The presence of Aβ seeds, and not age per se, is critical to the initiation of Aβ deposition in the brain (2024)

  • Journal List
  • HHS Author Manuscripts
  • PMC3297471

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

The presence of Aβ seeds, and not age per se, is critical to the initiation of Aβ deposition in the brain (1)

Link to Publisher's site

Acta Neuropathol. Author manuscript; available in PMC 2013 Jan 1.

Published in final edited form as:

Acta Neuropathol. 2012 Jan; 123(1): 31–37.

Published online 2011 Nov 20. doi:10.1007/s00401-011-0912-1

PMCID: PMC3297471

NIHMSID: NIHMS358565

PMID: 22101366

Tsuyoshi Hamaguchi,1,2 Yvonne S. Eisele,1,2 Nicholas H. Varvel,1,2 Bruce T. Lamb,3 Lary C. Walker,4 and Mathias Jucker1,2

Author information Copyright and License information PMC Disclaimer

The publisher's final edited version of this article is available at Acta Neuropathol

Abstract

The deposition of the β-amyloid (Aβ) peptide in senile plaques and cerebral Aβ-amyloid angiopathy can be seeded in β-amyloid precursor protein (APP)-transgenic mice by the intracerebral infusion of brain extracts containing aggregated Aβ. Previous studies of seeded β-amyloid induction have used relatively short incubation periods to dissociate seeded β-amyloid induction from endogenous β-amyloid deposition of the host, thus precluding the analysis of the impact of age and extended incubation periods on the instigation and spread of Aβ lesions in brain. In the present study using R1.40 APP-transgenic mice (which do not develop endogenous Aβ deposition up to 15 months of age) we show that: (i) seeding at 9 months of age does not induce more Aβ deposition than seeding at 3 months of age, provided that the incubation period (6 months) is the same; and (ii) very long-term (12 months) incubation after a focal application of the seed results in the emergence of Aβ deposits throughout the forebrain. These findings indicate that the presence of Aβ seeds, and not the age of the host per se, is critical to the initiation of Aβ aggregation in the brain, and that Aβ deposition, actuated in one brain area, eventually spreads throughout the brain.

Keywords: Alzheimer, amyloid, prion, seeding, senile plaques, transgenic mouse

Introduction

The deposition of β-amyloid (Aβ) peptide in senile plaques and cerebral Aβ angiopathy (CAA) can be induced in the brains of β-amyloid precursor protein (APP)-transgenic (tg) mice by the infusion of Aβ-rich brain extracts from patients with Alzheimer’s disease (AD) or from aged APP tg mice [11,18,5,29]. Mechanistically, the seeding of Aβ deposition resembles the transmission of prion disease in that a misfolded, multimeric form of the disease-associated protein appears to instigate aggregation by corruptive molecular templating [10,25,28]. Aβ deposition also has been induced in wild-type marmosets [1,22], a nonhuman primate that produces human-type sequence Aβ [9].

Advancing age is the greatest risk factor for developing AD [21], but how age promotes the development of Aβ plaques and CAA remains uncertain [3]. Previous studies of the seeded induction of Aβ deposition in mice have employed Tg2576 and APP23 mouse models that spontaneously form amyloid plaques and CAA beginning sometime around 8 to 10 months of age [11,18]. To unambiguously identify the seeded Aβ deposits in the absence of endogenous and spontaneously generated lesions, these studies typically used relatively short incubation periods of 3–6 months after intracerebral administration [5,11,16,18], and 6–7 months after intraperitoneal administration [6]. After longer periods, distinguishing seeded deposits from those that normally form in these models becomes increasingly problematic, thus hampering the analysis of the impact of advancing age and extended incubation periods on the instigation and spread of Aβ lesions in the brain.

In the present study, we used a genomic, hom*ozygous APP-tg mouse model (R1.40 mice) that begins to exhibit Aβ deposition only in late adulthood [13] to examine (i) whether seeding is more effective when initiated in older animals, compared to younger animals, and (ii) the long-term effects of focally administered Aβ seeds on the evolution and spread of β-amyloid lesions in the brain.

Materials and methods

Mice

Male, hemizygous R1.40 APP-tg mice on a B6.129 background were purchased from The Jackson Laboratory (Bar Harbor, ME, USA) and bred with C57BL/6J mice. hom*ozygous mice were then generated by cross-breeding the hemizygous offspring. R1.40 mice carry a yeast artificial chromosome containing the genomic copy of human APP harboring the K670N-M671L mutation [15]. R1.40 APP-tg mice in the present study were hom*ozygous and were maintained under specific pathogen-free conditions. The experimental procedures were carried out in accordance with the veterinary office regulations of Baden-Württemberg (Germany) and approved by the local Animal Care and Use Committees.

Preparation of brain extracts

Extracts were prepared from 22–25 month-old, Aβ-depositing APP23 mice (Tg extract), or from age-matched, non-transgenic (wildtype [Wt]) mice (Wt extract) as previously described [18]. The forebrains were obtained and frozen on dry ice and stored at −80°C. Samples were hom*ogenized at 10% (w/v) in PBS, vortexed, sonicated 3 × 5 s and centrifuged at 3000xg for 5 min. The supernatant was used as the seeding agent. Aβ levels were estimated with electrochemiluminescence-linked immunoassay, revealing Aβ concentrations of 10–20 ng/μl.

Stereotaxic injection of brain extracts

Mice were anaesthetized with a mixture of fentanyl (0.05 mg/kg body weight), midazolam (5 mg/kg body weight) and medetomidine (0.50 mg/kg body weight). Bilateral stereotaxic injections of extract were made with a Hamilton syringe into the hippocampus (2.5 μl) and overlying neocortex (1.0 μl). Extract was injected into the hippocampus (AP −2.5 mm, L +/−2.0mm, DV −1.8mm) over a 2-minute period, and the needle was kept in place for an additional 2 minutes. The needle was raised to the overlying neocortex (AP −2.5mm, L +/−2.0mm, DV −1.0mm) and the additional 1 μl of extract was injected over a 1-minute period. After 2 more minutes, the needle was slowly withdrawn before suturing. After surgery an antidote (flumazenil 0.5 mg/kg body weight, atipamezole 2.5 mg/kg body weight, naloxone 1.2 mg/kg body weight) was administered. A total of 38 mice were used (see figure captions for details). Two mice were excluded from the analysis because the injections missed the dorsal hippocampus; one mouse was excluded because of hydrocephalus; two mice were excluded due to lack of seeding. The excluded mice were dispersed over the experimental groups.

Tissue processing, histological and stereological analysis

The mice were transcardially perfused with PBS under deep anesthesia (ketamine 400 mg/kg, xylazine 40 mg/kg). Brains were harvested, hemidissected, and the left hemispheres were processed as described [6]. The sections were immunostained using polyclonal antibody CN3 (1:1,000; raised against residues 1–16 of human Aβ) [6] and visualized using standard immunoperoxidase procedures with the Vectastain Elite ABC Kit (Vector Laboratories, Burlingame, CA).

The areal density of Aβ-immunoreactive lesions was quantitated in random-systematic sets of every 12th section through the neocortex and hippocampus. Pia-associated vessels were not counted. Stereological analysis of CN3-positive staining was performed using a Zeiss microscope equipped with a motorized x-y-z stage coupled to a video-microscopy system and the Stereo Investigator software (MicroBrightField, Inc, Williston, VT) as described [18].

Adjacent sets of sections were stained with Congo red according to standard protocols and viewed under cross-polarized light and analyzed. The following additional antibodies were used: rabbit polyclonal antibody against cow glial fibrillary acidic protein (GFAP) (Dako, Glostrup, Denmark); rabbit polyclonal antibody against ionized calcium binding adapter molecule 1 (Iba1) (Wako; Richmond, VA).

Immunoblotting and electrochemiluminescence-linked immunoassay

The right hemisphere of PBS-perfused mice was hom*ogenized at 10% w/v in buffer (50mM Tris [PH 8.0], 150mM NaCl, 5mM EDTA, and Complete Mini protease inhibitor co*cktail [Roche Diagnostics GmbH, Penzberg, Germany]). To assess Aβ and APP levels, 4–12% NuPage Bis-Tris mini gels using NuPage LDS sample buffer were used (Invitrogen, Carlsbad, CA), followed by protein transfer onto nitrocellulose membranes and probing with antibody 6E10 (Covance). As an internal control, monoclonal anti-GAPDH antibody (Hy Test Ltd, Turku, Finland) was used. Densitometric values of band intensities were analysed using ImageJ 1.44 (http://rsbweb.nih.gov/ij/).

Statistical analysis

All values are expressed as means ±SEMs. Statistical analyses were performed using SPSS 16.0 software (SPSS Japan, Tokyo, Japan) and Statview 5.0.1 (SAS Institute Inc, Cary, NC), with the significance threshold set at p≤0.05, two-tailed.

Results

Human APP expression and the quantity of human Aβ were studied in the brains of R1.40 APP-tg mice at 3, 9, and 15 months of age. No significant differences among the three age groups were found (Fig. 1). Immunohistochemical analysis did not detect any Aβ deposits in the brains of the mice at 3 and 9 months, while in two of six 15 month-old mice, occasional Aβ deposits were found in the frontal cortex (Fig. 1).

Open in a separate window

Fig. 1

Steady-state levels of APP and Aβ are similar in pre-depositing R1.40 mice

(a) Human APP and Aβ levels in the brains of R1.40 mice at multiple ages. (3 months [n=6; 3 male, 3 female], 9 months [n=6; 3 male, 3 female] and 15 months [n=6; 3 male, 3 female]). Shown are immunoblots, using antibody 6E10, of three mice/age group; GAPDH served as a loading control. (b) Quantification of protein levels in all mice revealed no difference among the groups (one-way ANOVA for APP and Aβ; both p>0.05). Three samples in each age group were applied to 1 gel, and 2 gels were used in total. Densitometric values of band intensities were analysed using ImageJ 1.44, and percentages relative to the amount in 3month-old mice were calculated from the average value at 3 months of age. (c) No Aβ deposition was found in the brains of 3-and 9-month-old R1.40 mice by immunohistochemical analysis, while in two of six 15-month-old mice, infrequent Aβ deposits were present in the frontal cortex. Scale bar = 500 μm.

To determine whether the age of the host is important for the seeded induction of β amyloidosis, R1.40 APP-tg mice received injections of Aβ-containing brain extracts (Tg extract) or wildtype (Wt) extract into the hippocampus and overlying neocortex at 3 months of age and were analysed at 9 months of age; a second group was seeded at 9 months of age and analysed at 15 months of age (Fig. 2). As an additional control to study the long-term effects of seeding, a third group was injected at 3 months of age and sacrificed at 15 months of age (Fig. 2). The results revealed quantitatively similar induction of Aβ deposition in the hippocampus and overlying neocortex of the Tg extract-seeded mice in Groups 1 and 2, i.e., the degree of seeding after a 6-month incubation period was similar whether the mice were infused at 3 months of age or at 9 months of age (Fig. 3). In both groups, the amyloid was Congo red-negative and diffuse in nature. Wt extract-inoculated controls did not show Aβ induction (Fig. 3).

Open in a separate window

Fig. 2

Study design

Group 1 was intracerebrally seeded at 3 months of age, and analysed at 9 months; Group 2 was seeded at 9 months of age and analysed at 15 months; Group 3 was seeded at 3 months of age and analysed at 15 months.

Open in a separate window

Fig. 3

Older mice and younger mice show similar levels of induced Aβ deposition. However, long incubation periods allow for generation of widespread Aβ aggregation and glial cell response

(a) Induction of Aβ deposition was evident in all three groups of R1.40 mice that were intracerebrally seeded with the Tg extract, but not with the Wt extract. While the β-amyloid induction was similar in groups 1 and 2, Aβ load was greatest in Group 3. Adjacent sections were double-stained with Congo red and GFAP or Iba-1. While in groups 1 and 2, all induced β-amyloid was Congo red-negative, group 3 showed at least some Congo red-positive deposits surrounded by darkly stained and hypertrophic GFAP-positive astrocytes (insert, left) and Iba1-positive microglia (insert, right). (b) Quantitative stereological analysis. Group 1 (WT extract: n=5 [3 female, 2 male], Tg extract: n=7 [4 female, 3 male]); Group 2 (WT extract injected: n=6 [2 female, 4 male], Tg extract: n=6 [2 female, 4 male]); Group 3 (WT extract: n=5 [2 female, 3 male], Tg extract: n=4 [3 female, 1 male]). 3-way ANOVA for Gender × Group × Extract revealed significant main effects for Group and Extract (p<0.001) but no significant effect of gender (p>0.05). There was a significant Group × Extract interaction (F[2,21]=89.23; p<0.001) and subsequent Scheffé post hoc analyses were used to pinpoint group differences (*; p<0.05, **; p<0.01, ***; p<0.001). Scale bar = 500 μm and 10 μm.

When R1.40 APP-tg mice were intracerebrally injected with Tg extract at 3 months of age and allowed to incubate for 12 months, extensive Aβ deposition was apparent in the 15month-old mice that exceeded the quantity of lesions after 6 months of incubation by approximately 6-fold (Fig. 3). Although most of the induced amyloid again was Congo Red-negative, some of the Aβ deposits in the vicinity of the injection site (dorsal hippocampus) were Congo red-positive and surrounded by darkly stained, GFAP-positive hypertrophic astrocytes and Iba1-positive enlarged microglia (Fig. 3). Strikingly, in 12-month incubation mice, Aβ deposition had spread throughout the entire neocortex and hippocampus, with additional deposition in the thalamus and septal nuclei, and was more widespread compared to that in the 6-month incubation groups (Fig. 4). Both parenchymal and vessel-associated Aβ deposits were observed (Fig. 4).

Open in a separate window

Fig. 4

Longer incubation times result in extensive spreading of Aβ deposition in the forebrain

(a) Twelve months after intracerebral injections of the Tg extract, copious Aβ was deposited in the brain (shown are coronal sections of the mouse presented in Fig. 3; the coordinates from bregma A–P are indicated). Note that Aβ deposition was widespread throughout the neocortex and hippocampus, although Tg extract was injected only locally into the hippocampus and overlying cortex at AP −2.5mm. (b) Aβ load in each section from anterior to posterior is shown for the 12-month incubation mice (group 3) and compared to the 6-month incubation mice (group 1 &2) (section 1 to section 11; approximate location from bregma A–P is as follows, section 1= AP +2.2mm, section 5=AP +0.1mm, section 8= AP −1.8mm, section 11=AP −3.3mm). Note that the induced Aβ is highest around the injection site (corresponding to sections 9&10) in all groups, and that the Aβ deposits have spread throughout the brain, including the most anterior parts. Scale bar = 500 μm.

Discussion

Previous studies have shown that brain-derived Aβ aggregates can induce Aβ lesions in the brains of susceptible mice [11,16,18,5,6,29]. In the present study, we used the genomic-based R1.40 APP-tg model as host. These animals exhibit endogenous Aβ deposition at approximately 15 months of age [13], thus allowing for the analysis of extended incubation periods in Aβ extract-inoculated mice. Our results reveal that intracerebral injections of Aβ-containing brain extract also induce Aβ deposition in this model, although the Aβ induction after a 6-month incubation period was less than that seen in APP23 mice after 4 months of incubation [16,18]. This observation is consistent with the 3-fold overexpression of human APP in R1.40 mice compared to 7-fold overexpression in APP23 animals [26,15], and supports the view that the induction of Aβ deposition is dependent on the concentration of Aβ in the inoculum as well its production by the host [8,18].

We found that a 6-month incubation period induced similar levels of Aβ aggregation in the brains of young (3 months) and older (9 months) R1.40 APP-tg mice, indicating that the aged brain does not provide a more favorable environment for the induction of Aβ deposition once Aβ seeds are present. Our results, while surprising in light of the view that age is a prominent risk factor for amyloid lesions [7,3,27], may be attributed to the fact that the brain levels of Aβ are constant in pre-depositing R1.40 mice between 3 months and 15 months of age [17]. Unchanging Aβ levels prior to Aβ pathology also have been reported in other AD mouse models [12,14]. These findings suggest that age may increase the risk of AD by impairing the ability of cells to dispose pathogenic seeds [20,2]. In addition, it cannot be excluded that a similar experiment in which seeding is compared in a young host vs. a host at an age closer to the end of its mean life span would yield different results.

The most striking finding to emerge from the inoculations in R1.40 mice was the presence of Aβ deposits in much of the forebrain after the 12 month incubation period. Previous work has shown that the focal infusion of Aβ seeds into the hippocampus is sufficient to induce Aβ deposition throughout the entire hippocampus and even in connected brain areas; however, induction of lesions in widespread regions of the forebrain has not been previously observed [5,10]. The mechanism of this extensive spreading of Aβ deposition is not clear, but implies that seeds not only migrate within brain structures and along defined neuronal pathways, but may also disseminate along perivascular fluid drainage channels, by vascular transport, or by simple diffusion through the brain parenchyma [10]. The recent finding that small, soluble Aβ species are particularly potent inducers of β-amyloidosis [16], similar to the strong infectivity of small, non-fibrillar prion particles [24], underscores potential mechanistic similarities in the induction and spread of Aβ and PrP aggregates in the brain.

Birefringence under crossed-polarizing filters is indicative of the amyloid-like nature of proteinaceous deposits after staining with the dye Congo red [30]. In seeded R1.40 APP-tg mice, the duration of the incubation period influenced the degree to which the resulting Aβ deposits were congophilic. Whereas the 6-month incubation period, starting at either 3 or 9 months of age, did not result in the induction of congophilic lesions, Congo red-positive plaques were observed after the 12-month incubation period. However, congophilic deposits were only observed near the injection site, indicating that diffuse pathology occurs first and eventually develops into Congo red-positive (i.e., amyloid) plaques. Because the cytological changes associated with Aβ deposition are most strongly associated with amyloid per se [4] rather than with diffuse deposits, the lack of congophilic amyloid induction after the 6-month incubation period did not allow us to determine whether the amyloid-associated cytopathology (neuritic dystrophy and glial activation) differs between young (9 months) and older (15 months) hosts [7,3].

From a translational perspective, the present findings support the widely held notion that therapies directed at mitigating the formation of Aβ pathology should begin early in the pathogenic cascade that leads to AD [19,23]. The exposure to Aβ seeds profoundly influences the onset and degree of β-amyloid pathology in APP-tg mouse models. While there is not yet direct evidence that exogenous seeds are involved in the initiation of AD, it is likely that endogenously generated seeds become increasingly prevalent with age due to the decline of proteostatic processes [20,3]. The neutralization or removal of endogenous Aβ seeds thus remains a promising but elusive therapeutic goal for interfering with the pathogenesis of AD.

Acknowledgments

This work was supported by grants from the Competence Network on Degenerative Dementias (BMBF-01GI0705), the BMBF in the frame of ERA-Net NEURON (MIPROTRAN), NIH RR-00165, P50AG025688, the Alzheimer’s Association (NIRG-10173099) and the CART Foundation. TH is recipient of a postdoctoral fellowship from the Alexander von Humboldt Foundation (Bonn, Germany). We gratefully acknowledge helpful discussions and experimental help of Franziska Langer, Götz Heilbronner, Ulrike Obermüller, Jörg Odenthal, Stephan Kaeser, Michael Hruscha and Andrea Bosch (Tübingen, Germany) and Harry LeVine III (Lexington, KY, USA).

Footnotes

Authors’ Contribution: T.H., Y.S.E., M.J. designed the study, and T.H. performed the research; T.H., Y.S.E., N.H.V., L.C.W., M.J analysed the data; B.T. L. provided mouse tissue and advice; T.H., N.H.V., L.C.W. and M.J. wrote the manuscript.

Conflict of Interest The authors declare that they have no conflict of interest.

References

1. Baker HF, Ridley RM, duch*en LW, Crow TJ, Bruton CJ. Evidence for the experimental transmission of cerebral beta-amyloidosis to primates. Int J Exp Pathol. 1993;74 (5):441–454. [PMC free article] [PubMed] [Google Scholar]

2. Balch WE, Morimoto RI, Dillin A, Kelly JW. Adapting proteostasis for disease intervention. Science. 2008;319(5865):916–919. doi:10.1126/science.1141448. 319/5865/916 [pii] [PubMed] [CrossRef] [Google Scholar]

3. Dillin A, Cohen E. Ageing and protein aggregation-mediated disorders: from invertebrates to mammals. Philos Trans R Soc Lond B Biol Sci. 2011;366(1561):94–98. doi:10.1098/rstb.2010.0271. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

4. Duyckaerts C, Delatour B, Potier MC. Classification and basic pathology of Alzheimer disease. Acta Neuropathol. 2009;118(1):5–36. doi:10.1007/s00401-009-0532-1. [PubMed] [CrossRef] [Google Scholar]

5. Eisele YS, Bolmont T, Heikenwalder M, Langer F, Jacobson LH, Yan ZX, Roth K, Aguzzi A, Staufenbiel M, Walker LC, Jucker M. Induction of cerebral beta-amyloidosis: intracerebral versus systemic Abeta inoculation. Proc Natl Acad Sci U S A. 2009;106(31):12926–12931. doi:10.1073/pnas.0903200106. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

6. Eisele YS, Obermüller U, Heilbronner G, Baumann F, Kaeser SA, Wolburg H, Walker LC, Staufenbiel M, Heikenwalder M, Jucker M. Peripherally applied Abeta-containing inoculates induce cerebral beta-amyloidosis. Science. 2010;330(6006):980–982. doi:10.1126/science.1194516. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

7. Geula C, Wu CK, Saroff D, Lorenzo A, Yuan M, Yankner BA. Aging renders the brain vulnerable to amyloid beta-protein neurotoxicity. Nat Med. 1998;4 (7):827–831. [PubMed] [Google Scholar]

8. Harper JD, Lansbury PT., Jr Models of amyloid seeding in Alzheimer’s disease and scrapie: mechanistic truths and physiological consequences of the time-dependent solubility of amyloid proteins. Annu Rev Biochem. 1997;66:385–407. doi:10.1146/annurev.biochem.66.1.385. [PubMed] [CrossRef] [Google Scholar]

9. Heuer E, Rosen RF, Cintron A, Walker LC. Nonhuman primate models of Alzheimer-like cerebral proteopathy. Curr Pharm Des. 2011 in press. [PMC free article] [PubMed] [Google Scholar]

10. Jucker M, Walker LC. Pathogenic protein seeding in Alzheimer’s disease and other neurodegenerative disorders. Ann Neurol. 2011;70:532–540. doi:10.1002/ana.22615. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

11. Kane MD, Lipinski WJ, Callahan MJ, Bian F, Durham RA, Schwarz RD, Roher AE, Walker LC. Evidence for seeding of beta -amyloid by intracerebral infusion of Alzheimer brain extracts in beta -amyloid precursor protein-transgenic mice. J Neurosci. 2000;20 (10):3606–3611. [PMC free article] [PubMed] [Google Scholar]

12. Kawarabayashi T, Younkin LH, Saido TC, Shoji M, Ashe KH, Younkin SG. Age-dependent changes in brain, CSF, and plasma amyloid (beta) protein in the Tg2576 transgenic mouse model of Alzheimer’s disease. J Neurosci. 2001;21 (2):372–381. [PMC free article] [PubMed] [Google Scholar]

13. Kulnane LS, Lamb BT. Neuropathological characterization of mutant amyloid precursor protein yeast artificial chromosome transgenic mice. Neurobiol Dis. 2001;8(6):982–992. doi:10.1006/nbdi.2001.0446. [PubMed] [CrossRef] [Google Scholar]

14. Kuo YM, Beach TG, Sue LI, Scott S, Layne KJ, Kokjohn TA, Kalback WM, Luehrs DC, Vishnivetskaya TA, Abramowski D, Sturchler-Pierrat C, Staufenbiel M, Weller RO, Roher AE. The evolution of A beta peptide burden in the APP23 transgenic mice: implications for A beta deposition in Alzheimer disease. Mol Med. 2001;7 (9):609618. [PMC free article] [PubMed] [Google Scholar]

15. Lamb BT, Call LM, Slunt HH, Bardel KA, Lawler AM, Eckman CB, Younkin SG, Holtz G, Wagner SL, Price DL, Sisodia SS, Gearhart JD. Altered metabolism of familial Alzheimer’s disease-linked amyloid precursor protein variants in yeast artificial chromosome transgenic mice. Hum Mol Genet. 1997;6 (9):1535–1541. [PubMed] [Google Scholar]

16. Langer F, Eisele YS, Fritschi SK, Staufenbiel M, Walker LC, Jucker M. Soluble Abeta seeds are potent inducers of cerebral beta-amyloid deposition. J Neurosci. 2011;31:14488–14495. [PMC free article] [PubMed] [Google Scholar]

17. Lehman EJ, Kulnane LS, Gao Y, Petriello MC, Pimpis KM, Younkin L, Dolios G, Wang R, Younkin SG, Lamb BT. Genetic background regulates beta-amyloid precursor protein processing and beta-amyloid deposition in the mouse. Hum Mol Genet. 2003;12(22):2949–2956. doi:10.1093/hmg/ddg322. [PubMed] [CrossRef] [Google Scholar]

18. Meyer-Luehmann M, Coomaraswamy J, Bolmont T, Kaeser S, Schaefer C, Kilger E, Neuenschwander A, Abramowski D, Frey P, Jaton AL, Vigouret JM, Paganetti P, Walsh DM, Mathews PM, Ghiso J, Staufenbiel M, Walker LC, Jucker M. Exogenous induction of cerebral beta-amyloidogenesis is governed by agent and host. Science. 2006;313(5794):1781–1784. doi:10.1126/science.1131864. [PubMed] [CrossRef] [Google Scholar]

19. Perrin RJ, fa*gan AM, Holtzman DM. Multimodal techniques for diagnosis and prognosis of Alzheimer’s disease. Nature. 2009;461(7266):916–922. doi:10.1038/nature08538. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

20. Powers ET, Morimoto RI, Dillin A, Kelly JW, Balch WE. Biological and chemical approaches to diseases of proteostasis deficiency. Annu Rev Biochem. 2009;78:959–991. doi:10.1146/annurev.biochem.052308.114844. [PubMed] [CrossRef] [Google Scholar]

21. Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nat Rev Neurol. 2011;7(3):137–152. doi:10.1038/nrneurol.2011.2. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

22. Ridley RM, Baker HF, Windle CP, Cummings RM. Very long term studies of the seeding of beta-amyloidosis in primates. J Neural Transm. 2006;113(9):1243–1251. doi:10.1007/s00702-005-0385-2. [PubMed] [CrossRef] [Google Scholar]

23. Selkoe DJ. Resolving controversies on the path to Alzheimer’s therapeutics. Nat Med. 2011;17(9):1060–1065. doi:10.1038/nm.2460. [PubMed] [CrossRef] [Google Scholar]

24. Silveira JR, Raymond GJ, Hughson AG, Race RE, Sim VL, Hayes SF, Caughey B. The most infectious prion protein particles. Nature. 2005;437(7056):257–261. doi:10.1038/nature03989. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

25. Soto C, Estrada L, Castilla J. Amyloids, prions and the inherent infectious nature of misfolded protein aggregates. Trends Biochem Sci. 2006;31(3):150–155. doi:10.1016/j.tibs.2006.01.002. [PubMed] [CrossRef] [Google Scholar]

26. Sturchler-Pierrat C, Abramowski D, Duke M, Wiederhold KH, Mistl C, Rothacher S, Ledermann B, Burki K, Frey P, Paganetti PA, Waridel C, Calhoun ME, Jucker M, Probst A, Staufenbiel M, Sommer B. Two amyloid precursor protein transgenic mouse models with Alzheimer disease-like pathology. Proc Natl Acad Sci U S A. 1997;94 (24):13287–13292. [PMC free article] [PubMed] [Google Scholar]

27. Thal DR, Capetillo-Zarate E, Del Tredici K, Braak H. The development of amyloid beta protein deposits in the aged brain. Sci Aging Knowledge Environ. 2006;2006(6):re1. doi:10.1126/sageke.2006.6.re1. 2006/6/re1 [pii] [PubMed] [CrossRef] [Google Scholar]

28. Walker LC, Levine H, 3rd, Mattson MP, Jucker M. Inducible proteopathies. Trends Neurosci. 2006;29(8):438–443. doi:10.1016/j.tins.2006.06.010. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

29. Watts JC, Giles K, Grillo SK, Lemus A, DeArmond SJ, Prusiner SB. Bioluminescence imaging of Abeta deposition in bigenic mouse models of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2011;108(6):2528–2533. doi:10.1073/pnas.1019034108. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

30. Westermark GT, Johnson KH, Westermark P. Staining methods for identification of amyloid in tissue. Methods Enzymol. 1999;309:3–25. [PubMed] [Google Scholar]

The presence of Aβ seeds, and not age per se, is critical to the initiation of Aβ deposition in the brain (2024)
Top Articles
Read Remarried Empress - MGJinx.com
A town's name recalls the massacre of Indigenous people. Will changing it bring healing?
Funny Roblox Id Codes 2023
Golden Abyss - Chapter 5 - Lunar_Angel
Www.paystubportal.com/7-11 Login
Joi Databas
DPhil Research - List of thesis titles
Shs Games 1V1 Lol
Evil Dead Rise Showtimes Near Massena Movieplex
Steamy Afternoon With Handsome Fernando
Which aspects are important in sales |#1 Prospection
Detroit Lions 50 50
18443168434
Zürich Stadion Letzigrund detailed interactive seating plan with seat & row numbers | Sitzplan Saalplan with Sitzplatz & Reihen Nummerierung
Grace Caroline Deepfake
978-0137606801
Nwi Arrests Lake County
Justified Official Series Trailer
London Ups Store
Committees Of Correspondence | Encyclopedia.com
Pizza Hut In Dinuba
Jinx Chapter 24: Release Date, Spoilers & Where To Read - OtakuKart
How Much You Should Be Tipping For Beauty Services - American Beauty Institute
Free Online Games on CrazyGames | Play Now!
Sizewise Stat Login
VERHUURD: Barentszstraat 12 in 'S-Gravenhage 2518 XG: Woonhuis.
Jet Ski Rental Conneaut Lake Pa
Unforeseen Drama: The Tower of Terror’s Mysterious Closure at Walt Disney World
Ups Print Store Near Me
C&T Wok Menu - Morrisville, NC Restaurant
How Taraswrld Leaks Exposed the Dark Side of TikTok Fame
Dashboard Unt
Access a Shared Resource | Computing for Arts + Sciences
Speechwire Login
Healthy Kaiserpermanente Org Sign On
Restored Republic
3473372961
Craigslist Gigs Norfolk
Litter-Robot 3 Pinch Contact & DFI Kit
Moxfield Deck Builder
Senior Houses For Sale Near Me
Whitehall Preparatory And Fitness Academy Calendar
Trivago Myrtle Beach Hotels
Anya Banerjee Feet
Birmingham City Schools Clever Login
Thotsbook Com
Vérificateur De Billet Loto-Québec
Funkin' on the Heights
Vci Classified Paducah
Www Pig11 Net
Ty Glass Sentenced
Latest Posts
Article information

Author: Ouida Strosin DO

Last Updated:

Views: 6337

Rating: 4.6 / 5 (76 voted)

Reviews: 91% of readers found this page helpful

Author information

Name: Ouida Strosin DO

Birthday: 1995-04-27

Address: Suite 927 930 Kilback Radial, Candidaville, TN 87795

Phone: +8561498978366

Job: Legacy Manufacturing Specialist

Hobby: Singing, Mountain biking, Water sports, Water sports, Taxidermy, Polo, Pet

Introduction: My name is Ouida Strosin DO, I am a precious, combative, spotless, modern, spotless, beautiful, precious person who loves writing and wants to share my knowledge and understanding with you.