Synopsis
Type 2 diabetes, similarly to dementia, disproportionately affects the elderly. Diabetes has consistently been associated with risk of dementia—both Alzheimer’s disease (AD) and more strongly and consistently vascular dementia (VaD)—mild cognitive impairment (MCI), and cognitive decline suggesting that cognitive compromise is a deleterious manifestation of diabetes. This review summarizes observational studies and clinical trials of diabetes medications—insulin, peroxisome proliferator-activated receptor-γ, insulinotropic glucagon-like peptide-1, and metformin—and their respective associations and effects on cognitive outcomes. Despite biological plausibility, results from most human clinical trials have failed to show any efficacy in treating AD symptomatology and pathology. Clinical trials targeting vascular-related outcomes, diabetic patients, or cognitively normal elderly at risk for dementia, may provide greater cognitive benefits.
Diabetes disproportionately affects the elderly. Among U.S. residents aged 65 years and older, 26.9% had diabetes in 2010 and 50% had diabetes or pre-diabetes between 2005 and 2008 (Centers for Disease Control and Prevention 2011). According to the Alzheimer’s association, one in nine people age 65 and older (11%) has AD and about one-third of people age 85 and older (32%) have AD, and the prevalence of all forms of dementia is even higher (Alzheimer’s association 2013). This co-occurrence may reflect simply two simultaneous age-related events, possibly sharing one or more causal pathways, or may reflect a causative relationship between the conditions.
Diabetes is a risk factor for dementia and cognitive decline
Diabetes has consistently been associated with risk of dementia, mild cognitive impairment (MCI) and cognitive decline. A systematic review of effects of diabetes on cognitive dysfunction has suggested that the latter should be considered among the chronic consequences and disabling manifestations of diabetes (Cukierman et al. 2005). Furthermore, increased risks of dementia and AD were also associated with borderline diabetes, independent of the future development of diabetes (Xu et al. 2007). Diabetes, or impaired fasting glucose, may be present in up to 80% of persons with Alzheimer’s disease (AD) (Janson et al. 2004), and the 2010 NIH Consensus Development Conference Statement on Preventing Alzheimer’s Disease and Cognitive Decline listed diabetes first as a risk factor (Daviglus et al. 2010). This co-existence of diabetes (or diabetic markers) with cognitive dysfunctions have lead some investigators to propose that AD constitutes a brain-specific form of diabetes, i.e. Type 3 diabetes (de la Monte et al. 2006).
Our group has recently shown that diabetic elderly with the earliest signs of cognitive compromise have a faster rate of cognitive decline than non-diabetic elderly (Ravona-Springer et al. 2010). Similar associations between diabetes and cognitive compromise have been reported in numerous studies. Among the cognitive domains that have been associated with diabetes are attention, executive functions, perceptual/processing speed, verbal memory, and working memory, as well as global cognitive functioning (measured by the mini-mental state examination [MMSE]) (Arvanitakis et al. 2004; Gilmour 2011; Gregg et al. 2000; Hassing et al. 2004a; Hassing et al. 2004b; Knopman et al. 2001; Logroscino et al. 2004; Nandipati et al. 2012). Our recent study has also demonstrated that poor glycemic control is associated with cognitive decline even in non-diabetic individuals (Ravona-Springer et al. 2012). Such association, however, was not observed in elderly aged 85 years or older (van den Berg et al. 2006), possibly due to a survivors effect. Moreover, within a population of patients who were already demented in the time of the study, diabetics showed slower global cognitive decline than non-diabetics (Sanz et al. 2009), although functional status, measured by Activities of Daily Leaving (ADL), continuously declined (Sanz et al. 2012). One explanation for this observation may be a floor effect, or that the medical attention that this population receives is helpful in preventing cognitive deterioration.
MCI is characterized by memory complaints without loss of function in daily activities (Petersen et al. 1999). Diabetes has been related to a 40% higher risk of MCI, both amnestic and nonamnestic (Luchsinger et al. 2007). Two other studies reported a trend towards increased risk of MCI in a diabetic elderly population (Solfrizzi et al. 2004), and in a sample of postmenopausal women (Yaffe et al. 2004), but these changes, however, were statistically non-significant. A more recent study may shed some light on these inconsistencies: the frequency of diabetes was similar in elderly subjects with and without MCI, but MCI was associated with diabetes onset before the age of 65, diabetes duration of 10 years or longer, treatment with insulin, and the presence of diabetes complications (Roberts et al. 2008).
According to the DSM-IV (Text Revision), dementia is characterized by the development of multiple cognitive deficits that must include memory impairment and other cognitive disturbances (American Psychiatric Association 2000). Our prior research showed that subjects with diabetes in midlife had a 3-fold increased risk of dementia three decades later (Schnaider Beeri et al. 2004). Increased risk of dementia in diabetic patients was reported in other studies as well (Leibson et al. 1997; Ott et al. 1999; Peila et al. 2002; Whitmer et al. 2005; Xu et al. 2004), but not all (Yaffe et al. 2004). Furthermore, diabetes seems to increase the risk of certain subtypes of dementia in different extents. The most common subtypes of dementia are Alzheimer’s disease (AD) and vascular dementia (VaD). According to the National Institute of Neurological and Communicative Disorders and Stroke–Alzheimer’s Disease and Related Disorders Association (NINCDS-ADRDA), in order to fulfill research criteria for probable AD, a patient must present a significant episodic memory impairment and at least one supportive biomarker (such as medial temporal lobe atrophy) (Dubois et al. 2007). Even VaD itself is not a single disease, but a group of syndromes based on varying vascular mechanisms. These include dementias related to multiple infarcts, small vessel ischaemic disease, Alzheimer’s disease with cerebrovascular disease (sometimes known as ‘mixed dementia’), and others (O’Brien 2006). Diabetes was reported to increase the risk of AD by 45–90% (Arvanitakis et al. 2004; Leibson et al. 1997; Ott et al. 1999; Wang et al. 2012), but the risk for VaD was consistently and substantially higher, with increases ranging from 100% to 160% (Hassing et al. 2002; MacKnight et al. 2002; Xu et al. 2004), suggesting that diabetes is more closely associated with VaD than with AD.
Taken together, these findings suggest an association between diabetes and cognitive decline. Some of the studies evaluated diabetes in midlife, decades before dementia ascertainment, supporting the notion that cognitive impairment is a consequence of diabetes.
The effects of diabetes treatments on memory and cognition
The research of different pharmacological and non-pharmacological treatments of diabetes, and the known biological mechanisms of those treatments, may help us in understanding this association. Insulin and oral hypoglycemics are the most common treatments for diabetes. Diabetes medications have been associated with improved cognitive functioning, and have been demonstrated to affect AD markers, such as neuritic plaques (Beeri et al. 2008), as well as vascular integrity (Kalaria 2009). The SALSA study had reported that diabetic patients on antidiabetic monotherapy (insulin or oral), and more so on any combination therapy, had less cognitive decline, especially among those with a longer duration of the disease (Wu et al. 2003). Our previous postmortem study examined the association between AD neuropathology and diabetes medications in several brain regions that support cognitive functioning: hippocampus, entorhinal cortex, amygdala, and several neocortical regions. In each region, the study demonstrated substantially lower neuritic plaque density for the diabetic group taking both insulin and other antidiabetic medication, compared to diabetic patients on monotherapy or on no therapy (Beeri et al. 2008). Here we will briefly review the association of some of the most common antidiabetic treatments with cognitive functioning.
The association of circulating/CSF insulin levels and insulin administration with cognition
Produced nearly exclusively by the pancreas, insulin is readily transported into the CNS across the blood-brain barrier (BBB) using a saturable, receptor-mediated process. Insulin receptors are highly concentrated in the olfactory bulb, cerebral cortex, hippocampus, hypothalamus, and cerebellum (reviewed by (Banks et al. 2012)). Localization of insulin receptors in hippocampus and medial temporal cortex is consistent with evidence that insulin influences memory (Cholerton et al. 2011). Conditions such as insulin resistance—in which insulin is chronically elevated—may eliminate the salutary effects of insulin on cognition and other brain function (Craft 2006). Elevating insulin levels, through excess production (endogenously or provoked exogenously), excess administration of insulin or reduced clearance, will typically result in a down-regulation of insulin signaling pathways (reviewed by (White 2003)). One consequence of insulin resistance and chronic excess insulin (hyperinsulinemia) is reduced insulin transport into brain, which ultimately produces brain-insulin deficiency (Baura et al. 1996), and may attenuate the many beneficial influences of insulin. Support for this notion may be seen in recently published results from the PIVUS study: insulin resistance was negatively correlated with verbal cognitive performance, brain size, and temporal lobe gray matter volume, and these correlations were not affected by diabetic status or cognitive status (Benedict et al. 2012). Those findings are similar to an earlier report on pre-diabetic and diabetic patients, in which greater insulin resistance was associated with reduced glucose metabolic rate in frontal, parietotemporal, and cingulate regions, compatible with an AD pattern (Baker et al. 2011). Those individuals also showed a more diffuse and extensive activation patterns, and recalled fewer items on a delayed memory test (Baker et al. 2011). Since type 2 diabetic patients and approximately half of all adults over the age of 60, regardless of diabetic status, are insulin resistant (Craft 2006), studying the effects of insulin on cognition is of great importance.
Craft has pioneered the study of the role of insulin regulation in AD and cognitive decline. In normal physiological conditions, insulin administered at optimal (i.e. low) doses facilitates memory, as was demonstrated by direct administration of insulin to the brain in rodents (Park et al. 2000), and intravenous insulin administration in humans (Craft et al. 2003). The latter study also showed that higher levels of insulin were needed for achieving effective memory facilitation in ApoE ε4-negative AD patients, supporting the notion of insulin resistance in this subgroup (Craft et al. 2003). The authors suggested that for these patients, factors relating to insulin resistance may be important drivers of AD pathogenesis.
For memory-impaired adults, insulin was found to improve memory functions also when administered intranasally at acute dosage (Reger et al. 2008a) and chronic treatment for 3 weeks (Reger et al. 2008b) or 4 months (Craft et al. 2012). A recently published analysis of results from this trial further suggest sex and ApoE genotype differences in the response to the treatment: whereas women showed improved memory only when administered the lower dose, men showed cognitive improvement also for the higher dose, and this sex difference was most apparent for ApoE ε4-negative individuals (Claxton et al. 2013). Importantly, sex differences were also observed in the association between cognitive functioning and circulating insulin: men with non-amnestic or amnestic MCI had higher fasting plasma insulin than cognitively normal men, while women with amnestic MCI had lower fasting plasma insulin than cognitively normal women (Cholerton et al. 2012).
Insulin may exert its effects on cognition and AD risk through modulation of the β-amyloid peptide (Aβ). Aβ, a metabolite of the amyloid precursor protein (APP), aggregates in extracellular depositions, neuritic plaques, which constitute one of the hallmarks of AD pathology (Braak and Braak 1997). The close bidirectional relationship between insulin and Aβ is described in detail in a recently published review article (Craft et al. 2013). Subchronic elevations of CNS insulin concentrations by intranasal administration have been associated with reduced circulating concentrations of Aβ (Reger et al. 2008b). Very similar to the attenuation of the positive effects of insulin on memory facilitation, excessive insulin elevations through intravenous infusion increased levels of the Aβ in cerebrospinal fluid (CSF), most notably in older subjects (Watson et al. 2003). Importantly, greater increases in CSF Aβ levels attenuated the insulin-mediated memory facilitation (Watson et al. 2003). Moreover, insulin administration was found to reduce plasma APP concentrations, particularly in ApoE ε4-negative individuals, both in cognitively intact subjects (Boyt et al. 2000) and AD patients (Craft et al. 2000). The effective insulin dose in reducing plasma APP was higher for ApoE ε4-negative AD patients than for normal adults and ApoE ε4-positive AD patients (Craft et al. 2003). Finally, insulin administration was associated with inflammation, a key factor in the pathogenesis of AD (Rogers and Shen 2000). Anti-inflammatory effects were observed with low doses of insulin (Dandona 2002), but excessive hyperinsulinemia exacerbates inflammation (Krogh-Madsen et al. 2004). Craft’s research group has demonstrated that intravenous infusion of insulin to levels associated with insulin resistance increased CSF inflammatory markers (Fishel et al. 2005).
Peroxisome proliferator-activated receptor-γ and AD
These observations led to the investigation of how therapeutic strategies, originally aimed at treating diabetes, may also benefit elderly with a wide range of cognitive impairments, including AD. Agonists to the peroxisome proliferator-activated receptor-γ (PPAR-γ) are known to improve insulin sensitivity, decrease circulating insulin, and increase insulin-mediated glucose uptake with minimal risk of hypoglycemia (Olefsky 2000). In addition, PPAR-γ agonists were found to inhibit inflammation, and specifically the Aβ-stimulated secretion of pro-inflammatory products and the Aβ-stimulated expression of the cytokine genes (Combs et al. 2000), making them good candidates for therapeutic agents in treating AD. The beneficial effects of PPAR-γ agonists have been demonstrated in a line of studies on transgenic AD mice, typically displaying widespread microglial activation, age-related amyloid deposits, and dystrophic neurites. Tg2576 mice that were treated with chronic oral administration of ibuprofen, an efficient activator of PPAR-γ (Lehmann et al. 1997), have shown marked reduction in Aβ deposits (Lim et al. 2000). Positive effects of PPAR-γ agonists on AD mice were also shown for two compounds, rosiglitazone and pioglitazone, which are commonly prescribed for diabetics. Rosiglitazone treated Tg2576 mice showed age-dependent reversal of cognitive deficits (Rodriguez-Rivera et al. 2011), but with no evidence of reduction in Aβ deposits (Pedersen et al. 2006). In APPswe/PS1dE9 mice the drug improved spatial memory, decreased insoluble Aβ1–42, and decreased plaque number in the hippocampus (O’Reilly and Lynch 2012). Transgenic mice carrying the Swedish (K670N/M671L) and Indian (V717F) AD mutations of human APP showed rescue of memory impairments, removal of amyloid plaques in the hippocampus and entorhinal cortex, and decreased phosphorylated tau protein following rosiglitazone treatment (Escribano et al. 2010). Pioglitazone administered to the latter transgenic mice fully restored cerebrovascular reactivity, albeit it failed to improve spatial memory or to reduce Aβ plaque load (Nicolakakis et al. 2008). In triple transgenic AD mice, however, long-term pioglitazone treatment improved cognition and decreased hippocampal Aβ and tau deposits (Searcy et al. 2012).
Despite the findings from animal studies in favor of PPAR-γ agonists for the treatment of AD symptomatology and pathology, results from human clinical trials have been rather disappointing.. In a placebo-controlled, double-blind, parallel-group pilot study, rosiglitazone treated amnestic MCI and early AD patients showed preservation of some cognitive functions, whereas placebo-assigned subjects showed the expected memory decline (Watson et al. 2005). This cognitive maintenance was not consistent throughout the trial period, and was not observed in all functions. Also important to note is that the study group did not include patients with moderate or severe AD. A larger trial did not observe overall cognitive benefit of rosiglitazone in mild-to-moderate AD patients, although improvement was noted in ApoE ε4-negative subjects on a task of general cognitive function at the highest dose (Risner et al. 2006). Subsequent Phase III trials found no evidence of statistically or clinically significant efficacy of rosiglitazone in cognition or global function, regardless of genotype, when used as monotherapy (Gold et al. 2010) or as adjunctive therapy to AChE inhibitors (Harrington et al. 2011). Pioglitazone produced similar results. Trials of mild-AD patients with diabetes reported improved general cognition, improved verbal memory, and improved cerebral blood flow in the parietal lobe, following six months of treatment (Hanyu et al. 2009; Sato et al. 2011). Nevertheless, a trial of non-diabetic patients meeting research criteria for probable AD, showed no improvements on cognitive and functional measures (Geldmacher et al. 2011).
The lack of efficacy in these trials may be due to several reasons, including the complicated effects that inflammation exerts in AD pathogenesis, treatment at the wrong stage of the disease, or inappropriate dosing. Importantly, there is a debate on whether rosiglitazone effectively crosses the BBB in rodents: intraperitoneal administration to gerbils (Sheu et al. 2011) or oral delivery to mice (Strum et al. 2007) resulted in effective and rapid penetration to the brain, while intravenous administration to rats resulted in low brain uptake (Festuccia et al. 2008); the ability of pioglitazone to cross the BBB is less controversial (Grommes et al. 2013; Maeshiba et al. 1997). In addition, human trials raised some safety concerns regarding the effects of rosiglitazone on cardiovascular functioning and heart failure in diabetic patients (Mannucci et al. 2010). Combined, these findings suggest that there is continuous and consistent evidence of the beneficial effects of insulin and insulin senstitizing therapies on cognition, however, the emphasis on insulin in treating AD should be shifted towards a different approach.
Is glucagon-like peptide-1 a promising novel approach in treatment of AD?
One such pharmacologic approach to insulin resistance involves the use of insulinotropic glucagon-like peptide-1 (GLP-1), a hormone that facilitates insulin release under high blood sugar conditions, and does not affect blood sugar levels in non-diabetic people, enhances insulin signaling, and protects neurons from toxic effects (Holscher 2010). GLP-1 agonists bind GLP-1 receptor that is coupled to a second messenger pathway via G proteins (Green et al. 2004), and improve dyslipidemia, blood pressure, and other diabetes-associated vascular conditions (Sivertsen et al. 2012). GLP-1 is proposed to play a role in a regulatory mechanism involved in the actions of GLUT1 glucose transporters and glucose metabolism, by ensuring less fluctuation of brain glucose levels in response to alterations in plasma glucose (Gejl et al. 2012). The neuroprotective actions of GLP-1 have been demonstrated in in vivo and in vitro studies. GLP-1 has been documented to induce neurite outgrowth, reduce apoptosis, protect neurons from oxidative stress, protect synaptic plasticity and memory formation from the detrimental effects of Aβ, and reduce plaque formation and the inflammation response in the brains of mouse models of AD (reviewed by (Holscher 2010)). Intraperitoneal injection of GLP-1 receptor agonist to AD transgenic mice reduced hippocampal amyloid burden, while improving spatial memory (Bomfim et al. 2012).
The association between AD and the non-insulin antidiabetic therapy metformin
Metformin is a biguanide (Bailey and Turner 1996) that lowers blood glucose levels by increasing hepatic and muscle cell insulin sensitivity, by decreasing intestinal glucose absorption (Tian et al. 2004), and crosses the BBB readily (Labuzek et al. 2010). Metformin improved insulin sensitivity and decreased insulin levels in persons without diabetes (Kitabchi et al. 2005) and was therefore proposed as a new target for research in the context of cognitive compromise. In vitro, metformin significantly decreased phosphorylated tau and ameliorated Aβ1–42 levels in neuronal insulin resistance and AD-associated cell cultures (Gupta et al. 2011). Counter-intuitively, in primary cortical culture models, metformin significantly increases the generation of both intracellular and extracellular Aβ species, but in combined use with insulin, metformin enhances insulin’s effect in reducing Aβ levels (Chen et al. 2009). Finally, in murine primary neurons from wild-type and human tau transgenic mice, metformin reduced tau phosphorylation (Kickstein et al. 2010). In vivo, the association between metformin and AD-like neuropathology was examined in obese, leptin-resistant mice. Metformin attenuated the increase of phosphorylated tau and the reduction of the synaptic protein synaptophysin, but did no better than saline in decreasing Aβ levels, and did not attenuate the impairments of spatial learning and memory (Li et al. 2012). Metformin successfully attenuated cognitive deficits of diabetic rats (Bhutada et al. 2011), and high-fat diet fed rats (Pintana et al. 2012).
To date, we do not have adequate clinical data on the efficacy of metformin in preventing or treating AD and other dementia related disorders in humans. Nevertheless, a study of elderly diabetic individuals showed that metformin attenuated the decline in global cognitive function but not in verbal memory, an effect that was similar to that of a diet regime, and inferior to that of a combined treatment with rosiglitazone (Abbatecola et al. 2010). In addition, the association between metformin and risk of dementia was examined in two large cohort-based studies, and yielded contradictory results. A study of individuals aged 65 or older found no evidence that use of metformin is associated with lower risk of developing AD (Imfeld et al. 2012). Furthermore, the findings even suggested that long-term use of metformin may be associated with a slightly higher risk of developing AD, compared to nonuse of this drug, and such a finding was not seen for use of other antidiabetic drugs (Imfeld et al. 2012). In contrast, a study of Taiwanese aged 50 years or older, found that the use of metformin in the treatment of diabetes significantly decreased the risk of dementia, compared with no medication (Hsu et al. 2011). More research, particularly clinical placebo-controlled trials, is needed to clarify the potential benefits of metformin to cognitive health.
Summary
Based on current evidence, there is little doubt that diabetes and cognitive compromise are closely related. This relationship is manifested in a wide range of cognitive impairments, starting from cognitive decline, through mild cognitive impairment, and frank clinical dementia. The underlying mechanisms of these relationship are, however, still elusive. It seems that diabetes is more strongly and more consistently associated with vascular forms of cognitive impairment, rather than with AD-like neurodegenerative forms. Moreover, the different therapeutic strategies show some cognitive benefits, particularly intranasal insulin, which do not seem to be specific to AD: treatments seem less effective in attenuation of AD neuropathology in animal models and in alleviating cognitive dysfunctions in elderly who already succumbed to AD. Clinical trials have primarily targeted AD-related outcomes (e.g. Aβ accumulation, memory deficits, conversion rate to AD), while evidence suggests that vascular-related outcomes might be more promising. In addition, based on current basic science evidence, it is sensible to assume that mechanisms underlying diabetes complications might underlie dementia in non-diabetics as well. Nevertheless, some of the findings presented suggest that diabetes medications may be more beneficial to diabetic patients rather than to non-diabetics. Finally, both AD and vascular neuropathology are thought to begin to develop and accumulate decades before manifestation of clinical symptoms. Thus, prevention clinical trials utilizing diabetes medication aimed against the development of cognitive compromise, may be more effective for individuals at risk but initially cognitively normal, both diabetics and non-diabetics.
KEY POINTS.
Diabetes and cognitive compromise are closely related. This relationship is manifested in a wide range of cognitive impairments, starting from cognitive decline, through mild cognitive impairment, and frank clinical dementia.
Diabetes is more strongly and more consistently associated with vascular forms of cognitive impairment, rather than with AD-like neurodegenerative forms.
Both AD and vascular neuropathology are thought to begin to develop and accumulate decades before manifestation of clinical symptoms. Thus, prevention clinical trials utilizing diabetes medication aimed against the development of cognitive compromise, may be more effective for individuals at risk but initially cognitively normal, both diabetics and non-diabetics.
Acknowledgments
Acknowledgement and funding: This study was supported by NIA grants R01 AG034087 (Beeri), P50 AG05138 (Sano), the Ira T. Hirschl Award (Beeri), and by an award from the Helen Bader Foundation (Beeri).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Abbatecola AM, Lattanzio F, Molinari AM, Cioffi M, Mansi L, Rambaldi P, DiCioccio L, Cacciapuoti F, Canonico R, Paolisso G. Rosiglitazone and cognitive stability in older individuals with type 2 diabetes and mild cognitive impairment. Diabetes Care. 2010;33(8):1706–1711. doi: 10.2337/dc09-2030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alzheimer’s association. 2013 Alzheimer’s disease facts and figures. Alzheimers Dement. 2013;9(2):208–245. doi: 10.1016/j.jalz.2013.02.003. [DOI] [PubMed] [Google Scholar]
- American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 4. Washington, DC: American Psychiatric Press; 2000. Text Revision (DSM-IV-TR) [Google Scholar]
- Arvanitakis Z, Wilson RS, Bienias JL, Evans DA, Bennett DA. Diabetes mellitus and risk of Alzheimer disease and decline in cognitive function. Arch Neurol. 2004;61(5):661–666. doi: 10.1001/archneur.61.5.661. [DOI] [PubMed] [Google Scholar]
- Bailey CJ, Turner RC. Metformin. N Engl J Med. 1996;334(9):574–579. doi: 10.1056/NEJM199602293340906. [DOI] [PubMed] [Google Scholar]
- Baker LD, Cross DJ, Minoshima S, Belongia D, Watson GS, Craft S. Insulin resistance and Alzheimer-like reductions in regional cerebral glucose metabolism for cognitively normal adults with prediabetes or early type 2 diabetes. Arch Neurol. 2011;68(1):51–57. doi: 10.1001/archneurol.2010.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banks WA, Owen JB, Erickson MA. Insulin in the brain: there and back again. Pharmacol Ther. 2012;136(1):82–93. doi: 10.1016/j.pharmthera.2012.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baura GD, Foster DM, Kaiyala K, Porte D, Jr, Kahn SE, Schwartz MW. Insulin transport from plasma into the central nervous system is inhibited by dexamethasone in dogs. Diabetes. 1996;45(1):86–90. doi: 10.2337/diab.45.1.86. [DOI] [PubMed] [Google Scholar]
- Beeri MS, Schmeidler J, Silverman JM, Gandy S, Wysocki M, Hannigan CM, Purohit DP, Lesser G, Grossman HT, Haroutunian V. Insulin in combination with other diabetes medication is associated with less Alzheimer neuropathology. Neurology. 2008;71(10):750–757. doi: 10.1212/01.wnl.0000324925.95210.6d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benedict C, Brooks SJ, Kullberg J, Burgos J, Kempton MJ, Nordenskjold R, Nylander R, Kilander L, Craft S, Larsson EM, Johansson L, Ahlstrom H, Lind L, Schioth HB. 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(3):488–494. doi: 10.2337/dc11-2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhutada P, Mundhada Y, Bansod K, Tawari S, Patil S, Dixit P, Umathe S, Mundhada D. Protection of cholinergic and antioxidant system contributes to the effect of berberine ameliorating memory dysfunction in rat model of streptozotocin-induced diabetes. Behav Brain Res. 2011;220(1):30–41. doi: 10.1016/j.bbr.2011.01.022. [DOI] [PubMed] [Google Scholar]
- Bomfim TR, Forny-Germano L, Sathler LB, Brito-Moreira J, Houzel JC, Decker H, Silverman MA, Kazi H, Melo HM, McClean PL, Holscher C, Arnold SE, Talbot K, Klein WL, Munoz DP, Ferreira ST, De Felice FG. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease- associated Abeta oligomers. J Clin Invest. 2012;122(4):1339–1353. doi: 10.1172/JCI57256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boyt AA, Taddei TK, Hallmayer J, Helmerhorst E, Gandy SE, Craft S, Martins RN. The effect of insulin and glucose on the plasma concentration of Alzheimer’s amyloid precursor protein. Neuroscience. 2000;95(3):727–734. doi: 10.1016/s0306-4522(99)00458-3. [DOI] [PubMed] [Google Scholar]
- Braak H, Braak E. Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol Aging. 1997;18(4):351–357. doi: 10.1016/s0197-4580(97)00056-0. [DOI] [PubMed] [Google Scholar]
- Centers for Disease Control and Prevention. National diabetes fact sheet: national estimates and general information on diabetes and prediabetes in the United States, 2011. 2011. [Google Scholar]
- Chen Y, Zhou K, Wang R, Liu Y, Kwak YD, Ma T, Thompson RC, Zhao Y, Smith L, Gasparini L, Luo Z, Xu H, Liao FF. Antidiabetic drug metformin (GlucophageR) increases biogenesis of Alzheimer’s amyloid peptides via up-regulating BACE1 transcription. Proc Natl Acad Sci U S A. 2009;106(10):3907–3912. doi: 10.1073/pnas.0807991106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cholerton B, Baker LD, Craft S. Insulin resistance and pathological brain ageing. Diabet Med. 2011;28(12):1463–1475. doi: 10.1111/j.1464-5491.2011.03464.x. [DOI] [PubMed] [Google Scholar]
- Cholerton B, Baker LD, Trittschuh EH, Crane PK, Larson EB, Arbuckle M, Saucedo HH, McCurry SM, Bowen JD, McCormick WC, Craft S. Insulin and sex interactions in older adults with mild cognitive impairment. J Alzheimers Dis. 2012;31 (2):401–410. doi: 10.3233/JAD-2012-120202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Claxton A, Baker LD, Wilkinson CW, Trittschuh EH, Chapman D, Watson GS, Cholerton B, Plymate SR, Arbuckle M, Craft S. 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 doi: 10.3233/JAD-122308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Combs CK, Johnson DE, Karlo JC, Cannady SB, Landreth GE. Inflammatory mechanisms in Alzheimer’s disease: inhibition of beta-amyloid-stimulated proinflammatory responses and neurotoxicity by PPARgamma agonists. J Neurosci. 2000;20(2):558–567. doi: 10.1523/JNEUROSCI.20-02-00558.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Craft S. Insulin resistance syndrome and Alzheimer disease: pathophysiologic mechanisms and therapeutic implications. Alzheimer Dis Assoc Disord. 2006;20(4):298–301. doi: 10.1097/01.wad.0000213866.86934.7e. [DOI] [PubMed] [Google Scholar]
- Craft S, Asthana S, Cook DG, Baker LD, Cherrier M, Purganan K, Wait C, Petrova A, Latendresse S, Watson GS, Newcomer JW, Schellenberg GD, Krohn AJ. Insulin dose-response effects on memory and plasma amyloid precursor protein in Alzheimer’s disease: interactions with apolipoprotein E genotype. Psychoneuroendocrinology. 2003;28(6):809–822. doi: 10.1016/s0306-4530(02)00087-2. [DOI] [PubMed] [Google Scholar]
- Craft S, Asthana S, Schellenberg G, Baker L, Cherrier M, Boyt AA, Martins RN, Raskind M, Peskind E, Plymate S. Insulin effects on glucose metabolism, memory, and plasma amyloid precursor protein in Alzheimer’s disease differ according to apolipoprotein-E genotype. Ann N Y Acad Sci. 2000;903:222–228. doi: 10.1111/j.1749-6632.2000.tb06371.x. [DOI] [PubMed] [Google Scholar]
- Craft S, Baker LD, Montine TJ, Minoshima S, Watson GS, Claxton A, Arbuckle M, Callaghan M, Tsai E, Plymate SR, Green PS, Leverenz J, Cross D, Gerton B. Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial. Arch Neurol. 2012;69(1):29–38. doi: 10.1001/archneurol.2011.233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Craft S, Cholerton B, Baker LD. Insulin and Alzheimer’s disease: untangling the web. J Alzheimers Dis. 2013;33(Suppl 1):S263–275. doi: 10.3233/JAD-2012-129042. [DOI] [PubMed] [Google Scholar]
- Cukierman T, Gerstein HC, Williamson JD. Cognitive decline and dementia in diabetes--systematic overview of prospective observational studies. Diabetologia. 2005;48 (12):2460–2469. doi: 10.1007/s00125-005-0023-4. [DOI] [PubMed] [Google Scholar]
- Dandona P. Endothelium, inflammation, and diabetes. Curr Diab Rep. 2002;2(4):311–315. doi: 10.1007/s11892-002-0019-0. [DOI] [PubMed] [Google Scholar]
- Daviglus ML, Bell CC, Berrettini W, Bowen PE, Connolly ES, Jr, Cox NJ, Dunbar-Jacob JM, Granieri EC, Hunt G, McGarry K, Patel D, Potosky AL, Sanders-Bush E, Silberberg D, Trevisan M. National Institutes of Health State-of-the-Science Conference statement: preventing alzheimer disease and cognitive decline. Ann Intern Med. 2010;153(3):176–181. doi: 10.7326/0003-4819-153-3-201008030-00260. [DOI] [PubMed] [Google Scholar]
- de la Monte SM, Tong M, Lester-Coll N, Plater M, Jr, Wands JR. Therapeutic rescue of neurodegeneration in experimental type 3 diabetes: relevance to Alzheimer’s disease. J Alzheimers Dis. 2006;10(1):89–109. doi: 10.3233/jad-2006-10113. [DOI] [PubMed] [Google Scholar]
- Dubois B, Feldman HH, Jacova C, Dekosky ST, Barberger-Gateau P, Cummings J, Delacourte A, Galasko D, Gauthier S, Jicha G, Meguro K, O’Brien J, Pasquier F, Robert P, Rossor M, Salloway S, Stern Y, Visser PJ, Scheltens P. Research criteria for the diagnosis of Alzheimer’s disease: revising the NINCDS-ADRDA criteria. Lancet Neurol. 2007;6(8):734–746. doi: 10.1016/S1474-4422(07)70178-3. [DOI] [PubMed] [Google Scholar]
- Escribano L, Simon AM, Gimeno E, Cuadrado-Tejedor M, Lopez de Maturana R, Garcia-Osta A, Ricobaraza A, Perez-Mediavilla A, Del Rio J, Frechilla D. Rosiglitazone rescues memory impairment in Alzheimer’s transgenic mice: mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacology. 2010;35(7):1593–1604. doi: 10.1038/npp.2010.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Festuccia WT, Oztezcan S, Laplante M, Berthiaume M, Michel C, Dohgu S, Denis RG, Brito MN, Brito NA, Miller DS, Banks WA, Bartness TJ, Richard D, Deshaies Y. Peroxisome proliferator-activated receptor-gamma-mediated positive energy balance in the rat is associated with reduced sympathetic drive to adipose tissues and thyroid status. Endocrinology. 2008;149(5):2121–2130. doi: 10.1210/en.2007-1553. [DOI] [PubMed] [Google Scholar]
- Fishel MA, Watson GS, Montine TJ, Wang Q, Green PS, Kulstad JJ, Cook DG, Peskind ER, Baker LD, Goldgaber D, Nie W, Asthana S, Plymate SR, Schwartz MW, Craft S. Hyperinsulinemia provokes synchronous increases in central inflammation and beta-amyloid in normal adults. Arch Neurol. 2005;62(10):1539–1544. doi: 10.1001/archneur.62.10.noc50112. [DOI] [PubMed] [Google Scholar]
- Gejl M, Egefjord L, Lerche S, Vang K, Bibby BM, Holst JJ, Mengel A, Moller N, Rungby J, Brock B, Gjedde A. Glucagon-like peptide-1 decreases intracerebral glucose content by activating hexokinase and changing glucose clearance during hyperglycemia. J Cereb Blood Flow Metab. 2012;32(12):2146–2152. doi: 10.1038/jcbfm.2012.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geldmacher DS, Fritsch T, McClendon MJ, Landreth G. A randomized pilot clinical trial of the safety of pioglitazone in treatment of patients with Alzheimer disease. Arch Neurol. 2011;68(1):45–50. doi: 10.1001/archneurol.2010.229. [DOI] [PubMed] [Google Scholar]
- Gilmour H. Cognitive performance of Canadian seniors. Health Rep. 2011;22(2):27–31. [PubMed] [Google Scholar]
- Gold M, Alderton C, Zvartau-Hind M, Egginton S, Saunders AM, Irizarry M, Craft S, Landreth G, Linnamagi U, Sawchak S. Rosiglitazone monotherapy in mild-to-moderate Alzheimer’s disease: results from a randomized, double-blind, placebo-controlled phase III study. Dement Geriatr Cogn Disord. 2010;30(2):131–146. doi: 10.1159/000318845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green BD, Gault VA, Flatt PR, Harriott P, Greer B, O’Harte FP. Comparative effects of GLP-1 and GIP on cAMP production, insulin secretion, and in vivo antidiabetic actions following substitution of Ala8/Ala2 with 2-aminobutyric acid. Arch Biochem Biophys. 2004;428(2):136–143. doi: 10.1016/j.abb.2004.05.005. [DOI] [PubMed] [Google Scholar]
- Gregg EW, Yaffe K, Cauley JA, Rolka DB, Blackwell TL, Narayan KM, Cummings SR. Is diabetes associated with cognitive impairment and cognitive decline among older women? Study of Osteoporotic Fractures Research Group. Arch Intern Med. 2000;160(2):174–180. doi: 10.1001/archinte.160.2.174. [DOI] [PubMed] [Google Scholar]
- Grommes C, Karlo JC, Caprariello A, Blankenship D, Dechant A, Landreth GE. The PPARgamma agonist pioglitazone crosses the blood-brain barrier and reduces tumor growth in a human xenograft model. Cancer Chemother Pharmacol. 2013;71(4):929–936. doi: 10.1007/s00280-013-2084-2. [DOI] [PubMed] [Google Scholar]
- Gupta A, Bisht B, Dey CS. Peripheral insulin-sensitizer drug metformin ameliorates neuronal insulin resistance and Alzheimer’s-like changes. Neuropharmacology. 2011;60(6):910–920. doi: 10.1016/j.neuropharm.2011.01.033. [DOI] [PubMed] [Google Scholar]
- Hanyu H, Sato T, Kiuchi A, Sakurai H, Iwamoto T. Pioglitazone improved cognition in a pilot study on patients with Alzheimer’s disease and mild cognitive impairment with diabetes mellitus. J Am Geriatr Soc. 2009;57(1):177–179. doi: 10.1111/j.1532-5415.2009.02067.x. [DOI] [PubMed] [Google Scholar]
- Harrington C, Sawchak S, Chiang C, Davies J, Donovan C, Saunders AM, Irizarry M, Jeter B, Zvartau-Hind M, van Dyck CH, Gold M. Rosiglitazone does not improve cognition or global function when used as adjunctive therapy to AChE inhibitors in mild-to-moderate Alzheimer’s disease: two phase 3 studies. Curr Alzheimer Res. 2011;8 (5):592–606. doi: 10.2174/156720511796391935. [DOI] [PubMed] [Google Scholar]
- Hassing LB, Grant MD, Hofer SM, Pedersen NL, Nilsson SE, Berg S, McClearn G, Johansson B. Type 2 diabetes mellitus contributes to cognitive decline in old age: a longitudinal population-based study. J Int Neuropsychol Soc. 2004a;10(4):599–607. doi: 10.1017/S1355617704104165. [DOI] [PubMed] [Google Scholar]
- Hassing LB, Hofer SM, Nilsson SE, Berg S, Pedersen NL, McClearn G, Johansson B. Comorbid type 2 diabetes mellitus and hypertension exacerbates cognitive decline: evidence from a longitudinal study. Age Ageing. 2004b;33(4):355–361. doi: 10.1093/ageing/afh100. [DOI] [PubMed] [Google Scholar]
- Hassing LB, Johansson B, Nilsson SE, Berg S, Pedersen NL, Gatz M, McClearn G. Diabetes mellitus is a risk factor for vascular dementia, but not for Alzheimer’s disease: a population-based study of the oldest old. Int Psychogeriatr. 2002;14(3):239–248. doi: 10.1017/s104161020200844x. [DOI] [PubMed] [Google Scholar]
- Holscher C. The role of GLP-1 in neuronal activity and neurodegeneration. Vitam Horm. 2010;84:331–354. doi: 10.1016/B978-0-12-381517-0.00013-8. [DOI] [PubMed] [Google Scholar]
- Hsu CC, Wahlqvist ML, Lee MS, Tsai HN. Incidence of dementia is increased in type 2 diabetes and reduced by the use of sulfonylureas and metformin. J Alzheimers Dis. 2011;24(3):485–493. doi: 10.3233/JAD-2011-101524. [DOI] [PubMed] [Google Scholar]
- Imfeld P, Bodmer M, Jick SS, Meier CR. Metformin, other antidiabetic drugs, and risk of Alzheimer’s disease: a population-based case-control study. J Am Geriatr Soc. 2012;60(5):916–921. doi: 10.1111/j.1532-5415.2012.03916.x. [DOI] [PubMed] [Google Scholar]
- Janson J, Laedtke T, Parisi JE, O’Brien P, Petersen RC, Butler PC. Increased risk of type 2 diabetes in Alzheimer disease. Diabetes. 2004;53(2):474–481. doi: 10.2337/diabetes.53.2.474. [DOI] [PubMed] [Google Scholar]
- Kalaria RN. Neurodegenerative disease: Diabetes, microvascular pathology and Alzheimer disease. Nat Rev Neurol. 2009;5(6):305–306. doi: 10.1038/nrneurol.2009.72. [DOI] [PubMed] [Google Scholar]
- Kickstein E, Krauss S, Thornhill P, Rutschow D, Zeller R, Sharkey J, Williamson R, Fuchs M, Kohler A, Glossmann H, Schneider R, Sutherland C, Schweiger S. Biguanide metformin acts on tau phosphorylation via mTOR/protein phosphatase 2A (PP2A) signaling. Proc Natl Acad Sci U S A. 2010;107(50):21830–21835. doi: 10.1073/pnas.0912793107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitabchi AE, Temprosa M, Knowler WC, Kahn SE, Fowler SE, Haffner SM, Andres R, Saudek C, Edelstein SL, Arakaki R, Murphy MB, Shamoon H. Role of insulin secretion and sensitivity in the evolution of type 2 diabetes in the diabetes prevention program: effects of lifestyle intervention and metformin. Diabetes. 2005;54(8):2404–2414. doi: 10.2337/diabetes.54.8.2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knopman D, Boland LL, Mosley T, Howard G, Liao D, Szklo M, McGovern P, Folsom AR. Cardiovascular risk factors and cognitive decline in middle-aged adults. Neurology. 2001;56(1):42–48. doi: 10.1212/wnl.56.1.42. [DOI] [PubMed] [Google Scholar]
- Krogh-Madsen R, Plomgaard P, Keller P, Keller C, Pedersen BK. Insulin stimulates interleukin-6 and tumor necrosis factor-alpha gene expression in human subcutaneous adipose tissue. Am J Physiol Endocrinol Metab. 2004;286(2):E234–238. doi: 10.1152/ajpendo.00274.2003. [DOI] [PubMed] [Google Scholar]
- Labuzek K, Suchy D, Gabryel B, Bielecka A, Liber S, Okopien B. Quantification of metformin by the HPLC method in brain regions, cerebrospinal fluid and plasma of rats treated with lipopolysaccharide. Pharmacol Rep. 2010;62(5):956–965. doi: 10.1016/s1734-1140(10)70357-1. [DOI] [PubMed] [Google Scholar]
- Lehmann JM, Lenhard JM, Oliver BB, Ringold GM, Kliewer SA. Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs. J Biol Chem. 1997;272(6):3406–3410. doi: 10.1074/jbc.272.6.3406. [DOI] [PubMed] [Google Scholar]
- Leibson CL, Rocca WA, Hanson VA, Cha R, Kokmen E, O’Brien PC, Palumbo PJ. The risk of dementia among persons with diabetes mellitus: a population-based cohort study. Ann N Y Acad Sci. 1997;826:422–427. doi: 10.1111/j.1749-6632.1997.tb48496.x. [DOI] [PubMed] [Google Scholar]
- Li J, Deng J, Sheng W, Zuo Z. Metformin attenuates Alzheimer’s disease-like neuropathology in obese, leptin-resistant mice. Pharmacol Biochem Behav. 2012;101(4):564–574. doi: 10.1016/j.pbb.2012.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim GP, Yang F, Chu T, Chen P, Beech W, Teter B, Tran T, Ubeda O, Ashe KH, Frautschy SA, Cole GM. Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer’s disease. J Neurosci. 2000;20(15):5709–5714. doi: 10.1523/JNEUROSCI.20-15-05709.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logroscino G, Kang JH, Grodstein F. Prospective study of type 2 diabetes and cognitive decline in women aged 70–81 years. BMJ. 2004;328(7439):548. doi: 10.1136/bmj.37977.495729.EE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luchsinger JA, Reitz C, Patel B, Tang MX, Manly JJ, Mayeux R. Relation of diabetes to mild cognitive impairment. Arch Neurol. 2007;64(4):570–575. doi: 10.1001/archneur.64.4.570. [DOI] [PubMed] [Google Scholar]
- MacKnight C, Rockwood K, Awalt E, McDowell I. Diabetes mellitus and the risk of dementia, Alzheimer’s disease and vascular cognitive impairment in the Canadian Study of Health and Aging. Dement Geriatr Cogn Disord. 2002;14(2):77–83. doi: 10.1159/000064928. [DOI] [PubMed] [Google Scholar]
- Maeshiba Y, Kiyota Y, Yamashita K, Yoshimura Y, Motohashi M, Tanayama S. Disposition of the new antidiabetic agent pioglitazone in rats, dogs, and monkeys. Arzneimittelforschung. 1997;47(1):29–35. [PubMed] [Google Scholar]
- Mannucci E, Monami M, Di Bari M, Lamanna C, Gori F, Gensini GF, Marchionni N. Cardiac safety profile of rosiglitazone: a comprehensive meta-analysis of randomized clinical trials. Int J Cardiol. 2010;143(2):135–140. doi: 10.1016/j.ijcard.2009.01.064. [DOI] [PubMed] [Google Scholar]
- Nandipati S, Luo X, Schimming C, Grossman HT, Sano M. Cognition in non-demented diabetic older adults. Curr Aging Sci. 2012;5(2):131–135. doi: 10.2174/1874609811205020131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicolakakis N, Aboulkassim T, Ongali B, Lecrux C, Fernandes P, Rosa-Neto P, Tong XK, Hamel E. Complete rescue of cerebrovascular function in aged Alzheimer’s disease transgenic mice by antioxidants and pioglitazone, a peroxisome proliferator-activated receptor gamma agonist. J Neurosci. 2008;28(37):9287–9296. doi: 10.1523/JNEUROSCI.3348-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Brien JT. Vascular cognitive impairment. Am J Geriatr Psychiatry. 2006;14(9):724–733. doi: 10.1097/01.JGP.0000231780.44684.7e. [DOI] [PubMed] [Google Scholar]
- O’Reilly JA, Lynch M. Rosiglitazone improves spatial memory and decreases insoluble Abeta(1–42) in APP/PS1 mice. J Neuroimmune Pharmacol. 2012;7(1):140–144. doi: 10.1007/s11481-011-9282-7. [DOI] [PubMed] [Google Scholar]
- Olefsky JM. Treatment of insulin resistance with peroxisome proliferator-activated receptor gamma agonists. J Clin Invest. 2000;106(4):467–472. doi: 10.1172/JCI10843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM. Diabetes mellitus and the risk of dementia: The Rotterdam Study. Neurology. 1999;53(9):1937–1942. doi: 10.1212/wnl.53.9.1937. [DOI] [PubMed] [Google Scholar]
- Park CR, Seeley RJ, Craft S, Woods SC. Intracerebroventricular insulin enhances memory in a passive-avoidance task. Physiol Behav. 2000;68(4):509–514. doi: 10.1016/s0031-9384(99)00220-6. [DOI] [PubMed] [Google Scholar]
- Pedersen WA, McMillan PJ, Kulstad JJ, Leverenz JB, Craft S, Haynatzki GR. Rosiglitazone attenuates learning and memory deficits in Tg2576 Alzheimer mice. Exp Neurol. 2006;199(2):265–273. doi: 10.1016/j.expneurol.2006.01.018. [DOI] [PubMed] [Google Scholar]
- Peila R, Rodriguez BL, Launer LJ. Type 2 diabetes, APOE gene, and the risk for dementia and related pathologies: The Honolulu-Asia Aging Study. Diabetes. 2002;51 (4):1256–1262. doi: 10.2337/diabetes.51.4.1256. [DOI] [PubMed] [Google Scholar]
- Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen E. Mild cognitive impairment: clinical characterization and outcome. Arch Neurol. 1999;56 (3):303–308. doi: 10.1001/archneur.56.3.303. [DOI] [PubMed] [Google Scholar]
- Pintana H, Apaijai N, Pratchayasakul W, Chattipakorn N, Chattipakorn SC. Effects of metformin on learning and memory behaviors and brain mitochondrial functions in high fat diet induced insulin resistant rats. Life Sci. 2012;91(11–12):409–414. doi: 10.1016/j.lfs.2012.08.017. [DOI] [PubMed] [Google Scholar]
- Ravona-Springer R, Luo X, Schmeidler J, Wysocki M, Lesser G, Rapp M, Dahlman K, Grossman H, Haroutunian V, Schnaider Beeri M. Diabetes is associated with increased rate of cognitive decline in questionably demented elderly. Dement Geriatr Cogn Disord. 2010;29(1):68–74. doi: 10.1159/000265552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ravona-Springer R, Moshier E, Schmeidler J, Godbold J, Akrivos J, Rapp M, Grossman HT, Wysocki M, Silverman JM, Haroutunian V, Beeri MS. Changes in glycemic control are associated with changes in cognition in non-diabetic elderly. J Alzheimers Dis. 2012;30(2):299–309. doi: 10.3233/JAD-2012-120106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reger MA, Watson GS, Green PS, Baker LD, Cholerton B, Fishel MA, Plymate SR, Cherrier MM, Schellenberg GD, Frey WH, 2nd, Craft S. Intranasal insulin administration dose-dependently modulates verbal memory and plasma amyloid-beta in memory-impaired older adults. J Alzheimers Dis. 2008a;13(3):323–331. doi: 10.3233/jad-2008-13309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reger MA, Watson GS, Green PS, Wilkinson CW, Baker LD, Cholerton B, Fishel MA, Plymate SR, Breitner JC, DeGroodt W, Mehta P, Craft S. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology. 2008b;70(6):440–448. doi: 10.1212/01.WNL.0000265401.62434.36. [DOI] [PubMed] [Google Scholar]
- Risner ME, Saunders AM, Altman JF, Ormandy GC, Craft S, Foley IM, Zvartau-Hind ME, Hosford DA, Roses AD. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer’s disease. Pharmacogenomics J. 2006;6(4):246–254. doi: 10.1038/sj.tpj.6500369. [DOI] [PubMed] [Google Scholar]
- Roberts RO, Geda YE, Knopman DS, Christianson TJ, Pankratz VS, Boeve BF, Vella A, Rocca WA, Petersen RC. Association of duration and severity of diabetes mellitus with mild cognitive impairment. Arch Neurol. 2008;65(8):1066–1073. doi: 10.1001/archneur.65.8.1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodriguez-Rivera J, Denner L, Dineley KT. Rosiglitazone reversal of Tg2576 cognitive deficits is independent of peripheral gluco-regulatory status. Behav Brain Res. 2011;216(1):255–261. doi: 10.1016/j.bbr.2010.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rogers J, Shen Y. A perspective on inflammation in Alzheimer’s disease. Ann N Y Acad Sci. 2000;924:132–135. doi: 10.1111/j.1749-6632.2000.tb05571.x. [DOI] [PubMed] [Google Scholar]
- Sanz C, Andrieu S, Sinclair A, Hanaire H, Vellas B. Diabetes is associated with a slower rate of cognitive decline in Alzheimer disease. Neurology. 2009;73(17):1359–1366. doi: 10.1212/WNL.0b013e3181bd80e9. [DOI] [PubMed] [Google Scholar]
- Sanz CM, Hanaire H, Vellas BJ, Sinclair AJ, Andrieu S. Diabetes mellitus as a modulator of functional impairment and decline in Alzheimer’s disease. The Real.FR cohort. Diabet Med. 2012;29(4):541–548. doi: 10.1111/j.1464-5491.2011.03445.x. [DOI] [PubMed] [Google Scholar]
- Sato T, Hanyu H, Hirao K, Kanetaka H, Sakurai H, Iwamoto T. Efficacy of PPAR-gamma agonist pioglitazone in mild Alzheimer disease. Neurobiol Aging. 2011;32 (9):1626–1633. doi: 10.1016/j.neurobiolaging.2009.10.009. [DOI] [PubMed] [Google Scholar]
- Schnaider Beeri M, Goldbourt U, Silverman JM, Noy S, Schmeidler J, Ravona-Springer R, Sverdlick A, Davidson M. Diabetes mellitus in midlife and the risk of dementia three decades later. Neurology. 2004;63(10):1902–1907. doi: 10.1212/01.wnl.0000144278.79488.dd. [DOI] [PubMed] [Google Scholar]
- Searcy JL, Phelps JT, Pancani T, Kadish I, Popovic J, Anderson KL, Beckett TL, Murphy MP, Chen KC, Blalock EM, Landfield PW, Porter NM, Thibault O. Long-term pioglitazone treatment improves learning and attenuates pathological markers in a mouse model of Alzheimer’s disease. J Alzheimers Dis. 2012;30(4):943–961. doi: 10.3233/JAD-2012-111661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sheu WH, Chuang HC, Cheng SM, Lee MR, Chou CC, Cheng FC. Microdialysis combined blood sampling technique for the determination of rosiglitazone and glucose in brain and blood of gerbils subjected to cerebral ischemia. J Pharm Biomed Anal. 2011;54(4):759–764. doi: 10.1016/j.jpba.2010.10.008. [DOI] [PubMed] [Google Scholar]
- Sivertsen J, Rosenmeier J, Holst JJ, Vilsboll T. The effect of glucagon-like peptide 1 on cardiovascular risk. Nat Rev Cardiol. 2012;9(4):209–222. doi: 10.1038/nrcardio.2011.211. [DOI] [PubMed] [Google Scholar]
- Solfrizzi V, Panza F, Colacicco AM, D’Introno A, Capurso C, Torres F, Grigoletto F, Maggi S, Del Parigi A, Reiman EM, Caselli RJ, Scafato E, Farchi G, Capurso A. Vascular risk factors, incidence of MCI, and rates of progression to dementia. Neurology. 2004;63(10):1882–1891. doi: 10.1212/01.wnl.0000144281.38555.e3. [DOI] [PubMed] [Google Scholar]
- Strum JC, Shehee R, Virley D, Richardson J, Mattie M, Selley P, Ghosh S, Nock C, Saunders A, Roses A. Rosiglitazone induces mitochondrial biogenesis in mouse brain. J Alzheimers Dis. 2007;11(1):45–51. doi: 10.3233/jad-2007-11108. [DOI] [PubMed] [Google Scholar]
- Tian J, Shi J, Bailey K, Lendon CL, Pickering-Brown SM, Mann DM. Association between apolipoprotein E e4 allele and arteriosclerosis, cerebral amyloid angiopathy, and cerebral white matter damage in Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2004;75(5):696–699. doi: 10.1136/jnnp.2003.012096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van den Berg E, de Craen AJ, Biessels GJ, Gussekloo J, Westendorp RG. The impact of diabetes mellitus on cognitive decline in the oldest of the old: a prospective population-based study. Diabetologia. 2006;49(9):2015–2023. doi: 10.1007/s00125-006-0333-1. [DOI] [PubMed] [Google Scholar]
- Wang KC, Woung LC, Tsai MT, Liu CC, Su YH, Li CY. Risk of Alzheimer’s disease in relation to diabetes: a population-based cohort study. Neuroepidemiology. 2012;38(4):237–244. doi: 10.1159/000337428. [DOI] [PubMed] [Google Scholar]
- Watson GS, Cholerton BA, Reger MA, Baker LD, Plymate SR, Asthana S, Fishel MA, Kulstad JJ, Green PS, Cook DG, Kahn SE, Keeling ML, Craft S. 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(11):950–958. doi: 10.1176/appi.ajgp.13.11.950. [DOI] [PubMed] [Google Scholar]
- Watson GS, Peskind ER, Asthana S, Purganan K, Wait C, Chapman D, Schwartz MW, Plymate S, Craft S. Insulin increases CSF Abeta42 levels in normal older adults. Neurology. 2003;60(12):1899–1903. doi: 10.1212/01.wnl.0000065916.25128.25. [DOI] [PubMed] [Google Scholar]
- White MF. Insulin signaling in health and disease. Science. 2003;302(5651):1710–1711. doi: 10.1126/science.1092952. [DOI] [PubMed] [Google Scholar]
- Whitmer RA, Sidney S, Selby J, Johnston SC, Yaffe K. Midlife cardiovascular risk factors and risk of dementia in late life. Neurology. 2005;64(2):277–281. doi: 10.1212/01.WNL.0000149519.47454.F2. [DOI] [PubMed] [Google Scholar]
- Wu JH, Haan MN, Liang J, Ghosh D, Gonzalez HM, Herman WH. Impact of antidiabetic medications on physical and cognitive functioning of older Mexican Americans with diabetes mellitus: a population-based cohort study. Ann Epidemiol. 2003;13 (5):369–376. doi: 10.1016/s1047-2797(02)00464-7. [DOI] [PubMed] [Google Scholar]
- Xu W, Qiu C, Winblad B, Fratiglioni L. The effect of borderline diabetes on the risk of dementia and Alzheimer’s disease. Diabetes. 2007;56(1):211–216. doi: 10.2337/db06-0879. [DOI] [PubMed] [Google Scholar]
- Xu WL, Qiu CX, Wahlin A, Winblad B, Fratiglioni L. Diabetes mellitus and risk of dementia in the Kungsholmen project: a 6-year follow-up study. Neurology. 2004;63(7):1181–1186. doi: 10.1212/01.wnl.0000140291.86406.d1. [DOI] [PubMed] [Google Scholar]
- Yaffe K, Blackwell T, Kanaya AM, Davidowitz N, Barrett-Connor E, Krueger K. Diabetes, impaired fasting glucose, and development of cognitive impairment in older women. Neurology. 2004;63(4):658–663. doi: 10.1212/01.wnl.0000134666.64593.ba. [DOI] [PubMed] [Google Scholar]