Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

A Reduced Astrocyte Response to β-Amyloid Plaques in the Ageing Brain Associates with Cognitive Impairment

  • Ryan Mathur,

    Affiliation Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, England, United Kingdom

  • Paul G. Ince,

    Affiliation Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, England, United Kingdom

  • Thais Minett,

    Affiliation Institute of Public Health, University of Cambridge, Cambridge, England, United Kingdom

  • Claire J. Garwood,

    Affiliation Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, England, United Kingdom

  • Pamela J. Shaw,

    Affiliation Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, England, United Kingdom

  • Fiona E. Matthews,

    Affiliation MRC Biostatistics Unit, Cambridge, England, United Kingdom

  • Carol Brayne,

    Affiliation Institute of Public Health, University of Cambridge, Cambridge, England, United Kingdom

  • Julie E. Simpson ,

    Contributed equally to this work with: Julie E. Simpson, Stephen B. Wharton

    julie.simpson@sheffield.ac.uk

    Affiliation Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, England, United Kingdom

  • Stephen B. Wharton ,

    Contributed equally to this work with: Julie E. Simpson, Stephen B. Wharton

    Affiliation Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, England, United Kingdom

  • on behalf of the MRC Cognitive Function and Ageing Neuropathology Study Group

    Membership of the MRC Cognitive Function and Ageing Neuropathology Study Group are listed on the CFAS website (www.cfas.ac.uk/pages/contacts/index.html).

Abstract

Aims

β-amyloid (Aβ) plaques are a key feature of Alzheimer’s disease pathology but correlate poorly with dementia. They are associated with astrocytes which may modulate the effect of Aβ-deposition on the neuropil. This study characterised the astrocyte response to Aβ plaque subtypes, and investigated their association with cognitive impairment.

Methods

Aβ plaque subtypes were identified in the cingulate gyrus using dual labelling immunohistochemistry to Aβ and GFAP+ astrocytes, and quantitated in two cortical areas: the area of densest plaque burden and the deep cortex near the white matter border (layer VI). Three subtypes were defined for both diffuse and compact plaques (also known as classical or core-plaques): Aβ plaque with (1) no associated astrocytes, (2) focal astrogliosis or (3) circumferential astrogliosis.

Results

In the area of densest burden, diffuse plaques with no astrogliosis (β = -0.05, p = 0.001) and with focal astrogliosis (β = -0.27, p = 0.009) significantly associated with lower MMSE scores when controlling for sex and age at death. In the deep cortex (layer VI), both diffuse and compact plaques without astrogliosis associated with lower MMSE scores (β = -0.15, p = 0.017 and β = -0.81, p = 0.03, respectively). Diffuse plaques with no astrogliosis in layer VI related to dementia status (OR = 1.05, p = 0.025). In the area of densest burden, diffuse plaques with no astrogliosis or with focal astrogliosis associated with increasing Braak stage (β = 0.01, p<0.001 and β = 0.07, p<0.001, respectively), and ApoEε4 genotype (OR = 1.02, p = 0.001 and OR = 1.10, p = 0.016, respectively). In layer VI all plaque subtypes associated with Braak stage, and compact amyloid plaques with little and no associated astrogliosis associated with ApoEε4 genotype (OR = 1.50, p = 0.014 and OR = 0.10, p = 0.003, respectively).

Conclusions

Reactive astrocytes in close proximity to either diffuse or compact plaques may have a neuroprotective role in the ageing brain, and possession of at least one copy of the ApoEε4 allele impacts the astroglial response to Aβ plaques.

Introduction

Neuropathologically Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterised by β-amyloid (Aβ) plaques (diffuse and compact dense-core) and intracellular tangles of hyperphosphorylated tau [1]. Aβ plaque formation is thought to progress from diffuse through to compact [2,3]. The relative frequency of the plaque subtypes changes during the progression of AD, with diffuse Aβ plaques being prevalent in the preclinical stages and compact plaques increasing in frequency as the disease progresses [4,5]. The Medical Research Council’s population-based Cognitive Function and Ageing Study (CFAS) has shown that Alzheimer-type pathology is the most common pathology associated with dementia in the ageing population [6], but that there is considerable overlap in the burden of plaques and tangles between individuals with or without dementia, especially in the oldest old [7,8], suggesting other factors contribute to the progression of cognitive decline.

Astrocytes, the most abundant glial cell, play a critical role in neuronal support and in maintaining homeostasis within the central nervous system (CNS) [9]. Activated glia surround and infiltrate Aβ plaques in AD [1012], however their exact role in the pathogenesis of age-related neuropathology remains unknown. While astrocytes have been shown to play a significant role in the degradation and clearance of Aβ suggesting a neuroprotective role [13], other studies have shown astrocyte activation results in the production of critical inflammatory mediators, suggesting they play a detrimental role in the progression of age-related neurodegenerative pathology [14]. Reactive astrocytes up-regulate glial fibrillary acidic protein (GFAP) expression in response to CNS insults [15,16]. Astrogliosis and astrocyte dystrophy are prominent features of several dementia pathologies, including AD and frontotemporal dementia, where the degree of astrocyte degeneration correlates with the severity of dementia [17]. Astrogliosis occurs at early stages of AD pathogenesis and treatment of cultured astrocytes with aggregated Aβ or with amyloid isolated from human AD brains has been shown to trigger astrogliosis [1820]. Recent studies have further characterised the astroglial response in AD, demonstrating an increase in plaque-associated GFAPα and GFAPδ isoforms, and although the number of astrocytes expressing the GFAP(+1) isoform correlates with AD progression, they are not associated with plaques [12]. Possession of the ApoEε4 allele, a major genetic risk factor for AD [21], is associated with an increased cortical Aβ plaque burden [2224] and astrocyte dysfunction [25]. ApoE4, primarily expressed by astrocytes in the brain, plays a role in the metabolism of amyloid [26,27], and has been shown to promote Aβ deposition [28].

The CFAS neuropathology cohort is population-based thus allowing unbiased assessment of pathologies in brain ageing and their relationships to cognitive impairment [68]. We have previously characterised the astrocyte phenotype in the CFAS cohort and demonstrated increased GFAP immunoreactivity associated with increasing Braak and Braak neurofibrillary tangle stage with some, but not all, Aβ plaques associated with GFAP+ astrocytes [29]. We hypothesised that the astrocyte response to Aβ deposits in the cingulate gyrus may modulate the effect of the amyloid plaque on surrounding brain tissue, and therefore on cognition. This region was selected as it contributes to spatial learning and memory [30], is associated with a high prevalence of Aβ pathology. It is involved in the intermediate stages of Aβ progression (Aβ phase 3/5), [31], is involved in the limbic stage of neurofibrillary tangle progression (Braak and Braak stage III-IV) and has projections to the entorhinal cortex, the area with the earliest NFT formation [32], and presents with metabolic and vascular changes before the development of AD [33,34]. Therefore the aim of this study was to examine the variation in the astrocyte response associated with both diffuse and compact Aβ plaques in the cingulate gyrus, and investigate their association with Braak and Braak stage, cognitive impairment, dementia status and ApoE genotype.

Materials and Methods

Human CNS cases

Human CNS material was obtained from the Medical Research Council Cognitive Function and Ageing Study (MRC CFAS) autopsy cohort, which has been described in detail previously [35,36]. Individuals selected for the assessment interviews were approached by a trained liaison officer for brain donation in each centre, who discussed the donation programme with the respondent and his or her family or carers, as appropriate. When an individual died and the research team was notified, the next of kin was approached to give consent for brain donation and retention. Limited or full necroscopy then proceeded if all permissions were obtained. Multi-centre research ethics committee (REC) approval for the current study was obtained from Cambridgeshire 1 Research Ethics Committee (REC reference number 10/H0304/61).

The study used all of the cases derived from one of the CFAS centres (Cambridge), thereby maintaining the unbiased, population-based nature of the study. Cortical blocks were sampled within 4–6 weeks following a standard protocol [37], from 109 formalin-fixed cases. Neuropathological lesions were assessed as part of the core CFAS neuropathology study using a modified protocol from the Consortium to Establish a Registry of Alzheimer’s Disease (CERAD) [38] (wwws.cfas.ac.uk). Braak and Braak staging was assessed by analysis of AT8 immunostaining of neurofibrillary tangles in the hippocampus and isocortical regions [32,39]. The cases were categorised into groups representing entorhinal stages (Braak stages 0–2; 27 cases), limbic stages (Braak stages 3–4; 50 cases) and isocortical stages (Braak stages 5–6; 22 cases). ApoE genotype was previously determined in the cohort [40,41].

Individuals in the study were regularly interviewed and underwent Geriatric Mental State-Automated Geriatric Examination for Computer-Assisted Taxonomy (GMS-AGECAT), Cambridge Mental Disorders of the Elderly Examination (CAMDEX), and mini mental state examination (MMSE) [7,8]. Dementia status at death was determined on the basis of all information available for each participant, as previously described [8,35]. Within this cohort, 68 participants had dementia, 39 had no dementia and 2 participants had an unknown dementia status at death due to the lack of information in the years preceding death (Table 1). Twenty-one participants with dementia and 9 participants with no dementia possessed at least one ApoEε4 allele.

thumbnail
Table 1. Demographic and cognitive profile of cases, according to dementia status.

https://doi.org/10.1371/journal.pone.0118463.t001

Immunohistochemistry

Immunohistochemistry of formalin-fixed, paraffin-embedded sections (5μm) from the cingulate cortex was performed using a standard avidin-biotin complex (ABC) method. Sections were deparaffinised, rehydrated to water and endogenous peroxidase activity quenched by placing the sections in 0.3% H2O2/methanol for 20min at room temperature (RT). Sections were subjected to antigen retrieval (0.01M tri-sodium citrate pH6.5, microwave 10min) followed by formic acid pre-treatment for 60min at RT. Following incubation with 1.5% normal serum for 30min at RT, the sections were incubated with anti-Aβ (Clone 6F/3D; DakoCytomation, UK) [42] at the optimal antibody dilution of 1:200 for 60min at RT. To visualise antibody binding, the horse-radish peroxidase avidin biotin complex was used (Vectastain Elite kit, Vector Laboratories, UK) with 3,3’-diaminodenzidine (DAB) as the chromagen (Vector Laboratories, UK; brown). Following incubation with the avidin-biotin blocking kit (Vector Laboratories, UK), sections were incubated overnight at 4°C with anti-GFAP (1:500; DakoCytomation, UK) [43], followed by the alkaline-phosphatase-conjugated avidin-biotin complex (Vectastain Elite kit, Vector Laboratories, UK), developed with alkaline phosphatase substrate 1 (Vector Laboratories, UK; red) and lightly counterstained with Mayer’s haematoxylin. Negative controls, either omission of the primary antibody or isotype controls, were included in every run.

Quantitative neuropathological analysis

Assessment of Aβ and GFAP immunoreactivity was conducted using a Nikon Eclipse Ni-U microscope and Nikon DS-Ri1 camera with NIS-Elements BR 4.20.01 64-bit microscope imaging software (Nikon, UK). Aβ plaques were subtyped based on the type of plaque (diffuse or compact) and the surrounding astrocyte reaction. Compact plaques are spherical in shape and are characterised by a dense central core of Aβ surrounded by a less compact peripheral halo, in contrast to the diffuse plaques which are usually not spherical and stain weakly for Aβ [44]. Compact plaques may be associated with tau-positive dystrophic neurites, which are then referred to as neuritic plaques. However, triple labelling with Aβ, GFAP and tau was not performed, therefore neuritic plaques containing dystrophic neurites were not assessed as part of this study. The frequency and grade (none, mild, moderate or severe) of both diffuse and compact Aβ plaques were assessed in single Aβ immunostained cingulate gyrus sections, based on the CERAD protocol [38].

Three subtypes were defined for both diffuse and compact plaques: (1) Aβ plaque with no associated astrocytes, (2) focal astrogliosis and (3) circumferential astrogliosis. Focal astrogliosis was defined as reactive astrocytes directly in contact with a plaque solely on one of its borders. Circumferential astrogliosis was defined as reactive astrocytes directly in contact with and completely surrounding a plaque. Areas of astrogliosis remote from plaques were also identified and defined as small (less than two distinct GFAP+ astrocytes) and large (greater than or equal to three distinct GFAP+ astrocytes). The number of each plaque subtype was quantitated in the area with the densest of amyloid burden under the 10x objective (field area 1275 x 925 μm2). Micro-plaques (<10μm in diameter) and plaques in layer I of the cortex were ignored and did not contribute to the count. Using an identical method, the number of each plaque subtype was also assessed in four fields of the deep cortex (layer VI) (5100 x 925 μm2), in areas remote from the area of densest amyloid burden.

Inter-rater reliability of Aβ plaque subtype quantitation

The number and subtype of Aβ plaques and non-plaque associated regions of astrogliosis were quantitated in the area of densest burden in a subset of 10 randomly selected cases by two independent observers (RM and JES), and the extent of agreement assessed by calculating Gwet’s AC2 coefficients [45]. The coefficient calculations were performed using Agreestat 2011.2 programme (Advanced Analytics, Gaithersburg, MD, USA), and the extent of agreement was assessed using the benchmark proposed by Landis and Koch [46], a coefficient >0.6 indicating substantial agreement and a value >0.8 near-perfect agreement.

There was near-perfect agreement in the scoring of diffuse plaques with no associated astrocytes (AC2 = 0.86, 95%CI(AC2) 0.62; 1.00), diffuse plaques with focal astrogliosis (AC2 = 0.88, 95%CI(AC2) 0.75; 1.00), compact plaques with no associated astrocytes (AC2 = 0.83, 95%CI(AC2) 0.61; 1.00), compact plaques with circumferential astrogliosis (AC2 = 0.86, 95%CI(AC2) 0.65; 1.00) and large areas of non-plaque associated astrogliosis (AC2 = 0.93, 95%CI(AC2) 0.76; 1.00). There was a substantial agreement in the scoring of diffuse plaques with circumferential astrogliosis (AC2 = 0.76, 95%CI(AC2) 0.36; 1.00), compact plaques with focal astrocytes (AC2 = 0.79, 95%CI(AC2) 0.42; 1.00) as well as small areas of non-plaque associated astrogliosis (AC2 = 0.75, 95%CI(AC2) 0.29; 1.00), confirming the reliability of the scoring method. Subsequent analyses of the number and subtype of Aβ plaques and non-plaque associated regions of astrogliosis in the cohort were performed on scores by RM.

Statistical Analysis

Statistical analyses were performed and graphs obtained using IBM SPSS Statistics 21 (Armonk, NY) and Stata Statistical Software 12 (College Station, TX). The association between Braak and Braak neurofibrillary tangle stage and the frequency of either diffuse or compact Aβ plaques was assessed using Kendall’s tau correlation coefficient. The relationships between plaque subtype with Braak and Braak stage and MMSE score were tested via multiple linear regression analysis where MMSE score and Braak and Braak stage were the dependent variables, whereas the relationships between plaque subtype with dementia status and ApoE genotype were verified using logistic regression, where dementia status and E4 ApoE genotype were the dependent variables. All the regression analyses were controlled by sex and age at death.

Results

Aβ plaque subtypes in the ageing brain

Diffuse and compact Aβ plaques were diverse in size, number and distribution throughout the cohort. The frequency of each grade (none, mild, moderate or severe) of diffuse and compact Aβ amyloid plaque in the cingulate gyrus is shown in Table 2. Both diffuse (τ = 0.333, p<0.001) and compact (τ = 0.259, p = 0.001) Aβ plaques associated with Braak and Braak neurofibrillary tangle stage.

thumbnail
Table 2. Frequency of diffuse and compact Aβ plaques in the cingulate gyrus, based on the CERAD protocol.

https://doi.org/10.1371/journal.pone.0118463.t002

Six distinct Aβ plaque subtypes were identified in the ageing cohort: (1) diffuse plaques with no associated astrocytes (Fig. 1A); (2) compact plaques with no associated astrocytes (Fig. 1B); (3) diffuse plaques with focal astrogliosis (Fig. 1C); (4) compact plaques with focal astrogliosis (Fig. 1D); (5) diffuse plaques with circumferential astrogliosis (Fig. 1E); (6) compact plaques with circumferential astrogliosis (Fig. 1F).

thumbnail
Fig 1. Aβ plaque subtypes in the ageing brain.

Six distinct Aβ plaque subtypes were identified in the ageing cohort: (a) diffuse plaques with no associated astrocytes; (b) compact plaques (also known as classical or core plaques) with no associated astrocytes; (c) diffuse plaques with focal astrogliosis; (d) compact plaques with focal astrogliosis; (e) diffuse plaques with circumferential astrogliosis; (f) compact plaques with circumferential astrogliosis, as indicated by the arrow. Scale bar represents 50μm

https://doi.org/10.1371/journal.pone.0118463.g001

Both small (Fig. 2A) and large areas of astrogliosis (Fig. 2B) were also detected in regions remote from plaques. Initial investigation of the cohort noted that clusters of plaques with associated reactive astrocytes were frequently observed in the deep cortex near the white matter border (layer VI) (Fig. 2C), distinct from the areas of densest amyloid burden in layers I-V (Fig. 2D), therefore Aβ plaque subtype was assessed in both regions.

thumbnail
Fig 2. Astrogliosis remote from Aβ plaques, Aβ plaques in layer VI and regions of densest plaque burden.

Both (a) small and (b) large areas of astrogliosis were detected in regions remote from Aβ plaques, as indicated by the arrow. (c) Clusters of plaques with associated reactive astrocytes were frequently observed in the deep cortex (layer VI) near the white matter border (WM), distinct from (d) the areas of densest amyloid burden. Scale bar represents 50μm (a) and 100μm (b-d).

https://doi.org/10.1371/journal.pone.0118463.g002

Association of Aβ plaque subtype with Braak stage

In the area of densest burden diffuse plaques with no astrogliosis (β = 0.01, p<0.001) or with focal astrogliosis (β = 0.07, p<0.001) significantly associated with increasing Braak and Braak neurofibrillary tangle stage (Table 3). In layer VI, diffuse plaques with no (β = 0.04, p<0.001), focal (β = 0.08, p<0.001) or circumferential astrogliosis (β = 0.2, p = 0.003), as well as compact plaques with no (β = 0.21, p = 0.001), focal (β = 0.38, p<0.001) or circumferential astrogliosis (β = 0.45, p = 0.001) significantly associated with Braak stage (Table 4). Large areas (β = -0.32, p = 0.043), but not small areas (β = 0.02, p = 0.413), of non-plaque associated astrogliosis significantly associated with Braak stage.

thumbnail
Table 3. Association of Aβ plaque subtype in the area of densest Aβ burden with Braak stage, general cognition (MMSE), dementia status and possession of ApoEε4 allele.

https://doi.org/10.1371/journal.pone.0118463.t003

thumbnail
Table 4. Association of Aβ plaque subtype in the deep cortex (layer VI) with Braak stage, general cognition (MMSE), dementia status and possession of ApoEε4 allele.

https://doi.org/10.1371/journal.pone.0118463.t004

Aβ plaques with little or no associated astrogliosis correlate with cognitive impairment

In the area of densest burden, diffuse Aβ plaques with no astrogliosis (β = -0.05, p = 0.001) or with focal astrogliosis (β = -0.27, p = 0.009) significantly associated with lower MMSE scores (Table 3). In layer VI, both diffuse plaques and compact plaques without astrogliosis significantly associated with lower MMSE scores (β = -0.15, p = 0.017 and β = -0.81, p = 0.03, respectively) (Table 4). Only diffuse plaques with no astrogliosis in the deep cortex significantly related to dementia status (p = 0.025) (Table 3). Neither small nor large areas of non-plaque associated astrogliosis associated with either MMSE scores (β = -0.24, p = 0.114 and β = -0.25, p = 0.791, respectively) or dementia status (p = 0.248 and p = 0.558).

Association of Aβ plaque subtype with ApoE genotype

Possession of at least one ApoEε4 allele was significantly associated with a greater number of diffuse plaques with no (OR = 1.02, p = 0.001) or focal astrogliosis (OR = 1.10, p = 0.016) in the region of densest burden (Table 3), and with compact plaques with no (OR = 1.77, p = 0.003) or focal astrogliosis (OR = 1.50, p = 0.014) in layer VI (Table 4). Neither small (OR = 0.92, p = 0.313) nor large (OR = 0.89, p = 0.693) areas of non-plaque associated astrogliosis associated with ApoE genotype. A summary of the major Aβ plaque subtype associations with Braak stage, general cognition (MMSE) and possession of the ApoEε4 allele is shown in Table 5.

thumbnail
Table 5. Summary of the major Aβ plaque subtype associations with Braak stage, general cognition (MMSE) and possession of the ApoEε4 allele.

https://doi.org/10.1371/journal.pone.0118463.t005

Discussion

Several studies have demonstrated an association between Aβ plaques and astrogliosis; however whether these reactive astrocytes are actively contributing to ongoing neurodegenerative processes or play a neuroprotective role is highly debated [11,13,14,47]. The results of the current study demonstrate that astrogliosis associated with both diffuse and compact plaques in the area of densest burden and in the deep cortex (layer VI) negatively relates to cognitive impairment, and that possession of at least one copy of the ApoEε4 allele impacts the astroglial response to Aβ plaques.

The amyloid cascade hypothesis is currently the major theory of AD pathogenesis [48], however therapies based on removal of A have, to date, been disappointing [49]. Population-based studies have shown a weak association of amyloid pathologies with dementia [36,50], suggesting other factors contribute to cognitive impairment in the ageing brain. An additional possibility may be population variation in the response to Aβ deposits, with some individuals better able to prevent toxic effects on the surrounding neuropil. Inter-individual variation in the astrocyte response to Aβ deposition may be a part of this, and furthermore, ApoE genotype may be one regulator of the astrocyte response.

Amyloid plaque development starts in the superficial layers of the cortex and extends to the deep cortex as pathology progresses [51]. Increased levels of diffuse plaques without astrogliosis, but not plaque number, in both areas of densest burden and cortical layer VI demonstrated a significant association with lower MMSE scores, suggesting astrocytes play a neuroprotective role when associated with amyloid deposits. Diffuse, but not compact, plaques with no astrogliosis in layer VI strongly associated with dementia status.

In contrast to compact plaques which contain the fibrillar form of Aβ, diffuse plaques contain pre-fibrillary Aβ and may represent a precursor in plaque development [52]. The different composition of plaques may result in differences in toxicity, as intermediate forms of amyloid are considered as one of the most neurotoxic species of Aβ [1]. Intraneuronal accumulation of Aβ, rather than extracellular Aβ deposition may contribute to neuronal dysfunction and drive AD pathology [5355]. Future studies aimed at specifically assessing the association between astrogliosis and intraneuronal Aβ are required. We cannot demonstrate how reactive astrocytes might promote neuronal survival but they have been shown to protect neurones by regulating extracellular ion concentrations and neurotransmitter recycling [56], secreting neurotrophic factors [57], and to modulate Aβ-mediated neurotoxicity in vitro, safeguarding against neuronal dystrophy and synaptic loss [58]. Furthermore, astrocytes can degrade, internalise and clear Aβ [13,28,59], and have been shown to regulate microglial phagocytosis of compact plaque cores [20]. Our data suggests that astrocytes may form a protective barrier around amyloid plaques, demarcating the area for Aβ degradation, phagocytosis and a local inflammatory reaction. This would predict that plaques that are not insulated by astrocytes have a greater toxic effect on surrounding brain tissue. However, further investigations are required to confirm and characterise the neuroprotective role of astrocytes in response to Aβ plaque formation.

In the area of densest amyloid burden diffuse plaques with no, or focally, associated reactive astrocytes, but not with circumferential astrogliosis, demonstrated a significant association with Braak stage while all compact plaque subtypes showed a significant association with Braak stage, confirming the increasing burden of amyloid plaques in the cingulate gyrus mirrors the progression of tau pathology in the ageing brain [60]. The lack of association between diffuse plaques with circumferential astrogliosis and Braak stage suggests that the astrocyte reaction to plaque formation occurs at the earliest stages of tangle pathology, and does not directly parallel amyloid deposition [11]. Large regions of non-plaque associated astrogliosis showed no relation to dementia status or cognitive impairment, but did significantly associate with increasing Braak stage, supporting studies which suggest these astrogliotic lesions parallel neurofibrillary tangle progression and react to the burden of neurofibrillary tangles in the ageing brain [11,61]. AD is characterised by early damage to synapses [62,63] and dendritic atrophy [64]. Further work is required to investigate if the regions of non-plaque associated astrogliosis detected in this study reflect astrocyte reaction to dendritic degeneration and synaptic loss [60,65].

Possession of a single copy of the ApoEε4 allele is associated with a significant increased risk of developing AD [21,26], increased numbers of Aβ plaques [24], increased accumulation of intraneuronal Aβ [66] and elevated levels of astrogliosis [67]. In contrast to previous reports that ApoEε4 genotype does not impact glial responses to plaques in AD studies [47], the current findings demonstrate that in the area of densest Aβ burden diffuse amyloid plaques with little or no associated astrogliosis are significantly higher in ApoEε4 carriers in the ageing population. Although studies have demonstrated a significant correlation between compact plaques and ApoE genotype in AD [68,69], only compact plaques with no or focal astrogliosis in layer VI correlated with ApoE genotype. We have previously shown that astrocyte dysfunction in association with the progression of Alzheimer-type pathology is an early event for ApoEε4 carriers in this ageing cohort [25], and propose that this ApoEε4-associated astrocyte dysfunction may explain the lack of association with plaques with circumferential astrogliosis, and the significant association between increased levels of plaques with no or little astrogliosis and ApoE genotype.

In addition to an astrocyte response to Aβ plaques, microglial activation is also a prominent feature of AD pathology [70,71], and is associated with the degradation and clearance of Aβ [72,73]. While the activation and recruitment of microglia may occur in tandem with astrogliosis, studies have shown that reactive astrocytes and activated microglia respond differently to Aβ plaque formation and development [47], with activated microglia associated with proliferation and the secretion of pro-inflammatory cytokines [74]. Furthermore, CNS injury is associated with crosstalk between astrocytes and microglia involving a cytokine network, which regulates glial activation and impacts neuronal survival [75]. Future studies assessing microglial activation in addition to astrogliosis will enable a detailed characterisation of the glial response to Aβ plaque formation in the ageing brain.

The current study examined the astrocyte response to amyloid plaques solely in the cingulate gyrus, a region associated with metabolic and vascular changes in the very early stages of AD [33,34]. Expanding the investigation to include additional brain regions is essential to provide further validation to the findings reported here. In this study, the detection of plaques without associated astrocytes in a single section may have failed to detect a focal astrocyte response associated with larger Aβ plaques which span several sections. Quantitation of the number and subtype of plaques in serial sections, as opposed to a single field, would enable a three-dimensional astrocyte response to Aβ plaques to be determined. Further investigation into areas of reactive astrocytes remote from Aβ plaques should be performed to enable clearer definition of these lesions with respect to other local pathological features including synaptic loss, dendritic atrophy, tau pathology and microglial activation, as discussed above.

The current population-based study of the astrocyte response to amyloid plaques demonstrates clear relationships between Aβ plaque subtypes and cognitive impairment. Our findings may indicate a neuroprotective role of plaque-associated astrocytes, and suggest that astrogliosis may attenuate the neurotoxic effects of Aβ in the ageing brain. These findings encourage future studies to confirm the neuroprotective role of plaque-associated astrocytes and elucidate the precise mechanism(s) which may aid in the development of novel therapeutic strategies.

Conclusions

Reactive astrocytes in close proximity to either diffuse or compact plaques may have a neuroprotective role in the ageing brain, and possession of at least one copy of the ApoEε4 allele impacts the astroglial response to Aβ plaques.

Acknowledgments

CFAS would like to acknowledge the essential contribution of the liaison officers, the general practitioners, their staff, and nursing and residential home staff. We are grateful to our respondents and their families for their generous gift to medical research, which has made this study possible.

Author Contributions

Conceived and designed the experiments: SBW PGI. Performed the experiments: RM JES. Analyzed the data: TM FM SBW. Wrote the paper: RM JES SBW PGI TM CJG PJS FEM CB.

References

  1. 1. Querfurth HW, LaFerla FM. Alzheimer's disease. N Engl J Med. 2010;362: 329–344. pmid:20107219
  2. 2. Rozemuller JM, Eikelenboom P, Stam FC, Beyreuther K, Masters CL. A4 protein in Alzheimer's disease: primary and secondary cellular events in extracellular amyloid deposition. J Neuropathol Exp Neurol. 1989;48: 674–691. pmid:2677252
  3. 3. Tagliavini F, Giaccone G, Frangione B, Bugiani O. Preamyloid deposits in the cerebral cortex of patients with Alzheimer's disease and nondemented individuals. Neurosci Lett. 1988;93: 191–196. pmid:3241644
  4. 4. Dickson TC, Vickers JC. The morphological phenotype of beta-amyloid plaques and associated neuritic changes in Alzheimer's disease. Neuroscience. 2001;105: 99–107. pmid:11483304
  5. 5. Thal DR, Rub U, Schultz C, Sassin I, Ghebremedhin E, Del Tredici K. Sequence of Abeta-protein deposition in the human medial temporal lobe. J Neuropathol Exp Neurol. 2000;59: 733–748. pmid:10952063
  6. 6. Matthews F, Brayne C. The incidence of dementia in England and Wales: findings from the five identical sites of the MRC CFA Study. PLoS Med. 2005;2: e193. pmid:16111436
  7. 7. CFANS. Pathological correlates of late-onset dementia in a multicentre, community-based population in England and Wales. Neuropathology Group of the Medical Research Council Cognitive Function and Ageing Study (MRC CFAS). Lancet. 2001;357: 169–175. pmid:11213093
  8. 8. Savva GM, Wharton SB, Ince PG, Forster G, Matthews FE, Brayne C. Age, neuropathology, and dementia. N Engl J Med. 2009;360: 2302–2309. pmid:19474427
  9. 9. Maragakis NJ, Rothstein JD. Mechanisms of Disease: astrocytes in neurodegenerative disease. Nat Clin Pract Neurol. 2006;2: 679–689. pmid:17117171
  10. 10. Itagaki S, McGeer PL, Akiyama H, Zhu S, Selkoe D. Relationship of microglia and astrocytes to amyloid deposits of Alzheimer disease. J Neuroimmunol. 1989;24: 173–182. pmid:2808689
  11. 11. Serrano-Pozo A, Mielke ML, Gomez-Isla T, Betensky RA, Growdon JH, Frosch MP, et al. Reactive glia not only associates with plaques but also parallels tangles in Alzheimer's disease. Am J Pathol 2011;179: 1373–1384. pmid:21777559
  12. 12. Kamphuis W, Middeldorp J, Kooijman L, Sluijs JA, Kooi EJ, Moeton M, et al. Glial fibrillary acidic protein isoform expression in plaque related astrogliosis in Alzheimer's disease. Neurobiol Aging. 2014;35: 492–510. pmid:24269023
  13. 13. Thal DR. The role of astrocytes in amyloid beta-protein toxicity and clearance. Exp Neurol. 2012;236: 1–5. pmid:22575598
  14. 14. Medeiros R, LaFerla FM. Astrocytes: conductors of the Alzheimer disease neuroinflammatory symphony. Exp Neurol. 2013;239: 133–138. pmid:23063604
  15. 15. Rodriguez JJ, Olabarria M, Chvatal A, Verkhratsky A. Astroglia in dementia and Alzheimer's disease. Cell Death Differ. 2009;16: 378–385. pmid:19057621
  16. 16. Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. 2010;119: 7–35. pmid:20012068
  17. 17. Broe M, Kril J, Halliday GM. Astrocytic degeneration relates to the severity of disease in frontotemporal dementia. Brain. 2004;127: 2214–2220. pmid:15282215
  18. 18. Nagele RG, D'Andrea MR, Lee H, Venkataraman V,Wang HY. Astrocytes accumulate A beta 42 and give rise to astrocytic amyloid plaques in Alzheimer disease brains. Brain Res. 2003;971: 197–209. pmid:12706236
  19. 19. Kersaitis C, Halliday GM, Kril JJ. Regional and cellular pathology in frontotemporal dementia: relationship to stage of disease in cases with and without Pick bodies. Acta Neuropathol. 2004;108: 515–523. pmid:15368070
  20. 20. DeWitt DA, Perry G, Cohen M, Doller C, Silver J. Astrocytes regulate microglial phagocytosis of senile plaque cores of Alzheimer's disease. Exp Neurol. 1998;149: 329–340. pmid:9500964
  21. 21. Bertram L, McQueen MB, Mullin K, Blacker D, Tanzi RE. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet. 2007;39: 17–23. pmid:17192785
  22. 22. McNamara MJ, Gomez-Isla T, Hyman BT. Apolipoprotein E genotype and deposits of Abeta40 and Abeta42 in Alzheimer disease. Arch Neurol. 1998;55: 1001–1004. pmid:9678319
  23. 23. Schmechel DE, Saunders AM, Strittmatter WJ, Crain BJ, Hulette CM, Joo SH, et al. Increased amyloid beta-peptide deposition in cerebral cortex as a consequence of apolipoprotein E genotype in late-onset Alzheimer disease. Proc Natl Acad Sci USA. 1993;90: 9649–9653. pmid:8415756
  24. 24. Tiraboschi P, Hansen LA, Masliah E, Alford M, Thal LJ, Corey-Bloom J. Impact of APOE genotype on neuropathologic and neurochemical markers of Alzheimer disease. Neurology. 2004;62: 1977–1983. pmid:15184600
  25. 25. Simpson JE, Ince PG, Shaw PJ, Heath PR, Raman R, Garwood CJ, et al. Microarray analysis of the astrocyte transcriptome in the aging brain: relationship to Alzheimer's pathology and APOE genotype. Neurobiol Aging. 2011;32: 1795–1807. pmid:21705112
  26. 26. Kim J, Basak JM, Holtzman DM. The role of apolipoprotein E in Alzheimer's disease. Neuron. 2009;63: 287–303. pmid:19679070
  27. 27. Verghese PB, Castellano JM, Holtzman DM. Apolipoprotein E in Alzheimer's disease and other neurological disorders. Lancet Neurol. 2011;10: 241–252. pmid:21349439
  28. 28. Koistinaho M, Lin S, Wu X, Esterman M, Koger D, Hanson J, et al. Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat Med. 2004;10: 719–726. pmid:15195085
  29. 29. Simpson JE, Ince PG, Lace G, Forster G, Shaw PJ, Matthews F, et al. Astrocyte phenotype in relation to Alzheimer-type pathology in the ageing brain. Neurobiol Aging. 2010;31: 578–590. pmid:18586353
  30. 30. Sutherland RJ, Whishaw IQ, Kolb B. Contributions of cingulate cortex to two forms of spatial learning and memory. J Neurosci. 1988;8: 1863–1872. pmid:3385478
  31. 31. Thal DR, Rub U, Orantes M, Braak H. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58: 1791–1800. pmid:12084879
  32. 32. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82: 239–259. pmid:1759558
  33. 33. Huang C, Wahlund LO, Svensson L, Winblad B, Julin P. Cingulate cortex hypoperfusion predicts Alzheimer's disease in mild cognitive impairment. BMC Neurol. 2002;2: 9. pmid:12227833
  34. 34. Minoshima S, Giordani B, Berent S, Frey KA, Foster NL, Kuhl DE. Metabolic reduction in the posterior cingulate cortex in very early Alzheimer's disease. Ann Neurol. 1997;42: 85–94. pmid:9225689
  35. 35. CFANS. Cognitive function and dementia in six areas of England and Wales: the distribution of MMSE and prevalence of GMS organicity level in the MRC CFA Study. The Medical Research Council Cognitive Function and Ageing Study (MRC CFAS). Psychol Med. 1998;28: 319–335. pmid:9572090
  36. 36. Wharton SB, Brayne C, Savva GM, Matthews FE, Forster G, Simpson J, et al. Epidemiological neuropathology: the MRC Cognitive Function and Aging Study experience. J Alzheimers Dis. 2011;25: 359–372. pmid:21422529
  37. 37. Ince PG, McArthur FK, Bjertness E, Torvik A, Candy JM, Edwardson J. Neuropathological diagnoses in elderly patients in Oslo: Alzheimer's disease, Lewy body disease, vascular lesions. Dementia. 1995;6: 162–168. pmid:7620529
  38. 38. Mirra SS. The CERAD neuropathology protocol and consensus recommendations for the postmortem diagnosis of Alzheimer's disease: a commentary. Neurobiol Aging. 1997;18: S91–94. pmid:9330994
  39. 39. Braak H, Alafuzoff I, Arzberger T, Kretzschmar H, Del Tredici K. Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol. 2006;112: 389–404. pmid:16906426
  40. 40. Nicoll JA, Savva GM, Stewart J, Matthews FE, Brayne C, Ince PG. Association between APOE genotype, neuropathology and dementia in the older population of England and Wales. Neuropathol Appl Neurobiol. 2011;37: 285–294. pmid:20880354
  41. 41. Yip AG, Brayne C, Easton D, Rubinsztein DC. Apolipoprotein E4 is only a weak predictor of dementia and cognitive decline in the general population. J Med Genet. 2002;39: 639–643. pmid:12205106
  42. 42. Akiyama H, Mori H, Saido T, Kondo H, Ikeda K, McGeer PL. Occurrence of the diffuse amyloid beta-protein (Abeta) deposits with numerous Abeta-containing glial cells in the cerebral cortex of patients with Alzheimer's disease. Glia. 1999;25: 324–331. pmid:10028915
  43. 43. Oh D, Prayson RA. Evaluation of epithelial and keratin markers in glioblastoma multiforme: an immunohistochemical study. Arch Pathol Lab Med. 1999;123: 917–920. pmid:10506444
  44. 44. Greenfield JG, Love S, Louis DN, Ellison DP. Greenfield's neuropathology. London: Hodder Arnold. 2008.
  45. 45. Gwet KL. Computing inter-rater reliability and its variance in the presence of high agreement. Br J Math Stat Psychol. 2008;61: 29–48. pmid:18482474
  46. 46. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics. 1977;33: 159–174. pmid:843571
  47. 47. Serrano-Pozo A, Muzikansky A, Gomez-Isla T, Growdon JH, Betensky RA, Frosch MP, et al. Differential relationships of reactive astrocytes and microglia to fibrillar amyloid deposits in Alzheimer disease. J Neuropathol Exp Neurol. 2013;72: 462–471. pmid:23656989
  48. 48. Hardy J. The amyloid hypothesis for Alzheimer's disease: a critical reappraisal. J Neurochem. 2009;110: 1129–1134. pmid:19457065
  49. 49. Holmes C, Boche D, Wilkinson D, Yadegarfar G, Hopkins V, Bayer A, et al. Long-term effects of Abeta42 immunisation in Alzheimer's disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet. 2008;372: 216–223. pmid:18640458
  50. 50. Fotuhi M, Hachinski V, Whitehouse PJ. Changing perspectives regarding late-life dementia. Nat Rev Neurol. 2009;5: 649–658. pmid:19918254
  51. 51. Romito-DiGiacomo RR, Menegay H, Cicero SA, Herrup K. Effects of Alzheimer's disease on different cortical layers: the role of intrinsic differences in Abeta susceptibility. J Neurosci. 2007;27: 8496–8504. pmid:17687027
  52. 52. Morgan C, Colombres M, Nunez MT, Inestrosa NC. Structure and function of amyloid in Alzheimer's disease. Prog Neurobiol. 2004;74: 323–349. pmid:15649580
  53. 53. Gouras GK, Almeida CG, Takahashi RH. Intraneuronal Abeta accumulation and origin of plaques in Alzheimer's disease. Neurobiol Aging. 2005;26: 1235–1244. pmid:16023263
  54. 54. Wirths O, Multhaup G, Bayer TA. A modified beta-amyloid hypothesis: intraneuronal accumulation of the beta-amyloid peptide—the first step of a fatal cascade. J Neurochem. 2004;91: 513–520. pmid:15485483
  55. 55. LaFerla FM, Green KN, Oddo S. Intracellular amyloid-beta in Alzheimer's disease. Nat Rev Neurosci. 2007;8: 499–509. pmid:17551515
  56. 56. Parpura V, Heneka MT, Montana V, Oliet SH, Schousboe A, Haydon PG, et al. Glial cells in (patho)physiology. J Neurochem. 2012;121: 4–27. pmid:22251135
  57. 57. Gozes I, Bassan M, Zamostiano R, Pinhasov A, Davidson A, Giladi E, et al. A novel signaling molecule for neuropeptide action: activity-dependent neuroprotective protein. Ann N Y Acad Sci. 1999;897: 125–135. pmid:10676441
  58. 58. Paradisi S, Sacchetti B, Balduzzi M, Gaudi S, Malchiodi-Albedi F. Astrocyte modulation of in vitro beta-amyloid neurotoxicity. Glia. 2004;46: 252–260. pmid:15048848
  59. 59. Belanger M, Magistretti PJ. The role of astroglia in neuroprotection. Dialogues Clin Neurosci. 2009;11: 281–295. pmid:19877496
  60. 60. Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT. Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med. 2011;1: a006189. pmid:22229116
  61. 61. Ikeda K, Akiyama H, Haga C, Haga S. (1992) Evidence that neurofibrillary tangles undergo glial modification. Acta Neuropathol. 1992;85: 101–104. pmid:1285492
  62. 62. Masliah E. The role of synaptic proteins in Alzheimer's disease. Ann N Y Acad Sci. 2000;924: 68–75. pmid:11193804
  63. 63. Crews L, Masliah E. Molecular mechanisms of neurodegeneration in Alzheimer's disease. Hum Mol Genet. 2010;19: R12–20. pmid:20413653
  64. 64. Grutzendler J, Helmin K, Tsai J, Gan WB. Various dendritic abnormalities are associated with fibrillar amyloid deposits in Alzheimer's disease. Ann N Y Acad Sci. 2007;1097: 30–39. pmid:17413007
  65. 65. Knobloch M, Mansuy IM. Dendritic spine loss and synaptic alterations in Alzheimer's disease. Mol Neurobiol. 2008;37: 73–82. pmid:18438727
  66. 66. Christensen DZ, Schneider-Axmann T, Lucassen PJ, Bayer TA, Wirths O. Accumulation of intraneuronal Abeta correlates with ApoE4 genotype. Acta Neuropathol. 2010;119: 555–566. pmid:20217101
  67. 67. Overmyer M, Helisalmi S, Soininen H, Laakso M, Riekkinen P Sr, Alafuzoff I. Astrogliosis and the ApoE genotype. an immunohistochemical study of postmortem human brain tissue. Dement Geriatr Cogn Disord. 1999;10: 252–257. pmid:10364641
  68. 68. Olichney JM, Hansen LA, Galasko D, Saitoh T, Hofstetter CR, Katzman R, et al. The apolipoprotein E epsilon 4 allele is associated with increased neuritic plaques and cerebral amyloid angiopathy in Alzheimer's disease and Lewy body variant. Neurology. 1996;47: 190–196. pmid:8710076
  69. 69. Sparks DL, Scheff SW, Liu H, Landers T, Danner F, Coyne CM, et al. Increased density of senile plaques (SP), but not neurofibrillary tangles (NFT), in non-demented individuals with the apolipoprotein E4 allele: comparison to confirmed Alzheimer's disease patients. J Neurol Sci. 1996;138: 97–104. pmid:8791246
  70. 70. Wyss-Coray T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med. 2006;12: 1005–1015. pmid:16960575
  71. 71. Perry VH, Nicoll JA, Holmes C. Microglia in neurodegenerative disease. Nat Rev Neurol. 2010;6: 193–201. pmid:20234358
  72. 72. Rogers J, Strohmeyer R, Kovelowski CJ, Li R. Microglia and inflammatory mechanisms in the clearance of amyloid beta peptide. Glia. 2002;40: 260–269. pmid:12379913
  73. 73. Yang CN, Shiao YJ, Shie FS, Guo BS, Chen PH, Cho CY, et al. Mechanism mediating oligomeric Abeta clearance by naive primary microglia. Neurobiol Dis. 2011;42: 221–230. pmid:21220023
  74. 74. Marlatt MW, Bauer J, Aronica E, van Haastert ES, Hoozemans JJ, Joels M, et al. Proliferation in the Alzheimer hippocampus is due to microglia, not astroglia, and occurs at sites of amyloid deposition. Neural Plast. 2014: 693851. pmid:25215243
  75. 75. Giulian D, Li J, Li X, George J, Rutecki PA. The impact of microglia-derived cytokines upon gliosis in the CNS. Dev Neurosci. 1994;16: 128–136. pmid:7535679