291

Journal of Alzheimer’s Disease 32 (2012) 291–305 DOI 10.3233/JAD-2012-120571 IOS Press

Amyloid-␤ and Tau Pathology of Alzheimer’s Disease Induced by Diabetes in a Rabbit Animal Model Claudine L. Bitela , Chinnaswamy Kasinathanb , Rajesh H. Kaswalab , William L. Kleinc and Peter H. Frederiksea,b,∗ a Department

of Pharmacology and Physiology and Rutgers-UMDNJ Integrative Neurosciences Program, UMDNJ-New Jersey Medical School, Newark, NJ, USA b Department of Oral Biology, UMDNJ-New Jersey Medical School, Newark, NJ, USA c Department of Neurobiology and Physiology, Northwestern University, Evanston, IL, USA

Accepted 6 June 2012

Abstract. Alzheimer’s disease (AD) is the major age-dependent disease of the brain, but what instigates late-onset AD is not yet clear. Epidemiological, animal model, and cell biology findings suggest links between AD and diabetes. Although AD pathology is accelerated by diabetes in mice engineered to accumulate human-sequence amyloid-␤ (A␤) peptides, they do not adequately model non-inherited AD. We investigated AD-type pathology induced solely by diabetes in genetically unmodified rabbits which generate human-sequence A␤ peptides. After 15 weeks, alloxan-treated diabetic rabbits with expected high blood glucose showed ∼5-fold increase in A␤40 /A␤42 in cortex and hippocampus, and significantly, generated A␤-derived assemblies found in human AD. Deposits of these putative pathogenic toxins were detected by A␤/A␤ oligomer antibodies in brain parenchyma and surrounding vasculature, also co-localizing with markedly elevated levels of RAGE. Soluble brain extracts showed diabetesinduced buildup of A␤ oligomers on dot-blots. Phospho-tau also was clearly elevated, overlapping with ␤III-tubulin along neuronal tracts. Indications of retina involvement in AD led to examination of AD-type pathology in diabetic retinas and showed A␤ accumulation in ganglion and inner nuclear cell layers using A␤/oligomer antibodies, and RAGE again was elevated. Our study identifies emergence of AD pathology in brain and retina as a major consequence of diabetes; implicating dysfunctional insulin signaling in late-onset AD, and a potential relationship between A␤-derived neurotoxins and retinal degeneration in aging and diabetes, as well as AD. AD-type pathology demonstrated in genetically unmodified rabbits calls attention to the considerable potential of the model for investigation of AD pathogenesis, diagnostics, and therapeutics. Keywords: Alzheimer’s disease, amyloid-␤, brain, diabetes, oligomers, retina

INTRODUCTION Alzheimer’s disease (AD) is a progressive brain disorder constituting the major cause of dementia as well as the sixth leading cause of death [1]. Its molecular ∗ Correspondence

to: Peter H. Frederikse, PhD, Pharmacology and Physiology and Rutgers-UMDNJ Integrative Neuroscience Program, UMDNJ New Jersey Medical School, 185 South Orange Avenue MSB, H645, Newark, NJ 07103, USA. Tel.: +1 973 972 1686 lab-4785; Fax: +1 973 972 7950; E-mail: [email protected].

basis is linked to accumulation of pathogenic toxins [2, 3]. What is not yet evident, except for a very small subpopulation of patients with familial AD (FAD), is how this accumulation of toxins is instigated. In the 12% of patients with FAD, various point mutations lead to elevated production of the amyloid-␤ (A␤) peptide [4], ordinarily a physiological metabolite, but which at elevated concentrations drives its self-association into gain-of-function toxic assemblies. These toxins comprise the well-known insoluble amyloid plaques

ISSN 1387-2877/12/$27.50 © 2012 – IOS Press and the authors. All rights reserved

292

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

and the more recently discovered toxic soluble A␤ oligomers, currently a focus of interest for their role early in dementia [3, 5]. Overproduction of A␤ in FAD has been modeled in rodents expressing human transgenes, which give rise to plaques, toxic oligomers, and ensuing behavioral dysfunction and neuropathology [6]. Transgenic animals have limited value, however, for elucidating mechanisms that generate these toxins in sporadic AD (SAD), which comprises the great majority of cases. Current models also have been illsuited for predicting whether investigational new drugs will be successful in treating SAD [7, 8]. An emerging possibility now garnering attention is that initiation and development of SAD, at least in some cases, may be related to diabetes [9]. Recent etiological data have identified type 2 diabetes as an AD risk factor [10]. Because AD brain is relatively insulin resistant [10, 11], and insulin treatment can enhance memory function in some AD patients [12, 13], it also has been suggested that AD could be regarded as a type 3 diabetes [14]. This is consistent with findings that brain insulin has a role in synaptic plasticity as well as energy metabolism [15]. At the molecular level, synaptic insulin receptors are greatly down-regulated by toxic A␤ oligomer binding [2], a loss that has been attributed to their impact on receptor trafficking [16, 17]. Reciprocally, lack of insulin signaling renders neurons more vulnerable to oligomer synaptotoxicity [18, 19], suggestive of a vicious cycle [20]. Whether deficiencies in insulin signaling could act at even earlier stages, as an instigator of AD pathogenesis, is uncertain. Because insulin plays a role in clearance of A␤ monomers and oligomers at synapses, it is feasible that failed brain insulin signaling could promote formation of A␤ synaptic insulin receptor complexes leading to the down-regulation of insulin receptors by these A␤-derived toxins. Consistent with this possibility, diabetes causes the onset of amyloid pathology to develop earlier in transgenic mouse AD models [21]. In the current study, the relationship between diabetes and the onset of AD has been examined in a non-transgenic animal to segregate the intrinsic effects of insulin deficiency from effects also requiring genetic manipulations. Our experiments used genetically unmodified rabbits which, unlike rodents, produce A␤ peptides identical in sequence to those expressed in human brain [22]. Diabetes induced due to degeneration of pancreatic beta-cells with alloxan [23, 24] was found to produce AD-type neuropathology in both brain and retina of wild-type rabbits, with significant accumulation of A␤ monomers and oligomers and

of aberrant phospho-tau. Results strongly support the hypothesis that diabetes can act as a primary factor in inducing early-stage AD phenotype. Results further suggest the potential of the wild type rabbit as a model for aspects of sporadic AD, a new resource for investigating disease initiation and treatment strategies.

MATERIALS AND METHODS Induction of diabetes in rabbits New Zealand white rabbits (Covance) were used according to NIH guidelines and University approved protocols. Seven rabbits were included per group, and males only were used to limit hormonal considerations at this stage of our studies. The study was repeated with a second group of rabbits and replicated our findings. 3-4 month-old rabbits were housed at room temperature with a 12 h light/12 h dark cycle and fed 260 gm/day of a standard pellet diet with free access to dH2 O. Rabbits were given 150 mg/kg alloxan (Sigma) in a single bolus injection through an ear vein catheter. Alloxan is structurally related to streptozotocin, and both toxic glucose analogues selectively enter pancreatic ␤-cells via GLUT2 glucose transporters [23, 25]. Alloxan predominantly produces reactive oxygen species consistent with its 1-2 min half-life and rapid clearance, with GLUT1 expressed at the blood brain barrier and GLUT3 in neurons [26, 27]. Fasting blood glucose was measured with a glucometer four times per week. Rabbits maintaining >350 mg/dl blood glucose remained in the study, and rabbits with lower glucose values were removed. Rabbits weighed ∼2.75 kg at the outset of our study. Diabetic rabbits gained ∼225 gm (s.d. 180 gm), and controls gained ∼500 gm (s.d. 50 gm), with no rabbits losing weight. Retina and brain were examined after 15 wks, similar to related studies of diabetic complications in alloxan-treated rabbits [e.g., 28]. Brains were removed for study and bisected for tissue fixation or immersed in liquid nitrogen for biochemical studies. Enucleated eyes were fixed in buffered formalin for preparation of histological sections. Serum cholesterol at the end of the 15 wk trial showed a range of increases in diabetic animals when compared with controls (Antech Diagnostics, NY): Cholesterol in normal controls 21–40 mg/dl (S.D. 11), showed increases in diabetic rabbit serum that varied from 39 to 498 mg/dl (S.D. 101), and appeared consistent with related studies in alloxan-treated rabbits [29].

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

Immunochemistry and immunofluorescence Paraffin sections prepared from paraformaldehyde fixed brains and eyes were de-waxed and endogenous peroxidases were quenched in 3% H2 O2 in 10% MeOH for 5 min. Sections were blocked in PBS, 0.01% tween-20, 3% BSA for 60 min, or PBS with 10% normal serum corresponding to the 2◦ antibody for immunofluorescence. HRP conjugated 2◦ antibodies (Vector Labs) or fluor-conjugated 2o antibodies (Invitrogen) were used to visualize immune complexes. Antibodies used included: mAb anti-A␤ 6E10 and 4G8 (Covance), rabbit mAb anti-A␤42 (Invitrogen), and anti-A␤40 (Genscript). Anti-A␤ oligomer antibodies included NU1, NU4 [30, 31], A11, and OC (Invitrogen, gift of C. Glabe, UC Irvine) [32], as well as mouse anti-A␤ oligomer IgM (Agrisera). Additional antibodies included anti-RAGE (Santa Cruz, Genscript), mAb ␤III-tubulin (Sigma), and GAPDH (Abcam). Immunoblots used equal amounts of protein samples resolved by MW (NuPAGE, Invitrogen) blotted to filters and probed with antibodies using supplied protocols. Immune complexes were visualized with HRP-conjugated 2◦ antibodies (Jackson) and chemiluminescence kits (Amersham).

293

4 ␮g and 1.5 ␮g of the first PBS soluble fraction and final RIPA insoluble fraction were spotted onto nitrocellulose filters, and probed with A11, OC or 4G8 antibodies, or stained for total protein with amido black. RESULTS Aβ40 and Aβ42 peptides increased in the cortex and hippocampus of diabetic rabbit brains Fifteen weeks after onset of reduced insulin production and sustained hyperglycemia, we used standard ELISAs to measure A␤40 and A␤42 peptides in tissue samples from diabetic and normal control brains. Both A␤ forms increased several fold in the cortex and hippocampus in diabetic rabbit brains in total protein samples solubilized in 5M guanidine buffer (Fig. 1),

ELISA assays A␤ was quantified in cortex and hippocampus tissues using ELISA assays and protocols specific for A␤40 or A␤42 and peptide standards provided by the supplier (Covance). Equal brain total protein samples solubilized in 5M guanidine were assayed by colorimetric quantification of HRP product with a spectrometer plate reader. ELISAs were performed 3X to measure A␤40 and A␤42 for each sample. For analysis of ELISA data, p-values were calculated using a 2-tailed ANOVA statistical test. Immuno dot-blots Small soluble A␤ oligomeric forms were assayed using nitrocellulose filter dot blot assays as described by Tomic et al. [3]. A␤ peptides and soluble oligomers were extracted from ∼60 mg brain tissue by repeated pipetting in 4 volumes of ice-cold non-denaturing PBS [A buffer: PBS, 0.02% NaN3 , pH 7.4 and protease inhibitors (Calbiochem)], and centrifuged at 100K×G for 1 h. After taking supernatants for assay, the pellets were resuspended in 4 volumes RIPA buffer (Sigma) and centrifuged again as above. This RIPA insoluble pellet was again resuspended in A buffer for assay.

Fig. 1. A␤40 and A␤42 peptides increased significantly in brain cortex and hippocampus of genetically un-modified diabetic rabbits. A␤40 and A␤42 peptides were assayed separately by ELISA in total protein samples from cortex and hippocampus in four diabetic and four control rabbits. A␤40 cortex: p < 0.03, A␤40 hippocampus: p < 0.005, A␤42 cortex: p < 0.05, A␤42 hippocampus: p < 0.03.

294

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

Fig. 2. Pathological A␤ accumulations in the hippocampus and cortex in diabetic rabbit brain tissues, and normoglycemic control brains detected via immunohistochemistry. A␤40 and A␤42 showed similar distributions in histological sections in cortex and hippocampus from genetically unmodified diabetic rabbits using peptide-specific antibodies. Brain tissues were examined 15 weeks after induction of reduced insulin production and onset of hyperglycemia. Photographs are representative of seven diabetic and seven control rabbits in each of two experimental trials.

and more deleterious A␤42 peptides increased to a greater extent than A␤40 in both regions. A␤40 was ∼3–5-fold higher in diabetic brains, whereas A␤42 was detected at lower levels in control brain tissue and increased >5-fold in the cortex and hippocampus in diabetic animals. When we examined diabetic brains in situ for hallmark A␤ accumulations similar to what occurs in AD brains, we observed A␤ deposits in many areas in the cortex and hippocampus from wild-type (wt) diabetic rabbits. In contrast, little or no evidence of A␤ deposits was observed in control rabbit brains using mAb anti-A␤ 6E10 and 4G8 antibodies or with anti-A␤42 peptide-specific antibodies recognizing a Cterminus epitope (Fig. 2). Deposits were observed in the brain parenchyma and associated with capillary walls in the cortex and hippocampus similar to what has been described in human AD brains and transgenic (Tg) AD mouse models [33, 34]. A␤ deposits varied in size from small deposits that appeared to be contained within a cell or lysis of a single cell, to larger deposits and may suggest bystander effects on adjacent cells. A␤ was also seen in accumulations surrounding cell nuclei in apparently intact neurons in different brain regions. When we compared the distribution of specific A␤ peptide forms in diabetic brains with anti-A␤42

and A␤40 specific antibodies, we observed similar distributions of A␤ deposits with both antibodies in the cortex and hippocampus of diabetic brains (Fig. 2), and again found little or no evidence of A␤ deposits in either of these brain regions in euglycemic control 7-8 month-old rabbit brains. Spontaneous Aβ conversion to oligomeric forms in vivo in wt diabetic rabbit brains with evidence of Aβ oligomer accumulation in brain The ability of ∼4 kDa A␤ monomers to form small diffusible A␤ oligomers which can be ∼60 kDa or greater in size has been characterized by a number of laboratories [3, 30, 31]. To date, human-sequence A␤ peptides have been used to produce oligomers that elicit production of conformation-specific antibodies which recognize A␤ oligomers. Observations of substantially increased human-sequence A␤ peptides in diabetic rabbit brains indicated significant potential for oligomer formation in vivo. To examine A␤ oligomers in diabetic brains, we probed brain sections with a number of anti-A␤ oligomer antibodies produced in different laboratories. Similar to findings with anti-A␤ peptide antibodies above, few or no oligomers were

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

detected in brain tissue or surrounding vessels in normoglycemic control brains using NU1 or NU4 mAb antibodies, or A11 or OC anti-A␤ oligomer rabbit antibodies [32] that have been shown capable of preferentially detecting higher order A␤ structures (Fig. 3). In contrast, a similar distribution of A␤ oligomer deposits as shown above using anti-A␤ detection was detected throughout the cortex and hippocampus of diabetic brains, and associated with vessel walls of the surrounding microvasculature using anti-A␤ oligomer antibodies (Fig. 3). Deposits which we could orient similarly in adjacent histological sections were identified with anti-A␤ and A␤ oligomer antibodies. In addition to A␤/ADDL accumulations in the cortex and hippocampus, similar deposits were also observed in other brain areas including the ventromedial hypothalamus (not shown). This brain region has also been shown to be affected in human AD brains [35, 36] and may have particular relevance in diabetes in light of its previously described roles in glucose sensing and regulating glucose homeostasis [37]. To further characterize A␤ peptide and ADDL distributions, we used in situ immunfluorescence with A␤42 specific antibodies and mouse IgM mAb anti-oligomer (OMAB) antibodies [38] to examine brain pathology (Fig. 3 C). These antibodies also demonstrated substantial overlap in detection of A␤ peptides and oligomers in the cortex and hippocampus of diabetic animals. Our results indicate significant ADDL formation occurs in diabetic rabbit brains that we could detect using mouse and rabbit IgG as well as IgM anti-A␤ oligomer antibodies. Immuno dot-blot assays of brain proteins detected increased PBS extractable oligomeric Aβ in diabetic brains Recent studies showed that in contrast to harsh guanidine denaturing buffers used in ELISAs above, oligomeric A␤ forms in brain tissue are extracted by repeated pipetting in phosphate buffered saline (PBS). A␤ oligomers obtained with these methods were shown to increase in human AD brains when compared with non-demented controls, detected on dot-blots using A␤ oligomer-specific antibodies [3]. To test these findings in the rabbit model, and to provide further evidence of A␤ conversion to oligomeric forms in diabetic brains, we used a similar dot-blot immunodetection protocol to analyze A␤ higher order assemblies eluted from diabetic and control rabbit brain tissues (Fig. 4). Aliquots of non-denatured proteins in PBS extracted brain tissue were compared with protein material in a detergent insoluble pellet

295

fraction after ultracentrifugation, by spotting protein samples onto filters which were then were probed with anti-oligomer A11 and OC antibodies, and antiA␤ 4G8. After immunohistochemical detection, we observed increased levels of soluble oligomeric A␤ forms in diabetic brains using OC as well as A11 anti-A␤ oligomer antibodies. Increased RAGE expression and localized accumulation with Aβ/oligomers occurs in cortical and hippocampal regions and vessels in diabetic brains Advanced glycation end-products (AGEs) result from non-enzymatic addition of sugar units to a variety of macromolecules and increase significantly with hyperglycemia in diabetes. These increases have been shown to be matched by increased RAGE scavenger receptors [39–41]. To examine these effects in diabetic rabbit brains, we measured RAGE expression on immunoblots and observed a significant increase from low baseline expression in diabetic brains (Fig. 5), in contrast to little apparent overall change in IR␣ or GAPDH. We note the GAPDH doublet on immunoblots was also indicated by the antibody supplier. When we examined RAGE protein in situ, we observed that RAGE accumulation was also readily detected in deposits in the cortex and hippocampus in diabetic brains but not control brain tissue, as were also observed in surrounding vascular structures (Fig. 5). Increased phosphorylated tau occurs in diabetic brains and was detected predominantly along neuronal tracts co-localizing with βIII-tubulin Tau pathology is also a well-characterized fundamental neuropathological characteristic of AD. Phosphorylation at tau serine and threonine residues associated with AD and other neurodegenerative conditions also increase in response to oxidative and osmotic stress [42, 43]. In addition, tau phosphorylation was found to correlate with A␤ oligomer toxicity [44]. We examined tau phosphorylation in brain samples from diabetic and control brains, probing for phosphorylated threonine 205 and phospho-serines 396 and 404 on immunoblots (Fig. 6). We observed increased phosphorylation at residues threonine 205 and serine 404 in brain proteins from diabetic rabbits. When we next examined the localization of phosphorylated tau protein in situ in diabetic brains, we observed increased phospho-tau that corresponded with axonal tracts in the cortex of diabetic animals

296

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

Fig. 3. Accumulation of A␤ peptides and A␤ oligomers detected in diabetic rabbit brain. A) Immunohistochemical detection of A␤ oligomers (NU4) in pathological deposits in control (Ctl, bar: 200 ␮m) and diabetic cortex (Diab, bar: 100 ␮m). B) Detection of A␤ (6E10, A␤42 ) and A␤ oligomers (NU4, A11) in diabetic cortex and hippocampus (upper bar: 50 ␮m, lower bar: 200 ␮m). C) Immunofluorescence assay of A␤ oligomers (IgM mA␤) and A␤42 in cortex and hippocampus (upper bar: 25 ␮m, lower bar: 50 ␮m). Photographs are representative of seven diabetic and seven control rabbits.

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

297

Fig. 4. Dot-blot analysis of soluble A␤ oligomeric forms in diabetic and control brain samples identified greater A␤ oligomers in diabetic brains. Brain tissue samples from two control and two diabetic rabbits homogenized in PBS were centrifuged at 100K×g. Pellets were re-suspended in RIPA buffer and centrifuged as above. 4 ␮g and 1.5 ␮g of 1st PBS supernatant (S) or final RIPA insoluble pellet resuspended in PBS (P) were spotted onto nitrocellulose filters and probed with A11 and OC anti-A␤ oligomer antibodies, or stained with Amido Black to visualize total proteins. Data is representative of seven diabetic and seven control rabbits used in two experimental trials.

Fig. 5. Increased RAGE receptor expression in diabetic brain with local accumulation in cortex and hippocampus of diabetic rabbits. Upper right panels show immunoblot analysis of RAGE, IR␣, and GAPDH expression in control and diabetic brain tissue samples. Graph below shows relative densitometry band densities (Dark bar: control. Light bar: diabetic). The remaining panels shows IHC detection of RAGE in control hippocampus (bar = 100 ␮m), diabetic hippocampus (bar: 50 ␮m) and below: Control cortex (bar: 100 ␮m), and diabetic hippocampus (bar: 200 ␮m), Higher magnification view is shown in lower right panel (bar: 50 ␮m). Photographs are representative of two experimental trials that used seven diabetic and seven control rabbits.

using immunohistochemical and immunofluorescence methods (Fig. 7). However, we found little or no evidence of tangles in diabetic brains. To provide further evidence of intra-neuronal phosphorylated tau, we examined the distribution of neuronal ␤III-tubulin (tubb3) along neuronal tracts. We detected an overlapping distribution of phospho-tau and tubb3 in neurons

present in the cortex and hippocampus of diabetic brains (Fig. 7). In the hippocampus, phospho-tau ser404 and thr-205 was observed mainly in neurons surrounding nuclei present in the CA3 area in diabetic rabbits. The present findings of tau hyperphosphorylation with few neurofibrillary tangles formed in diabetic rabbit brains agrees with a recent report that identified

298

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

Fig. 6. Increased tau phosphorylation is detected in diabetic brains. Immunoblots contain equal samples of total brain protein samples from control and diabetic rabbits, resolved by MW. Individual filters were probed with of anti-Tau (tau5), and antibodies recognizing tau phosphorylated at ser-404, ser-396, and thr-205. Graph below shows relative densitometry band densities (Dark bar: control. Light bar: diabetic).

links between insulin resistance and tau AD brain pathology in humans [10]. Corresponding Aβ, Aβ oligomers, and RAGE accumulation and pathology is produced in the retina of diabetic rabbits largely in outer ganglion cell and inner nuclear layers The mammalian retina contains stratified layers of several neural cell types interspersed with Muller glial cells. The present diabetes model predicts that AD-related neurodegeneration and A␤ pathology is

coordinately produced in these neural retina layers together with AD brain pathology described above in diabetic rabbits. To examine retinas in diabetic rabbits, we prepared whole eye histological sections from diabetic and control animals and probed them with anti-A␤ and anti-ADDL antibodies also used above to examine the brain. Analysis of retina morphology at 15 weeks after diabetes onset did not show obvious changes in structure at this time point (Fig. 8). Diabetic retinas probed with mAb 4G8 as well as anti-A␤42 antibodies that recognize a C-terminus epitope on 42 amino acid A␤ peptides both identified A␤ accumulation predominantly in the outer ganglion cell layer (GCL) and inner nuclear layer (INL), and to a lesser degree the inner plexiform layer of the retina (Fig. 8). This distribution of A␤ peptides in diabetic rabbit retinas showed a strikingly similarity to A␤ accumulation reported on in retinas from 14 month-old Tg2576 mice by others [45]. We next probed retinas with mouse (NU2, NU4) and rabbit (A11) anti-A␤ oligomer conformation specific antibodies and observed a similar distribution of A␤ oligomer accumulation with each of these antibodies that overlapped with our detection of A␤ in diabetic retina. ADDLs were detected in the GCL adjacent to the vitreous of the eye, as well as the INL neural layers. The distribution of A␤ and A␤ oligomers in these GCL and INL retinal layers also suggested perinuclear accumulations occurred in the cells, similar to their perinuclear detection in neurons in the brain above. We next probed eye sections with anti-RAGE antibodies, and detected RAGE accumulation in diabetic retinas that was not present in control eyes. RAGE deposits overlapped with the distribution of A␤ and A␤ oligomers in GCL and INL layers of the retina. However, we also identified increased RAGE in photoreceptor cells, and more strongly at the border with the outer nuclear layer of the retina. DISCUSSION At present, factors contributing to pathological increases of A␤ in disease and, as importantly, to the accumulation of its toxic derivatives, are not well understood. This is largely due to the absence of physiological, wild-type animal models. The present study addressed this gap by investigating AD-type neuropathology in genetically unmodified rabbits. Rabbit was chosen as it shares physiological production of human-sequence A␤ peptides throughout the body [22]. Our findings show that AD-type CNS neuropathology is instigated in rabbit by alloxan-induced diabetes. Loss of insulin production and consequent

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

299

Fig. 7. Increased detection of phosphorylated tau protein is observed along axonal tracts in brain cortex of diabetic rabbit brains. A) Immunohistochemical detection of phospho-tau (p-ser404 and p-thr205 in control and diabetic cortex. B) Immunofluorescence detection of phopho-Tau p-ser404 (green) in diabetic cortex. For comparison, anti-␤III-tubulin (red) detected in diabetic cortex shows an overlapping distribution along axonal tracts in diabetic brains. Photographs are representative of seven diabetic and seven control rabbits.

hyperglycemia led to large increases of both A␤40 and A␤42 peptides in the CNS, in hippocampus, cortex, and retina. Phosphorylated tau characteristic of AD pathology also increased. Significantly, diabetic brains in our study manifested the spontaneous accumulation of soluble A␤ oligomers, which are widely implicated in AD-related memory loss and brain cell damage [46–48]. These oligomers, which were assayed here on dot blots of PBS extractable protein from brain tissue and observed in situ in several brain regions using mouse and rabbit IgG and IgM anti-oligomer antibodies, have not previously been detected in a

non-transgenic experimental animal. Results identify rabbit as a new resource to investigate mechanisms that cause accumulation of AD-linked oligomers and related facets of neuropathology. The new findings provide strong support for the hypothesis that systemic insulin deficiency and hyperglycemia are contributing factors to the onset of neuropathology in sporadic AD [48–52], the major form of the disease [53]. Pathological accumulation of A␤ peptides also notably manifests in diabetic rabbit retina. As in brain, there is substantial conversion to A␤ oligomers. The pathology in retina, like brain, is evident at

300

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

Fig. 8. A␤ peptide, A␤ oligomers and RAGE accumulate in retinas of diabetic rabbits, and corresponding with brain pathology described above. Antibodies included anti-RAGE, anti-A␤42 , anti-A␤ mAb 4G8, anti-A␤ oligomer NU2, NU4, and A11. Retinal layers numbered at the upper-left as 1–5 indicate 1: ganglion cell layer, 2: inner plexiform layer, 3: inner nuclear layer, 4: outer nuclear layer, 5: photoreceptor layer.

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

15 weeks after alloxan treatment, which produced hyperglycemia in the animals studied (>350 mg/dl). The distribution of A␤ peptide and oligomer accumulation in diabetic rabbits is remarkably similar to the pattern of A␤ accumulation reported in retinas in Tg2576 mice carrying the human A␤PP transgene [44]. Overlapping distributions of A␤ and oligomers with RAGE deposits were consistently detected in the ganglion cell layer adjacent to the vitreous, and inner nuclear cell layers of the retina. This distribution in rabbit agrees with analysis of human retinas chronically exposed to hyperglycemia [54, 55]. The present findings with diabetic brain and retina are consistent with our earlier study of muscle of alloxan-treated rabbits [56], which showed A␤ accumulation similar to the A␤ muscle pathology linked with aging and muscle disease in humans. Given that A␤ oligomers have been identified in human muscle degenerative disease [57], and that oligomers are firmly linked to neuronal damage in brain [58–63], the current findings suggest these toxins likely also contribute to diabetic retinopathy. Our findings may have added significance in light of studies suggesting that retinal changes may inform about AD onset and progression in humans [64, 65]. Analogous to findings reported for AD brain [33, 66], the immunohistochemistry of vessel walls in diabetic brain showed indications of oligomer deposits. Whether the deposits represent early stages of cerebral amyloid angiopathy due to diabetes is not yet known. More advanced A␤ deposition localized adjacent to vessels walls in the brain has been reported in Tg AD mouse models [34]. Together, the observations are consistent with clinical indications that a majority of AD cases in older patients represent a combination of neurodegeneration and cerebral angiopathy, which may be typical in late-onset AD [67]. We noted here a large diabetes-induced increase in RAGE expression and accumulation in vessels and brain parenchyma, along with the overlap of RAGE with A␤ distribution. These findings are consistent with the hypothesis that RAGE- A␤ binding could have a role in promoting and influencing A␤ accumulation and localization in diabetic brains [39]. Diabetic rabbit brain also showed increased tau phosphorylation but not tau tangles. It previously has been established that there is increased brain phosphotau linked to insulin resistance in human diabetes [68]. The mechanism likely involves dysfunctional control over the GSK3␤ signaling pathway, which acts as a tau kinase [69–71]. Another possibility is stimulation of tau phosphorylation by the diabetes-induced A␤ oligomers. A␤ oligomers appear to be upstream

301

of tau pathology, as oligomers stimulate AD-type tau hyperphosphorylation in cultured neurons [19, 72], and oligomer-specific antibodies reduce tau pathology in transgenic mice [73]. Tau, moreover, is required to mediate aspects of A␤ oligomer-induced neuronal damage [74, 75]. Interestingly, type 1 diabetes in STZtreated wild-type mice and rats consistently manifests tau hyperphosphorylation in brain, but unlike what was found with rabbits, the diabetic rodents show little or no A␤ pathology [76–78]. As seen here, diabetes can instigate major elements of AD neuropathology. Whether there are specific distinctions between diabetes-dependent and other sporadic forms of AD neuropathology involving A␤ peptides and tau proteins remains to be determined. Significantly, the impact of diabetes is likely exacerbated by cellular interactions between neuronal insulin signaling and A␤ oligomer synaptotoxicity [79, 80]. Insulin signaling greatly reduces the binding and toxicity of oligomers [2, 81, 82], so if such signaling were to be lost, neurons would become vulnerable to oligomer exposure [16, 82]. Moreover, once attached to neurons, oligomers cause insulin receptors to be eliminated from surface membranes, and contributes to neuronal insulin resistance, also observed in some AD patients [10, 11, 83]. These phenomena have the potential to create a vicious cycle in which diabetes induces oligomer accumulation, neuron vulnerability to oligomers is elevated because of deficiencies in their insulin signaling, and oligomers exacerbate the deficiency by making neurons even more insulin resistant. Although exploratory efforts are underway to treat AD with insulin and insulin enhancers [83–86], such a vicious cycle indicates it may be difficult to affect ADrelated cognitive failures with single-modality insulin treatments. It has been proposed that minimal efficacy may depend on co-treatments in which insulin enhancement is supplemented with antibodies to help reduce oligomer levels. In conclusion, the current observations highlight the potential of the rabbit as a genetically unmodified animal model to investigate mechanisms leading to sporadic, late-onset AD. The present study provides evidence that one such AD-promoting mechanism is diabetes. Results show that untreated diabetes, in the absence of any other contributing factors, can instigate AD-type neuropathology, including tau phosphorylation and A␤ oligomer accumulation, over a relatively short period. The consequences of this pathology for cognitive function are open to future investigation, as rabbit affords experimental access to mechanisms of memory formation at the synaptic [87] and circuit

302

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

[88] levels. The rabbit thus offers potential value for investigating mechanisms of progressive dementia as well as disease onset. Because of growing awareness that transgenic mouse may have restricted value as general AD models [89, 90], the various attributes of the rabbit model indicate it may prove to be a useful new system for use in therapeutic research and development strategies. Goals of such strategies would include treatments for milder cognitive impairments induced by diabetes as well as for the catastrophic forms of dementia that develop in sporadic, late-onset AD. ACKNOWLEDGMENTS We are grateful to Paula Pierce, Excalibur pathology for assistance in preparation of histological sections and consultation in immunohistological and immunofluorescence studies. We also thank Dr. Bruce Scharf, Director of the UMDNJ Comparative Medicine Resource Facility for advice regarding implementation of our animal protocol, and thank Vir Singh and Yicheng Feng for technical assistance. We also acknowledge NIH grant R01 EY015855 and Neuroscience Education and Research Foundation grant to PHF, and NIH grant R01 AG022547 to WLK. Authors’ disclosures available online (http://www.jalz.com/disclosures/view.php?id=1381). REFERENCES [1] [2]

[3]

[4]

[5] [6]

[7]

[8]

Thies W, Bleiler L (2011) 2011 Alzheimer’s disease facts and figures. Alzheimers Dement 7, 208-244. De Felice FG, Vieira MN, Bomfim TR, Decker H, Velasco PT, Lambert MP, Viola KL, Zhao WQ, Ferreira ST, Klein WL (2009) Protection of synapses against Alzheimer’s-linked toxins: Insulin signaling prevents the pathogenic binding of Abeta oligomers. Proc Natl Acad Sci U S A 106, 19711976. Tomic JL, Pensalfini A, Head E, Glabe CG (2009) Soluble fibrillar oligomer levels are elevated in Alzheimer’s disease brain and correlate with cognitive dysfunction. Neurobiol Dis 35, 352-358. Gandy S (2005) The role of cerebral amyloid beta accumulation in common forms of Alzheimer disease. J Clin Invest 115, 1121-1129. Ono K, Yamada M (2011) Low-n oligomers as therapeutic targets of Alzheimer’s disease. J Neurochem 117, 19-28. Spires TL, Hyman BT (2005) Transgenic models of Alzheimer’s disease: Learning from animals. NeuroRx 2, 423437. Duff K, Suleman F (2004) Transgenic mouse models of Alzheimer’s disease: How useful have they been for therapeutic development? Brief Funct Genomic Proteomic 3, 47-59. Savonenko AV, Melnikova T, Hiatt A, Li T, Worley PF, Troncoso JC, Wong PC, Price DL (2012) Alzheimer’s therapeutics: Translation of preclinical science to clinical drug development. Neuropsychopharmacology 37, 261-277.

[9]

[10]

[11] [12]

[13]

[14]

[15]

[16]

[17]

[18]

[19]

[20]

[21]

[22]

[23]

Akter K, Lanza EA, Martin SA, Myronyuk N, Rua M, Raffa RB (2011) Diabetes mellitus and Alzheimer’s disease: Shared pathology and treatment? Br J Clin Pharmacol 71, 365-376. Matsuzaki T, Sasaki K, Tanizaki Y, Hata J, Fujimi K, Matsui Y, Sekita A, Suzuki SO, Kanba S, Kiyohara Y, Iwaki T (2010) Insulin resistance is associated with the pathology of Alzheimer disease: The Hisayama study. Neurology 75, 764-770. de la Monte SM (2009) Insulin resistance and Alzheimer’s disease. BMB Rep 42, 475-481. Craft S, Dagogo-Jack SE, Wiethop BV, Murphy C, Nevins RT, Fleischman S, Rice V, Newcomer JW, Cryer PE (1993) Effects of hyperglycemia on memory and hormone levels in dementia of the Alzheimer type: A longitudinal study. Behav Neurosci 107, 926-940. Craft S (2005) Insulin resistance syndrome and Alzheimer’s disease: Age- and obesity-related effects on memory, amyloid, and inflammation. Neurobiol Aging 26(Suppl 1), 65-69. Kroner Z (2009) The relationship between Alzheimer’s disease and diabetes: Type 3 diabetes? Altern Med Rev 14, 373-379. Neumann KF, Rojo L, Navarrete LP, Farias G, Reyes P, Maccioni RB (2008) Insulin resistance and Alzheimer’s disease: molecular links and clinical implications. Curr Alzheimer Res 5, 438-447. Zhao WQ, Lacor PN, Chen H, Lambert MP, Quon MJ, Krafft GA, Klein WL (2009) Insulin receptor dysfunction impairs cellular clearance of neurotoxic oligomeric A{beta}. J Biol Chem 284, 18742-18753. Zhao WQ, Santini F, Breese R, Ross D, Zhang XD, Stone DJ, Ferrer M, Townsend M, Wolfe AL, Seager MA, Kinney GG, Shughrue PJ, Ray WJ (2010) Inhibition of calcineurin-mediated endocytosis and alpha-amino-3hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors prevents amyloid beta oligomer-induced synaptic disruption. J Biol Chem 285, 7619-7632. De Felice FG, Velasco PT, Lambert MP, Viola K, Fernandez SJ, Ferreira ST, Klein WL (2007) Abeta oligomers induce neuronal oxidative stress through an N-methyl-Daspartate receptor-dependent mechanism that is blocked by the Alzheimer drug memantine. J Biol Chem 282, 1159011601. De Felice FG, Wu D, Lambert MP, Fernandez SJ, Velasco PT, Lacor PN, Bigio EH, Jerecic J, Acton PJ, Shughrue PJ, Chen-Dodson E, Kinney GG, Klein WL (2008) Alzheimer’s disease-type neuronal tau hyperphosphorylation induced by A beta oligomers. Neurobiol Aging 29, 1334-1347. 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, De Felice FG (2012) An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease- associated Abeta oligomers. J Clin Invest 122, 1339-1353. Jolivalt CG, Hurford R, Lee CA, Dumaop W, Rockenstein E, Masliah E (2010) Type 1 diabetes exaggerates features of Alzheimer’s disease in APP transgenic mice. Exp Neurol 223, 422-431. Johnstone EM, Chaney MO, Norris FH, Pascual R, Little SP (1991) Conservation of the sequence of the Alzheimer’s disease amyloid peptide in dog, polar bear and five other mammals by cross-species polymerase chain reaction analysis. Brain Res Mol Brain Res 10, 299-305. Munday R, Ludwig K, Lenzen S (1993) The relationship between the physicochemical properties and the biological

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

[24]

[25]

[26]

[27]

[28]

[29]

[30]

[31]

[32]

[33]

[34]

[35] [36]

[37]

[38]

[39]

effects of alloxan and several N-alkyl substituted alloxan derivatives. J Endocrinol 139, 153-163. Maiese K, Chong ZZ, Shang YC (2007) Mechanistic insights into diabetes mellitus and oxidative stress. Curr Med Chem 14, 1729-1738. Szkudelski T (2001) The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physiol Res 50, 537-546. Boado RJ (2001) Amplification of blood-brain barrier GLUT1 glucose transporter gene expression by brain-derived peptides. Neurosci Res 40, 337-342. Maher F, Simpson IA (1994) The GLUT3 glucose transporter is the predominant isoform in primary cultured neurons: assessment by biosynthetic and photoaffinity labelling. Biochem J 301(Pt 2), 379-384. Al-Azzawie HF, Alhamdani MS (2006) Hypoglycemic and antioxidant effect of oleuropein in alloxan-diabetic rabbits. Life Sci 78, 1371-1377. Lenich CM, Chobanian AV, Brecher P, Zannis VI (1991) Effect of dietary cholesterol and alloxan-diabetes on tissue cholesterol and apolipoprotein E mRNA levels in the rabbit. J Lipid Res 32, 431-438. Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, Morgan TE, Rozovsky I, Trommer B, Viola KL, Wals P, Zhang C, Finch CE, Krafft GA, Klein WL (1998) Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci U S A 95, 6448-6453. Lublin AL, Gandy S (2010) Amyloid-beta oligomers: Possible roles as key neurotoxins in Alzheimer’s Disease. Mt Sinai J Med 77, 43-49. Glabe CG (2004) Conformation-dependent antibodies target diseases of protein misfolding. Trends Biochem Sci 29, 542547. Masters CL, Multhaup G, Simms G, Pottgiesser J, Martins RN, Beyreuther K (1985) Neuronal origin of a cerebral amyloid: Neurofibrillary tangles of Alzheimer’s disease contain the same protein as the amyloid of plaque cores and blood vessels. EMBO J 4, 2757-2763. Kumar-Singh S, Pirici D, McGowan E, Serneels S, Ceuterick C, Hardy J, Duff K, Dickson D, Van Broeckhoven C (2005) Dense-core plaques in Tg2576 and PSAPP mouse models of Alzheimer’s disease are centered on vessel walls. Am J Pathol 167, 527-543. Braak H, Braak E (1991) Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 82, 239-259. Standaert DG, Lee VM, Greenberg BD, Lowery DE, Trojanowski JQ (1991) Molecular features of hypothalamic plaques in Alzheimer’s disease. Am J Pathol 139, 681-691. Song Z, Levin BE, McArdle JJ, Bakhos N, Routh VH (2001) Convergence of pre- and postsynaptic influences on glucosensing neurons in the ventromedial hypothalamic nucleus. Diabetes 50, 2673-2681. Lindhagen-Persson M, Brannstrom K, Vestling M, Steinitz M, Olofsson A (2010) Amyloid-beta oligomer specificity mediated by the IgM isotype–implications for a specific protective mechanism exerted by endogenous auto-antibodies. PloS one 5, e13928. Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, Welch D, Manness L, Lin C, Yu J, Zhu H, Ghiso J, Frangione B, Stern A, Schmidt AM, Armstrong DL, Arnold B, Liliensiek B, Nawroth P, Hofman F, Kindy M, Stern D, Zlokovic B (2003) RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain. Nat Med 9, 907-913.

[40]

[41]

[42]

[43]

[44]

[45]

[46]

[47] [48]

[49]

[50]

[51] [52]

[53] [54]

[55] [56]

[57]

303

Sturchler E, Galichet A, Weibel M, Leclerc E, Heizmann CW (2008) Site-specific blockade of RAGE-Vd prevents amyloid-beta oligomer neurotoxicity. J Neurosci 28, 51495158. Takeda S, Sato N, Uchio-Yamada K, Sawada K, Kunieda T, Takeuchi D, Kurinami H, Shinohara M, Rakugi H, Morishita R (2010) Diabetes-accelerated memory dysfunction via cerebrovascular inflammation and Abeta deposition in an Alzheimer mouse model with diabetes. Proc Natl Acad Sci U S A 107, 7036-7041. Stoothoff WH, Johnson GV (2001) Hyperosmotic stressinduced apoptosis and tau phosphorylation in human neuroblastoma cells. J Neurosci Res 65, 573-582. Yoon SY, Park JS, Choi JE, Choi JM, Lee WJ, Kim SW, Kim DH (2010) Rosiglitazone reduces tau phosphorylation via JNK inhibition in the hippocampus of rats with type 2 diabetes and tau transfected SH-SY5Y cells. Neurobiol Dis 40, 449-455. Tomiyama T, Matsuyama S, Iso H, Umeda T, Takuma H, Ohnishi K, Ishibashi K, Teraoka R, Sakama N, Yamashita T, Nishitsuji K, Ito K, Shimada H, Lambert MP, Klein WL, Mori H (2010) A mouse model of amyloid beta oligomers: Their contribution to synaptic alteration, abnormal tau phosphorylation, glial activation, and neuronal loss in vivo. J Neurosci 30, 4845-4856. Dutescu RM, Li QX, Crowston J, Masters CL, Baird PN, Culvenor JG (2009) Amyloid precursor protein processing and retinal pathology in mouse models of Alzheimer’s disease. Graefes Arch Clin Exp Ophthalmol 247, 1213-1221. Haass C, Selkoe DJ (2007) Soluble protein oligomers in neurodegeneration: Lessons from the Alzheimer’s amyloid beta-peptide. Nat Rev Mol Cell Biol 8, 101-112. Schnabel J (2011) Amyloid: little proteins, big clues. Nature 475, S12-S14. Ferreira ST, Klein WL (2011) The Abeta oligomer hypothesis for synapse failure and memory loss in Alzheimer’s disease. Neurobiol Learn Mem 96, 529-543. Holscher C (2011) Diabetes as a risk factor for Alzheimer’s disease: Insulin signalling impairment in the brain as an alternative model of Alzheimer’s disease. Biochem Soc Trans 39, 891-897. Kopf D, Frolich L (2009) Risk of incident Alzheimer’s disease in diabetic patients: A systematic review of prospective trials. J Alzheimers Dis 16, 677-685. Carlsson CM (2010) Type 2 diabetes mellitus, dyslipidemia, and Alzheimer’s disease. J Alzheimers Dis 20, 711-722. Cole GM, Frautschy SA (2007) The role of insulin and neurotrophic factor signaling in brain aging and Alzheimer’s disease. Exp Gerontol 42, 10-21. Alzheimer’s Association, What is Alzheimer’s? http:// www.alz.org/alzheimers disease what is alzheimers.asp. Barile GR, Pachydaki SI, Tari SR, Lee SE, Donmoyer CM, Ma W, Rong LL, Buciarelli LG, Wendt T, Horig H, Hudson BI, Qu W, Weinberg AD, Yan SF, Schmidt AM (2005) The RAGE axis in early diabetic retinopathy. Invest Ophthalmol Vis Sci 46, 2916-2924. Zong H, Ward M, Stitt AW (2011) AGEs, RAGE, and diabetic retinopathy. Curr Diab Rep 11, 244-252. Bitel CL, Feng Y, Souayah N, Frederikse PH (2010) Increased expression and local accumulation of the prion protein, Alzheimer Abeta peptides, superoxide dismutase 1, and nitric oxide synthases 1 & 2 in muscle in a rabbit model of diabetes. BMC Physiol 10, 18. Nogalska A, D’Agostino C, Engel WK, Klein WL, Askanas V (2010) Novel demonstration of amyloid-beta oligomers in

304

[58]

[59]

[60]

[61]

[62]

[63]

[64]

[65]

[66]

[67]

[68]

[69]

[70]

[71]

[72]

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model sporadic inclusion-body myositis muscle fibers. Acta Neuropathol 120, 661-666. Gong Y, Chang L, Viola KL, Lacor PN, Lambert MP, Finch CE, Krafft GA, Klein WL (2003) Alzheimer’s diseaseaffected brain: Presence of oligomeric A beta ligands (ADDLs) suggests a molecular basis for reversible memory loss. Proc Natl Acad Sci U S A 100, 10417-10422. Lacor PN, Buniel MC, Furlow PW, Clemente AS, Velasco PT, Wood M, Viola KL, Klein WL (2007) Abeta oligomerinduced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer’s disease. J Neurosci 27, 796-807. Cleary JP, Walsh DM, Hofmeister JJ, Shankar GM, Kuskowski MA, Selkoe DJ, Ashe KH (2005) Natural oligomers of the amyloid-beta protein specifically disrupt cognitive function. Nat Neurosci 8, 79-84. Walsh DM, Klyubin I, Shankar GM, Townsend M, Fadeeva JV, Betts V, Podlisny MB, Cleary JP, Ashe KH, Rowan MJ, Selkoe DJ (2005) The role of cell-derived oligomers of Abeta in Alzheimer’s disease and avenues for therapeutic intervention. Biochem Soc Trans 33, 1087-1090. Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, Gallagher M, Ashe KH (2006) A specific amyloid-beta protein assembly in the brain impairs memory. Nature 440, 352-357. Ma QL, Yang F, Rosario ER, Ubeda OJ, Beech W, Gant DJ, Chen PP, Hudspeth B, Chen C, Zhao Y, Vinters HV, Frautschy SA, Cole GM (2009) Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: suppression by omega-3 fatty acids and curcumin. J Neurosci 29, 9078-9089. Berisha F, Feke GT, Trempe CL, McMeel JW, Schepens CL (2007) Retinal abnormalities in early Alzheimer’s disease. Invest Ophthalmol Vis Sci 48, 2285-2289. Ohno-Matsui K (2011) Parallel findings in age-related macular degeneration and Alzheimer’s disease. Prog Retin Eye Res 30, 217-238. Ishibashi K, Tomiyama T, Nishitsuji K, Hara M, Mori H (2006) Absence of synaptophysin near cortical neurons containing oligomer Abeta in Alzheimer’s disease brain. J Neurosci Res 84, 632-636. Fotuhi M, Hachinski V, Whitehouse PJ (2009) Changing perspectives regarding late-life dementia. Nat Rev Neurol 5, 649-658. Liu Y, Liu F, Grundke-Iqbal I, Iqbal K, Gong CX (2011) Deficient brain insulin signalling pathway in Alzheimer’s disease and diabetes. J Pathol 225, 54-62. Takashima A, Noguchi K, Sato K, Hoshino T, Imahori K (1993) Tau protein kinase I is essential for amyloid betaprotein-induced neurotoxicity. Proc Natl Acad Sci U S A 90, 7789-7793. Takashima A, Yamaguchi H, Noguchi K, Michel G, Ishiguro K, Sato K, Hoshino T, Hoshi M, Imahori K (1995) Amyloid beta peptide induces cytoplasmic accumulation of amyloid protein precursor via tau protein kinase I/glycogen synthase kinase-3 beta in rat hippocampal neurons. Neurosci Lett 198, 83-86. Sperber BR, Leight S, Goedert M, Lee VM (1995) Glycogen synthase kinase-3 beta phosphorylates tau protein at multiple sites in intact cells. Neurosci Lett 197, 149-153. Lambert MP, Stevens G, Sabo S, Barber K, Wang G, Wade W, Krafft G, Snyder S, Holzman TF, Klein WL (1994) Beta/A4-evoked degeneration of differentiated SH-SY5Y human neuroblastoma cells. J Neurosci Res 39, 377-385.

[73]

[74]

[75]

[76]

[77]

[78]

[79]

[80]

[81]

[82]

[83]

[84]

[85]

[86]

[87]

Oddo S, Caccamo A, Tran L, Lambert MP, Glabe CG, Klein WL, LaFerla FM (2006) Temporal profile of amyloid-beta (Abeta) oligomerization in an in vivo model of Alzheimer disease. A link between Abeta and tau pathology. J Biol Chem 281, 1599-1604. Rapoport M, Dawson HN, Binder LI, Vitek MP, Ferreira A (2002) Tau is essential to beta -amyloid-induced neurotoxicity. Proc Natl Acad Sci U S A 99, 6364-6369. Reifert J, Hartung-Cranston D, Feinstein SC (2011) Amyloid beta-mediated cell death of cultured hippocampal neurons reveals extensive Tau fragmentation without increased full-length tau phosphorylation. J Biol Chem 286, 2079720811. Clodfelder-Miller BJ, Zmijewska AA, Johnson GV, Jope RS (2006) Tau is hyperphosphorylated at multiple sites in mouse brain in vivo after streptozotocin-induced insulin deficiency. Diabetes 55, 3320-3325. Planel E, Tatebayashi Y, Miyasaka T, Liu L, Wang L, Herman M, Yu WH, Luchsinger JA, Wadzinski B, Duff KE, Takashima A (2007) Insulin dysfunction induces in vivo tau hyperphosphorylation through distinct mechanisms. J Neurosci 27, 13635-13648. Ke YD, Delerue F, Gladbach A, Gotz J, Ittner LM (2009) Experimental diabetes mellitus exacerbates tau pathology in a transgenic mouse model of Alzheimer’s disease. PLoS One 4, e7917. Zhao WQ, Townsend M (2009) Insulin resistance and amyloidogenesis as common molecular foundation for type 2 diabetes and Alzheimer’s disease. Biochim Biophys Acta 1792, 482-496. Lee CC, Kuo YM, Huang CC, Hsu KS (2009) Insulin rescues amyloid beta-induced impairment of hippocampal long-term potentiation. Neurobiol Aging 30, 377-387. Townsend M, Mehta T, Selkoe DJ (2007) Soluble Abeta inhibits specific signal transduction cascades common to the insulin receptor pathway. J Biol Chem 282, 3330533312. Zhao WQ, De Felice FG, Fernandez S, Chen H, Lambert MP, Quon MJ, Krafft GA, Klein WL (2009) Amyloid beta oligomers induce impairment of neuronal insulin receptors. FASEB J 22, 246-260. Craft S, Asthana S, Schellenberg G, Baker L, Cherrier M, Boyt AA, Martins RN, Raskind M, Peskind E, Plymate S (2000) 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 903, 222-228. Li Y, Duffy KB, Ottinger MA, Ray B, Bailey JA, Holloway HW, Tweedie D, Perry T, Mattson MP, Kapogiannis D, Sambamurti K, Lahiri DK, Greig NH (2010) GLP-1 receptor stimulation reduces amyloid-beta peptide accumulation and cytotoxicity in cellular and animal models of Alzheimer’s disease. J Alzheimers Dis 19, 1205-1219. 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 (2012) Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: A pilot clinical trial. Arch Neurol 69, 29-38. McClean PL, Parthsarathy V, Faivre E, Holscher C (2011) The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J Neurosci 31, 65876594. Bliss TV, Lomo T (1973) Long-lasting potentiation of synaptic transmission in the dentate area of the anaesthetized rabbit

C.L. Bitel et al. / AD Pathology in a Rabbit Diabetes Model

[88]

following stimulation of the perforant path. J Physiol 232, 331-356. Weible AP, Weiss C, Disterhoft JF (2007) Connections of the caudal anterior cingulate cortex in rabbit: Neural circuitry participating in the acquisition of trace eyeblink conditioning. Neuroscience 145, 288-302.

[89] [90]

305

Balducci C, Forloni G (2011) APP transgenic mice: Their use and limitations. Neuromolecular Med 13, 117-137. Zahs KR, Ashe KH (2010) ’Too much good news’ - are Alzheimer mouse models trying to tell us how to prevent, not cure, Alzheimer’s disease? Trends Neurosci 33, 381-389.

Bitel - amyloid beta.pdf

in a Rabbit Animal Model. Claudine L. Bitela .... spectrometer plate reader. ELISAs were performed 3X ... Bitel - amyloid beta.pdf. Bitel - amyloid beta.pdf. Open.

1MB Sizes 0 Downloads 163 Views

Recommend Documents

Amyloid insulin interaction with erythrocytes
ing purification, production, storage, and particularly during the use of infusion pumps .... Fluorescence spectroscopy was measured using Hitachi model 650-40 ...

Amyloid Toxicity In Skeletal Myoblasts
Skeletal muscle disorder, Inclusion-body myositis (IBM) has been known for ... of IBM, the interaction of amyloid fibrils with skeletal myoblast cells (SMC) has ...

Studying the dissociation of Amyloid Forming region in ...
colonization and pathogenesis (1). ... 2. Methods: 1) Preparation of the protein structure: The crystal structure of ... the structure for 150 ns at 300 K in the NPT.

pdf-0750\amyloid-and-related-disorders-surgical-pathology-and ...
... plasma cell dyscrasia/multiple. Page 3 of 9. pdf-0750\amyloid-and-related-disorders-surgical-pathol ... t-clinical-pathology-by-maria-m-picken-md-phd-fasn.pdf.

Imaging Distinct Conformational States of Amyloid
Aug 3, 2007 - and Technology, 7491 Trondheim, Norway, ¶Division of Cell Biology, Linköping ... center of these plaques showed disordered conformations of the fibrils, and the ex- .... tion of amyloid fibrils was verified by ThT binding (data.

Unlocking the Atomic-Level Details of Amyloid Fibril Growth through ...
College Dublin, Belfield, Dublin, Ireland. Molecular fibrils formed by aggregated .... statistical mechanics-based computa- tional algorithms that can significantly.

Usefulness of C-reactive protein, serum amyloid A ...
using commercial Analytical Profile Index (API) tests. (API-Staph, API-Strep, ... software. The significance of differences was set at P. 0.05 and P. 0.01. 3. Results.