Skip to main content
Erschienen in: CNS Drugs 1/2021

Open Access 01.01.2021 | Review Article

Intranasal Insulin for Alzheimer’s Disease

verfasst von: Manfred Hallschmid

Erschienen in: CNS Drugs | Ausgabe 1/2021

Abstract

Brain insulin signaling contributes to memory function and might be a viable target in the prevention and treatment of memory impairments including Alzheimer’s disease. This short narrative review explores the potential of central nervous system (CNS) insulin administration via the intranasal pathway to improve memory performance in health and disease, with a focus on the most recent results. Proof-of-concept studies and (pilot) clinical trials in individuals with mild cognitive impairment or Alzheimer’s disease indicate that acute and prolonged intranasal insulin administration enhances memory performance, and suggest that brain insulin resistance is a pathophysiological factor in Alzheimer’s disease with or without concomitant metabolic dysfunction. Intranasally administered insulin is assumed to trigger improvements in synaptic plasticity and regional glucose uptake as well as alleviations of Alzheimer’s disease neuropathology; additional contributions of changes in hypothalamus-pituitary-adrenocortical axis activity and sleep-related mechanisms are discussed. While intranasal insulin delivery has been conclusively demonstrated to be effective and safe, the recent outcomes of large-scale clinical studies underline the need for further investigations, which might also yield new insights into sex differences in the response to intranasal insulin and contribute to the optimization of delivery devices to grasp the full potential of intranasal insulin for Alzheimer’s disease.
Key Points
Insulin acting in the brain is a relevant neuromodulator that contributes to cognitive function via mechanisms that are still to be fully explored.
CNS insulin delivery via the nose improves memory performance in healthy individuals but also patients with Alzheimer’s disease who are assumed to be less sensitive to the brain insulin signal.
Mixed results of larger scale clinical trials call for further research on the preconditions and mechanisms of the memory effect of intranasal insulin as well as for the optimization of delivery approaches.

1 Introduction: Insulin in the Brain

Almost 50 million people worldwide lived with dementia in 2015 according to estimates based on over 200 studies, with expected increases to 75 million by 2030 and 132 million by 2050 [1]. Recent assumptions that the incidence and prevalence of dementia may remain stable or even decline offer a glimmer of hope [2], but the high total number of afflicted people and the severity of dementia-associated impairments in the daily life of patients and their families underline the magnitude of the challenge, which also poses a considerable financial burden on global health systems (estimated to have amounted to US$818 billion in 2015 [3]). Alzheimer’s disease (AD) is the major cause of dementia, but there are still no causal treatments for this debilitating disease (cholinesterase inhibitors and memantine are used for symptomatic relief at early stages). The progressive loss of cognitive and functional abilities in AD is associated with the accumulation of aberrant, misfolded, and aggregated oligomeric amyloid beta (Aβ) peptides and hyperphosphorylated tau, but the etiology of AD remains poorly understood [4].
Recent research indicates that insulin action in the brain might be a key factor in its pathogenesis as well as a target of interventions to prevent and treat this devastating ailment. Although compared with other fields of neuroscience, central nervous system (CNS) insulin signaling is a relatively young topic, the last 30 years have greatly advanced our understanding of the mechanisms and functions of insulin’s role in the brain and for the brain. The presence of insulin receptors in rat brains was first demonstrated by Havrankova et al. in 1978 [5]; not much later, insulin receptors were also detected in the human brain [6]. Insulin concentrations in cerebrospinal fluid (CSF) and plasma are correlated, but insulin concentrations are much lower in CSF [7]. It is assumed that the bulk of brain insulin has its source in peripheral insulin crossing the blood–brain barrier (BBB) by a saturable receptor-mediated transport mechanism [8]. Some indicators of local insulin production in the cerebral cortex have been found in animals [9, 10] and there are reports of insulin transcription in human brain tissue [11], but the assumption that insulin is released in decisive amounts within the brain still lacks coherent evidence [12].
As the brain does not essentially rely on insulin to regulate its energy supply [13, 14], the function of CNS insulin receptors first remained elusive; today, it is known that the neuropeptide affects a broad range of functions including peripheral energy and glucose homeostasis [15, 16], growth [17] and, notably, neuronal plasticity [18]. Stephen Woods and his team were the first to perform seminal studies showing that insulin, which circulates within the bloodstream in proportion to body fat stores, acts in the brain to reduce food intake [19]. This finding was repeatedly replicated [20, 21] and insulin is now regarded as an important adiposity signal that provides feedback from the body periphery to CNS circuitries that control energy intake [22]. Unsurprisingly for a signal of such obvious relevance for metabolism, research activities first focused on this aspect of brain insulin signaling. In the meantime, however, it has become clear that insulin’s CNS function pertains to cognitive processes, suggesting that brain insulin action also constitutes a neuroendocrine link between metabolism and cognition and might be a suitable target in the treatment both of metabolic and cognitive disorders [23].
In patients with obesity and/or type 2 diabetes mellitus, who experience variable degrees of peripheral insulin resistance (i.e., a decrease or lack of effective insulin signaling), the brain is likewise less sensitive to insulin, which supports the notion that relative brain insulin resistance or a lack of insulin in the CNS is a key factor in dysfunctional metabolic control [24]. As will be discussed in this review, it is likely that impaired brain insulin sensitivity moreover contributes to memory impairments including AD; the potential of insulin in the prevention and therapy of AD is illustrated by evidence that insulin delivery to the CNS improves cognitive function in healthy individuals and, moreover, patients with cognitive impairments or AD.
In this regard, the intranasal (IN) approach to increase the availability of insulin in the CNS is of particular interest because it has been put to successful use in most of the more recent investigations that this narrative review focuses on. The search strategy pivoted around PubMed results in English language with the terms “intranasal”, “brain”, “insulin”, “cognition”, “memory”, and “AD” retrieved until September 2020 and the reference lists of the respective publications, with a focus on work published since 2017. Note that the relevance of brain insulin signaling (and respective beneficial effects of IN insulin) pertains to neurological and psychiatric conditions such as vascular cognitive impairment [25], Parkinson’s disease [26, 27], traumatic brain injury [28], Huntington’s disease [29, 30], depression [31], and addictive behavior [32], which are outside the scope of the present paper.

2 Intranasal Insulin Administration to the CNS

The BBB, an endothelial layer of cells and tight junctions, separates the vessels perfusing the CNS from its environment, thereby shielding the brain against toxins and infections while allowing gas and ion exchange. It regulates the entry and exit of molecules into and out of the brain and, moreover, serves as a communication interface that is endowed with receptors and transporters for hormonal signals including insulin [33]. The BBB is passively permeable to molecules of approximately < 400 Da in size and with fewer than eight to ten hydrogen bonds; in addition, it enables the active, often saturable transport of bigger molecules [34]. With a molecular weight of 5808 Da, insulin is too large to cross the BBB passively and therefore depends on active transport mechanisms to enter the brain [35]. Insulin concentrations in the CSF increase after intravenous infusion in men [7], but the efficiency of blood-to-CSF transport is limited by conditions such as increases in body weight [36]. In experiments in animals, insulin is routinely administered to the CNS by, for example, direct intracerebroventricular [19] or hypothalamic infusion [37]. Systemic insulin administration to investigate CNS effects of the hormone has long been the method of choice in experiments in humans [e.g., 3841], but this approach comes with some important drawbacks. The decrease in blood glucose concentrations induced by systemic insulin infusion below certain thresholds inevitably impairs cognition [42] and, moreover, activates endocrine (stress) axes that can affect brain function [43]. Insulin-induced hypoglycemia can be prevented by simultaneous glucose infusion that, however, may itself exert a biasing impact on (cognitive) brain functions. Euglycemic–hyperinsulinemic clamps are moreover time and labor intensive and do not permit the differentiation between direct brain effects and effects mediated via peripheral pathways.
The IN route of insulin administration overcomes these methodological impediments. The first US patent on IN administration to bypass the BBB and target the brain was filed by William H. Frey II in 1989 [44], followed by a second patent on IN insulin to treat AD and Parkinson’s disease [45] and proof-of-concept demonstrations in animals [e.g., 4648]. Experiments in Sprague-Dawley rats relying on gamma counting and high-resolution phosphor imaging of tissue sections suggest that after IN administration insulin-like growth factor-1 quickly activates multiple sites within the brain and spinal cord [48]. It has likewise been shown that intranasally administered neuropeptides reach brain structures that are relevant for cognitive function [49]. Considering that the intra-neuronal transport of neuropeptides from the nasal cavity to the olfactory bulb takes several hours [50], it is assumed that intranasally administered peptides primarily travel along extra-neuronal routes, i.e., through intercellular clefts of the olfactory epithelium situated on the superior turbinate and opposite the nasal septum [51, 52]; additional transport along trigeminal nerve branches to brainstem regions has been demonstrated [48, 53]. Studies in humans indicate that intranasally administered insulin can bypass the BBB and reach the CNS within 1 h after administration [54]. Systemic absorption after IN insulin administration is negligible at moderate doses [54] and seems to trigger side effects such as increases in cortisol and growth hormone only when cumulative doses exceed around 200 IU [55]. Therefore, it is also unlikely that BBB transport after absorption into the bloodstream is a major contributor to brain uptake and functional impact of IN insulin. The IN pathway moreover extends insulin’s half-life by minimizing hepatic first-pass elimination [56]. It may also be possible to target specific areas of the brain, especially those near the administration site [57]. Because of its easy methodology and favorable safety profile [58] (see Sect. 4.3), the IN method of insulin administration to the brain offers a non-invasive, easy-to-use approach that has now been widely applied in experimental settings of preclinical but also clinical research (for in-depth reviews on the IN administration of insulin and other peptides see, for example, [52, 59, 60]). Indeed, it seems that besides oxytocin [61], insulin is the hormone with the most promising evidence of functional effectivity after IN delivery.

3 Intranasal Insulin and Memory

3.1 Intranasal Insulin-Induced Memory Improvements in Humans Without Cognitive Impairments

Beneficial cognitive effects of CNS insulin administration via the IN route have been demonstrated in a series of studies in healthy humans [6266]. Eight weeks of IN insulin administration (4 × 40 IU/day vs diluent) to young men and women [63] improved the delayed recall of a list of 30 words encoded 1 week earlier, a measure of hippocampus-dependent declarative memory. In contrast, immediate word recall 3 min after encoding and non-declarative memory functions remained unaffected [63]. The improvement in declarative memory could even be intensified by administering the rapid-acting insulin analog insulin aspart [64]; insulin aspart has a reduced tendency to self-associate but shares the receptor binding profile of regular insulin [67]. In acute paradigms, preliminary evidence for sex-dependent insulin effects on memory function was obtained because women, but not men, improved performance on declarative and working memory tasks after receiving 160 IU of insulin compared to placebo (diluent) [62]. In subsequent experiments, IN insulin in comparison to placebo (diluent) administration before nocturnal sleep tended to improve the acquisition of word-pairs on the subsequent evening in women, with opposite effects in men [65]. In an acute experiment that only included healthy male participants [66], IN insulin compared with placebo (diluent) enhanced the odor-cued recall of spatial memory, while an impairing effect of IN insulin (vs diluent) on olfactory sensitivity was observed in young healthy women but not men [68]. Although experimental indicators of a preponderance of metabolic effects of IN insulin in men rather than women [62, 69] buttress the assumption of a sex difference in the functional response to IN insulin, studies in larger samples of male and female participants with cognitive impairments have only yielded sporadic evidence [70]. Animal experiments suggest that insulin’s CNS impact is modified by estrogen signaling [71], but related studies in humans do not support the assumption that estrogen may boost the sensitivity to the memory effect of insulin [72]. Systematic investigations into sex differences in brain insulin effects, underlying mechanisms, or possible implications for the prevention and treatment of AD are currently lacking. This is somewhat surprising considering that the age-specific prevalence of AD is higher in women [1]—in the USA, two thirds of individuals with AD are women [73]—and that the risk of AD in carriers of the ɛ4 variant of the apolipoprotein E gene (apoE ε4), a risk factor for sporadic AD [74] whose frequency does not differ between men and women, is four times higher in women than men aged between 65 and 75 years [75] (for further AD-related sex differences, see [76]). Moreover, women have a greater risk of developing systemic insulin resistance [77].
In accordance with the results in normal-weight individuals [63], obese men who were administered IN insulin compared with placebo (diluent) for 8 weeks according to the same paradigm likewise displayed improvements in declarative memory [78]. Electrophysiological evidence for the impact of IN inulin on brain function was obtained in experiments relying on magnetoencephalography [79] or measuring scalp-recorded event-related [80] and direct current brain potentials [81]. In the latter study, a largely comparable negative shift in direct current potentials was observed within minutes after IN and intravenous bolus administration of insulin that was assumed to reflect changes in extracellular ionic concentrations due to glial activity [82]. These findings suggest a rapid effect of insulin on brain activity in humans and, moreover, that IN insulin delivery is able to mimic the brain impact of intravenously administered and, presumably, endogenous insulin.
Brain insulin may not only modulate cognitive but also emotional functions. The 8-week paradigm of IN insulin administration induced an improvement in self-rated rated mood in normal-weight [63] as well as obese participants [78]. In mice, IN insulin enhances object memory and induces anxiolytic behavioral effects [83], whereas lentivirus-mediated downregulation of hypothalamic insulin receptor expression in rats elicits depressive and anxiety-like behaviors [84]. Impaired glucose tolerance due to diet-induced obesity likewise abrogates the memory-improving and anxiolytic impact of insulin [83]. Taken together, these findings suggest that impaired CNS insulin signaling might contribute to the association between metabolic disorders such as obesity and diabetes and cognitive impairments as well as dysphoria [85].

3.2 Mechanisms of the Enhancing Effect of Insulin on Cognition

In-vitro studies and experiments in animals, but also humans, have enabled insights into a number of possible mechanisms behind the improving cognitive impact of (intranasal) insulin. Insulin activates its receptor by binding to extracellular α-subunits and triggering the dimerization of intracellular β-subunits, thereby inducing receptor autophosphorylation. The two most relevant signaling pathways activated by insulin are the insulin-insulin receptor substrate (IRS)-Akt pathway (recruiting IRS1 or IRS2) and the mitogen-activated protein kinase pathway. The insulin-IRS-Akt pathway mediates the glucoregulatory action of insulin in muscle, adipose, and liver tissue and further downstream processes in all cell types, while the mitogen-activated protein kinase pathway regulates transcription factors such as CREB and c-Fos (see [86] for details). Brain insulin receptors are expressed in high densities in the olfactory bulb, hypothalamus, and cerebellum and in regions that enable memory formation such as the hippocampus and connected limbic brain structures [87, 88]. Neuronal insulin receptors are expressed both pre- and post-synaptically and neuronal insulin signaling relies on the insulin-IRS-Akt as well as the mitogen-activated protein kinase pathway [89]. Insulin has been demonstrated to contribute to a broad range of neuronal signaling mechanisms, including but not limited to catecholamine release and uptake, ion channel trafficking, and the regulation of receptors for neurotransmitters, i.e., γ-aminobutyric acid, N-methyl-d-aspartate (NMDA), and α-amino‑3‑hydroxy‑5‑methyl‑4‑isoxazole propionic acid (AMPA) [90]. It also contributes to activity-dependent processes of synaptic plasticity, i.e., long-term potentiation and long-term depression [91]. More details on insulin signaling pathways in the brain can be found elsewhere [86, 92].
The establishment of memory traces in the hippocampus depends both on long-term depression and long-term potentiation [93]. Supporting the assumption that insulin improves memory by modulating these plastic processes, insulin was found to induce glutamatergic AMPA receptor internalization leading to long-term depression [94], and moreover to phosphorylate AMPA receptors leading to overexpression of PKMζ [95]. The downregulation of hippocampal insulin receptor function impairs long-term potentiation and spatial memory [96]. Insulin also potentiates NMDA receptor activity via delivery of NMDA receptors to the cell surface [97] and NMDA receptor phosphorylation [98], processes that may induce long-lasting meta-plastic changes. In addition to effects on synaptic plasticity [99], there is some evidence that insulin benefits regional brain glucose uptake by activating the neuronal glucose transporter type 4 and enhances glycogen uptake in regions such as the basal forebrain, hippocampus, amygdala, and cortex [100, 101] (for reviews see [102, 103]), in particular under conditions of high cognitive demand [104, 105]. In experiments in healthy humans relying on 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) measurements during intravenous insulin infusion while endogenous insulin production was suppressed by somatostatin, whole-brain glucose utilization was found to be stimulated by insulin [106], whereas experiments using 1H-magnetic resonance spectroscopy yielded no effect of insulin infusion on brain glucose [14]. Neuronal glucose uptake is mostly regulated via glucose transporter type 3, which is generally assumed not to depend on insulin [107, 108]; however, recent in vitro experiments indicate that 4 days of insulin receptor activation up-regulate glucose transporter type 3 membrane expression in hippocampal neurons [109]. Insulin may also support brain energy supply via effects on astrocytes [110] and other glia cells including oligodentrocytes (for a review, see [86]). Moreover, IN insulin has been found to improve regional vasoreactivity alongside visuospatial memory function [111]. On a systems level, IN insulin administration was observed to increase the concentrations of high-energy phosphate compounds, i.e., adenosine triphosphate and phosphocreatine, in the motor cortex as assessed by 31P-magnetic resonance spectroscopy, an effect that was positively related to the subsequent insulin-induced suppression in food intake [112]. Intranasal insulin can moreover trigger enhancements in functional connectivity between prefrontal regions and the hippocampal formation that benefit memory formation [113].
Recent evidence points to sleep- and stress-related mechanisms as further potential mediators of improving cognitive effects of IN insulin. The impact on hypothalamic-pituitary-adrenal (HPA) axis activity of 160 IU of IN insulin administered before sleep was assessed in a study that included young and elderly healthy men and women [114]. In comparison with the young participants, the elderly subjects showed signs of increased cortisol concentrations during early sleep, when HPA axis secretion typically reaches its circadian nadir. Intranasal insulin compared to placebo (diluent) dampened cortisol levels in the first night-half in the elderly, but not in the young participants in a sex-independent manner. Reductions in HPA axis activity upon IN insulin vs placebo (diluent) were also observed in awake young men exposed to a psychosocial stress test [115], as well as under resting conditions after 8 weeks of daily administration [63, 78]. Attenuating effects of brain insulin on HPA axis activity have been assumed to be mediated by enhanced corticosteroid feedback processing in the hippocampus [116]. In healthy elderly humans, cortisol was found to acutely reduce FDG-PET-assessed glucose utilization in the hippocampus [117], and increases in HPA axis activity are associated with an increased risk for metabolic and cognitive impairments including AD [118120]. It is of particular interest that insulin may co-regulate HPA axis activity in association with circadian and sleep-related mechanisms because the consolidation of memory content strongly benefits from sleep: neuronal ensembles that encode information during wakefulness are reactivated during subsequent sleep, thereby strengthening respective memory representations [121]. Accordingly, impaired sleep may predispose to or accelerate cognitive impairments including AD [122].
While IN insulin delivery before sleep does not affect polysomnographically assessed sleep architecture or subjective sleep quality [114], electroencephalogram delta power during the second 90 min of non-rapid-eye-movement (NREM) sleep was found to be enhanced by insulin compared with a placebo (diluent) in young healthy men [65]. Nocturnal insulin secretion is entrained to NREM sleep phases [123]. In rats, peripheral and intracerebroventricular administration of insulin increases the time spent in NREM sleep [124], whereas the hormone seems to have the opposite effect on REM sleep [124]. The enhancement of electroencephalogram delta power by IN insulin coincided with a pronounced, but statistically unrelated insulin-induced increase in growth hormone levels that was independent of the participant’s sex [65]. Participants also encoded declarative and procedural memory contents (word-pairs and, respectively, finger tapping sequences) before IN insulin administration in the evening. Insulin compared to placebo did not directly alter the retrieval of memory contents acquired before sleep, but generally impaired the acquisition of interfering memory contents on the next day (although, as described above, the female participants displayed a trend to improved learning of new word-pairs in the insulin vs placebo condition). These results suggest that sleep-associated memory consolidation may not be a primary mediator of insulin’s acute memory-improving effect in healthy subjects. Still, that IN insulin reduces the interfering influence of encoding new information on the subsequent day may be taken as an indicator that processes of active forgetting during sleep [125] are inhibited by insulin. Insulin-induced improvements in sleep electroencephalogram delta power may support the clearance of metabolic waste that is linked to slow-wave activity [126]; notably, slow-wave activity during NREM sleep has also been found to be negatively correlated with tau pathology and Aβ deposition in the brain of cognitively healthy aging humans [127].
A role for sleep-related mechanisms in cognitive improvements due to IN insulin would also be in line with observations in healthy male subjects that longer term daily IN administration of 160 IU of insulin vs placebo before nocturnal sleep, but not in the morning, induces slight improvements in declarative memory, i.e., delayed recall of words learned 1 week earlier [128], which also suggests that timing might be a critical determinant of IN insulin effects. This effect appeared to be more pronounced after 5 weeks compared with the end of treatment after 8 weeks, but all in all remained rather modest; interestingly, post-hoc median-split analyses suggested that participants with relatively high compared with those with relatively low systemic insulin sensitivity (reflected by homeostatic model assessment insulin resistance) benefitted to a greater extent [128]. While the mechanisms described in this paragraph are assumed to mediate the functional impact of boosting the physiological brain insulin signal in healthy adults, additional mechanisms likely come into play in individuals who exhibit impairments in memory performance, not least because such impairments are assumed to stem from reduced CNS insulin sensitivity.

4 Intranasal Insulin and Impaired Memory

4.1 Intranasal Insulin Effects in Humans with Mild Cognitive Impairment and AD

Interest in the role of brain insulin signaling in the development of AD and in methods to improve insulin action in the CNS to prevent disease progression has intensified in recent years [e.g., 129131]. This interest has been stoked by pioneering studies conducted by Suzanne Craft and colleagues indicating that the beneficial effects of IN insulin on declarative memory outlined above are not restricted to healthy participants but can also be found in people with mild cognitive impairment (MCI) or (early) AD (see [132] for a systematic review covering relevant research up to October 2017).
In a study in 23 men and women with AD and 14 aged-matched healthy controls who were all non-diabetic, intravenous insulin in comparison with placebo improved story recall, a measure of declarative memory function, and selective attention assessed with the Stroop interference test [133]. Subsequent trials made use of the IN paradigm. In a comparison of 13 adult men and women with early AD and 13 men and women with MCI, matched with 35 controls, the acute effect of IN insulin was investigated in three conditions (placebo [saline], 20 IU and 40 IU of insulin administered 15 min before cognitive assessments) [134]. The cognitive test battery assessed verbal declarative memory (story recall and word-list recall), visual working memory (self-ordered pointing task), selective attention (Stroop test), and visual search. Intranasal insulin compared with placebo improved both measures of recall only in memory-impaired apoE ε4-negative participants, whereas healthy controls did not benefit and memory-impaired apoE ε4 carriers even showed signs of insulin-induced deterioration of word-list recall. Follow-up studies found comparable patterns: apoE ε4-negative participants with memory impairments benefited from acute IN insulin vs placebo (saline) delivery in terms of memory improvement whereas apoE ε4 carriers demonstrated a relative decline [135]. Adults with MCI including amnestic symptoms (e.g., due to AD) who were treated with IN insulin for 3 weeks (2 × 20 IU/day, n = 13) showed significantly increased story recall compared with participants treated with a placebo (saline; n = 12) [136]. The observation of apoE ε4-dependent differences in the impact of IN insulin raises the possibility that brain insulin signaling may only be impaired, and therefore a particularly worthwhile target of interventions, in patients without the apoE ε4 allele [137], which has received further support in subsequent trials [41, 70] (for conflicting data see e.g., 138).
In a pilot clinical trial lasting 4 months [139], women and men diagnosed with MCI or mild-to-moderate AD received 40 IU of regular insulin, placebo (saline), or 40 IU of insulin detemir (each n = 12), a long-acting insulin analog with relatively high lipophilicity that has been assumed to exert stronger effects on brain functions than regular insulin [138, 140]. Cognitive tests included delayed story recall, the Alzheimer Disease Assessment Scale-cognitive subscale 12 (ADAS-Cog-12 [141]), and the Dementia Severity Rating Scale [142]. Intranasal delivery of regular insulin compared with placebo improved memory scores after 2 and 4 months of treatment and was associated with preserved magnetic resonance imaging (MRI)-assessed brain volumes in the left superior parietal cortex, right middle cingulum, left cuneus, and right parahippocampal gyrus. Surprisingly, insulin detemir administration remained without effects. In a related 4-month trial [143], male and female adults with amnestic MCI or mild-to-moderate AD received placebo (saline; n = 30) or 20 IU (n = 36) or 40 IU (n = 38) of regular insulin/day. In comparisons with the placebo group, story recall after a delay of 20 min was enhanced in the 20-IU but not in the 40-IU group, while caregiver-rated functional ability was preserved in both insulin-treated groups; moreover, the progression of hypometabolism assessed via FDG-PET was dampened in both insulin groups. Findings like these suggest that there may be an optimal regimen of IN insulin administration between doses that are too low and, notably, too high, i.e., a inverted U-shaped function of beneficial insulin effects. This assumption has received support in acute experiments by Suzanne Craft’s group [135] and might imply that above a certain threshold (which is yet to be identified) insulin may impair cognitive function, potentially by inducing inflammatory effects (see Sect. 5) [144].
The results of the first multi-site phase II/III clinical trial of IN insulin for MCI and AD, conducted at 27 sites of the Alzheimer’s Therapeutic Research Institute and including 289 participants (155 of them men) between 55 and 85 years of age with a diagnosis of amnestic MCI or AD, have been recently published [145]. The ViaNase device (Kurve Technology), which had been effectively used in previous studies on IN insulin [138, 139, 143], proved unreliable in the first 49 participants because of problems with a newly added electronic timer. Therefore, the remaining 240 participants (designated the primary intention-to-treat population) received a daily dose of 40 IU of insulin or placebo (diluent) with the I109 Precision Olfactory Delivery device (Impel NeuroPharma) for 12 months followed by a 6-month open-label extension phase. Mean score change on the ADAS-Cog-12 [141], evaluated at 3-month intervals, was the primary outcome measure. In contrast to the promising effects discussed above, no differences between insulin and placebo were observed in the primary measure or in other clinical (e.g., Alzheimer Disease Cooperative Study Activities of Daily Living Scale for MCI, ADLMCI [146]) or CSF parameters (e.g., Aβ42 and Aβ40, total tau protein, tau p-181, CSF insulin concentrations). Very small reductions in hippocampal and entorhinal cortex volume were identified by MRI in the insulin- compared with the placebo-treated participants. Interestingly, in secondary analyses of the participants who used the ViaNase device, signs of improved ADAS-Cog-12 scores were observed in the insulin (n = 23) compared with the placebo group (n = 22) during the blinded as well as during the open-label extension phase along with increased Aβ42–Aβ40 and Aβ42 to total tau ratios as well as an insulin-induced decrease in enthorinal cortex volume. Considering that the participants were allowed to receive background therapy such as cholinesterase inhibitors or memantine, these improvements have been judged to be clinically relevant [25].

4.2 Brain Insulin Resistance in AD and Related Memory Impairments

Given that the CNS administration (via the IN pathway) of insulin, a major factor in the control of peripheral glucose homeostasis, ameliorates cognitive function in amnestic patients, it is not surprising that impairments in systemic and brain insulin sensitivity have been found to be interrelated and that they may jointly contribute to the pathogenesis and progression of AD. “Brain insulin resistance,” defined as the failure of brain cells to respond to insulin [24, 86], on a functional level implies that the CNS insulin signal does not effectively support cognitive processes (or the control of metabolism), and could involve downregulation or failure of insulin receptors as well as impairments of downstream signaling. Brain insulin resistance may be a cooccurrence or, potentially, a consequence of peripheral insulin resistance, which is for example in line with Fernanda de Felice’s cumulative hypothesis that the additive impact of unhealthy lifestyles (e.g., low physical activity, inadequate nutrition) eventually results in defects of brain metabolism and brain insulin signaling that trigger cognitive decline [92]. Notably, impairments in peripheral insulin signaling in individuals with AD were suggested more than 25 years ago [147]. Frazier and colleagues have recently come up with an inspiring account of research into brain insulin resistance, putting forward the idea that whereas brain insulin signaling may be impaired in AD, type 2 diabetes, and aging, insulin sensitivity per se may be preserved in these conditions [103]. Indicators of brain insulin resistance have also been found in the relative absence of systemic insulin resistance (see below). As pointed out recently [25], however, it is unclear whether insulin resistance can develop in the brain independently from systemic insulin resistance. Additionally, brain insulin resistance so far has only been determined in relation to supposedly normal insulin effects on the brain, whereas discrete functional, neurophysiological, or neuroimaging-derived criteria have not been established [25]. A number of cognitive domains have been consistently observed to be affected in individuals with type 2 diabetes (e.g., memory, psychomotor speed, executive function, processing speed, verbal fluency, attention [137]) and respective organ deficits include white matter lesions [148] as well as ischemic impairments, cerebral atrophy, and cortical hypometabolism [86]. In animal experiments, chronic hyperinsulinemia as found in obesity and diabetes was demonstrated to decrease the number of insulin receptors at the BBB [35], thereby attenuating brain insulin uptake. Aggregation of advanced glycation end-products due to hyperglycemia likewise compromises BBB functionality [149]. Such impairments might contribute to the increased incidence of AD in patients with metabolic impairments like diabetes that is indicated by epidemiological as well as experimental findings [e.g., 150, 151] (for reviews see [152, 153]), and that may have unfavorable therapeutic consequences when it comes to diabetes self-management [154]. A recently completed clinical trial (NCT02415556) has investigated the impact of long-term administration (24 weeks and 24 weeks follow-up) of IN insulin (40 IU/day vs saline) on measures of cognition (e.g., spatial working memory, paired associate learning), daily functionality, and gait speed in adults with type 2 diabetes and controls of aged 50–85 years [155]; its results are expected to potentially identify clinical phenotypes that predict the response to IN insulin.
Using high spatial resolution, arterial spin labeling MRI at rest and during mild hypercapnia, Frosch and colleagues [156] compared lean controls and obese or overweight adults with and without insulin resistance and found a reduction in cerebrovascular reactivity to mild hypercapnia in obesity compared with normal weight. In the obese subjects with insulin resistance, cerebrovascular reactivity and insulin sensitivity as reflected by QUICKI values [157] were significantly related, suggesting that impairments in cerebrovascular reactivity might precede full-blown diabetes and eventually result in a vicious circle of central and peripheral insulin resistance. Notably, individuals with systemic insulin resistance also display a decrease in hippocampal volume [158] and hippocampal atrophy, a marker of neurodegeneration [159]. Hyperphosphorylated tau in CSF and brain parenchyma [160, 161] and increased deposition of Aβ [162, 163] have been found to be associated with signs of insulin resistance in some studies. Although this and related evidence [164] points to an association of systemic insulin resistance or type 2 diabetes and molecular symptoms of neurodegenerative diseases, many studies have failed to establish such a relationship [e.g., [165] (for in-depth discussions of in-vivo and post-mortem studies as well as genetic risk factors, see [25, 86]). Recent investigations that assessed brain Aβ accumulation via 11C-Pittsburgh compound B (PiB)-PET scans in 41 individuals with type 2 diabetes of the Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) likewise revealed only weak indicators of a relationship between blood markers of insulin resistance and Aβ deposition [166].
There is some experimental support for the assumption that brain insulin resistance may contribute to the development of AD independent of systemic failures in insulin signaling (as in type 2 diabetes) [e.g., 92, 167169]. Post-mortem analyses of the brains of patients with AD have indicated decreases in messenger RNA and protein expression of insulin and insulin receptors as well as insulin-like growth factor-1 and insulin-like growth factor-2 along with signs of reduced downstream insulin signaling mechanisms that were related to disease markers of AD [167]. Such changes may trigger negative consequences for neuronal repair, dendritic sprouting, and differentiation [170] and impair neuronal plasticity via detrimental effects on glutamatergic and cholinergic pathways [137, 171]. In subsequent and very sophisticated analyses of post-mortem hippocampal tissues from elderly individuals with or without AD, without a history of diabetes, indicators of dysregulation of insulin signaling pathways were detected [168]: in a novel ex vivo stimulation paradigm, insulin signaling cascades were strongly impaired in the hippocampal tissue of patients compared with controls matched for age and sex, and these impairments were negatively related to scores of cognition and memory. In further post-mortem analyses of insulin signaling in the middle frontal gyrus cortex in 150 individuals (mean age at death, 87 years, 48% women), there were no differences between individuals with or without diabetes in IRS1 phosphorylation (pS307IRS1/total IRS1) and Akt phosphorylation (pT308Akt1/total Akt1); the latter was highly significantly associated with composite scores of AD pathology [172]. (In contrast to the previous findings from the same group [168], IRS1 serine phosphorylation was not found to be associated with cognitive AD pathology in this sample.) The concentration of insulin in the CSF of patients with AD appears to be an unresolved issue as some reports have indicated increased [173] or, on the contrary, reduced levels [174, 175], whereas other findings point to normal concentrations [176, 177]; the respective contribution of potential impairments in insulin production within the CNS is an intriguing, albeit debated issue [9, 11, 86, 103]. Deteriorations in the clearance and degradation of Aβ due to insulin resistance are discussed as a mechanism that increases the risk of AD [178] and may be improved by insulin administration [179181]. In 3×Tg-AD mice, a rodent model of AD, IN insulin compared with placebo administration for 2 months improved measures of short-term memory (spatial learning in the Morris water maze test and novel object recognition), ameliorated depressive-like behavior (assessed by the tail suspension and the forced swim test), and decreased markers of disease pathology, i.e., tau phosphorylation in the hippocampus and frontal cortex as well as hippocampal concentrations of Aβ oligomers and 3-nitrotyrosine [182]. These findings extend previous observations in animal experiments (e.g., [83, 183]). Brain insulin resistance has also been assumed to be influenced by genetic factors in addition to and beyond apoE ɛ4. For example, subjects with the FTO gene polymorphism rs8050136 as well as carriers of the Gly972Arg polymorphism of IRS1 exhibit a decreased cerebrocortical response to intravenous insulin [184, 185].

4.3 Effectiveness and Safety of Intranasal Insulin for AD

Only one study so far has presented straightforward evidence for CSF uptake of insulin after IN delivery in humans [54]. Although studies in animals conclusively support the assumption that IN administered substances (including insulin) are readily transported to the brain compartment [59], further experimental corroboration of the bioavailability of IN insulin, not least in patients with AD and related disorders as well as elderly individuals, would be welcome evidence for the effectiveness of IN insulin delivery. Nevertheless, respective experiments on other peptides such as oxytocin [186] corroborate the feasibility of IN peptide administration. Considering the lack of effects on primary outcome measures in the recent multi-site phase II/III clinical trial of IN insulin for MCI and AD [145], the currently available devices for IN drug delivery may benefit from further optimization [187]. The device used in that trial, which relies on a liquid hydrofluoroalkane propellant to eject a metered dose of insulin through a nose tip and achieved very high adherence rates, had not been previously tested in patients with AD but proved effective in animal experiments [59]. In this context, it should be noted that CSF increases after IN delivery of insulin [54] and a plethora of functional effects [6265, 69, 72, 81, 114, 128, 188] in humans were observed in experiments that used a simple spray atomizer to initiate nose-to-brain transport of insulin. (Pharmacokinetic considerations notwithstanding, the same can be said of IN oxytocin [189]). Thus, it seems worthwhile to ponder if delivery devices that include more advanced, but maybe less robust or reliable, hardware or electronic components are essential to achieve successful brain uptake of IN administered hormones. While specifically targeting the upper third of the nasal cavity to optimally reach the olfactory epithelium is certainly a worthwhile idea [59], functional MRI assessments of regional cerebral blood flow corroborate the effectiveness of basic nasal spray devices [190]. However, considering that advanced age [191] and cognitive impairments including AD [192] are associated with olfactory impairments that may be exacerbated by nasal membrane atrophy and nasal obstructions, efforts to improve the bioavailability of intranasally administered drugs are warranted. Relying on, for example, the use of nanoparticle carriers [193], cell-penetrating peptides [194], focused ultrasound [195], and other absorption enhancers [196], they have yielded promising results and might be expected to enhance the nasal uptake of insulin while maintaining the safety profile and low systemic exposure associated with IN administration.
Insulin treatment did not increase CSF insulin concentrations regardless of the administration device in the phase II/III trial, but the measurements were made at single time-points during baseline and after 12 months of administration; the authors conclude that direct (CSF- or imaging-derived) proof of the ability of an IN device to target the CNS should best be collected before its use in clinical trials [145]. As a side note, it is worth mentioning a peculiar feature of IN insulin. All experiments in healthy participants and clinical cohorts described herein used insulin formulations (e.g., Novolin R, Humulin R, Levemir) that contain m-cresol (meta-cresol), an excipient with a distinct “coal tar” smell that is highly noticeable (and sometimes reported to be unpleasant) during IN use. In experiments with a crossover design [e.g., 6365, 114, 128], it seems therefore mandatory to administer a diluent/carrier solution in the placebo condition to prevent premature unblinding. Although this precaution might appear of lesser relevance for parallel studies that expose participants to only one treatment [e.g., 70, 136, 138, 139, 143], it is conceivable that the intense smell of insulin solutions elicits stronger expectancy effects than a non-odorous placebo, with potential implications for cognitive outcomes (perhaps even in respective animal studies). In the recent phase II/III trial, this potential confounder was excluded by using a diluent for the placebo [145].
The principal effectiveness to enhance memory function of boosting brain insulin signaling by IN insulin delivery in healthy participants, but also individuals with MCI or AD has been demonstrated in the studies discussed above. While signs of a modulating effect of apoE-ε4 on the neurofunctional impact of IN insulin in patients with AD have been repeatedly found ([134, 135, 138, 139]; see above) and animal experiments hint at potentially underlying mechanisms [197], systematic investigations in humans are needed to clarify the relevance of apoE-ε4 in the response to IN insulin [198], also with regard to the role of brain glucose metabolism. Experiments relying on FDG-PET in middle-aged adults at risk of developing AD revealed an association between systemic insulin resistance and lower glucose metabolism in the left temporal medial lobe that predicted impaired immediate and delayed memory performance, but did not interact with apoE-ε4 status; however, carriers of one or two ε4 alleles displayed decreased global glucose metabolism [199]. Mice carrying the apoE ɛ4 variant in comparison with controls carrying the ɛ2 allele, which is assumed to be protective, show reduced BBB glucose transport [200], suggesting that the higher AD risk in carriers of apoE ɛ4 may in part derive from reduced glucose transport into the brain [201]. Against the background of these and related reports of impaired brain glucose metabolism in AD ([e.g. [202, 203]), it might be speculated that insulin-induced enhancements of cognitive function in memory-impaired patients that occur within minutes at least in part derive from increases in cerebral glucose metabolism. However, considering that the absence of apoE ɛ4 appears to be a prerequisite for the cognitive impact of IN insulin, additional glucose-independent mechanisms are likely; it has also been argued that enhanced glucose uptake may mediate the acute effects of IN insulin whereas prolonged treatment may be necessary to induce improvements in synaptic plasticity [204]. In recent analyses of plasma samples obtained before and after 4 months of IN insulin vs saline administration to participants with MCI [205], favorable cognitive outcomes (ADAS-Cog) in response to the 20-IU dose of IN insulin [143] were mirrored by changes in neuronal extracellular vesicle biomarkers of insulin resistance (pS312-IRS-1, pY-IRS-1), which are known to be increased in patients with type 2 diabetes or AD and discussed as an easily accessible marker of brain insulin resistance [25]. This outcome, which appeared to be restricted to apoE ε4 non-carriers, suggests the engagement of the neuronal insulin cascade.
A meta-analysis of the efficacy and acceptability of antidiabetic agents (IN insulin, pioglitazone, rosiglitazone, metformin, and liraglutide) for MCI and AD that comprised 19 studies published until January 2018 found that antidiabetic treatments overall improved cognitive performance [206]. Thus, approaches to overcome CNS insulin resistance might for example make use of the insulin-sensitizing effects of glucagon-like peptide-1 [207] or of metformin that is routinely prescribed for type 2 diabetes [208]. Metformin enhanced memory and decreased the concentrations of Aβ, hyperphosphorylated tau, and activated microglia in AD mouse models along with signs of improved insulin signaling in the brain [209, 210]. On the background of promising metformin effects on memory performance in individuals with MCI but without diabetes [211], a phase II trial (NCT04098666) in patients with MCI or AD is ongoing. While initial studies also boded well for the use of the peroxisome proliferator-activated receptor-ƴ agonist rosiglitazone [212], subsequent clinical trials did not indicate primary endpoint improvements in AD [213]. Moreover, a recent multi-site trial of piaglitazone in healthy participants aged 65 years or older with a high genotype-determined risk of developing cognitive impairments due to AD was terminated early for a lack of efficacy (NCT01931566 [214]). It should also be noted that lifestyle interventions to improve dietary habits [215] and increase physical activity [216] hold some promise to ameliorate cognitive impairments and AD, possibly via enhancements in brain insulin signaling.
The safety profile of IN insulin has been systematically reviewed [58] (see [132, 217] for further reports). In 38 studies on acute IN insulin administration that included 1092 participants, no adverse events or cases of hypoglycemia were reported. Eighteen studies used long-term administration, with durations between 21 days and 9.7 years and a combined number of 832 participants. The only symptomatic case of hypoglycemia in these studies was reported after administration of a placebo spray [218]. It was concluded that irritation of the nasal mucosa is the most commonly reported side effect, and that the IN route for insulin administration is safe and well tolerated both during acute and chronic use. These findings were corroborated in related meta-analyses [206] and the most recent trial on IN insulin [145] that found no indicators of clinically relevant adverse events as a result of the daily administration of 40 IU of insulin with two different administration devices.

5 Concluding Remarks

Some caveats should be mentioned. Considering the hyperinsulinemia that accompanies peripheral insulin resistance, it might be argued that the (relative) reduction of CSF insulin observed in obese individuals [36] and, in some experiments, in patients with AD [174, 175], represents a protective mechanism limiting CNS hyperinsulinemia and potentially detrimental sequelae of cellular insulin resistance in CNS pathways. This speculative assumption is in line with the observations of dose-dependent effects of IN insulin administration on memory function discussed above: acute IN insulin administration to individuals with AD improved verbal memory recall at lower (20 IU) but not higher doses (up to 60 IU); in carriers of the apoE ε4 allele, higher doses were even found to compromise memory performance [135]. Acute moderate euglycemic hyperinsulinemia in healthy individuals has been found to increase markers of CNS inflammation and Aβ formation [144], both of which increase the risk to develop cognitive impairments. However, pro-inflammatory in vitro effects on glial cells were found to vanish at higher insulin concentrations [219] and IN insulin decreased neuroinflammation and hippocampal lesion volume in a rat model of traumatic brain injury [28] (see [220, 221] for a discussion of insulin signaling and inflammatory processes in neurodegenerative disorders). The assumption that CNS hyperinsulinemia might promote brain insulin resistance is supported by in vitro experiments indicating that prolonged (4–24 h) exposure of hypothalamic cells to high insulin concentrations inactivate and degrade insulin receptors and IRS-1 [222]. Therefore, and against the background of the outcomes of most recent larger trials [145], it will be critical to investigate if the beneficial effects of acute and prolonged IN insulin administration can be corroborated and eventually put to use in the clinical setting, or if exogenous insulin delivery implies the risk of “induced brain insulin resistance.” Moreover, there are many open questions regarding the mechanisms underlying and the implications of impaired brain insulin signaling in cognitive and metabolic disorders. They concern the relationship between AD and diabetes and brain concentrations of insulin, the factors that mediate cognitive impairments in metabolic disorders and, not least, the question whether neurodegeneration in AD can negatively affect the CNS control of systemic energy metabolism and contribute to systemic insulin resistance [86].
With regard to the use of IN insulin to prevent or counteract neurodegenerative disorders, future research may focus on a number of unresolved major issues:
  • Considering that (long-term) IN insulin delivery alone might be associated with gradual downregulation of CNS insulin sensitivity, may its combination with insulin sensitizers such as metformin be superior in boosting cognitive function? Should IN insulin be administered after improvements in (CNS) insulin sensitivity have been achieved in patients with cognitive impairments and metabolic comorbidities via conventional means such as lifestyle intervention, so that resulting gains in brain functions can be preserved?
  • Which delivery approaches and devices are optimally suited to enable nose-to-brain transport of insulin and other drugs, particularly in the clinical setting? Which absorption enhancers are best equipped to maximize brain permeation of IN insulin, and which doses, dosing schedules, insulin formulations, or insulin analogs are needed for the optimization of the memory effect?
  • To which extent do mechanisms related to olfaction and sensory perception contribute to memory improvements after IN insulin delivery? Do sleep-related and circadian neurophysiological and neuroendocrine processes and stress-related psychoneuroendocrine factors modulate the impact of IN insulin in a (clinically) relevant manner?
  • Does the cognitive (as well as metabolic) response to IN insulin critically depend on age and sex, and if so, how can future treatment approaches relying on IN insulin be tailored to the individual needs of patients?
In sum, while the bulk of experimental work outlined in this review underlines the effectiveness of IN insulin to improve memory function, there is still some work to be done to avoid pitfalls and fulfill the potential of IN insulin for AD.

Declarations

Funding

Open Access funding enabled and organized by Projekt DEAL. This work was supported by grants from the German Federal Ministry of Education and Research (BMBF) to the German Center for Diabetes Research (DZD e.V.; 01GI0925).

Conflicts of interest/Competing interests

Manfred Hallschmid has received honoraria and/or travel reimbursements from Boehringer-Ingelheim, Germany, Lilly UK, and NovoNordisk, Denmark. These relationships had no effect on the preparation of the article.

Ethics approval

Not applicable.
Not applicable.
Not applicable.

Availability of data and material

Not applicable.

Code availability

Not applicable.

Authors’ contributions

MH performed the literature search and conceived, drafted, and revised the work.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Neurologie

Kombi-Abonnement

Mit e.Med Neurologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes, den Premium-Inhalten der neurologischen Fachzeitschriften, inklusive einer gedruckten Neurologie-Zeitschrift Ihrer Wahl.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
2.
Zurück zum Zitat Wu Y-T, Beiser AS, Breteler MMB, Fratiglioni L, Helmer C, Hendrie HC, et al. The changing prevalence and incidence of dementia over time: current evidence. Nat Rev Neurol. 2017;13:327–39.PubMed Wu Y-T, Beiser AS, Breteler MMB, Fratiglioni L, Helmer C, Hendrie HC, et al. The changing prevalence and incidence of dementia over time: current evidence. Nat Rev Neurol. 2017;13:327–39.PubMed
3.
Zurück zum Zitat Wimo A, Guerchet M, Ali G-C, Wu Y-T, Prina AM, Winblad B, et al. The worldwide costs of dementia 2015 and comparisons with 2010. Alzheimers Dement. 2017;13:1–7.PubMedPubMedCentral Wimo A, Guerchet M, Ali G-C, Wu Y-T, Prina AM, Winblad B, et al. The worldwide costs of dementia 2015 and comparisons with 2010. Alzheimers Dement. 2017;13:1–7.PubMedPubMedCentral
4.
Zurück zum Zitat Scheltens P, Blennow K, Breteler MMB, de Strooper B, Frisoni GB, Salloway S, et al. Alzheimer’s disease. Lancet. 2016;388:505–17.PubMed Scheltens P, Blennow K, Breteler MMB, de Strooper B, Frisoni GB, Salloway S, et al. Alzheimer’s disease. Lancet. 2016;388:505–17.PubMed
5.
Zurück zum Zitat Havrankova J, Roth J, Brownstein M. Insulin receptors are widely distributed in the central nervous system of the rat. Nature. 1978;272:827–9.PubMed Havrankova J, Roth J, Brownstein M. Insulin receptors are widely distributed in the central nervous system of the rat. Nature. 1978;272:827–9.PubMed
6.
Zurück zum Zitat Sara VR, Hall K, Von Holtz H, Humbel R, Sjögren 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.PubMed Sara VR, Hall K, Von Holtz H, Humbel R, Sjögren 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.PubMed
7.
Zurück zum Zitat Wallum BJ, Taborsky GJ, Porte D, Figlewicz DP, Jacobson L, Beard JC, et al. Cerebrospinal fluid insulin levels increase during intravenous insulin infusions in man. J Clin Endocrinol Metab. 1987;64:190–4.PubMed Wallum BJ, Taborsky GJ, Porte D, Figlewicz DP, Jacobson L, Beard JC, et al. Cerebrospinal fluid insulin levels increase during intravenous insulin infusions in man. J Clin Endocrinol Metab. 1987;64:190–4.PubMed
8.
Zurück zum Zitat Woods S, Seeley R, Baskin D, Schwartz M. Insulin and the blood-brain barrier. Curr Pharm Des. 2003;9:795–800.PubMed Woods S, Seeley R, Baskin D, Schwartz M. Insulin and the blood-brain barrier. Curr Pharm Des. 2003;9:795–800.PubMed
9.
Zurück zum Zitat Molnar G, Farago N, Kocsis AK, Rozsa M, Lovas S, Boldog E, et al. GABAergic neurogliaform cells represent local sources of insulin in the cerebral cortex. J Neurosci. 2014;34:1133–7.PubMedPubMedCentral Molnar G, Farago N, Kocsis AK, Rozsa M, Lovas S, Boldog E, et al. GABAergic neurogliaform cells represent local sources of insulin in the cerebral cortex. J Neurosci. 2014;34:1133–7.PubMedPubMedCentral
10.
Zurück zum Zitat Hrytsenko O, Wright JR, Morrison CM, Pohajdak B. Insulin expression in the brain and pituitary cells of tilapia (Oreochromis niloticus). Brain Res. 2007;1135:31–40.PubMed Hrytsenko O, Wright JR, Morrison CM, Pohajdak B. Insulin expression in the brain and pituitary cells of tilapia (Oreochromis niloticus). Brain Res. 2007;1135:31–40.PubMed
11.
Zurück zum Zitat Steen E, Terry BM, Rivera JE, Cannon JL, Neely TR, Tavares R, et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease: is this type 3 diabetes? J Alzheimer’s Dis. 2005;7:63–80. Steen E, Terry BM, Rivera JE, Cannon JL, Neely TR, Tavares R, et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease: is this type 3 diabetes? J Alzheimer’s Dis. 2005;7:63–80.
12.
Zurück zum Zitat Gray SM, Meijer RI, Barrett EJ. Insulin regulates brain function, but how does it get there? Diabetes. 2014;63:3992–7.PubMedPubMedCentral Gray SM, Meijer RI, Barrett EJ. Insulin regulates brain function, but how does it get there? Diabetes. 2014;63:3992–7.PubMedPubMedCentral
13.
Zurück zum Zitat Hom FG, Goodner CJ, Berrie MA. A (3H)2-deoxyglucose method for comparing rates of glucose metabolism and insulin responses among rat tissues in vivo: validation of the model and the absence of an insulin effect on brain. Diabetes. 1984;33:141–52.PubMed Hom FG, Goodner CJ, Berrie MA. A (3H)2-deoxyglucose method for comparing rates of glucose metabolism and insulin responses among rat tissues in vivo: validation of the model and the absence of an insulin effect on brain. Diabetes. 1984;33:141–52.PubMed
14.
Zurück zum Zitat Seaquist ER, Damberg GS, Tkac I, Gruetter R. The effect of insulin on in vivo cerebral glucose concentrations and rates of glucose transport/metabolism in humans. Diabetes. 2001;50:2203–9.PubMed Seaquist ER, Damberg GS, Tkac I, Gruetter R. The effect of insulin on in vivo cerebral glucose concentrations and rates of glucose transport/metabolism in humans. Diabetes. 2001;50:2203–9.PubMed
15.
Zurück zum Zitat Bruning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban PC, et al. Role of brain insulin receptor in control of body weight and reproduction. Science. 2000;289:2122–5.PubMed Bruning JC, Gautam D, Burks DJ, Gillette J, Schubert M, Orban PC, et al. Role of brain insulin receptor in control of body weight and reproduction. Science. 2000;289:2122–5.PubMed
16.
Zurück zum Zitat Pocai A, Lam TK, Gutierrez-Juarez R, Obici S, Schwartz GJ, Bryan J, et al. Hypothalamic K (ATP) channels control hepatic glucose production. Nature. 2005;434:1026–231.PubMed Pocai A, Lam TK, Gutierrez-Juarez R, Obici S, Schwartz GJ, Bryan J, et al. Hypothalamic K (ATP) channels control hepatic glucose production. Nature. 2005;434:1026–231.PubMed
17.
Zurück zum Zitat Rulifson EJ, Kim SK, Nusse R. Ablation of insulin-producing neurons in files: growth and diabetic phenotypes. Science. 2002;296:1118–20.PubMed Rulifson EJ, Kim SK, Nusse R. Ablation of insulin-producing neurons in files: growth and diabetic phenotypes. Science. 2002;296:1118–20.PubMed
18.
Zurück zum Zitat Spinelli M, Fusco S, Grassi C. Brain insulin resistance and hippocampal plasticity: mechanisms and biomarkers of cognitive decline. Front Neurosci. 2019;13:1–13. Spinelli M, Fusco S, Grassi C. Brain insulin resistance and hippocampal plasticity: mechanisms and biomarkers of cognitive decline. Front Neurosci. 2019;13:1–13.
19.
Zurück zum Zitat Woods SC, Lotter EC, McKay LD, Porte D. Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature. 1979;282:503–5.PubMed Woods SC, Lotter EC, McKay LD, Porte D. Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature. 1979;282:503–5.PubMed
20.
Zurück zum Zitat Air EL, Benoit SC, Clegg DJ, Seeley RJ, Woods SC. Insulin and leptin combine additively to reduce food intake and body weight in rats. Endocrinology. 2002;143:2449–52.PubMed Air EL, Benoit SC, Clegg DJ, Seeley RJ, Woods SC. Insulin and leptin combine additively to reduce food intake and body weight in rats. Endocrinology. 2002;143:2449–52.PubMed
21.
Zurück zum Zitat Brown LM, Clegg DJ, Benoit SC, Woods SC. Intraventricular insulin and leptin reduce food intake and body weight in C57BL/6J mice. Physiol Behav. 2006;89:687–91.PubMed Brown LM, Clegg DJ, Benoit SC, Woods SC. Intraventricular insulin and leptin reduce food intake and body weight in C57BL/6J mice. Physiol Behav. 2006;89:687–91.PubMed
22.
Zurück zum Zitat Morton GJ, Meek TH, Schwartz MW. Neurobiology of food intake in health and disease. Nat Rev Neurosci. 2014;15:367–78.PubMedPubMedCentral Morton GJ, Meek TH, Schwartz MW. Neurobiology of food intake in health and disease. Nat Rev Neurosci. 2014;15:367–78.PubMedPubMedCentral
23.
Zurück zum Zitat Hallschmid M, Schultes B. Central nervous insulin resistance: a promising target in the treatment of metabolic and cognitive disorders? Diabetologia. 2009;52:2264–9.PubMed Hallschmid M, Schultes B. Central nervous insulin resistance: a promising target in the treatment of metabolic and cognitive disorders? Diabetologia. 2009;52:2264–9.PubMed
24.
Zurück zum Zitat Kullmann S, Heni M, Hallschmid M, Fritsche A, Preissl H, Häring H-U. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiol Rev. 2016;96:1169–209.PubMed Kullmann S, Heni M, Hallschmid M, Fritsche A, Preissl H, Häring H-U. Brain insulin resistance at the crossroads of metabolic and cognitive disorders in humans. Physiol Rev. 2016;96:1169–209.PubMed
25.
Zurück zum Zitat Kellar D, Craft S. Brain insulin resistance in Alzheimer’s disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol. 2020;19:758–66.PubMed Kellar D, Craft S. Brain insulin resistance in Alzheimer’s disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol. 2020;19:758–66.PubMed
26.
Zurück zum Zitat Pang Y, Lin S, Wright C, Shen J, Carter K, Bhatt A, et al. Intranasal insulin protects against substantia nigra dopaminergic neuronal loss and alleviates motor deficits induced by 6-OHDA in rats. Neuroscience. 2016;318:157–65.PubMed Pang Y, Lin S, Wright C, Shen J, Carter K, Bhatt A, et al. Intranasal insulin protects against substantia nigra dopaminergic neuronal loss and alleviates motor deficits induced by 6-OHDA in rats. Neuroscience. 2016;318:157–65.PubMed
27.
Zurück zum Zitat Novak P, Pimentel Maldonado DA, Novak V. Safety and preliminary efficacy of intranasal insulin for cognitive impairment in Parkinson disease and multiple system atrophy: a double-blinded placebo-controlled pilot study. PLoS ONE. 2019;14:e0214364.PubMedPubMedCentral Novak P, Pimentel Maldonado DA, Novak V. Safety and preliminary efficacy of intranasal insulin for cognitive impairment in Parkinson disease and multiple system atrophy: a double-blinded placebo-controlled pilot study. PLoS ONE. 2019;14:e0214364.PubMedPubMedCentral
28.
Zurück zum Zitat Brabazon F, Wilson CM, Jaiswal S, Reed J, Frey WH, Byrnes KR. Intranasal insulin treatment of an experimental model of moderate traumatic brain injury. J Cereb Blood Flow Metab. 2017;37:3203–18.PubMedPubMedCentral Brabazon F, Wilson CM, Jaiswal S, Reed J, Frey WH, Byrnes KR. Intranasal insulin treatment of an experimental model of moderate traumatic brain injury. J Cereb Blood Flow Metab. 2017;37:3203–18.PubMedPubMedCentral
29.
Zurück zum Zitat Rea S, Della-Morte D, Pacifici F, Capuani B, Pastore D, Coppola A, et al. Insulin and exendin-4 reduced mutated Huntingtin accumulation in neuronal cells. Front Pharmacol. 2020;11:779.PubMedPubMedCentral Rea S, Della-Morte D, Pacifici F, Capuani B, Pastore D, Coppola A, et al. Insulin and exendin-4 reduced mutated Huntingtin accumulation in neuronal cells. Front Pharmacol. 2020;11:779.PubMedPubMedCentral
30.
Zurück zum Zitat Craft S, Watson GS. Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol. 2004;3:169–78.PubMed Craft S, Watson GS. Insulin and neurodegenerative disease: shared and specific mechanisms. Lancet Neurol. 2004;3:169–78.PubMed
31.
Zurück zum Zitat Grigolon RB, Brietzke E, Mansur RB, Idzikowski MA, Gerchman F, De Felice FG, et al. Association between diabetes and mood disorders and the potential use of anti-hyperglycemic agents as antidepressants. Prog Neuropsychopharmacol Biol Psychiatry. 2019;95:109720.PubMed Grigolon RB, Brietzke E, Mansur RB, Idzikowski MA, Gerchman F, De Felice FG, et al. Association between diabetes and mood disorders and the potential use of anti-hyperglycemic agents as antidepressants. Prog Neuropsychopharmacol Biol Psychiatry. 2019;95:109720.PubMed
32.
Zurück zum Zitat Kashyap B, Hanson LR, Frey WH II. Intranasal insulin: a treatment strategy for addiction. Neurotherapeutics. 2020;17:105–15.PubMedPubMedCentral Kashyap B, Hanson LR, Frey WH II. Intranasal insulin: a treatment strategy for addiction. Neurotherapeutics. 2020;17:105–15.PubMedPubMedCentral
33.
Zurück zum Zitat Banks WA. Role of the blood-brain barrier in the evolution of feeding and cognition. Ann NY Acad Sci. 2012;1264:13–9.PubMed Banks WA. Role of the blood-brain barrier in the evolution of feeding and cognition. Ann NY Acad Sci. 2012;1264:13–9.PubMed
34.
Zurück zum Zitat Banks WA. The source of cerebral insulin. Eur J Pharmacol. 2004;490:5–12.PubMed Banks WA. The source of cerebral insulin. Eur J Pharmacol. 2004;490:5–12.PubMed
35.
Zurück zum Zitat Schwartz MW, Sipols A, Kahn SE, Lattemann DF, Taborsky GJ, Bergman RN, et al. Kinetics and specificity of insulin uptake from plasma into cerebrospinal fluid. Am J Physiol Metab. 1990;259:E378. Schwartz MW, Sipols A, Kahn SE, Lattemann DF, Taborsky GJ, Bergman RN, et al. Kinetics and specificity of insulin uptake from plasma into cerebrospinal fluid. Am J Physiol Metab. 1990;259:E378.
36.
Zurück zum Zitat Kern W, Benedict C, Schultes B, Plohr F, Moser A, Born J, et al. Low cerebrospinal fluid insulin levels in obese humans. Diabetologia. 2006;49:2790–2.PubMed Kern W, Benedict C, Schultes B, Plohr F, Moser A, Born J, et al. Low cerebrospinal fluid insulin levels in obese humans. Diabetologia. 2006;49:2790–2.PubMed
37.
Zurück zum Zitat Van Dijk G, De Groote C, Chavez M, Van Der Werf Y, Steffens AB, Strubbe JH. Insulin in the arcuate nucleus of the hypothalamus reduces fat consumption in rats. Brain Res. 1997;777:147–52.PubMed Van Dijk G, De Groote C, Chavez M, Van Der Werf Y, Steffens AB, Strubbe JH. Insulin in the arcuate nucleus of the hypothalamus reduces fat consumption in rats. Brain Res. 1997;777:147–52.PubMed
38.
Zurück zum Zitat Kern W, Lieb K, Kerner W, Born J, Fehm HL. Differential effects of human and pork insulin-induced hypoglycemia on neuronal functions in humans. Diabetes. 1990;39:1091–8.PubMed Kern W, Lieb K, Kerner W, Born J, Fehm HL. Differential effects of human and pork insulin-induced hypoglycemia on neuronal functions in humans. Diabetes. 1990;39:1091–8.PubMed
39.
Zurück zum Zitat Rotte M, Baerecke C, Pottag G, Klose S, Kanneberg E, Heinze H-J, et al. Insulin affects the neuronal response in the medial temporal lobe in Humans. Neuroendocrinology. 2005;81:49–55.PubMed Rotte M, Baerecke C, Pottag G, Klose S, Kanneberg E, Heinze H-J, et al. Insulin affects the neuronal response in the medial temporal lobe in Humans. Neuroendocrinology. 2005;81:49–55.PubMed
40.
Zurück zum Zitat Benedict L, Nelson CA, Schunk E, Sullwold K, Seaquist ER. Effect of insulin on the brain activity obtained during visual and memory tasks in healthy human subjects. Neuroendocrinology. 2006;83:20–6.PubMed Benedict L, Nelson CA, Schunk E, Sullwold K, Seaquist ER. Effect of insulin on the brain activity obtained during visual and memory tasks in healthy human subjects. Neuroendocrinology. 2006;83:20–6.PubMed
41.
Zurück zum Zitat Craft S, Asthana S, Cook DG, Baker LD, Cherrier M, Purganan K, et al. Insulin dose-response effects on memory and plasma amyloid precursor protein in Alzheimer’s disease: interactions with apolipoprotein E genotype. Psychoneuroendocrinology. 2003;28:809–22.PubMed Craft S, Asthana S, Cook DG, Baker LD, Cherrier M, Purganan K, et al. Insulin dose-response effects on memory and plasma amyloid precursor protein in Alzheimer’s disease: interactions with apolipoprotein E genotype. Psychoneuroendocrinology. 2003;28:809–22.PubMed
42.
Zurück zum Zitat Mitrakou A, Ryan C, Veneman T, Mokan M, Jenssen T, Kiss I, et al. Hierarchy of glycemic thresholds for counterregulatory hormone secretion, symptoms, and cerebral dysfunction. Am J Physiol Metab. 1991;260:E67-74. Mitrakou A, Ryan C, Veneman T, Mokan M, Jenssen T, Kiss I, et al. Hierarchy of glycemic thresholds for counterregulatory hormone secretion, symptoms, and cerebral dysfunction. Am J Physiol Metab. 1991;260:E67-74.
43.
Zurück zum Zitat Tesfaye N, Seaquist ER. Neuroendocrine responses to hypoglycemia. Ann NY Acad Sci. 2010;1212:12–28.PubMed Tesfaye N, Seaquist ER. Neuroendocrine responses to hypoglycemia. Ann NY Acad Sci. 2010;1212:12–28.PubMed
44.
Zurück zum Zitat Frey WHI. Neurologic agents for nasal administration to the brain. PCT International Patent WO91/07947; 1991. Frey WHI. Neurologic agents for nasal administration to the brain. PCT International Patent WO91/07947; 1991.
45.
Zurück zum Zitat Frey WHI. Method for administering insulin to the brain. US Patent 6,313,093 B1; 2001. Frey WHI. Method for administering insulin to the brain. US Patent 6,313,093 B1; 2001.
46.
Zurück zum Zitat Thorne RG, Frey WH. Delivery of neurotrophic factors to the central nervous system: pharmacokinetic considerations. Clin Pharmacokinet. 2001;40:907–46.PubMed Thorne RG, Frey WH. Delivery of neurotrophic factors to the central nervous system: pharmacokinetic considerations. Clin Pharmacokinet. 2001;40:907–46.PubMed
47.
Zurück zum Zitat Frey WH, Liu J, Chen X, Thorne RG, Fawcett JR, Ala TA, et al. Delivery of 125 I-NGF to the brain via the olfactory route. Drug Deliv. 1997;4:87–92. Frey WH, Liu J, Chen X, Thorne RG, Fawcett JR, Ala TA, et al. Delivery of 125 I-NGF to the brain via the olfactory route. Drug Deliv. 1997;4:87–92.
48.
Zurück zum Zitat Thorne RG, Pronk GJ, Padmanabhan V, Frey WH. Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration. Neuroscience. 2004;127:481–96.PubMed Thorne RG, Pronk GJ, Padmanabhan V, Frey WH. Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration. Neuroscience. 2004;127:481–96.PubMed
49.
Zurück zum Zitat Hanson LR, Frey WH. Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci. 2008;9:S5.PubMedPubMedCentral Hanson LR, Frey WH. Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci. 2008;9:S5.PubMedPubMedCentral
50.
Zurück zum Zitat Thorne RG, Emory CR, Ala TA, Frey WH. Quantitative analysis of the olfactory pathway for drug delivery to the brain. Brain Res. 1995;692:278–82.PubMed Thorne RG, Emory CR, Ala TA, Frey WH. Quantitative analysis of the olfactory pathway for drug delivery to the brain. Brain Res. 1995;692:278–82.PubMed
51.
Zurück zum Zitat Illum L. Transport of drugs from the nasal cavity to the central nervous system. Eur J Pharm Sci. 2000;11:1–18.PubMed Illum L. Transport of drugs from the nasal cavity to the central nervous system. Eur J Pharm Sci. 2000;11:1–18.PubMed
52.
Zurück zum Zitat Crowe TP, Greenlee MHW, Kanthasamy AG, Hsu WH. Mechanism of intranasal drug delivery directly to the brain. Life Sci. 2018;195:44–52.PubMed Crowe TP, Greenlee MHW, Kanthasamy AG, Hsu WH. Mechanism of intranasal drug delivery directly to the brain. Life Sci. 2018;195:44–52.PubMed
53.
Zurück zum Zitat Lochhead JJ, Kellohen KL, Ronaldson PT, Davis TP. Distribution of insulin in trigeminal nerve and brain after intranasal administration. Sci Rep. 2019;9:2621.PubMedPubMedCentral Lochhead JJ, Kellohen KL, Ronaldson PT, Davis TP. Distribution of insulin in trigeminal nerve and brain after intranasal administration. Sci Rep. 2019;9:2621.PubMedPubMedCentral
54.
Zurück zum Zitat Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci. 2002;5:514–6.PubMed Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci. 2002;5:514–6.PubMed
55.
Zurück zum Zitat Ott V, Lehnert H, Staub J, Wönne K, Born J, Hallschmid M. Central nervous insulin administration does not potentiate the acute glucoregulatory impact of concurrent mild hyperinsulinemia. Diabetes. 2015;64:760–5.PubMed Ott V, Lehnert H, Staub J, Wönne K, Born J, Hallschmid M. Central nervous insulin administration does not potentiate the acute glucoregulatory impact of concurrent mild hyperinsulinemia. Diabetes. 2015;64:760–5.PubMed
56.
Zurück zum Zitat Djupesland PG, Messina JC, Mahmoud RA. The nasal approach to delivering treatment for brain diseases: an anatomic, physiologic, and delivery technology overview. Ther Deliv. 2014;5:709–33.PubMed Djupesland PG, Messina JC, Mahmoud RA. The nasal approach to delivering treatment for brain diseases: an anatomic, physiologic, and delivery technology overview. Ther Deliv. 2014;5:709–33.PubMed
57.
Zurück zum Zitat Van de Bittner GC, Van de Bittner KC, Wey H-Y, Rowe W, Dharanipragada R, Ying X, et al. Positron emission tomography assessment of the intranasal delivery route for orexin A. ACS Chem Neurosci. 2018;9:358–68.PubMed Van de Bittner GC, Van de Bittner KC, Wey H-Y, Rowe W, Dharanipragada R, Ying X, et al. Positron emission tomography assessment of the intranasal delivery route for orexin A. ACS Chem Neurosci. 2018;9:358–68.PubMed
58.
Zurück zum Zitat Schmid V, Kullmann S, Gfrörer W, Hund V, Hallschmid M, Lipp H, et al. Safety of intranasal human insulin: a review. Diabetes Obes Metab. 2018;20:1563–77.PubMed Schmid V, Kullmann S, Gfrörer W, Hund V, Hallschmid M, Lipp H, et al. Safety of intranasal human insulin: a review. Diabetes Obes Metab. 2018;20:1563–77.PubMed
59.
Zurück zum Zitat Lochhead JJ, Thorne RG. Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev. 2012;64:614–28.PubMed Lochhead JJ, Thorne RG. Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev. 2012;64:614–28.PubMed
60.
Zurück zum Zitat Pires A, Fortuna A, Alves G, Falcão A. Intranasal drug delivery: how, why and what for? J Pharm Pharm Sci. 2009;12:288.PubMed Pires A, Fortuna A, Alves G, Falcão A. Intranasal drug delivery: how, why and what for? J Pharm Pharm Sci. 2009;12:288.PubMed
61.
Zurück zum Zitat Spetter MS, Hallschmid M. Current findings on the role of oxytocin in the regulation of food intake. Physiol Behav. 2017;176:31–9.PubMed Spetter MS, Hallschmid M. Current findings on the role of oxytocin in the regulation of food intake. Physiol Behav. 2017;176:31–9.PubMed
62.
Zurück zum Zitat 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;93:1339–44.PubMed 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;93:1339–44.PubMed
63.
Zurück zum Zitat Benedict C, Hallschmid M, Hatke A, Schultes B, Fehm HL, Born J, et al. Intranasal insulin improves memory in humans. Psychoneuroendocrinology. 2004;29:1326–34.PubMed Benedict C, Hallschmid M, Hatke A, Schultes B, Fehm HL, Born J, et al. Intranasal insulin improves memory in humans. Psychoneuroendocrinology. 2004;29:1326–34.PubMed
64.
Zurück zum Zitat Benedict C, Hallschmid M, Schmitz K, Schultes B, Ratter F, Fehm HL, et al. Intranasal insulin improves memory in humans: Superiority of insulin aspart. Neuropsychopharmacology. 2007;32:239–43.PubMed Benedict C, Hallschmid M, Schmitz K, Schultes B, Ratter F, Fehm HL, et al. Intranasal insulin improves memory in humans: Superiority of insulin aspart. Neuropsychopharmacology. 2007;32:239–43.PubMed
65.
Zurück zum Zitat Feld GB, Wilhem I, Benedict C, Rüdel B, Klameth C, Born J, et al. Central nervous insulin signaling in sleep-associated memory formation and neuroendocrine regulation. Neuropsychopharmacology. 2016;41:1540–50.PubMed Feld GB, Wilhem I, Benedict C, Rüdel B, Klameth C, Born J, et al. Central nervous insulin signaling in sleep-associated memory formation and neuroendocrine regulation. Neuropsychopharmacology. 2016;41:1540–50.PubMed
66.
Zurück zum Zitat Brünner YF, Kofoet A, Benedict C, Freiherr J. Central insulin administration improves odor cued reactivation of spatial memory in young men. J Clin Endocrinol Metab. 2015;100:212–9.PubMed Brünner YF, Kofoet A, Benedict C, Freiherr J. Central insulin administration improves odor cued reactivation of spatial memory in young men. J Clin Endocrinol Metab. 2015;100:212–9.PubMed
67.
Zurück zum Zitat Kurtzhals P, Schaffer L, Sorensen A, Kristensen C, Jonassen I, Schmid C, et al. Correlations of receptor binding and metabolic and mitogenic potencies of insulin analogs designed for clinical use. Diabetes. 2000;49:999–1005.PubMed Kurtzhals P, Schaffer L, Sorensen A, Kristensen C, Jonassen I, Schmid C, et al. Correlations of receptor binding and metabolic and mitogenic potencies of insulin analogs designed for clinical use. Diabetes. 2000;49:999–1005.PubMed
68.
Zurück zum Zitat Rodriguez-Raecke R, Brünner YF, Kofoet A, Mutic S, Benedict C, Freiherr J. Odor sensitivity after intranasal insulin application is modulated by gender. Front Endocrinol (Lausanne). 2018;9:580.PubMedPubMedCentral Rodriguez-Raecke R, Brünner YF, Kofoet A, Mutic S, Benedict C, Freiherr J. Odor sensitivity after intranasal insulin application is modulated by gender. Front Endocrinol (Lausanne). 2018;9:580.PubMedPubMedCentral
69.
Zurück zum Zitat Hallschmid M, Benedict C, Schultes B, Fehm HL, Born J, Kern W. Intranasal insulin reduces body fat in men but not in women. Diabetes. 2004;53:3024–9.PubMed Hallschmid M, Benedict C, Schultes B, Fehm HL, Born J, Kern W. Intranasal insulin reduces body fat in men but not in women. Diabetes. 2004;53:3024–9.PubMed
70.
Zurück zum Zitat Claxton A, Baker LD, Wilkinson CW, Trittschuh EH, Chapman D, Watson GS, et al. Sex and ApoE genotype differences in treatment response to two doses of intranasal insulin in adults with mild cognitive impairment or alzheimer’s disease. J Alzheimers Dis. 2013;35:789–97.PubMedPubMedCentral Claxton A, Baker LD, Wilkinson CW, Trittschuh EH, Chapman D, Watson GS, et al. Sex and ApoE genotype differences in treatment response to two doses of intranasal insulin in adults with mild cognitive impairment or alzheimer’s disease. J Alzheimers Dis. 2013;35:789–97.PubMedPubMedCentral
71.
Zurück zum Zitat Clegg DJ, Brown LM, Woods SC, Benoit SC. Gonadal hormones determine sensitivity to central leptin and insulin. Diabetes. 2006;55:978–87.PubMed Clegg DJ, Brown LM, Woods SC, Benoit SC. Gonadal hormones determine sensitivity to central leptin and insulin. Diabetes. 2006;55:978–87.PubMed
72.
Zurück zum Zitat Krug R, Benedict C, Born J, Hallschmid M. Comparable sensitivity of postmenopausal and young women to the effects of intranasal insulin on food intake and working memory. J Clin Endocrinol Metab. 2010;95:E468–72.PubMed Krug R, Benedict C, Born J, Hallschmid M. Comparable sensitivity of postmenopausal and young women to the effects of intranasal insulin on food intake and working memory. J Clin Endocrinol Metab. 2010;95:E468–72.PubMed
73.
Zurück zum Zitat Alzheimer’s Association. . Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2019;2019(15):321–87. Alzheimer’s Association. . Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2019;2019(15):321–87.
74.
Zurück zum Zitat Raber J, Huang Y, Ashford JW. ApoE genotype accounts for the vast majority of AD risk and AD pathology. Neurobiol Aging. 2004;25:641–50.PubMed Raber J, Huang Y, Ashford JW. ApoE genotype accounts for the vast majority of AD risk and AD pathology. Neurobiol Aging. 2004;25:641–50.PubMed
75.
Zurück zum Zitat Neu SC, Pa J, Kukull W, Beekly D, Kuzma A, Gangadharan P, et al. Apolipoprotein E genotype and sex risk factors for Alzheimer disease: a meta-analysis. JAMA Neurol. 2017;74:1178–89.PubMedPubMedCentral Neu SC, Pa J, Kukull W, Beekly D, Kuzma A, Gangadharan P, et al. Apolipoprotein E genotype and sex risk factors for Alzheimer disease: a meta-analysis. JAMA Neurol. 2017;74:1178–89.PubMedPubMedCentral
76.
Zurück zum Zitat Mauvais-Jarvis F, Bairey Merz N, Barnes PJ, Brinton RD, Carrero J, DeMeo DL, et al. Sex and gender: modifiers of health, disease, and medicine. Lancet. 2020;396:565–82.PubMedPubMedCentral Mauvais-Jarvis F, Bairey Merz N, Barnes PJ, Brinton RD, Carrero J, DeMeo DL, et al. Sex and gender: modifiers of health, disease, and medicine. Lancet. 2020;396:565–82.PubMedPubMedCentral
77.
Zurück zum Zitat Mittendorfer B. Insulin resistance: sex matters. Curr Opin Clin Nutr Metab Care. 2005;8:367–72.PubMed Mittendorfer B. Insulin resistance: sex matters. Curr Opin Clin Nutr Metab Care. 2005;8:367–72.PubMed
78.
Zurück zum Zitat 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. 2008;32:275–82. 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. 2008;32:275–82.
79.
Zurück zum Zitat Stingl KT, Kullmann S, Guthoff M, Heni M, Fritsche A, Preissl H. Insulin modulation of magnetoencephalographic resting state dynamics in lean and obese subjects. Front Syst Neurosci. 2010;4:157.PubMedPubMedCentral Stingl KT, Kullmann S, Guthoff M, Heni M, Fritsche A, Preissl H. Insulin modulation of magnetoencephalographic resting state dynamics in lean and obese subjects. Front Syst Neurosci. 2010;4:157.PubMedPubMedCentral
80.
Zurück zum Zitat Kern W, Born J, Schreiber H, Fehm HL. Central nervous system effects of intranasally administered insulin during euglycemia in men. Diabetes. 1999;48:557–63.PubMed Kern W, Born J, Schreiber H, Fehm HL. Central nervous system effects of intranasally administered insulin during euglycemia in men. Diabetes. 1999;48:557–63.PubMed
81.
Zurück zum Zitat Hallschmid M, Schuttes B, Marshall L, Mölle M, Kern W, Bredthauer J, et al. Transcortical direct current potential shift reflects immediate signaling of systemic insulin to the human brain. Diabetes. 2004;53:2202–8.PubMed Hallschmid M, Schuttes B, Marshall L, Mölle M, Kern W, Bredthauer J, et al. Transcortical direct current potential shift reflects immediate signaling of systemic insulin to the human brain. Diabetes. 2004;53:2202–8.PubMed
82.
Zurück zum Zitat Kum W, Zhu SQ, Ho SK, Young JD, Cockram CS. Effect of insulin on glucose and glycogen metabolism and leucine incorporation into protein in cultured mouse astrocytes. Glia. 1992;6:264–8.PubMed Kum W, Zhu SQ, Ho SK, Young JD, Cockram CS. Effect of insulin on glucose and glycogen metabolism and leucine incorporation into protein in cultured mouse astrocytes. Glia. 1992;6:264–8.PubMed
83.
Zurück zum Zitat Marks DR, Tucker K, Cavallin MA, Mast TG, Fadool DA. Awake intranasal insulin delivery modifies protein complexes and alters memory, anxiety, and olfactory behaviors. J Neurosci. 2009;29:6734–51.PubMedPubMedCentral Marks DR, Tucker K, Cavallin MA, Mast TG, Fadool DA. Awake intranasal insulin delivery modifies protein complexes and alters memory, anxiety, and olfactory behaviors. J Neurosci. 2009;29:6734–51.PubMedPubMedCentral
84.
Zurück zum Zitat Grillo CA, Piroli GG, Kaigler KF, Wilson SP, Wilson MA, Reagan LP. Downregulation of hypothalamic insulin receptor expression elicits depressive-like behaviors in rats. Behav Brain Res. 2011;222:230–5.PubMedPubMedCentral Grillo CA, Piroli GG, Kaigler KF, Wilson SP, Wilson MA, Reagan LP. Downregulation of hypothalamic insulin receptor expression elicits depressive-like behaviors in rats. Behav Brain Res. 2011;222:230–5.PubMedPubMedCentral
85.
Zurück zum Zitat Ott V, Benedict C, Schultes B, Born J, Hallschmid M. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism. Diabetes Obes Metab. 2012;14:214–21.PubMed Ott V, Benedict C, Schultes B, Born J, Hallschmid M. Intranasal administration of insulin to the brain impacts cognitive function and peripheral metabolism. Diabetes Obes Metab. 2012;14:214–21.PubMed
86.
Zurück zum Zitat Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang H-Y, Ahima RS, et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol. 2018;14:168–81.PubMedPubMedCentral Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang H-Y, Ahima RS, et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol. 2018;14:168–81.PubMedPubMedCentral
87.
Zurück zum Zitat Marks JL, Porte D, Stahl WL, Basking DG. Localization of insulin receptor mrna in rat brain by in situ hybridization. Endocrinology. 1990;127:3234–6.PubMed Marks JL, Porte D, Stahl WL, Basking DG. Localization of insulin receptor mrna in rat brain by in situ hybridization. Endocrinology. 1990;127:3234–6.PubMed
88.
Zurück zum Zitat Unger JW. Insulin receptors and signal transduction proteins in the hypothalamo- hypophyseal system: a review on morphological findings and functional implications. Histol Histopathol. 1998;13:1215–24.PubMed Unger JW. Insulin receptors and signal transduction proteins in the hypothalamo- hypophyseal system: a review on morphological findings and functional implications. Histol Histopathol. 1998;13:1215–24.PubMed
89.
Zurück zum Zitat Nelson TJ, Sun M-K, Hongpaisan J, Alkon DL. Insulin, PKC signaling pathways and synaptic remodeling during memory storage and neuronal repair. Eur J Pharmacol. 2008;585:76–87.PubMed Nelson TJ, Sun M-K, Hongpaisan J, Alkon DL. Insulin, PKC signaling pathways and synaptic remodeling during memory storage and neuronal repair. Eur J Pharmacol. 2008;585:76–87.PubMed
90.
Zurück zum Zitat Gralle M. The neuronal insulin receptor in its environment. J Neurochem. 2017;140:359–67.PubMed Gralle M. The neuronal insulin receptor in its environment. J Neurochem. 2017;140:359–67.PubMed
91.
Zurück zum Zitat Van Der Heide LP, Kamal A, Artola A, Gispen WH, Ramakers GMJ. Insulin modulates hippocampal activity-dependent synaptic plasticity in a N-methyl-d-aspartate receptor and phosphatidyl-inositol-3-kinase-dependent manner. J Neurochem. 2005;94:1158–66.PubMed Van Der Heide LP, Kamal A, Artola A, Gispen WH, Ramakers GMJ. Insulin modulates hippocampal activity-dependent synaptic plasticity in a N-methyl-d-aspartate receptor and phosphatidyl-inositol-3-kinase-dependent manner. J Neurochem. 2005;94:1158–66.PubMed
92.
Zurück zum Zitat De Felice FG. Alzheimer’s disease and insulin resistance: translating basic science into clinical applications. J Clin Invest. 2013;123:531–9.PubMedPubMedCentral De Felice FG. Alzheimer’s disease and insulin resistance: translating basic science into clinical applications. J Clin Invest. 2013;123:531–9.PubMedPubMedCentral
93.
Zurück zum Zitat Goh JJ, Manahan-Vaughan D. Role of inhibitory autophosphorylation of calcium/calmodulin-dependent kinase II (αCAMKII) in persistent (>24 h) hippocampal LTP and in LTD facilitated by novel object-place learning and recognition in mice. Behav Brain Res. 2015;285:79–88.PubMed Goh JJ, Manahan-Vaughan D. Role of inhibitory autophosphorylation of calcium/calmodulin-dependent kinase II (αCAMKII) in persistent (>24 h) hippocampal LTP and in LTD facilitated by novel object-place learning and recognition in mice. Behav Brain Res. 2015;285:79–88.PubMed
94.
Zurück zum Zitat Man H-Y, Lin JW, Ju WH, Ahmadian G, Liu L, Becker LE, et al. Regulation of AMPA receptor–mediated synaptic transmission by clathrin-dependent receptor internalization. Neuron. 2000;25:649–62.PubMed Man H-Y, Lin JW, Ju WH, Ahmadian G, Liu L, Becker LE, et al. Regulation of AMPA receptor–mediated synaptic transmission by clathrin-dependent receptor internalization. Neuron. 2000;25:649–62.PubMed
95.
Zurück zum Zitat Adzovic L, Domenici L. Insulin induces phosphorylation of the AMPA receptor subunit GluR1, reversed by ZIP, and over-expression of Protein Kinase M zeta, reversed by amyloid beta. J Neurochem. 2014;131:582–7.PubMed Adzovic L, Domenici L. Insulin induces phosphorylation of the AMPA receptor subunit GluR1, reversed by ZIP, and over-expression of Protein Kinase M zeta, reversed by amyloid beta. J Neurochem. 2014;131:582–7.PubMed
96.
Zurück zum Zitat Grillo CA, Piroli GG, Lawrence RC, Wrighten SA, Green AJ, Wilson SP, et al. Hippocampal insulin resistance impairs spatial learning and synaptic plasticity. Diabetes. 2015;64:3927–36.PubMedPubMedCentral Grillo CA, Piroli GG, Lawrence RC, Wrighten SA, Green AJ, Wilson SP, et al. Hippocampal insulin resistance impairs spatial learning and synaptic plasticity. Diabetes. 2015;64:3927–36.PubMedPubMedCentral
97.
Zurück zum Zitat Skeberdis VA, Lan JY, Zheng X, Zukin RS, Bennett MVL. Insulin promotes rapid delivery of N-methyl-D-aspartate receptors to the cell surface by exocytosis. Proc Natl Acad Sci U S A. 2001;98:3561–6.PubMedPubMedCentral Skeberdis VA, Lan JY, Zheng X, Zukin RS, Bennett MVL. Insulin promotes rapid delivery of N-methyl-D-aspartate receptors to the cell surface by exocytosis. Proc Natl Acad Sci U S A. 2001;98:3561–6.PubMedPubMedCentral
98.
Zurück zum Zitat Christie JM, Wenthold RJ, Monaghan DT. Insulin causes a transient tyrosine phosphorylation of NR2A and NR2B NMDA receptor subunits in rat hippocampus. J Neurochem. 1999;72:1523–8.PubMed Christie JM, Wenthold RJ, Monaghan DT. Insulin causes a transient tyrosine phosphorylation of NR2A and NR2B NMDA receptor subunits in rat hippocampus. J Neurochem. 1999;72:1523–8.PubMed
99.
Zurück zum Zitat Chiu S-L, Chen C-M, Cline HT. Insulin receptor signaling regulates synapse number, dendritic plasticity, and circuit function in vivo. Neuron. 2008;58:708–19.PubMedPubMedCentral Chiu S-L, Chen C-M, Cline HT. Insulin receptor signaling regulates synapse number, dendritic plasticity, and circuit function in vivo. Neuron. 2008;58:708–19.PubMedPubMedCentral
100.
Zurück zum Zitat Ashrafi G, Wu Z, Farrell RJ, Ryan TA. GLUT4 Mobilization supports energetic demands of active synapses. Neuron. 2017;93(606–15):e3. Ashrafi G, Wu Z, Farrell RJ, Ryan TA. GLUT4 Mobilization supports energetic demands of active synapses. Neuron. 2017;93(606–15):e3.
101.
Zurück zum Zitat Pearson-Leary J, Jahagirdar V, Sage J, McNay EC. Insulin modulates hippocampally-mediated spatial working memory via glucose transporter-4. Behav Brain Res. 2018;338:32–9.PubMed Pearson-Leary J, Jahagirdar V, Sage J, McNay EC. Insulin modulates hippocampally-mediated spatial working memory via glucose transporter-4. Behav Brain Res. 2018;338:32–9.PubMed
102.
Zurück zum Zitat Cunnane SC, Trushina E, Morland C, Prigione A, Casadesus G, Andrews ZB, et al. Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing. Nat Rev Drug Discov. 2020;19:609–33.PubMedPubMedCentral Cunnane SC, Trushina E, Morland C, Prigione A, Casadesus G, Andrews ZB, et al. Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing. Nat Rev Drug Discov. 2020;19:609–33.PubMedPubMedCentral
103.
Zurück zum Zitat Frazier HN, Ghoweri AO, Anderson KL, Lin RL, Porter NM, Thibault O. Broadening the definition of brain insulin resistance in aging and Alzheimer’s disease. Exp Neurol. 2019;313:79–87.PubMed Frazier HN, Ghoweri AO, Anderson KL, Lin RL, Porter NM, Thibault O. Broadening the definition of brain insulin resistance in aging and Alzheimer’s disease. Exp Neurol. 2019;313:79–87.PubMed
104.
105.
Zurück zum Zitat McNay EC, Fries TM, Gold PE. Decreases in rat extracellular hippocampal glucose concentration associated with cognitive demand during a spatial task. Proc Natl Acad Sci U S A. 2000;97:2881–5.PubMedPubMedCentral McNay EC, Fries TM, Gold PE. Decreases in rat extracellular hippocampal glucose concentration associated with cognitive demand during a spatial task. Proc Natl Acad Sci U S A. 2000;97:2881–5.PubMedPubMedCentral
106.
Zurück zum Zitat Bingham EM, Hopkins D, Smith D, Pernet A, Hallett W, Reed L, et al. The role of insulin in human brain glucose metabolism: an 18Fluoro-deoxyglucose positron emission tomography study. Diabetes. 2002;51:3384–90.PubMed Bingham EM, Hopkins D, Smith D, Pernet A, Hallett W, Reed L, et al. The role of insulin in human brain glucose metabolism: an 18Fluoro-deoxyglucose positron emission tomography study. Diabetes. 2002;51:3384–90.PubMed
107.
Zurück zum Zitat Duelli R, Kuschinsky W. Brain glucose transporters: Relationship to local energy demand. Physiology. 2001;16:71–6. Duelli R, Kuschinsky W. Brain glucose transporters: Relationship to local energy demand. Physiology. 2001;16:71–6.
108.
Zurück zum Zitat Ferreira JM, Burnett AL, Rameau GA. Activity-dependent regulation of surface glucose transporter-3. J Neurosci. 2011;31:1991–9.PubMedPubMedCentral Ferreira JM, Burnett AL, Rameau GA. Activity-dependent regulation of surface glucose transporter-3. J Neurosci. 2011;31:1991–9.PubMedPubMedCentral
109.
Zurück zum Zitat Frazier HN, Ghoweri AO, Anderson KL, Lin R-L, Popa GJ, Mendenhall MD, et al. Elevating insulin signaling using a constitutively active insulin receptor increases glucose metabolism and expression of GLUT3 in hippocampal neurons. Front Neurosci. 2020;14:668.PubMedPubMedCentral Frazier HN, Ghoweri AO, Anderson KL, Lin R-L, Popa GJ, Mendenhall MD, et al. Elevating insulin signaling using a constitutively active insulin receptor increases glucose metabolism and expression of GLUT3 in hippocampal neurons. Front Neurosci. 2020;14:668.PubMedPubMedCentral
110.
Zurück zum Zitat Garwood CJ, Ratcliffe LE, Morgan SV, Simpson JE, Owens H, Vazquez-Villaseñor I, et al. Insulin and IGF1 signalling pathways in human astrocytes in vitro and in vivo; characterisation, subcellular localisation and modulation of the receptors. Mol Brain. 2015;8:51.PubMedPubMedCentral Garwood CJ, Ratcliffe LE, Morgan SV, Simpson JE, Owens H, Vazquez-Villaseñor I, et al. Insulin and IGF1 signalling pathways in human astrocytes in vitro and in vivo; characterisation, subcellular localisation and modulation of the receptors. Mol Brain. 2015;8:51.PubMedPubMedCentral
111.
Zurück zum Zitat Novak V, Milberg W, Hao Y, Munshi M, Novak P, Galica A, et al. Enhancement of vasoreactivity and cognition by intranasal insulin in type 2 diabetes. Diabetes Care. 2014;37:751–9.PubMedPubMedCentral Novak V, Milberg W, Hao Y, Munshi M, Novak P, Galica A, et al. Enhancement of vasoreactivity and cognition by intranasal insulin in type 2 diabetes. Diabetes Care. 2014;37:751–9.PubMedPubMedCentral
112.
Zurück zum Zitat Jauch-Chara K, Friedrich A, Rezmer M, Melchert UH, Scholand-Engler GH, Hallschmid M, et al. Intranasal insulin suppresses food intake via enhancement of brain energy levels in humans. Diabetes. 2012;61:2261–8.PubMedPubMedCentral Jauch-Chara K, Friedrich A, Rezmer M, Melchert UH, Scholand-Engler GH, Hallschmid M, et al. Intranasal insulin suppresses food intake via enhancement of brain energy levels in humans. Diabetes. 2012;61:2261–8.PubMedPubMedCentral
113.
Zurück zum Zitat Zhang H, Hao Y, Manor B, Novak P, Milberg W, Zhang J, et al. Intranasal insulin enhanced resting-state functional connectivity of hippocampal regions in type 2 diabetes. Diabetes. 2015;64:1025–34.PubMed Zhang H, Hao Y, Manor B, Novak P, Milberg W, Zhang J, et al. Intranasal insulin enhanced resting-state functional connectivity of hippocampal regions in type 2 diabetes. Diabetes. 2015;64:1025–34.PubMed
114.
Zurück zum Zitat Thienel M, Wilhelm I, Benedict C, Born J, Hallschmid M. Intranasal insulin decreases circulating cortisol concentrations during early sleep in elderly humans. Neurobiol Aging. 2017;54:170–4.PubMed Thienel M, Wilhelm I, Benedict C, Born J, Hallschmid M. Intranasal insulin decreases circulating cortisol concentrations during early sleep in elderly humans. Neurobiol Aging. 2017;54:170–4.PubMed
115.
Zurück zum Zitat Bohringer A, Schwabe L, Richter S, Schachinger H. Intranasal insulin attenuates the hypothalamic-pituitary-adrenal axis response to psychosocial stress. Psychoneuroendocrinology. 2008;33:1394–400.PubMed Bohringer A, Schwabe L, Richter S, Schachinger H. Intranasal insulin attenuates the hypothalamic-pituitary-adrenal axis response to psychosocial stress. Psychoneuroendocrinology. 2008;33:1394–400.PubMed
116.
Zurück zum Zitat de Kloet ERR, Meijer OCC, de Nicola AFF, de Rijk RHH, Joëls M. Importance of the brain corticosteroid receptor balance in metaplasticity, cognitive performance and neuro-inflammation. Front Neuroendocrinol. 2018;49:124–45.PubMed de Kloet ERR, Meijer OCC, de Nicola AFF, de Rijk RHH, Joëls M. Importance of the brain corticosteroid receptor balance in metaplasticity, cognitive performance and neuro-inflammation. Front Neuroendocrinol. 2018;49:124–45.PubMed
117.
Zurück zum Zitat de Leon MJ, McRae T, Rusinek H, Convit A, De Santi S, Tarshish C, et al. Cortisol reduces hippocampal glucose metabolism in normal elderly, but not in Alzheimer’s disease. J Clin Endocrinol Metab. 1997;82:3251–9.PubMed de Leon MJ, McRae T, Rusinek H, Convit A, De Santi S, Tarshish C, et al. Cortisol reduces hippocampal glucose metabolism in normal elderly, but not in Alzheimer’s disease. J Clin Endocrinol Metab. 1997;82:3251–9.PubMed
118.
Zurück zum Zitat McEwen BS. The neurobiology of stress: from serendipity to clinical relevance. Brain Res. 2000;886:172–89.PubMed McEwen BS. The neurobiology of stress: from serendipity to clinical relevance. Brain Res. 2000;886:172–89.PubMed
119.
Zurück zum Zitat Incollingo Rodriguez AC, Epel ES, White ML, Standen EC, Seckl JR, Tomiyama AJ. Hypothalamic-pituitary-adrenal axis dysregulation and cortisol activity in obesity: a systematic review. Psychoneuroendocrinology. 2015;62:301–18.PubMed Incollingo Rodriguez AC, Epel ES, White ML, Standen EC, Seckl JR, Tomiyama AJ. Hypothalamic-pituitary-adrenal axis dysregulation and cortisol activity in obesity: a systematic review. Psychoneuroendocrinology. 2015;62:301–18.PubMed
120.
Zurück zum Zitat Popp J, Wolfsgruber S, Heuser I, Peters O, Hüll M, Schröder J, et al. Cerebrospinal fluid cortisol and clinical disease progression in MCI and dementia of Alzheimer’s type. Neurobiol Aging. 2015;36:601–7.PubMed Popp J, Wolfsgruber S, Heuser I, Peters O, Hüll M, Schröder J, et al. Cerebrospinal fluid cortisol and clinical disease progression in MCI and dementia of Alzheimer’s type. Neurobiol Aging. 2015;36:601–7.PubMed
121.
Zurück zum Zitat Klinzing JG, Niethard N, Born J. Mechanisms of systems memory consolidation during sleep. Nat Neurosci. 2019;22:1598–610.PubMed Klinzing JG, Niethard N, Born J. Mechanisms of systems memory consolidation during sleep. Nat Neurosci. 2019;22:1598–610.PubMed
122.
Zurück zum Zitat Cedernaes J, Osorio RS, Varga AW, Kam K, Schiöth HB, Benedict C. Candidate mechanisms underlying the association between sleep-wake disruptions and Alzheimer’s disease. Sleep Med Rev. 2017;31:102–11.PubMed Cedernaes J, Osorio RS, Varga AW, Kam K, Schiöth HB, Benedict C. Candidate mechanisms underlying the association between sleep-wake disruptions and Alzheimer’s disease. Sleep Med Rev. 2017;31:102–11.PubMed
123.
Zurück zum Zitat Kern W, Offenheuser S, Born J, Fehm HL. Entrainment of ultradian oscillations in the secretion of insulin and glucagon to the nonrapid eye movement/rapid eye movement sleep rhythm in humans. J Clin Endocrinol Metab. 1996;81:1541–7.PubMed Kern W, Offenheuser S, Born J, Fehm HL. Entrainment of ultradian oscillations in the secretion of insulin and glucagon to the nonrapid eye movement/rapid eye movement sleep rhythm in humans. J Clin Endocrinol Metab. 1996;81:1541–7.PubMed
124.
Zurück zum Zitat Sangiah S, Caldwell DF, Villeneuve MJ, Clancy JJ. Sleep: sequential reduction of paradoxical (REM) and elevation of slow-wave (NREM) sleep by a non-convulsive dose of insulin in rats. Life Sci. 1982;31:763–9.PubMed Sangiah S, Caldwell DF, Villeneuve MJ, Clancy JJ. Sleep: sequential reduction of paradoxical (REM) and elevation of slow-wave (NREM) sleep by a non-convulsive dose of insulin in rats. Life Sci. 1982;31:763–9.PubMed
125.
Zurück zum Zitat Hardt O, Nader K, Nadel L. Decay happens: the role of active forgetting in memory. Trends Cogn Sci. 2013;17:111–20.PubMed Hardt O, Nader K, Nadel L. Decay happens: the role of active forgetting in memory. Trends Cogn Sci. 2013;17:111–20.PubMed
126.
Zurück zum Zitat Fultz NE, Bonmassar G, Setsompop K, Stickgold RA, Rosen BR, Polimeni JR, et al. Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science. 2019;366:628–31.PubMedPubMedCentral Fultz NE, Bonmassar G, Setsompop K, Stickgold RA, Rosen BR, Polimeni JR, et al. Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science. 2019;366:628–31.PubMedPubMedCentral
127.
Zurück zum Zitat Lucey BP, McCullough A, Landsness EC, Toedebusch CD, McLeland JS, Zaza AM, et al. Reduced non–rapid eye movement sleep is associated with tau pathology in early Alzheimer’s disease. Sci Transl Med. 2019;11:eaau6550.PubMedPubMedCentral Lucey BP, McCullough A, Landsness EC, Toedebusch CD, McLeland JS, Zaza AM, et al. Reduced non–rapid eye movement sleep is associated with tau pathology in early Alzheimer’s disease. Sci Transl Med. 2019;11:eaau6550.PubMedPubMedCentral
128.
Zurück zum Zitat Ritze Y, Kern W, Ebner E-M, Jahn S, Benedict C, Hallschmid M. Metabolic and cognitive outcomes of subchronic once-daily intranasal insulin administration in healthy men. Front Endocrinol (Lausanne). 2018;9:663.PubMedPubMedCentral Ritze Y, Kern W, Ebner E-M, Jahn S, Benedict C, Hallschmid M. Metabolic and cognitive outcomes of subchronic once-daily intranasal insulin administration in healthy men. Front Endocrinol (Lausanne). 2018;9:663.PubMedPubMedCentral
129.
Zurück zum Zitat Benedict C, Grillo CA. Insulin resistance as a therapeutic target in the treatment of Alzheimer’s disease: a state-of-the-art review. Front Neurosci. 2018;12:215.PubMedPubMedCentral Benedict C, Grillo CA. Insulin resistance as a therapeutic target in the treatment of Alzheimer’s disease: a state-of-the-art review. Front Neurosci. 2018;12:215.PubMedPubMedCentral
130.
Zurück zum Zitat de Silva LNM, Gonçalves RA, Boehnke SE, Forny-Germano L, Munoz DP, De Felice FG. Understanding the link between insulin resistance and Alzheimer’s disease: insights from animal models. Exp Neurol. 2019;316:1–11. de Silva LNM, Gonçalves RA, Boehnke SE, Forny-Germano L, Munoz DP, De Felice FG. Understanding the link between insulin resistance and Alzheimer’s disease: insights from animal models. Exp Neurol. 2019;316:1–11.
131.
Zurück zum Zitat Stanley M, Macauley SL, Holtzman DM. Changes in insulin and insulin signaling in Alzheimer’s disease: cause or consequence? J Exp Med. 2016;213:1375–85.PubMedPubMedCentral Stanley M, Macauley SL, Holtzman DM. Changes in insulin and insulin signaling in Alzheimer’s disease: cause or consequence? J Exp Med. 2016;213:1375–85.PubMedPubMedCentral
132.
Zurück zum Zitat Avgerinos KI, Kalaitzidis G, Malli A, Kalaitzoglou D, Myserlis PG, Lioutas V-A. Intranasal insulin in Alzheimer’s dementia or mild cognitive impairment: a systematic review. J Neurol. 2018;265:1497–510.PubMedPubMedCentral Avgerinos KI, Kalaitzidis G, Malli A, Kalaitzoglou D, Myserlis PG, Lioutas V-A. Intranasal insulin in Alzheimer’s dementia or mild cognitive impairment: a systematic review. J Neurol. 2018;265:1497–510.PubMedPubMedCentral
133.
Zurück zum Zitat Craft S, Asthana S, Newcomer JW, Wilkinson CW, Tio Matos I, Baker LD, et al. Enhancement of memory in Alzheimer disease with insulin and somatostatin, but not glucose. Arch Gen Psychiatry. 1999;56:1135–40.PubMed Craft S, Asthana S, Newcomer JW, Wilkinson CW, Tio Matos I, Baker LD, et al. Enhancement of memory in Alzheimer disease with insulin and somatostatin, but not glucose. Arch Gen Psychiatry. 1999;56:1135–40.PubMed
134.
Zurück zum Zitat Reger MA, Watson GS, Frey WH, Baker LD, Cholerton B, Keeling ML, et al. Effects of intranasal insulin on cognition in memory-impaired older adults: modulation by APOE genotype. Neurobiol Aging. 2006;27:451–8.PubMed Reger MA, Watson GS, Frey WH, Baker LD, Cholerton B, Keeling ML, et al. Effects of intranasal insulin on cognition in memory-impaired older adults: modulation by APOE genotype. Neurobiol Aging. 2006;27:451–8.PubMed
135.
Zurück zum Zitat Reger MA, Watson GS, Green PS, Baker LD, Cholerton B, Fishel MA, et al. Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-β in memory-impaired older adults. J Alzheimers Dis. 2008;13:323–31.PubMedPubMedCentral Reger MA, Watson GS, Green PS, Baker LD, Cholerton B, Fishel MA, et al. Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-β in memory-impaired older adults. J Alzheimers Dis. 2008;13:323–31.PubMedPubMedCentral
136.
Zurück zum Zitat Reger MA, Watson GS, Green PS, Wilkinson CW, Baker LD, Cholerton B, et al. Intranasal insulin improves cognition and modulates β-amyloid in early AD. Neurology. 2008;70:440–8.PubMed Reger MA, Watson GS, Green PS, Wilkinson CW, Baker LD, Cholerton B, et al. Intranasal insulin improves cognition and modulates β-amyloid in early AD. Neurology. 2008;70:440–8.PubMed
138.
Zurück zum Zitat Claxton A, Baker LD, Hanson A, Trittschuh EH, Cholerton B, Morgan A, et al. Long-acting intranasal insulin detemir improves cognition for adults with mild cognitive impairment or early-stage Alzheimer’s Disease dementia. J Alzheimers Dis. 2015;44:897–906.PubMed Claxton A, Baker LD, Hanson A, Trittschuh EH, Cholerton B, Morgan A, et al. Long-acting intranasal insulin detemir improves cognition for adults with mild cognitive impairment or early-stage Alzheimer’s Disease dementia. J Alzheimers Dis. 2015;44:897–906.PubMed
139.
Zurück zum Zitat Craft S, Claxton A, Baker LD, Hanson AJ, Cholerton B, Trittschuh EH, et al. Effects of regular and long-acting insulin on cognition and Alzheimer’s disease biomarkers: a pilot clinical trial. J Alzheimers Dis. 2017;57:1325–34.PubMedPubMedCentral Craft S, Claxton A, Baker LD, Hanson AJ, Cholerton B, Trittschuh EH, et al. Effects of regular and long-acting insulin on cognition and Alzheimer’s disease biomarkers: a pilot clinical trial. J Alzheimers Dis. 2017;57:1325–34.PubMedPubMedCentral
140.
Zurück zum Zitat Hallschmid M, Jauch-Chara K, Korn O, Mölle M, Rasch B, Born J, et al. Euglycemic infusion of insulin detemir compared with human insulin appears to increase direct current brain potential response and reduces food intake while inducing similar systemic effects. Diabetes. 2010;59:1101–7.PubMedPubMedCentral Hallschmid M, Jauch-Chara K, Korn O, Mölle M, Rasch B, Born J, et al. Euglycemic infusion of insulin detemir compared with human insulin appears to increase direct current brain potential response and reduces food intake while inducing similar systemic effects. Diabetes. 2010;59:1101–7.PubMedPubMedCentral
141.
Zurück zum Zitat Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry. 1984;141:1356–64.PubMed Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzheimer’s disease. Am J Psychiatry. 1984;141:1356–64.PubMed
142.
Zurück zum Zitat Clark CM, Ewbank DC. Performance of the Dementia Severity Rating Scale: a caregiver questionnaire for rating severity in Alzheimer disease. Alzheimer Dis Assoc Disord. 1996;10:31–9.PubMed Clark CM, Ewbank DC. Performance of the Dementia Severity Rating Scale: a caregiver questionnaire for rating severity in Alzheimer disease. Alzheimer Dis Assoc Disord. 1996;10:31–9.PubMed
143.
Zurück zum Zitat Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS, Claxton A, et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol. 2012;69:29–38.PubMed Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS, Claxton A, et al. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol. 2012;69:29–38.PubMed
144.
Zurück zum Zitat Fishel MA, Watson GS, Montine TJ, Wang Q, Green PS, Kulstad JJ, et al. Hyperinsulinemia provokes synchronous increases in central inflammation and β-amyloid in normal adults. Arch Neurol. 2005;62:1539–44.PubMed Fishel MA, Watson GS, Montine TJ, Wang Q, Green PS, Kulstad JJ, et al. Hyperinsulinemia provokes synchronous increases in central inflammation and β-amyloid in normal adults. Arch Neurol. 2005;62:1539–44.PubMed
145.
Zurück zum Zitat Craft S, Raman R, Chow TW, Rafii MS, Sun C-K, Rissman RA, et al. Safety, efficacy, and feasibility of intranasal insulin for the treatment of mild cognitive impairment and Alzheimer disease dementia. JAMA Neurol. 2020;77:1099–109.PubMed Craft S, Raman R, Chow TW, Rafii MS, Sun C-K, Rissman RA, et al. Safety, efficacy, and feasibility of intranasal insulin for the treatment of mild cognitive impairment and Alzheimer disease dementia. JAMA Neurol. 2020;77:1099–109.PubMed
146.
Zurück zum Zitat Galasko D, Bennett D, Sano M, Ernesto C, Thomas R, Grundman M, et al. An inventory to assess activities of daily living for clinical trials in Alzheimer’s disease. Alzheimer Dis Assoc Disord. 1997;11:33–9. Galasko D, Bennett D, Sano M, Ernesto C, Thomas R, Grundman M, et al. An inventory to assess activities of daily living for clinical trials in Alzheimer’s disease. Alzheimer Dis Assoc Disord. 1997;11:33–9.
147.
Zurück zum Zitat Hoyer S, Muller D, Plaschke K. Desensitization of brain insulin receptor: effect on glucose/energy and related metabolism. J Neural Transm Suppl. 1994;44:259–68.PubMed Hoyer S, Muller D, Plaschke K. Desensitization of brain insulin receptor: effect on glucose/energy and related metabolism. J Neural Transm Suppl. 1994;44:259–68.PubMed
148.
Zurück zum Zitat van Harten B, de Leeuw F-E, Weinstein HC, Scheltens P, Biessels GJ. Brain imaging in patients with diabetes: a systematic review. Diabetes Care. 2006;29:2539–48.PubMed van Harten B, de Leeuw F-E, Weinstein HC, Scheltens P, Biessels GJ. Brain imaging in patients with diabetes: a systematic review. Diabetes Care. 2006;29:2539–48.PubMed
149.
Zurück zum Zitat Sasaki N, Fukatsu R, Tsuzuki K, Hayashi Y, Yoshida T, Fujii N, et al. Advanced glycation end products in Alzheimer’s disease and other neurodegenerative diseases. Am J Pathol. 1998;153:1149–55.PubMedPubMedCentral Sasaki N, Fukatsu R, Tsuzuki K, Hayashi Y, Yoshida T, Fujii N, et al. Advanced glycation end products in Alzheimer’s disease and other neurodegenerative diseases. Am J Pathol. 1998;153:1149–55.PubMedPubMedCentral
150.
Zurück zum Zitat Raffaitin C, Feart C, Le Goff M, Amieva H, Helmer C, Akbaraly TN, et al. Metabolic syndrome and cognitive decline in French elders: the Three-City Study. Neurology. 2011;76:518–25.PubMed Raffaitin C, Feart C, Le Goff M, Amieva H, Helmer C, Akbaraly TN, et al. Metabolic syndrome and cognitive decline in French elders: the Three-City Study. Neurology. 2011;76:518–25.PubMed
151.
Zurück zum Zitat Cukierman T, Gerstein HC, Williamson JD. Cognitive decline and dementia in diabetes: systematic overview of prospective observational studies. Diabetologia. 2005;48:2460–9.PubMed Cukierman T, Gerstein HC, Williamson JD. Cognitive decline and dementia in diabetes: systematic overview of prospective observational studies. Diabetologia. 2005;48:2460–9.PubMed
152.
153.
Zurück zum Zitat Assuncao N, Sudo FK, Drummond C, de Felice FG, Mattos P. Metabolic syndrome and cognitive decline in the elderly: a systematic review. PLoS ONE. 2018;13:e0194990.PubMedPubMedCentral Assuncao N, Sudo FK, Drummond C, de Felice FG, Mattos P. Metabolic syndrome and cognitive decline in the elderly: a systematic review. PLoS ONE. 2018;13:e0194990.PubMedPubMedCentral
154.
Zurück zum Zitat Punthakee Z, Miller ME, Launer LJ, Williamson JD, Lazar RM, Cukierman-Yaffee T, et al. Poor cognitive function and risk of severe hypoglycemia in type 2 diabetes: post hoc epidemiologic analysis of the ACCORD trial. Diabetes Care. 2012;35:787–93.PubMedPubMedCentral Punthakee Z, Miller ME, Launer LJ, Williamson JD, Lazar RM, Cukierman-Yaffee T, et al. Poor cognitive function and risk of severe hypoglycemia in type 2 diabetes: post hoc epidemiologic analysis of the ACCORD trial. Diabetes Care. 2012;35:787–93.PubMedPubMedCentral
155.
Zurück zum Zitat Galindo-Mendez B, Trevino JA, McGlinchey R, Fortier C, Lioutas V, Novak P, et al. Memory advancement by intranasal insulin in type 2 diabetes (MemAID) randomized controlled clinical trial: design, methods and rationale. Contemp Clin Trials. 2020;89:105934.PubMedPubMedCentral Galindo-Mendez B, Trevino JA, McGlinchey R, Fortier C, Lioutas V, Novak P, et al. Memory advancement by intranasal insulin in type 2 diabetes (MemAID) randomized controlled clinical trial: design, methods and rationale. Contemp Clin Trials. 2020;89:105934.PubMedPubMedCentral
156.
Zurück zum Zitat Frosch OH, Yau PL, Osorio RS, Rusinek H, Storey P, Convit A. Insulin resistance among obese middle-aged is associated with decreased cerebrovascular reactivity. Neurology. 2017;89:249–55.PubMedPubMedCentral Frosch OH, Yau PL, Osorio RS, Rusinek H, Storey P, Convit A. Insulin resistance among obese middle-aged is associated with decreased cerebrovascular reactivity. Neurology. 2017;89:249–55.PubMedPubMedCentral
157.
Zurück zum Zitat Katz A, Nambi SS, Mather K, Baron AD, Follmann DA, Sullivan G, et al. Quantitative Insulin Sensitivity Check Index: a simple, accurate method for assessing insulin sensitivity in humans. J Clin Endocrinol Metab. 2000;85:2402–10.PubMed Katz A, Nambi SS, Mather K, Baron AD, Follmann DA, Sullivan G, et al. Quantitative Insulin Sensitivity Check Index: a simple, accurate method for assessing insulin sensitivity in humans. J Clin Endocrinol Metab. 2000;85:2402–10.PubMed
158.
Zurück zum Zitat Benedict C, Brooks SJ, Kullberg J, Burgos J, Kempton MJ, Nordenskjold R, et al. Impaired insulin sensitivity as indexed by the HOMA score is associated with deficits in verbal fluency and temporal lobe gray matter volume in the elderly. Diabetes Care. 2012;35:488–94.PubMedPubMedCentral Benedict C, Brooks SJ, Kullberg J, Burgos J, Kempton MJ, Nordenskjold R, et al. Impaired insulin sensitivity as indexed by the HOMA score is associated with deficits in verbal fluency and temporal lobe gray matter volume in the elderly. Diabetes Care. 2012;35:488–94.PubMedPubMedCentral
159.
Zurück zum Zitat Convit A, Wolf OT, Tarshish C, de Leon MJ. Reduced glucose tolerance is associated with poor memory performance and hippocampal atrophy among normal elderly. Proc Natl Acad Sci U S A. 2003;100:2019–22.PubMedPubMedCentral Convit A, Wolf OT, Tarshish C, de Leon MJ. Reduced glucose tolerance is associated with poor memory performance and hippocampal atrophy among normal elderly. Proc Natl Acad Sci U S A. 2003;100:2019–22.PubMedPubMedCentral
160.
Zurück zum Zitat Yarchoan M, Toledo JB, Lee EB, Arvanitakis Z, Kazi H, Han L-Y, et al. Abnormal serine phosphorylation of insulin receptor substrate 1 is associated with tau pathology in Alzheimer’s disease and tauopathies. Acta Neuropathol. 2014;128:679–89.PubMedPubMedCentral Yarchoan M, Toledo JB, Lee EB, Arvanitakis Z, Kazi H, Han L-Y, et al. Abnormal serine phosphorylation of insulin receptor substrate 1 is associated with tau pathology in Alzheimer’s disease and tauopathies. Acta Neuropathol. 2014;128:679–89.PubMedPubMedCentral
161.
Zurück zum Zitat Laws SM, Gaskin S, Woodfield A, Srikanth V, Bruce D, Fraser PE, et al. Insulin resistance is associated with reductions in specific cognitive domains and increases in CSF tau in cognitively normal adults. Sci Rep. 2017;7:9766.PubMedPubMedCentral Laws SM, Gaskin S, Woodfield A, Srikanth V, Bruce D, Fraser PE, et al. Insulin resistance is associated with reductions in specific cognitive domains and increases in CSF tau in cognitively normal adults. Sci Rep. 2017;7:9766.PubMedPubMedCentral
162.
Zurück zum Zitat Ekblad LL, Johansson J, Helin S, Viitanen M, Laine H, Puukka P, et al. Midlife insulin resistance, APOE genotype, and late-life brain amyloid accumulation. Neurology. 2018;90:e1150–7.PubMedPubMedCentral Ekblad LL, Johansson J, Helin S, Viitanen M, Laine H, Puukka P, et al. Midlife insulin resistance, APOE genotype, and late-life brain amyloid accumulation. Neurology. 2018;90:e1150–7.PubMedPubMedCentral
163.
Zurück zum Zitat Willette AA, Johnson SC, Birdsill AC, Sager MA, Christian B, Baker LD, et al. Insulin resistance predicts brain amyloid deposition in late middle-aged adults. Alzheimers Dement. 2015;11(504–10):e1. Willette AA, Johnson SC, Birdsill AC, Sager MA, Christian B, Baker LD, et al. Insulin resistance predicts brain amyloid deposition in late middle-aged adults. Alzheimers Dement. 2015;11(504–10):e1.
164.
Zurück zum Zitat Kapogiannis D, Mustapic M, Shardell MD, Berkowitz ST, Diehl TC, Spangler RD, et al. Association of extracellular vesicle biomarkers with Alzheimer disease in the Baltimore Longitudinal Study of Aging. JAMA Neurol. 2019;76:1340–51.PubMedCentralPubMed Kapogiannis D, Mustapic M, Shardell MD, Berkowitz ST, Diehl TC, Spangler RD, et al. Association of extracellular vesicle biomarkers with Alzheimer disease in the Baltimore Longitudinal Study of Aging. JAMA Neurol. 2019;76:1340–51.PubMedCentralPubMed
165.
Zurück zum Zitat Thambisetty M, Metter EJ, Yang A, Dolan H, Marano C, Zonderman AB, et al. Glucose intolerance, insulin resistance, and pathological features of Alzheimer disease in the Baltimore Longitudinal Study of Aging. JAMA Neurol. 2013;70:1167.PubMedPubMedCentral Thambisetty M, Metter EJ, Yang A, Dolan H, Marano C, Zonderman AB, et al. Glucose intolerance, insulin resistance, and pathological features of Alzheimer disease in the Baltimore Longitudinal Study of Aging. JAMA Neurol. 2013;70:1167.PubMedPubMedCentral
166.
Zurück zum Zitat Pekkala T, Hall A, Mangialasche F, Kemppainen N, Mecocci P, Ngandu T, et al. Association of peripheral insulin resistance and other markers of type 2 diabetes mellitus with brain amyloid deposition in healthy individuals at risk of dementia. J Alzheimers Dis. 2020;76:1243–8.PubMedPubMedCentral Pekkala T, Hall A, Mangialasche F, Kemppainen N, Mecocci P, Ngandu T, et al. Association of peripheral insulin resistance and other markers of type 2 diabetes mellitus with brain amyloid deposition in healthy individuals at risk of dementia. J Alzheimers Dis. 2020;76:1243–8.PubMedPubMedCentral
167.
Zurück zum Zitat Rivera EJ, Goldin A, Fulmer N, Tavares R, Wands JR, de la Monte SM. Insulin and insulin-like growth factor expression and function deteriorate with progression of Alzheimer’s disease: link to brain reductions in acetylcholine. J Alzheimers Dis. 2005;8:247–68.PubMed Rivera EJ, Goldin A, Fulmer N, Tavares R, Wands JR, de la Monte SM. Insulin and insulin-like growth factor expression and function deteriorate with progression of Alzheimer’s disease: link to brain reductions in acetylcholine. J Alzheimers Dis. 2005;8:247–68.PubMed
168.
Zurück zum Zitat Talbot K, Wang H-Y, Kazi H, Han L-Y, Bakshi KP, Stucky A, et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Invest. 2012;122:1316–38.PubMedPubMedCentral Talbot K, Wang H-Y, Kazi H, Han L-Y, Bakshi KP, Stucky A, et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Invest. 2012;122:1316–38.PubMedPubMedCentral
169.
Zurück zum Zitat Moloney AM, Griffin RJ, Timmons S, O’Connor R, Ravid R, O’Neill C. Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease indicate possible resistance to IGF-1 and insulin signalling. Neurobiol Aging. 2010;31:224–43.PubMed Moloney AM, Griffin RJ, Timmons S, O’Connor R, Ravid R, O’Neill C. Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease indicate possible resistance to IGF-1 and insulin signalling. Neurobiol Aging. 2010;31:224–43.PubMed
170.
Zurück zum Zitat Logan S, Pharaoh GA, Marlin MC, Masser DR, Matsuzaki S, Wronowski B, et al. Insulin-like growth factor receptor signaling regulates working memory, mitochondrial metabolism, and amyloid-β uptake in astrocytes. Mol Metab. 2018;9:141–55.PubMedPubMedCentral Logan S, Pharaoh GA, Marlin MC, Masser DR, Matsuzaki S, Wronowski B, et al. Insulin-like growth factor receptor signaling regulates working memory, mitochondrial metabolism, and amyloid-β uptake in astrocytes. Mol Metab. 2018;9:141–55.PubMedPubMedCentral
171.
Zurück zum Zitat Trudeau F, Gagnon S, Massicotte G. Hippocampal synaptic plasticity and glutamate receptor regulation: influences of diabetes mellitus. Eur J Pharmacol. 2004;490:177–86.PubMed Trudeau F, Gagnon S, Massicotte G. Hippocampal synaptic plasticity and glutamate receptor regulation: influences of diabetes mellitus. Eur J Pharmacol. 2004;490:177–86.PubMed
172.
Zurück zum Zitat Arvanitakis Z, Wang H, Capuano AW, Khan A, Taïb B, Anokye-Danso F, et al. Brain insulin signaling, Alzheimer disease pathology, and cognitive function. Ann Neurol. 2020;88:513–25.PubMedPubMedCentral Arvanitakis Z, Wang H, Capuano AW, Khan A, Taïb B, Anokye-Danso F, et al. Brain insulin signaling, Alzheimer disease pathology, and cognitive function. Ann Neurol. 2020;88:513–25.PubMedPubMedCentral
173.
Zurück zum Zitat Fujisawa Y, Sasaki K, Akiyama K. Increased insulin levels after OGTT load in peripheral blood and cerebrospinal fluid of patients with dementia of Alzheimer type. Biol Psychiatry. 1991;30:1219–28.PubMed Fujisawa Y, Sasaki K, Akiyama K. Increased insulin levels after OGTT load in peripheral blood and cerebrospinal fluid of patients with dementia of Alzheimer type. Biol Psychiatry. 1991;30:1219–28.PubMed
174.
Zurück zum Zitat Craft S, Peskind E, Schwartz MW, Schellenberg GD, Raskind M, Porte D. Cerebrospinal fluid and plasma insulin levels in Alzheimer’s disease: relationship to severity of dementia and apolipoprotein E genotype. Neurology. 1998;50:164–8.PubMed Craft S, Peskind E, Schwartz MW, Schellenberg GD, Raskind M, Porte D. Cerebrospinal fluid and plasma insulin levels in Alzheimer’s disease: relationship to severity of dementia and apolipoprotein E genotype. Neurology. 1998;50:164–8.PubMed
175.
Zurück zum Zitat Gil-Bea FJ, Solas M, Solomon A, Mugueta C, Winblad B, Kivipelto M, et al. Insulin levels are decreased in the cerebrospinal fluid of women with prodomal Alzheimer’s disease. J Alzheimers Dis. 2010;22:405–13.PubMed Gil-Bea FJ, Solas M, Solomon A, Mugueta C, Winblad B, Kivipelto M, et al. Insulin levels are decreased in the cerebrospinal fluid of women with prodomal Alzheimer’s disease. J Alzheimers Dis. 2010;22:405–13.PubMed
176.
Zurück zum Zitat Geijselaers SLC, Aalten P, Ramakers IHGB, De Deyn PP, Heijboer AC, Koek HL, et al. Association of cerebrospinal fluid (CSF) insulin with cognitive performance and CSF biomarkers of Alzheimer’s Disease. J Alzheimer’s Dis. 2017;61:309–20. Geijselaers SLC, Aalten P, Ramakers IHGB, De Deyn PP, Heijboer AC, Koek HL, et al. Association of cerebrospinal fluid (CSF) insulin with cognitive performance and CSF biomarkers of Alzheimer’s Disease. J Alzheimer’s Dis. 2017;61:309–20.
177.
Zurück zum Zitat Molina JA, Jimenez-Jimenez FJ, Vargas C, Gomez P, de Bustos F, Gomez-Escalonilla C, et al. Cerebrospinal fluid levels of insulin in patients with Alzheimer’s disease. Acta Neurol Scand. 2002;106:347–50.PubMed Molina JA, Jimenez-Jimenez FJ, Vargas C, Gomez P, de Bustos F, Gomez-Escalonilla C, et al. Cerebrospinal fluid levels of insulin in patients with Alzheimer’s disease. Acta Neurol Scand. 2002;106:347–50.PubMed
178.
Zurück zum Zitat Craft S. The role of metabolic disorders in Alzheimer disease and vascular dementia: two roads converged. Arch Neurol. 2009;66:300–5.PubMedPubMedCentral Craft S. The role of metabolic disorders in Alzheimer disease and vascular dementia: two roads converged. Arch Neurol. 2009;66:300–5.PubMedPubMedCentral
179.
Zurück zum Zitat De Felice FG, Vieira MNN, Bomfim TR, Decker H, Velasco PT, Lambert MP, et al. Protection of synapses against Alzheimer’s-linked toxins: Insulin signaling prevents the pathogenic binding of Aβ oligomers. Proc Natl Acad Sci USA. 2009;106:1971–6.PubMedPubMedCentral De Felice FG, Vieira MNN, Bomfim TR, Decker H, Velasco PT, Lambert MP, et al. Protection of synapses against Alzheimer’s-linked toxins: Insulin signaling prevents the pathogenic binding of Aβ oligomers. Proc Natl Acad Sci USA. 2009;106:1971–6.PubMedPubMedCentral
180.
Zurück zum Zitat Franklin W, Krishnan B, Taglialatela G. Chronic synaptic insulin resistance after traumatic brain injury abolishes insulin protection from amyloid beta and tau oligomer-induced synaptic dysfunction. Sci Rep. 2019;9:8228.PubMedPubMedCentral Franklin W, Krishnan B, Taglialatela G. Chronic synaptic insulin resistance after traumatic brain injury abolishes insulin protection from amyloid beta and tau oligomer-induced synaptic dysfunction. Sci Rep. 2019;9:8228.PubMedPubMedCentral
181.
Zurück zum Zitat Kwon OH, Cho YY, Kim TW, Chung S. O-GlcNAcylation of amyloid-β protein precursor by insulin signaling reduces amyloid-β production. J Alzheimers Dis. 2019;69:1195–211.PubMed Kwon OH, Cho YY, Kim TW, Chung S. O-GlcNAcylation of amyloid-β protein precursor by insulin signaling reduces amyloid-β production. J Alzheimers Dis. 2019;69:1195–211.PubMed
182.
Zurück zum Zitat Barone E, Tramutola A, Triani F, Calcagnini S, Di Domenico F, Ripoli C, et al. Biliverdin reductase-A mediates the beneficial effects of intranasal insulin in Alzheimer disease. Mol Neurobiol. 2019;56:2922–43.PubMed Barone E, Tramutola A, Triani F, Calcagnini S, Di Domenico F, Ripoli C, et al. Biliverdin reductase-A mediates the beneficial effects of intranasal insulin in Alzheimer disease. Mol Neurobiol. 2019;56:2922–43.PubMed
183.
Zurück zum Zitat Guo Z, Chen Y, Mao Y-F, Zheng T, Jiang Y, Yan Y, et al. Long-term treatment with intranasal insulin ameliorates cognitive impairment, tau hyperphosphorylation, and microglial activation in a streptozotocin-induced Alzheimer’s rat model. Sci Rep. 2017;7:45971.PubMedPubMedCentral Guo Z, Chen Y, Mao Y-F, Zheng T, Jiang Y, Yan Y, et al. Long-term treatment with intranasal insulin ameliorates cognitive impairment, tau hyperphosphorylation, and microglial activation in a streptozotocin-induced Alzheimer’s rat model. Sci Rep. 2017;7:45971.PubMedPubMedCentral
184.
Zurück zum Zitat Tschritter O, Preissl H, Hennige AM, Stumvoll M, Porubska K, Frost R, et al. The cerebrocortical response to hyperinsulinemia is reduced in overweight humans: a magnetoencephalographic study. Proc Natl Acad Sci U S A. 2006;103:12103–8.PubMedPubMedCentral Tschritter O, Preissl H, Hennige AM, Stumvoll M, Porubska K, Frost R, et al. The cerebrocortical response to hyperinsulinemia is reduced in overweight humans: a magnetoencephalographic study. Proc Natl Acad Sci U S A. 2006;103:12103–8.PubMedPubMedCentral
185.
Zurück zum Zitat Tschritter O, Preissl H, Yokoyama Y, Machicao F, Häring H-U, Fritsche A. Variation in the FTO gene locus is associated with cerebrocortical insulin resistance in humans. Diabetologia. 2007;50:2602–3.PubMed Tschritter O, Preissl H, Yokoyama Y, Machicao F, Häring H-U, Fritsche A. Variation in the FTO gene locus is associated with cerebrocortical insulin resistance in humans. Diabetologia. 2007;50:2602–3.PubMed
186.
Zurück zum Zitat Striepens N, Kendrick KM, Hanking V, Landgraf R, Wüllner U, Maier W, et al. Elevated cerebrospinal fluid and blood concentrations of oxytocin following its intranasal administration in humans. Sci Rep. 2013;3:3440.PubMedPubMedCentral Striepens N, Kendrick KM, Hanking V, Landgraf R, Wüllner U, Maier W, et al. Elevated cerebrospinal fluid and blood concentrations of oxytocin following its intranasal administration in humans. Sci Rep. 2013;3:3440.PubMedPubMedCentral
187.
Zurück zum Zitat Schwarz B, Merkel OM. Nose-to-brain delivery of biologics. Ther Deliv. 2019;10:207–10.PubMed Schwarz B, Merkel OM. Nose-to-brain delivery of biologics. Ther Deliv. 2019;10:207–10.PubMed
188.
Zurück zum Zitat Hallschmid M, Higgs S, Thienel M, Ott V, Lehnert H. Postprandial administration of intranasal insulin intensifies satiety and reduces intake of palatable snacks in women. Diabetes. 2012;61:782–9.PubMedPubMedCentral Hallschmid M, Higgs S, Thienel M, Ott V, Lehnert H. Postprandial administration of intranasal insulin intensifies satiety and reduces intake of palatable snacks in women. Diabetes. 2012;61:782–9.PubMedPubMedCentral
190.
Zurück zum Zitat Wingrove J, Swedrowska M, Scherließ R, Parry M, Ramjeeawon M, Taylor D, et al. Characterisation of nasal devices for delivery of insulin to the brain and evaluation in humans using functional magnetic resonance imaging. J Control Release. 2019;302:140–7.PubMed Wingrove J, Swedrowska M, Scherließ R, Parry M, Ramjeeawon M, Taylor D, et al. Characterisation of nasal devices for delivery of insulin to the brain and evaluation in humans using functional magnetic resonance imaging. J Control Release. 2019;302:140–7.PubMed
191.
Zurück zum Zitat Schubert CR, Cruickshanks KJ, Klein BEK, Klein R, Nondahl DM. Olfactory impairment in older adults: five-year incidence and risk factors. Laryngoscope. 2011;121:873–8.PubMedPubMedCentral Schubert CR, Cruickshanks KJ, Klein BEK, Klein R, Nondahl DM. Olfactory impairment in older adults: five-year incidence and risk factors. Laryngoscope. 2011;121:873–8.PubMedPubMedCentral
192.
Zurück zum Zitat Roberts RO, Christianson TJH, Kremers WK, Mielke MM, Machulda MM, Vassilaki M, et al. Association between olfactory dysfunction and amnestic mild cognitive impairment and Alzheimer disease dementia. JAMA Neurol. 2016;73:93.PubMedPubMedCentral Roberts RO, Christianson TJH, Kremers WK, Mielke MM, Machulda MM, Vassilaki M, et al. Association between olfactory dysfunction and amnestic mild cognitive impairment and Alzheimer disease dementia. JAMA Neurol. 2016;73:93.PubMedPubMedCentral
193.
Zurück zum Zitat Su Y, Sun B, Gao X, Dong X, Fu L, Zhang Y, et al. Intranasal delivery of targeted nanoparticles loaded with miR-132 to brain for the treatment of neurodegenerative diseases. Front Pharmacol. 2020;11:1165.PubMedPubMedCentral Su Y, Sun B, Gao X, Dong X, Fu L, Zhang Y, et al. Intranasal delivery of targeted nanoparticles loaded with miR-132 to brain for the treatment of neurodegenerative diseases. Front Pharmacol. 2020;11:1165.PubMedPubMedCentral
194.
Zurück zum Zitat Kamei N, Takeda-Morishita M. Brain delivery of insulin boosted by intranasal coadministration with cell-penetrating peptides. J Control Release. 2015;197:105–10.PubMed Kamei N, Takeda-Morishita M. Brain delivery of insulin boosted by intranasal coadministration with cell-penetrating peptides. J Control Release. 2015;197:105–10.PubMed
195.
Zurück zum Zitat Ye D, Zhang X, Yue Y, Raliya R, Biswas P, Taylor S, et al. Focused ultrasound combined with microbubble-mediated intranasal delivery of gold nanoclusters to the brain. J Control Release. 2018;286:145–53.PubMedPubMedCentral Ye D, Zhang X, Yue Y, Raliya R, Biswas P, Taylor S, et al. Focused ultrasound combined with microbubble-mediated intranasal delivery of gold nanoclusters to the brain. J Control Release. 2018;286:145–53.PubMedPubMedCentral
196.
Zurück zum Zitat Kim NA, Thapa R, Jeong SH, Bae H, Maeng J, Lee K, et al. Enhanced intranasal insulin delivery by formulations and tumor protein-derived protein transduction domain as an absorption enhancer. J Control Release. 2019;294:226–36.PubMed Kim NA, Thapa R, Jeong SH, Bae H, Maeng J, Lee K, et al. Enhanced intranasal insulin delivery by formulations and tumor protein-derived protein transduction domain as an absorption enhancer. J Control Release. 2019;294:226–36.PubMed
197.
Zurück zum Zitat Zhao N, Liu C-C, Van Ingelgom AJ, Martens YA, Linares C, Knight JA, et al. Apolipoprotein E4 impairs neuronal insulin signaling by trapping insulin receptor in the endosomes. Neuron. 2017;96(115–29):e5. Zhao N, Liu C-C, Van Ingelgom AJ, Martens YA, Linares C, Knight JA, et al. Apolipoprotein E4 impairs neuronal insulin signaling by trapping insulin receptor in the endosomes. Neuron. 2017;96(115–29):e5.
198.
Zurück zum Zitat Rhea EM, Raber J, Banks WA. ApoE and cerebral insulin: trafficking, receptors, and resistance. Neurobiol Dis. 2020;137:104755.PubMedPubMedCentral Rhea EM, Raber J, Banks WA. ApoE and cerebral insulin: trafficking, receptors, and resistance. Neurobiol Dis. 2020;137:104755.PubMedPubMedCentral
199.
Zurück zum Zitat Willette AA, Bendlin BB, Starks EJ, Birdsill AC, Johnson SC, Christian BT, et al. Association of insulin resistance with cerebral glucose uptake in late middle-aged adults at risk for Alzheimer disease. JAMA Neurol. 2015;72:1013.PubMedPubMedCentral Willette AA, Bendlin BB, Starks EJ, Birdsill AC, Johnson SC, Christian BT, et al. Association of insulin resistance with cerebral glucose uptake in late middle-aged adults at risk for Alzheimer disease. JAMA Neurol. 2015;72:1013.PubMedPubMedCentral
200.
Zurück zum Zitat Alata W, Ye Y, St-Amour I, Vandal M, Calon F. Human apolipoprotein E ε4 expression impairs cerebral vascularization and blood-brain barrier function in mice. J Cereb Blood Flow Metab. 2015;35:86–94.PubMed Alata W, Ye Y, St-Amour I, Vandal M, Calon F. Human apolipoprotein E ε4 expression impairs cerebral vascularization and blood-brain barrier function in mice. J Cereb Blood Flow Metab. 2015;35:86–94.PubMed
201.
Zurück zum Zitat Verghese PB, Castellano JM, Holtzman DM. Apolipoprotein E in Alzheimer’s disease and other neurological disorders. Lancet Neurol. 2011;10:241–52.PubMedPubMedCentral Verghese PB, Castellano JM, Holtzman DM. Apolipoprotein E in Alzheimer’s disease and other neurological disorders. Lancet Neurol. 2011;10:241–52.PubMedPubMedCentral
202.
Zurück zum Zitat Mosconi L, Pupi A, De Leon MJ. Brain glucose hypometabolism and oxidative stress in preclinical Alzheimer’s disease. Ann NY Acad Sci. 2008;1147:180–95.PubMed Mosconi L, Pupi A, De Leon MJ. Brain glucose hypometabolism and oxidative stress in preclinical Alzheimer’s disease. Ann NY Acad Sci. 2008;1147:180–95.PubMed
203.
Zurück zum Zitat Mosconi L, Mistur R, Switalski R, Brys M, Glodzik L, Rich K, et al. Declining brain glucose metabolism in normal individuals with a maternal history of Alzheimer disease. Neurology. 2009;72:513–20.PubMedPubMedCentral Mosconi L, Mistur R, Switalski R, Brys M, Glodzik L, Rich K, et al. Declining brain glucose metabolism in normal individuals with a maternal history of Alzheimer disease. Neurology. 2009;72:513–20.PubMedPubMedCentral
204.
Zurück zum Zitat Gabbouj S, Ryhänen S, Marttinen M, Wittrahm R, Takalo M, Kemppainen S, et al. Altered insulin signaling in Alzheimer’s disease brain: sSpecial emphasis on PI3K-Akt pathway. Front Neurosci. 2019;13:1–8. Gabbouj S, Ryhänen S, Marttinen M, Wittrahm R, Takalo M, Kemppainen S, et al. Altered insulin signaling in Alzheimer’s disease brain: sSpecial emphasis on PI3K-Akt pathway. Front Neurosci. 2019;13:1–8.
205.
Zurück zum Zitat Mustapic M, Tran J, Craft S, Kapogiannis D. Extracellular vesicle biomarkers track cognitive changes following intranasal insulin in Alzheimer’s disease. J Alzheimer’s Dis. 2019;69:489–98. Mustapic M, Tran J, Craft S, Kapogiannis D. Extracellular vesicle biomarkers track cognitive changes following intranasal insulin in Alzheimer’s disease. J Alzheimer’s Dis. 2019;69:489–98.
206.
Zurück zum Zitat Cao B, Rosenblat JD, Brietzke E, Park C, Lee Y, Musial N, et al. Comparative efficacy and acceptability of antidiabetic agents for Alzheimer’s disease and mild cognitive impairment: a systematic review and network meta-analysis. Diabetes, Obes Metab. 2018;20:2467–71. Cao B, Rosenblat JD, Brietzke E, Park C, Lee Y, Musial N, et al. Comparative efficacy and acceptability of antidiabetic agents for Alzheimer’s disease and mild cognitive impairment: a systematic review and network meta-analysis. Diabetes, Obes Metab. 2018;20:2467–71.
207.
Zurück zum Zitat Batista AF, Bodart-Santos V, De Felice FG, Ferreira ST. Neuroprotective actions of glucagon-like peptide-1 (GLP-1) analogues in Alzheimer’s and Parkinson’s diseases. CNS Drugs. 2019;33:209–23.PubMed Batista AF, Bodart-Santos V, De Felice FG, Ferreira ST. Neuroprotective actions of glucagon-like peptide-1 (GLP-1) analogues in Alzheimer’s and Parkinson’s diseases. CNS Drugs. 2019;33:209–23.PubMed
208.
Zurück zum Zitat Koenig AM, Mechanic-Hamilton D, Xie SX, Combs MF, Cappola AR, Xie L, et al. Effects of the Insulin Sensitizer Metformin in Alzheimer Disease. Alzheimer Dis Assoc Disord. 2017;31:107–13.PubMedPubMedCentral Koenig AM, Mechanic-Hamilton D, Xie SX, Combs MF, Cappola AR, Xie L, et al. Effects of the Insulin Sensitizer Metformin in Alzheimer Disease. Alzheimer Dis Assoc Disord. 2017;31:107–13.PubMedPubMedCentral
209.
Zurück zum Zitat Ou Z, Kong X, Sun X, He X, Zhang L, Gong Z, et al. Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behav Immun. 2018;69:351–63.PubMed Ou Z, Kong X, Sun X, He X, Zhang L, Gong Z, et al. Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behav Immun. 2018;69:351–63.PubMed
210.
Zurück zum Zitat Farr SA, Roesler E, Niehoff ML, Roby DA, McKee A, Morley JE. Metformin improves learning and memory in the SAMP8 mouse model of Alzheimer’s disease. J Alzheimer’s Dis. 2019;68:1699–710. Farr SA, Roesler E, Niehoff ML, Roby DA, McKee A, Morley JE. Metformin improves learning and memory in the SAMP8 mouse model of Alzheimer’s disease. J Alzheimer’s Dis. 2019;68:1699–710.
211.
Zurück zum Zitat Luchsinger JA, Perez T, Chang H, Mehta P, Steffener J, Pradabhan G, et al. Metformin in amnestic mild cognitive impairment: results of a pilot randomized placebo controlled clinical trial. J Alzheimers Dis. 2016;51:501–14.PubMedPubMedCentral Luchsinger JA, Perez T, Chang H, Mehta P, Steffener J, Pradabhan G, et al. Metformin in amnestic mild cognitive impairment: results of a pilot randomized placebo controlled clinical trial. J Alzheimers Dis. 2016;51:501–14.PubMedPubMedCentral
212.
Zurück zum Zitat Watson GS. 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;13:950–8.PubMed Watson GS. 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;13:950–8.PubMed
213.
Zurück zum Zitat Harrington C, Sawchak S, Chiang C, Davies J, Donovan C, Saunders MA, et al. Rosiglitazone does not improve cognition or global function when used as adjunctive therapy to AChE inhibitors in mild-to-moderate Alzheimers disease: two phase 3 studies. Curr Alzheimer Res. 2011;8:592–606.PubMed Harrington C, Sawchak S, Chiang C, Davies J, Donovan C, Saunders MA, et al. Rosiglitazone does not improve cognition or global function when used as adjunctive therapy to AChE inhibitors in mild-to-moderate Alzheimers disease: two phase 3 studies. Curr Alzheimer Res. 2011;8:592–606.PubMed
214.
Zurück zum Zitat Burns DK, Chiang C, Welsh-Bohmer KA, Brannan SK, Culp M, O’Neil J, et al. The TOMMORROW study: design of an Alzheimer’s disease delay-of-onset clinical trial. Alzheimer’s Dement Transl Res Clin Interv. 2019;5:661–70. Burns DK, Chiang C, Welsh-Bohmer KA, Brannan SK, Culp M, O’Neil J, et al. The TOMMORROW study: design of an Alzheimer’s disease delay-of-onset clinical trial. Alzheimer’s Dement Transl Res Clin Interv. 2019;5:661–70.
215.
Zurück zum Zitat Solfrizzi V, Custodero C, Lozupone M, Imbimbo BP, Valiani V, Agosti P, et al. Relationships of dietary patterns, foods, and micro- and macronutrients with Alzheimer’s disease and late-life cognitive disorders: a systematic seview. J Alzheimers Dis. 2017;59:815–49.PubMed Solfrizzi V, Custodero C, Lozupone M, Imbimbo BP, Valiani V, Agosti P, et al. Relationships of dietary patterns, foods, and micro- and macronutrients with Alzheimer’s disease and late-life cognitive disorders: a systematic seview. J Alzheimers Dis. 2017;59:815–49.PubMed
216.
Zurück zum Zitat Stephen R, Hongisto K, Solomon A, Lönnroos E. Physical activity and Alzheimer’s disease: a systematic review. J Gerontol A Biol Sci Med Sci. 2017;72:733–9.PubMed Stephen R, Hongisto K, Solomon A, Lönnroos E. Physical activity and Alzheimer’s disease: a systematic review. J Gerontol A Biol Sci Med Sci. 2017;72:733–9.PubMed
217.
Zurück zum Zitat Gizurarson S. Anatomical and histological factors affecting intranasal drug and vaccine delivery. Curr Drug Deliv. 2012;9:566–82.PubMedPubMedCentral Gizurarson S. Anatomical and histological factors affecting intranasal drug and vaccine delivery. Curr Drug Deliv. 2012;9:566–82.PubMedPubMedCentral
218.
Zurück zum Zitat Kupila A, Sipilä J, Keskinen P, Simell T, Knip M, Pulkki K, et al. Intranasally administered insulin intended for prevention of type 1 diabetes: a safety study in healthy adults. Diabetes Metab Res Rev. 2003;19:415–20.PubMed Kupila A, Sipilä J, Keskinen P, Simell T, Knip M, Pulkki K, et al. Intranasally administered insulin intended for prevention of type 1 diabetes: a safety study in healthy adults. Diabetes Metab Res Rev. 2003;19:415–20.PubMed
219.
Zurück zum Zitat Spielman L, Bahniwal M, Little J, Walker D, Klegeris A. Insulin modulates in vitro secretion of cytokines and cytotoxins by human glial cells. Curr Alzheimer Res. 2015;12:684–93.PubMed Spielman L, Bahniwal M, Little J, Walker D, Klegeris A. Insulin modulates in vitro secretion of cytokines and cytotoxins by human glial cells. Curr Alzheimer Res. 2015;12:684–93.PubMed
220.
Zurück zum Zitat Hölscher C. Insulin signaling impairment in the brain as a risk factor in Alzheimer’s disease. Front Aging Neurosci. 2019;11:1–11. Hölscher C. Insulin signaling impairment in the brain as a risk factor in Alzheimer’s disease. Front Aging Neurosci. 2019;11:1–11.
221.
Zurück zum Zitat Shaughness M, Acs D, Brabazon F, Hockenbury N, Byrnes KR. Role of insulin in neurotrauma and neurodegeneration: a review. Front Neurosci. 2020;14:547175.PubMedPubMedCentral Shaughness M, Acs D, Brabazon F, Hockenbury N, Byrnes KR. Role of insulin in neurotrauma and neurodegeneration: a review. Front Neurosci. 2020;14:547175.PubMedPubMedCentral
222.
Zurück zum Zitat Mayer CM, Belsham DD. 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.PubMed Mayer CM, Belsham DD. 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.PubMed
Metadaten
Titel
Intranasal Insulin for Alzheimer’s Disease
verfasst von
Manfred Hallschmid
Publikationsdatum
01.01.2021
Verlag
Springer International Publishing
Erschienen in
CNS Drugs / Ausgabe 1/2021
Print ISSN: 1172-7047
Elektronische ISSN: 1179-1934
DOI
https://doi.org/10.1007/s40263-020-00781-x

Weitere Artikel der Ausgabe 1/2021

CNS Drugs 1/2021 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Thrombektomie auch bei großen Infarkten von Vorteil

16.05.2024 Ischämischer Schlaganfall Nachrichten

Auch ein sehr ausgedehnter ischämischer Schlaganfall scheint an sich kein Grund zu sein, von einer mechanischen Thrombektomie abzusehen. Dafür spricht die LASTE-Studie, an der Patienten und Patientinnen mit einem ASPECTS von maximal 5 beteiligt waren.

Schwindelursache: Massagepistole lässt Otholiten tanzen

14.05.2024 Benigner Lagerungsschwindel Nachrichten

Wenn jüngere Menschen über ständig rezidivierenden Lagerungsschwindel klagen, könnte eine Massagepistole der Auslöser sein. In JAMA Otolaryngology warnt ein Team vor der Anwendung hochpotenter Geräte im Bereich des Nackens.

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.