin and C-peptide concentrations decrease with aging (30%-45%) in all brain regions, while brain IR density decreases (38%-49%) in regionally specific manner, particularly stressed in frontal and parietal cortex, with IR tyrosine kinase activity also being decreased, particularly in the temporal region (31).

Additionally, IR signaling pathway in the brain interacts with signaling pathway of other factors, such as insulin growth factor I (IGF-I) and brain-derived neurotrophic factor (BDNF), the alteration of which may therefore contribute to brain insulin dysfunction. As previously mentioned, insulin plays a significant role in cognitive functions and significant decline in cognitive functions has been found with aging (22). Aging itself appears to be associated with decreased brain signaling of BDNF which is known to regulate synaptic plasticity, neurogenesis and neuronal survival in the adult brain (62). The receptor for BDNF belongs to tyrosine kinase superfamily and exerts its effects on neurones via a signal transduction mechanism similar to the insulin signaling pathway (30). When activated, BDNF receptor involves IRS-1 and -2 signaling pathway and stimulation of PI3 kinase which then activates PKB/Act. Activation of this pathway enhances learning and memory, can promote the survival of neurones in metabolic and oxidative stress, and also an important role in regulating glucose metabolism and possibly lifespan has been suggested (61). Alterations of BDNF and its overlapping with the part of insulin intracellular signaling may contribute to insulin resistant brain state in aging. Aging is also associated with a reduced activity of IGF-I in the CNS (91). In spite of relatively constant levels of mRNA in different brain regions, cortical IGF-I protein levels decrease significantly during aging process in rats (71). IGF-I and insulin share a common signal transduction mechanism involving IRS proteins and subsequent signaling pathway, moreover, IR and IGF-I receptors show relative promiscuity and can bind both IGF-I and insulin. There is ample evidence that IGF-I plays a role in cognitive functions, further supporting involvement of insulin signaling pathway dysfunction in age-related decline in cognitive functions (11,91).

There is a clear evidence that the function of the neuronal insulin/IR signal transduction is of a pivotal significance to maintain normal cerebral blood flow and oxidative energy metabolism, work of the endoplasmatic reticulum/Golgi apparatus and the cell cycle in terminally differentiated neurones no longer in the cell cycle (46). It has become evident that normal metabolism of both APP and tau-protein is a part of interactive processes controlled by the normal insulin/IR signal transduction. In physiological brain aging, the malfunction of this cascade starts, leading to various adverse effects that due to their multifolding and permanency may become severe and may increase the vulnerability of the aging brain, facilitating thus the generation of some age-related disorders, such as dementia and the late-onset sporadic Alzheimer’s disease (46).

INSULIN RESISTANT BRAIN STATE AND ALZHEIMER’S DISEASE

Alzheimer’s disease is the most common form of dementia among older adults. In spite of indistinguishable clinical dementia symptoms, there two types of origin-based Alzheimer’s disease. In a small proportion (familial early-onset Alzheimer’s disease), the disease is caused by missense mutations in three genes, resulting in permanent generation of APP derivative Aβ which aggregates forming amyloid and plaques, while in the great majority of people (late-onset Alzheimer’s disease), it is sporadic in origin with old age as main risk factor, where Aβ has not been proven to be necessary for the generation and development of the disease (47). Among other more or less known neurochemical alterations in the brain that are beyond the scope of this review, growing evidence has identified a potential association among Alzheimer’s disease, glucose metabolism, and insulin activity (47). Patients with Alzheimer’s disease may have decreased cerebrospinal fluid levels and decreased cerebrospinal fluid to plasma insulin ratio related to impaired transport of insulin across the BBB (21), but neuronal insulin signal transduction in this disease has been in the focus of the current research. In contrast to the more localised abnormalities in the early-onset type, the late-onset type of Alzheimer’s disease is associated with glucose utilization abnormalities distributed all over the cerebral cortex, and particularly in parietotemporal and frontal areas, in structures with both high glucose demands and high insulin sensitivity (43). Since it has become obvious that neuronal glucose metabolism is under the control of the neuronal insulin, the abnormalities in neuronal glucose metabolism in Alzheimer’s disease have been suggested to be caused at the level of insulin signal transduction (46).

The up-regulation of IR density observed in hippocampus and associated with reduced activity of IR tyrosine kinase in the brain of people with sporadic Alzheimer’s disease indicates a desensitization of the neuronal IR (21,31). Proposed mechanism of inhibition of neuronal IR in the late-onset sporadic Alzheimer’s disease is related to age-induced increase in cortisol and catecholamine levels that may compromise the phosphorylation of tyrosine residues in IR (39,47). This leads to a dysfunction of subsequent insulin signal transduction, which at the level of GSK-3 kinase (α and β subtypes) results in two main pathophysiological events. In the physiological condition, PKB/Act acts to phosphorylate GSK-3 at its serine 9 residue, thereby inactivating it. Insulin normally exerts a double-sided effect on Aβs, stimulating their neuronal release (mediated through GSK-3α kinase) and in the same time contributing to extraneuronal accumulation of Aβs by competing for insulin-degrading enzyme that degrades both insulin and Aβs (34). The net action of insulin is to increase extracellular levels of Aβs in the brain. Therefore, insulin resistance-induced disinhibition of GSK-3α function in the sporadic Alzheimer’s disease leads to increased storage of APP and Aβs in neurons which then undergo lysis to form amyloid plaques, one of the main pathological features of the Alzheimer’s disease (47). These Aβs in turn reduce the binding of insulin to its receptor and receptor autophosphorylation, which in the early-onset type of Alzheimer’s disease may be the cause of triggering the dysfunction of brain insulin signal transduction (103). Also, with the fall in extracellular APPs, its mediated functions in memory enhancement may be assumed to fail, contributing to cognitive deficits (66). On the other side, insulin resistance-induced disinhibition of GSK-3β function leads to uncontrolled hyperphosphorylation of tau-protein (47). Tau is a neuronal cytoskeletal protein that binds to microtubules and promotes tubulin polymerisation and stabilization. The binding of tau to microtubules is regulated through phosphorylation by protein kinases, including GSK-3β (47). In physiological condition, insulin inhibits GSK-3β and consequently reduces tau phosphorylation, promoting binding of tau to microtubules (44). Following brain insulin dysfunction hyperphosphorylated form of tau protein builds neurofibrillary tangles, the other important pathological feature of the Alzheimer’s disease.

This impairment of the insulin signal transduction causing insulin resistant brain state is similar to condition of systemic insulin resistance in non-neuronal tissues in type 2 diabetes mellitus, suggesting a hypothesis that sporadic Alzheimer’s disease is the brain equivalent of type 2 diabetes mellitus, in other words, a kind of “cerebral” diabetes (45). Comparable to type 2 diabetes mellitus, susceptibility genes in combination with the main risk factor aging dysregulate the function of insulin signaling cascade. In addition to insulin resistance, Alzheimer’s disease (familial and sporadic) and diabetes mellitus (type 1 and 2) share the similarity in fact that both diseases are heterogenic in origin and homogenic in clinical appearance of their respective subtypes (46). It has been proposed that whether type 2 diabetes or Alzheimer’s dementia develops as a consequence of loss of sensitivity to insulin, may depend on what target tissue, neuronal or non-neuronal, becomes resistant to it (17).

INSULIN RESISTANT BRAIN STATE AND DIABETES MELLITUS

Type 2 diabetes mellitus is associated with dysfunction of insulin intracellular signal transduction due to the cell does not couple insulin binding to IR with activation of the particular cell function, i.e. cell becomes resistant to the action of insulin. The intriguing question is whether peripheral insulin resistant state (type 2 diabetes) and central insulin resistant states (presented here as e.g. aging and sporadic Alzheimer’s disease) are two different, separated entities or just one condition that, like in the physics “Law of connected containers”, metaphorically speaking, can “overflow” from one container to another. In other words, does peripheral insulin resistance lead to development of central insulin resistance and vice versa? Due to insufficient level of current knowledge, answers to these questions could only be speculative ones.

Growing evidence indicates that these conditions may be connected; effects of diabetes and aging on the brain may interact, and both conditions may interact with the appearance of sporadic late-onset Alzheimer’s disease (8). Summarizing the aforementioned data, brain glucose metabolism and insulin signal transduction in the brain seem to be the interconnecting link. Decreased brain glucose metabolism and decreased brain insulin content have been demonstrated in diabetes (1) as well as in aging and in the sporadic Alzheimer’s disease, accompanied by dysfunction of brain IR signalling cascade (31). Disturbances in transducing the signal from IR to the intraneuronal structures in the brain can be caused by various factors that may inhibit the tyrosine phosphorylation of IRSs and PI3 kinase subunits, among which advanced glycation end products (AGEs) and oxidative stress were frequently mentioned (10,27,70). AGEs have been generated by glucose-induced reaction of glycation of long-lived macromolecules and tend to accumulate both in the periphery and in the brain with aging, as well as in diabetes and in Alzheimer’s disease (52,81,87). During the formation of AGEs free radicals are produced, and they all may interfere with the IR functioning and signal transduction by diminishing the tyrosine phosphorylation of IRS-1/2, which then is unable to activate PI3 kinase – PKB/Act pathway, leading to altered glucose/glycogen metabolism and insulin resistance (32). Contributing factor could also be oxidative stress mediated by free radicals (10). Preclinical and clinical data demonstrate that aging, diabetes and Alzheimer’s disease are all associated with increased oxidative stress both at the periphery and in the CNS (41,53,70). Chronic oxidative stress may cause chronic oxidizing of tyrosine phosphatase PTP-1B, enzyme that dephosphorylates IR tyrosine residues and reduces its activity, leading thus to insulin resistance (32). In the context of human aging, an association between the IR and the longevity can be drawn, since altered IR signaling and marked insulin resistance are associated with physiological aging. This may suggest that not only aging increases the risk for type 2 diabetes, but also that type 2 diabetes may lead to premature aging (32).

Increasing evidence supports the hypothesis that type 2 diabetes may increase the risk for Alzheimer’s disease. This involves not only those at the signal transduction level representing insulin resistance and those at the behavioural level representing cognitive deficits, but also at the morphological level demonstrating hippocampal and amygdalar atrophy on magnetic resonance imaging (which is a good, early marker of the degree of Alzheimer’s neuropathology) in type 2 diabetic patients (23), and at the structural level, demonstrating deposits of islet amyloid polypeptide in pancreas of diabetic patients, that share a 90% structural similarity with APP which forms deposits in brain of patients with Alzheimer’s disease. Conversely, patients with Alzheimer’s disease have an increased risk for aberrations in peripheral glucose metabolism exhibiting less efficient glucoregulation with slightly but significantly decreased basal arterial glucose concentration and increased plasma insulin concentration resembling partly to the type 2 diabetes condition (15,20). Another study also suggested that patients with Alzheimer’s disease may have abnormal insulin activity in the CNS as well as at the periphery (21).

Regardless the cause, insulin resistance in the brain leads to brain insulin dysfunction. Cognitive deficits, particularly manifested as memory and learning deficits, are well known and documented in relation to age and Alzheimer’s disease as well, and it has been demonstrated that insulin administration can facilitate memory in such individuals (100). Cognitive impairments associated with diabetes mellitus have been reported more consistently in type 2 diabetes characterised by increased insulin resistance, and individuals suffering from type 2 diabetes show an increased prevalence of dementia (19,79). Memory deficits are not usually evident in insulin-dependent type 1 diabetes in humans, and if they occur, are often associated with hypoglycaemia (80,88). In experimental type 1 (streptozotocin-induced) diabetes cognitive deficits have not been consistently reported (9). However, duration and severity dependent distinct changes in hippocampal synaptic plasticity, associated with deficits in NMDA-dependent long-term potentiation probably related to insulin dysfunction, have been reported in streptozotocin-diabetic animals (40).

Beside cognitive deficit discrepancies between type 1 and type 2 diabetes, in general similar changes of monoaminergic neurotransmission at the neurochemical level were observed in streptozotocin-diabetic animals (representing type 1 diabetes) and in streptozotocin-intracerebroventricularly (icv) treated animals (representing an experimental model for sporadic Alzheimer’s disease and brain type 2 diabetes) (54,55,93-96). However, while changes of monoamines and their metabolite content, monoamine turnover rate, as well of monoamine synaptic transporters and dopaminergic receptors were irreversible and tend to progress with the duration of streptozotocin-induced diabetes, in streptozotocin icv-treated animals they seem to be reversible. Bearing in mind that streptozotocin is selectively toxic for insulin producing/secreting cells (92), and that insulin (possibly co-localised in catecholamine-containing neurons) has a neuromodulatory role in synaptic monoaminergic transmission (83,102), these results may suggest similar, insulin-related mechanism of induction the neurochemical changes in brain (at least at the level of monoamine transmission), regardless whether peripheral or central insulin-producing cells have been affecting. The tendency of reversibility of streptozotocin icv-induced changes may suggest that either the damage of brain producing/secreting cells alone is not enough for brain insulin dysfunction to be persistent, or that brain cell damage can somehow be compensated (reversibility depending possibly on streptozotocin dose, or other mechanisms including molecules that share the signalling pathway with insulin, like IGF-1).

Molecular biology and genetic studies give support to the hypothesis that brain insulin dysfunction could have consequences spreading to the periphery, as well. Neuron-specific disruption of the IR gene in mice results in development of diet-sensitive obesity with increases in body fat and plasma leptin levels, insulin resistance, hyperinsulinemia and hypertriglyceridemia, features seen in type 2 diabetes (14). The phenotype of a knock-out mouse model lacking the IRS-2 is similar to model with neuron-specific deletion of IR characterized by increased food intake, body fat content and impaired hypothalamic control of reproduction (16,36). Very recent data indicated that conditional knockout of IRS-2 in mouse pancreas β cells and parts of brain including hypothalamus caused obesity and insulin resistance that progressed to diabetes (58).However, diabetes resolved when functional β cells expressing IRS-2 repopulated the pancreas, restoring sufficient β cell function to compensate for insulin resistance, supporting the aforementioned hypothesis about possible compensation of function/structure of damaged insulin producing/secreting cells in the brain. Furthermore, insulin infusion in the third cerebral ventricle suppresses glucose production at the periphery independent of circulating levels of insulin, which suggests that hypothalamic insulin resistance can contribute to hyperglycemia that may lead to type 2 diabetes (36,73). This indicates that a decrease in the IR number, defects in IR function and signaling, and insulin lack or resistance in the brain may lead to the development of type 2 diabetes even when pancreatic β-cells are normal.

CONCLUSION

Brain insulin resistant state is characterized by a dysfunction of the neuronal IR signaling cascade demonstrated in the PI3 kinase pathway, the consequences being manifested mostly as disturbed APP/Aβs metabolism and tau-protein hyperphosphorylation, cognitive deficits in learning and memory, and altered feeding behaviour. Accumulating data demonstrate that insulin resistant brain state is associated with conditions like aging, obesity, sporadic Alzheimer’s disease, and type 2 diabetes mellitus. Aging and sporadic Alzheimer’s disease may represent insulin resistance primarily in neuronal, non-classical target tissue, while obesity and type 2 diabetes may represent insulin resistance primarily in non-neuronal, classical insulin target tissue. Current knowledge suggests a bi-directional communication of central and peripheral insulin resistant state, respectively, one may increase the risk for development of another and vice versa. However, proposed mechanisms of insulin resistance, like AGEs accumulation and oxidative stress found both centrally and peripherally, do not offer plausible answer for the questions which tissue, neuronal or non-neuronal, becomes first resistant to the insulin, and why the central and peripheral insulin resistance do not develop simultaneously if their cause is present in both tissue types and body compartments. Various genetic, metabolic, nutritional and environmental factors certainly have an important role in this issue. However, the thin line that has to be crossed for both insulin resistant states to be developed and clinically manifested still remains to be discovered providing a challenging subject for further research.

Acknowledgements: Partly supported by The Ministry of Science, Technology and Sport, Republic of Croatia (No. 108134). Professor Peter Riederer and Professor Siegfried Hoyer are thanked for a fruitful discussion realised through a collaborative project “Stability Pact for South Eastern Europe” supported by Deutscher Akademischer Austausch Dienst (DAAD).

REFERENCES

1. AGARDH CD, LESNIAK MA, GERRITSEN GC, ROTH J 1986 The influence of plasma insulin concentrations on tissue insulin levels in rodents: a study of the diabetic chinese hamster and the ob/ob mouse. Metabolism 35: 244-249

2. ASHFORD ML, BODEN PR, TREHERNE JM 1990 Glucose-induced excitation of hypothalamic neurones is mediated by ATP-sensitive K+ channels. Pflugers Arch Eur J Physiol 415: 479-483

3. BANKS WA 2004 The source of cerebral insulin. Eur J Pharmacol 490: 5-12

4. BANKS WA, KASTIN AJ 1998 Differential permeability of the blood-brain barrier to two pancreatic peptides: insulin and amylin. Peptides 19: 883-889

5. BANKS WA KASTIN AJ, PAN W 1999 Uptake and degradation of blood-borne insulin by the olfactory bulb. Peptides 20: 373-378

6. BASKIN DG, PORTE Jr D, GUEST K, DORSA DM 1983 Regional concentrations of insulin in the rat brain. Endocrinology 112: 898-903

7. BENECKE H, FLIER JS, MOLLER DE 1992 Alternative spliced variants of the insulin receptor protein. Expression in normal and diabetic human tissues. J Clin Invest 89: 2066-2070

8. BIESSELS GJ, BRAVENBOER B, GISPEN WH 2004 Glucose, insulin and the brain: modulation of cognition and synaptic plasticity in health and disease: a preface. Eur J Pharmacol 490: 1-4

9. BIESSELS GJ, KAMAL A, RAMAKERS GM, URBAN IJ, SPRUIJT BM, ERKELENS DW, GISPEN WH 1996 Place learning and hippocampal synaptic plasticity in streptozotocin-induced diabetic rats. Diabetes 45: 1259-1266

10. BIESSELS GJ, VAN DER HEIDE LP, KAMAL A, BLEYS RL, GISPEN WH 2002 Ageing and diabetes: implications for brain function. Eur J Pharmacol 441: 1-14

11. BONDY CA, CHENG CM 2004 Signaling by insulin-like growth factor 1 in brain. Eur J Pharmacol 490: 25-31

12. BOYD Jr FT, RAIZADA MK 1983 Effects of insulin and tunicamycin on neuronal insulin receptors in culture. Am J Physiol 245: C283-C287

13. BREINER M, WEILAND M, BECKER W, MULLER-WIELAND D, STREICHER R, FABRY M, JOOST HG 1993 Heterogeneity of insulin receptors in rat tissues as detected with the partial agonist B29, B29’-suberoyl-insulin. Mol Pharmacol 44: 271-276

14. BRUNNING JC, GAUTAM D, BURKS DJ, GILLETTE J, SCHUBERT M, ORBAN PC, KLEIN R, KRONE W, MULLER-WIELAND D, KAHN CR 2000 Role of brain insulin receptor in control of body weight and reproduction. Science 289: 2122-2125

15. BUCHT G, ADOLFSSON R, LITHNER F, WINBLAD B 1983 Changes in blood glucose and insulin secretion in patients with senile dementia of Alzheimer type. Acta Med Scand 213: 367-392

16. BURKS DJ, DE MORA JF, SCHUBERT M, WITHERS DJ, MYERS MG, TOWERY HH, ALTAMURO SL, FLINT CL, WHITE MF 2000 IRS-2 pathways integrate female reproduction and energy homeostasis. Nature 407: 377-382

17. CARRO E, TORRES-ALEMAN I 2004 The role of insulin and insulin-like growth factor I in the molecular and cellular mechanisms underlying the pathology of Alzheimer’s disease. Eur J Pharmacol 490: 127-133

18. CARVALHO CR, BRENELLI SL, SILVA AC, NUNEZ AI, VELLOSO LA, STAAD MJ 1996 Effect of aging on insulin receptor, insulin receptor substrate-1 and phosphatidylinositol 3-kinase in liver and muscle rats. Endocrinology 137: 151-159

19. COSWAY R, STRACHAN MW, DOUGALL A, FRIER BM, DEARY IJ 2001 Cognitive function and information processing in type 2 diabetes. Diabet Med 18: 803-810

20. CRAFT S, NEWCOMER J, KANNE S, DAGOGO-JACK S, CRYER P, SHELINE Y, LUBY J, DAGOGO-JACK A, ALDERSON A 1996 Memory improvement following induced hyperinsulinemia in Alzheimer’s disease. Neurobiol Aging 17: 123-130

21. CRAFT S, PESKIND E, SCHWARTZ MW, SCHELLENBERG GD, RASKIND M, PORTE Jr D 1998 Cerebrospinal fluid and plasma insulin levels in Alzheimer’s disease: relationship to severity of dementia and apolipoprotein E genotype. Neurology 50: 164-168

22. DAL FORNO G, KAWAS CH 1995 Cognitive problems in the elderly. Curr Opin Neurol 8: 256-261

23. DEN HEIJER T, VERMEER SE, VAN DIJK EJ, PRINS ND, KOUDSTAAL PJ, HOFMAN A, BRETELER MMB 2003 Type 2 diabetes and atrophy of medial temporal lobe structures on brain MRI. Diabetologia 46: 1604-1610

24. DEVASKAR SU, GIDDINS SJ, RAJAKUMAR PA, CANAGHI LR, MENON RK, ZAHM DS 1994 Insulin gene expression and insulin synthesis in mammalian neuronal cells. J Biol Chem 269: 8445-8454

25. DIB K, WHITEHEAD JP, HUMOHREYS PJ, SOOS MA, BAYNES KCR, KUMAR S, HARVEY T, O’RAHILLY S 1998 Impaired activation of phosphoinositide 3-kinase by insulin in fibroblasts from patients with severe insulin resistance and pseudoacromegaly. J Clin Invest 101: 1111-1120

26. DOREULEE N, SERGEEVA OA, YANOVSKY Y, CHEPKOVA AN, SELBACH O, GODECKE A, SCHRADER J, HAAS HL 2003 Cortico-striatal synaptic plasticity in endothelial nitric oxide synthase deficient mice. Brain Res 964: 159-163

27. DRESNER A, LAURENT D, MARCUCCI M, GRIFFIN ME, DUFOUR S, CLINE GW, SLEZAK LA, ANDERSEN DK, HUNDAL RS, ROTHMAN DL, PETERSEN KF, SHULMAN GI 1999 Effects of free fatty acids on glucose transport and IRS-1-associated phosphatidylinositol 3-kinase activity. J Clin Invest 103: 253-259

28. DUELLI R, KUSCHINSKY W 2001 Brain glucose transporters; relationship to local energy demand. News Physiol Sci 16: 71-76

29. FOLLI F, BONFANTI L, RENARD E, KAHN CR, MERIGHI A 1994 Insulin receptor substrate-1 (IRS-1) distribution in the rat central nervous system. J Neurosci 14: 6412-6422

30. FOULSTONE EJ, TAVARE JM, GUNN-MOORE FJ 1999 Sustained phosphorylation and activation of protein kinase B correlates with brain-derived neurotrophic factor and insulin stimulated survival of cerebellar granule cells. Neurosci Lett 264: 125-128

31. FROLICH L, BLUM-DEGEN D, BERNSTEIN HG, ENGELSBERGER S, HUMRICH J, LAUFER S, MUSCHNER D, THALHEIMER A, TURK A, HOYER S, ZOCHLING R, BOISSL KW, JELLINGER K, RIEDERER P 1998 Brain insulin and insulin receptors in aging and sporadic Alzheimer’s disease. J Neural Transm 105: 423-438

32. FULOP T, LARBI A, DOUZIECH N 2003 Insulin receptor and ageing. Pathol Biol 51: 574-580

33. FULOP Jr T, NAGY J, WORUM I, FORIS G, MUDRI K, UDVARDY M 1987 Glucose intolerance and insulin resistance with aging. Studies on insulin receptors and post-receptor events. Arch Gerontol Geriatr 6: 107-115

34. GASPARINI L, GOURAS GK, WANG R, GROSS RS, BEAL MF, GREENGARD P, XU H 2001 Stimulation of beta-amyloid precursor protein trafficking by insulin reduces intraneuronal beta-amyloid and requires mitogen-activated protein kinase signalling. J Neurosci 21: 2561-2570

35. GEROZISSIS K 2003 Brain insulin: regulation, mechanisms of action and functions. Cell Mol Neurobiol 23: 1-25

36. GEROZISSIS K 2004 Brain insulin and feeding: a bi-directional communication. Eur J Pharmacol 490: 59-70

37. GEROZISSIS K, OROSCO M, ROUCH C, NICOLAIDIS S 1998 Brain insulin response to feeding in the rat is both macronutrient and area specific. Phyiol Behav 65: 271-275

38. GEROZISSIS K, ROUCH C, LEMIERRE S, NICOLAIDIS S, OROSCO M 2001 A potential role of central insulin in learning and memory related to feeding. Cell Mol Neurobiol 21: 389-401

39. GIORGINO F, ALMAHFOUZ A, GOODYEAR LJ, SMITH RJ 1993 Glucocorticoid regulation of insulin receptor and substrate IRS-1 tyrosine phosphorylation in rat skeletal muscle in vivo. J Clin Invest 91: 2020-2030

40. GISPEN WH, BIESSELS GJ 2000 Cognition and synaptic plasticity in diabetes mellitus. Trends Neurosci 23: 542-549

41. GSELL W, CONRAD R, HICKETHIER M, SOFIC E, FROLICH L, WICHART I, JELLINGER K, MOLL G, RANSMAYR G, BECKMANN H, RIEDERER P 1995 Decreased catalase activity but unchanged superoxide dismutase activity in brains of patients with dementia of Alzheimer type. J Neurochem 64: 1216-1223

42. HEINDREICH KA, ZAHNISER NR, BERHANU P, BRANDENBURG D, OLEFSKY JM 1983 Structural differences between insulin receptors in the brain and peripheral target tissues. J Biol Chem 258: 8527-8530

43. HENNEBERG N, HOYER S 1995 Desensitization of the neuronal insulin receptor: a new approach in the etiopathogenesis of late-onset sporadic dementia of the Alzheimer type (SDAT)? Arch Gerontol Geriatr 21: 63-74

44. HONG MF, LEE VMY 1997 Insulin and insulin-like growth factor-1 regulate tau phosphorylation in cultured human neurones. J Biol Chem 272: 19547-19553

45. HOYER S 1998 Is sporadic Alzheimer’s disease the brain type of non-insulin dependent diabetes mellitus? A challenging hypothesis, J Neural Transm 105: 415-422

46. HOYER S 2002 The brain insulin signal transduction system and sporadic (type II) Alzheimer’s disease: an update. J Neural Transm 109: 341-360

47. HOYER S 2004 Glucose metabolism and insulin receptor signal transduction in Alzheimer disease. Eur J Pharmacol 490: 115-125

48. HOYER S, PREM L, SORBI S, AMADUCCI L 1993 Stimulation of glycolitic key enzymes in cerebral cortex by insulin. Neuro Report 4: 991-993

49. JETTON T, LIANG Y, PETTEPHER CC 1994 Analysis of upstream glucokinase promoter activity in transgenic mice and identification of glucokinase in rare neuroendocrine cells in the brain and gut. J Biol Chem 269: 3641-3654

50. JOHNSTONE AM, PIROLA L, VAN OBBERGHEN E 2003 Molecular mechanisms of insulin receptor substrate protein-mediated modulation of insulin signalling. FEBS Letters 546: 32-36

51. JOOST HG 1995 Structural and functional heterogeneity of insulin receptors. Cell Signal 7: 85-91

52. KIMURA T, TAKAMATSU J, IKEDA K, KONDO A, MIAYAKAWA T, HORIUCHI S 1996 Accumulation of advanced glycation end products of the Maillard reaction with age in human hippocampal neurons. Neurosci Lett 208: 53-56

53. KUMAR JS, MENON VP 1993 Effect of diabetes on levels of lipid peroxides and glycolipids in rat brain. Metabolism 42: 1435-1439

54. LACKOVIĆ Z, ŠALKOVIĆ M 1990 Streptozocin and alloxan produce alterations in rat rain monoamines independently of pancreatic beta cells destruction. Life Sci 46: 49-54

55. LACKOVIĆ Z, ŠALKOVIĆ M, KUCI Z, RELJA M 1990 Effect of long lasting diabetes mellitus on rat and human brain monoamines. J Neurochem 54: 143-147

56. LEBOVITZ HE, BANERJI MA 2004 Treatment of insulin resistance in diabetes mellitus. Eur J Pharmacol 490: 135-146

57. LeLOUP C, ARLUISON M, KASSIS N, LEPETIT N, CARTIER N, FERRE P, PENICAUD L 1996 Discrete brain areas express the insulin-responsive glucose transporter GLUT4. Mol Brain Res 38: 45-53

58. LIN X, TAGUCHI A, PARK S, KUSHNER JA, LI F, LI Y, WHITE MF 2004 Dysregulation of insulin receptor substrate 2 in β cells and brain causes obesity and diabetes. J Clin Invest 114: 908-916

59. MAN HY, LIN JW, JU WH, AHMADAIN G, LIU L, BECKER LE, SHENG M, WANG YT 2000 Regulation of AMPA receptor-mediated synaptic transmission by clathrin-dependent receptor internalisation. Neuron 25: 649-662

60. MARKS JL, PORTE Jr D, BASKIN DG 1991 Localization of insulin and type I IGF receptors in rat brain by in vitro autoradiography and in situ hybridisation. Adv Exp Med Biol 293: 459-470

61. MATTSON MP, DUAN W, MASWOOD N 2002 How does the brain control lifespan? Age Res Rev 1: 155-165

62. MATTSON MP, MAUDSLEY S, MARTIN B 2004 BDNF and 5-HT: a dynamic duo in age-related neuronal plasticity and neurodegenerative disorders. TINS 27: 589-594

63. McCALL AL 1998 New findings and treatment strategies for hypoglycaemia and hypoglycaemia unawareness. Curr Opin Endocrinol Diabetes 5: 136-143

64. McCALL AL 2004 Cerebral glucose metabolism in diabetes mellitus. Eur J Pharmacol 490: 147-158

65. MESSARI SE, LeLOUP C, QUIGNON M, BRISORGUEIL M-J, PENICAUD L, ARLUISON M 1998 Immunocytochemical localization of the insulin-responsive glucose transporter 4 (Glut4) in the rat central nervous system. J Comp Neurol 399: 492-512

66. MEZIANE H, DODART JC, MATHIS C, LITTLE S, CLEMENS J, PAUL SM, UNGERER A 1998 Memory-enhancing effects of secreted forms of the beta-amyloid precursor protein in normal and amnestic mice. Proc Natl Acad Sci USA 95: 12683-12688

67. MOLERO JC, PEREZ C, MARTINEZ C, VILLAR M, ANDRES A, FERMIN Y, CARRASCOSA JM 2002 Activation of MAP kinase by insulin and vanadate in adipocytes from young and old rats. Mol Cell Endocrinol 189: 77-84

68. MONTAGNANI M, RAVICHANDRAN LV, CHEN H, ESPOSITO DL, QUON MJ 2002 Insulin receptor substrate-1 and phosphoinositide-dependent kinase-1 are required for insulin-stimulated production of nitric oxide in endothelial cells. Mol Endocrinol 16: 1931-1942

69. MOORADIAN AD 1997 Central nervous system complications of diabetes mellitus – a perspective from the blood-brain barrier. Brain Res Rev 23: 210-218

70. MUNCH G, SCHINZEL R, LOSKE C, WONG A, DURANY N, LI JJ, VLASSARA H, SMITH MA, PERRY G, RIEDERER P 1998 Alzheimer’s disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts. J Neural Transm 105: 439-461

71. NIBLOCK MM, BRUNSO-BECHTOLD JK, LYNCH CD, INGRAM RL, McSHANE T, SONNTAG WE 1998 Distribution and levels of insulin-like growth factor I mRNA across the life span in the Brown Norway x Fischer 344 rat brain. Brain Res 31: 79-86

72. NISWENDER KD, BASKIN DG, SCHWARTZ MW 2004 Insulin and its evolving partnership with leptin in the hypothalamic control of energy homeostasis. Trends Endocrinol Metab 15: 362-369

73. OBICI S, FENG Z, KARKANIAS G, BASKIN DG, ROSSETTI L 2002 Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats. Nat Neurosci 5: 566-572

74. OROSCO M, GEROZISSIS K 2001 Macronutrient-induced cascade of events leading to parallel changes in hypothalamic serotonin and insulin. Neurosci Biobehav Rev 25: 167-174

75. PARK CR 2001 Cognitive effects of insulin in the central nervous system. Neurosci Biobehav Rev 25: 311-323

76. PIROLI GG, GRILLO CA, HOSKIN EK, ZNAMENSKY V, KATZ EB, MILNER TA, McEWAN BS, CHARRON MJ, REAGAN LP 2002 Peripheral glucose administration stimulates the translocation of GLUT8 glucose transporter to the endoplasmic reticulum in the rat hippocampus. J Comp Neurol 452: 103-114

77. REAGAN LP 2002 Glucose, stress and hippocampal neuronal vulnerability. Int Rev Neurobiol 51: 289-324

78. RINAUDO MT, CURTO M, BRUNO R, MARINO C, ROSSETTI Y, MOSTERT M 1987 Evidence of an insulin generated pyruvate dehydrogenase stimulating factor in rat brain plasma membranes. Ital J Biochem 19: 909-913

79. RYAN CM, GECKLE M 2000 Why is learning and memory dysfunction in Type 2 diabetes limited to older adults? Diabetes Metab Res Rev 16: 308-315

80. RYAN CM, WILLIAMS TM 1993 Effects of insulin-dependent diabetes on learning and memory efficiency in adults. J Clin Exp Neuropsychol 15: 685-700

81. RYLE C, LEOW CK, DONAGHY M 1997 Nonenzymatic glycation of peripheral and central nervous system proteins in experimental diabetes mellitus. Muscle Nerve 20: 577-584

82. SANTOS MS, PEREIRA EM, CARVAHO AP 1999 Stimulation of immunoreactive insulin release by glucose in rat brain synaptosomes. Neurochem Res 24: 33-36

83. SCHULINGKAMP RJ, PAGANO TC, HUNG D, RAFFA RB 2000 Insulin receptors and insulin action in the brain: review and clinical implications. Neurosci Biobehav Rev 24: 855-872

84. SESTI G 2000 Insulin receptor variant forms and type 2 diabetes mellitus. Oharmacogenomics 1: 49-61

85. SKEBERDIES VA, LAN J, ZHENG X, ZUKIN RS, BENNETT MV 2001 Insulin promotes rapid delivery of N-methyl-D-aspartate receptors to the cell surface by exocytosis. Proc Natl Acad Sci USA 98: 3561-3566

86. SKYLER JS 2004 Diabetes mellitus: pathogenesis and treatment strategies. J Med Chem 47: 4113-4117

87. SMITH MA, SAYRE LM, MONNIER VM, PERRY G 1995 Radical AGEing in Alzheimer’s disease. Trends Neurosci 18: 172-176

88. SOMMERFIELD AJ, DEARY IJ, McAULAY V, FRIER BM 2003 Short-term, delayed, and working memory are impaired during hypoglycaemia in individuals with type 1 diabetes. Diabetes Care 26: 390-396

89. SPANSWICK D, SMITH MA, GROPPI VE, LOGAN SD, ASHFORD ML 1997 Leptin inhibits hypothalamic neurons by activation of ATP-sensitive potassium channels. Nature 390: 521-525

90. SPANSWICK D, SMITH MA, MIRSHAMSI S, ROUTH VH, ASHFORD ML 2000 Insulin activates ATP-sensitive K+ channels in hypothalamic neurons of lean, but not obese rats. Nat Neurosci 3: 757-758

91. SYTZE VAN DAM P, ALEMAN A 2004 Insulin-like growth factor-I, cognition and brain aging. Eur J Pharmacol 490: 87-95

92. SZKUDELSKI T 2001 The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physiol Res 50: 537-546

93. ŠALKOVIĆ M, LACKOVIĆ Z 1992 Brain D1 dopamine receptors in alloxan diabetes. Diabetes 41: 1119-1121

94. ŠALKOVIĆ M, SABOLIĆ I, LACKOVIĆ Z 1995 Striatal D1 and D2 receptors after intracerebroventricular applications of alloxan and streptozotocin in rat. J Neural Transm 100: 127-145

95. ŠALKOVIĆ-PETRIŠIĆ M, AUGOOD SJ, BICKNELL RJ 1997 Monoamine transporter gene expression in the central nervous system in diabetes mellitus. J Neurochem 68: 2435-2441

96. ŠALKOVIĆ-PETRIŠIĆ M, LACKOVIĆ Z 2003 Intracerebroventricular administration of betacytotoxics alters expression of brain monoamine transporter genes. J Neural Transm 110: 15-29

97. TREHERNE JM, ASHFORD MLJ 1992 Extracellular cations modulate the ATP sensitivity of ATP-K+ channels in rat ventromedial hypothalamic neurones. Proc R Soc Lond B Biol Sci 247: 121-124

98. VANNUCCI SJ, KOEHLER-STEC EM, LI K, REYNOLDS TH, CLARK R, SIMPSON IA 1998 GLUT4 glucose transporter expression in rodent brain: effects of diabetes. Brain Res 797: 1-11

99. WAN Q, XIONG ZG, MAN HY, ACKERLEY CA, BRAUNTON J, LU WY, BECKER LE, MacDONALD JF, WANG YT 1997 Recruitment of functional GABA(A) receptors to postsynaptic domains by insulin. Nature 388: 686-690

100. WATSON GS, CRAFT S 2004 Modulation of memory by insulin and glucose: neuropsychological observations in Alzheimer’s disease. Eur J Pharmacol 490: 97-113

101. WEI LT, MATSUMOTO H, RHOADS DE 1990 Release of immunoreactive insulin from rat brain synaptosomes under depolarising conditions. J Neutrochem 54: 1661-1665

102. WILCOX BJ, MATSUMOTO AM, DORSA DM, BASKIN DG 1989 Reduction of insulin binding in the arcuate nucleus of the rat hypothalamus after 6-hydroxy dopamine treatment. Brain Res 500: 149-155

103. XIE L, HELMERHORST E, TADDEL K, PLEWRIGHT B, VAN BRONSWIJK W, MARTINS R 2002 Alzheimer’s β-amyloid peptides compete for insulin binding to the insulin receptor. J Neurosci 22: 1-5

104. YOUNG WS 1986 Periventricular hypothalamic cells in the rat brain contain insulin mRNA. Neuropeptides 8: 93-97

105. ZHAO W-Q, CHEN H, QUON MJ, ALKON DL 2004 Insulin and the insulin receptor in experimental models of learning and memory. Eur J Pharmacol 490: 71-81

106. ZHAO WQ, CHEN H, XU H, MOORE E, MEIRI N, QUON MJ, ALKON DC 1999 Brain insulin receptors and spatial memory. J Biol Chem 274: 34893-34902

Fig. 1. Intracellular insulin receptor signaling pathway. Insulin binding to insulin receptor (IR) activates receptor tyrosine kinase (TK) activity, and among other molecules recruits insulin receptor substrate (IRS) protein. IRS becomes phosphorylated at tyrosine residues (Tyr-P) and capable of recruiting various signaling molecules including enzymes like phosphoinositide-3 kinase (PI3-K), or different adapters, among which one transduces the signal to the serine/threonine kinase MAPK (mitogen activated protein kinase) pathway via monomeric G protein Ras. Ras/MAPK pathway is related to insulin-induced effects on growth and proliferation. Downstream to the PI3-K pathway is protein kinase B (PKB/Act) that is related to insulin-induced metabolic events, like translocation of glucose transporter GLUT4 to the cell membrane to facilitate cellular glucose uptake. PKB/Act may also phosphorylate and thereby inactivate glycogen synthase kinase-3 (GSK-3), leading thus to activation of glycogen synthase and increased glycogen synthesis. In the brain, GSK-3 α is related to regulation of the metabolism of amyloid precursor protein (APP) and amyloid-β proteins (Aβs), while GSK-3 β balances the phosphorylation of tau protein. Amonth others, IR signal transduction may be reduced by decreasing the binding of insulin to IR via Aβs, by reducing the TK activity of IR via cortisol and catecholamine, or by inducing the serine/threonine phosphorylation (S/T-P) of IRS via free fatty acids (FFA), cytokines and inflammatory products like tumor necrosis factor α (TNF-α), or advanced glycation end products (AGEs). Simplified scheme modified according to Hoyer S, 2004 (47).

IR

Aβs

reduced insulin binding

cortisol, catecholamines... insulin

reduced TK activity TK activity

FFA, TNF-α, AGEs... insulin

reduced initiation of

insulin signal transduction insulin signal transduction

↓ ↓

PKB/Act

MAPK

↓ ↓

regulation of metabolic effects regulation of mitogenic effects

↓ ↓

GLUT4 translocation GSK-3 → glycogen synthesis

glucose uptake α β

APP/Aβ tau protein

metabolism phosphorylation

23

4