Skip to main content
Erschienen in: Translational Neurodegeneration 1/2018

Open Access 01.12.2018 | Review

Impact of the biological definition of Alzheimer’s disease using amyloid, tau and neurodegeneration (ATN): what about the role of vascular changes, inflammation, Lewy body pathology?

verfasst von: S. Gauthier, H. Zhang, K. P. Ng, T.A. Pascoal, P. Rosa-Neto

Erschienen in: Translational Neurodegeneration | Ausgabe 1/2018

Abstract

Background

The NIA-AA research framework proposes a biological definition of Alzheimer’s disease, where asymptomatic persons with amyloid deposition would be considered as having this disease prior to symptoms.

Discussion

Notwithstanding the fact that amyloid deposition in isolation is not associated with dementia, even the combined association of amyloid and tau pathology does not inevitably need to dementia over age 65. Other pathological factors may play a leading or an accelerating role in age-associated cognitive decline, including vascular small vessel disease, neuroinflammation and Lewy Body pathology.

Conclusion

Research should aim at understanding the interaction between all these factors, rather than focusing on them individually. Hopefully this will lead to a personalized approach to the prevention of brain aging, based on individual biological, genetic and cognitive profiles.

Background

The treatment of Alzheimer’s disease (AD) is currently symptomatic and based on neurotransmitter manipulation, akin to what has been achieved in Parkinson’s disease. Thus acetycholine activity is being increased by cholinesterase inhibitors, and glutamatergic activity is being dampened by memantine action on NMDA receptors. A modest but clinically detectable response is present in many patients using such drugs alone or in combination.
Unfortunately the next generation of drugs acting on AD core pathological factors such as amyloid deposition and phosphorylated tau aggregation has failed so far to delay disease progression, raising the issue of timing of these interventions along the continuum of AD neurodegeneration over time.
This review wants to highlight the facts that other pathological factors are at play in AD, and deserve consideration in the full diagnostic assessment of the patients, and for treatment. These factors are vascular changes, Lewy body pathology and neuroinflammation.

Classic pathology of AD

The clinical progression of AD is linked to specific neuropathological features, such as extracellular deposition of Aβ plaques, intracellular inclusions of tau protein in neurofibrillary tangles, and neuronal degeneration. The discovery and advance of disease biomarkers over the last decade have significantly advanced our understanding of the dynamic pathophysiological changes underlying AD and have allowed the detection of AD pathophysiology in vivo [1]. Given that the presence of AD pathophysiology has been found across a broad clinical spectrum including individuals asymptomatic and with mild cognitive symptoms, biomarkers now play an important role in characterizing the trajectory of AD pathophysiology and have been incorporated in the AD diagnostic research criteria [25]. These diagnostic research criterions recognize that the coexistence of abnormal Aβ and tau biomarkers better identify the preclinical and MCI individuals who will progress to dementia over relatively short time frames of three to 5 years.
Based on histopathological and genetic evidences, fibrillar Aβ, the main constituent of Aβ plaques, has been postulated as the major driving force leading to AD dementia (Aβ cascade hypothesis). According to this hypothesis, all the resulting pathological processes are due to an imbalance between Aβ production and clearance, which would then potentiate the spread of tauopathy, leading to neurodegeneration and cognitive decline. However, the lack of consistent association between Aβ and clinical progression, and the fact that amyloid pathology has been found in cognitively normal elderly individuals challenge the Aβ hypothesis in its original form.

Proposal for a new classification system

An unbiased biomarker classification system, A/T/N, which avoids the assumptions of the temporal ordering of AD biomarkers, has been proposed [6]. In this classification system where each biomarker category is binarized as either positive or negative, “A” represents Aβ biomarkers using amyloid PET or CSF Aβ42, “T” represents tau biomarkers using CSF p-tau or tau PET, and “N” represents neurodegeneration biomarkers using CSF p-tau, structural MRI or [18F]fluorodeoxyglucose PET (FDG). This descriptive classification aims to organize the multi-modality biomarker results at the individual person level in a way that is easy to adopt and interpret. Other brain pathological processes have been postulated as natural candidates to integrate this unbiased system. Studies under way are measuring simultaneously the amyloid, tau, and neuroinflammation in individuals, with follow-up over time to test the hypothesis that the coexistence of the brain pathological factors may accelerates AD clinical manifestations.
We argue that the A/T/N classification may be broadened to include other key pathological factors: vascular pathology, Lewy Body pathology and neuroinflammation.

Vascular changes

There is growing evidence that AD often coexists with cerebrovascular disease (CVD). They share many risk factors, leading to additive or synergistic effects on cognitive decline [7]. The APOE ε4 allele is the strongest genetic risk factor for late-onset AD, and APOE ε4 is also associated with increasing burden in MRI markers for both ischemic and hemorrhagic CVD [8]. There is increasingly robust relationship between other risk factors including hypertension, diabetes, atrial fibrillation, hypercholesterolemia, smoking, hyperhomocysteinaemia, age and obesity and AD, whereas there are possible protective effect of the ‘Mediterranean’ diet and physical exercise [914]. Although not all studies have found a correlation between vascular risk factors and AA [15, 16], it has been reported that the presence of vascular risk factors can predict the development of AD or the conversion from mild cognitive impairment (MCI) to AD [9, 17, 18].
Most AD patients have structural changes in their cerebral blood vessels. Imaging and pathological studies have demonstrated a high prevalence of arteriolosclerotic small vessel disease (SVD) in AD patients. Post-mortem and imaging studies demonstrate that arteriolar Aβ amyloid angiopathy, a sub-type of SVD, is more common in patients with AD than in elderly controls [1923]. The amyloid angiopathy mainly affects the leptomeningeal, cortical and capillary vessel walls, but sometimes the cerebellum, and occasionally the brainstem [12, 24]. In the autopsy studies, it suggests that AD is correlated with atherosclerosis of the Circle of Willis, and the severity of the atherosclerosis is associated with neuritic plaques and neurofibrillary tangles [2527].
An important component of CVD in AD is cerebral hypoperfusion, which can be present several years before the onset of clinical symptoms. The diffusion pattern of cerebral hypoperfusion is stereotyped in AD: the first affected area of is the precuneus, which has appeared 10 years before the onset of AD, followed by the cingulate gyrus and the lateral part of the parietal lobe, then the frontal and temporal lobes, and the eventually the cerebrum [12]. The main mechanism of cerebral hypoperfusion in AD may be non-structural [12]. In vivo and in vitro studies have shown cerebral hypoperfusion increases the production of Aβ and tau hyperphosphorylation, reduces the clearance of Aβ, then aggravates the progress of AD [2833]. There is good evidence that Aβ amyloid angiopathy and SVD are associated with infarction and cerebral hemorrhage in AD [3443]. The mechanisms may involve susceptibility to thrombosis, reduction of blood flow, impaired caliber regulation, and impaired function of the blood-brain barrier (BBB). Infarction or bleeding will reduce the threshold for the onset of AD, and is considered as an important risk factor for the clinical manifestations of AD [44, 45].
The links between vascular factors and AD have been clearly confirmed both clinically and pathologically. However, there is a lack of high-quality therapeutic research to examine the extent to which vascular risk changes alter the course of AD. Further longitudinal mechanisms and therapeutic studies are needed, especially to determine whether the treatment of vascular risk factors can prevent or delay the onset of AD.

Lewy body pathology

Although the accumulation of amyloid protein in plaques and tau protein in neurofibrillary tangles constitutes the core pathological feature of AD, the presence of abnormal brain aggregates of a third proteinopathy has been shown to be very prevalent in moderate and severe AD [4648]. Cytoplasmic inclusions of α-synuclein intraneuronally in Lewy bodies have been reported in up to 50% of sporadic AD cases and up to 60% of familial AD cases [4952]. In the context of AD, it is still unclear whether the overlap between Lewy bodies and the hallmark AD proteins occurs due to a mere co-occurrence of independent pathological processes or is the manifestation of interconnected pathological processes.
Histopathological studies have shown that Lewy bodies normally accumulate in a specific topographic brain pattern, starting in the brainstem and subsequently extending to the limbic and neocortical brain regions [47]. In contrast, in AD patients the Lewy bodies deposition concentrates in the amygdala with little deposition in the brainstem or neocortex [53]. This characteristic pattern of deposition has been called AD with amygdala Lewy bodies [47, 54]. Interestingly, the Lewy bodies in the amygdala normally overlap with tau accumulation [55] and neuronal loss [56], suggesting that pathological interaction between these pathologies may play a role in the progression of AD. The severity of the pathology in the amygdala correlates with disease duration [55, 56] and emotional and memory difficulties [57], which suggest that the aforementioned interaction plays a role in the AD clinical phenotype in this group of individuals [58]. Moreover, the cortical concentrations of Lewy bodies have been correlated with amyloid burden [59, 60] and neurofibrillary tangles [61, 62].
Postmortem observations focusing on the influence of Lewy bodies and the phonotypical presentation of AD have shown inconsistent results. These studies presented opposing results whether the presence of Lewy bodies in AD patients has an effect on the age of onset of symptoms, death [51, 6365], the likelihood of be an APOE ε4 carrier [51, 65, 66], parkinsonian symptoms [63, 64, 67, 68], cognitive impairment [63, 64], or visual hallucinations [64, 6870]. This disagreement arises in part, due to the fact that most of these studies have small sample sizes or limited range of AD phonotypical presentations. However, it is worth to mention that a well-powered multicenter study with a high sample size has reported that the onset of symptoms and death in AD individuals with Lewy bodies occurs at younger ages as compared to those without Lewy bodies, and that AD individuals with Lewy bodies have higher chance to be APOE ε4 carriers than AD individuals without Lewy bodies. Moreover, this study also suggested that hallucinations, motor disturbs, and sleep problems are more severe in AD individuals with Lewy bodies than in the ones without Lewy bodies [51].
The α-synuclein protein, which in the main constituent of the Lewy bodies, can be measured in the cerebrospinal fluid (CSF) of living people [71]. Some CSF studies have reported an increase in α-synuclein levels in patients with MCI and AD as compared to controls [7274], whereas other studies have shown no difference or reduced levels across the AD clinical spectrum [7577]. The levels of α-synuclein have shown positive correlation with CSF tau pathology in some studies [73, 77] and no correlation in another [78].
Although the characteristic topographic presentation and the frequency of Lewy bodies in AD suggest a potential common mechanism for AD and Lewy bodies, the divergence in results between studies indicates that further studies are imperative to clarify the role of Lewy body pathology in AD. One of the main limitations of the current studies is the absence of an imaging agent able to capture Lewy bodies in the living brain. Many groups are trying in developing such an imaging agent in order to provide the means to definitively clarify the dynamical changes of Lewy bodies in the human brain and its interplay with amyloid, tau, neuroinflammation, and vascular pathology.

Neuroinflammation

In addition to hallmark AD neuropathological features such as amyloid (Aβ) plaques, neurofibrillary tangles and neuronal degeneration, there is a growing body of evidence supporting neuroinflammation as an important player in the pathogenesis of AD [79, 80]. Neuropathological studies have shown the presence of activated microglia and inflammation related mediators in AD brains of low Braak stage [81], while genetic studies show that several genes that increase the risk of sporadic AD encode factors that regulate microglial clearance of misfolded proteins and inflammatory reaction, such as TREM2 and CD33 [82, 83]. Epidemiological studies further suggest that non-steroidal anti-inflammatory drugs (NSAIDS) can defer or prevent the onset of AD [84, 85]. Although subsequent clinical trials involving prednisolone and NSAIDS, such as the Alzheimer’s Disease Anti-inflammatory Prevention Trial (ADAPT), failed to show improvement in cognitive decline in AD patients or prevent AD progression in adults with a family history of dementia [86], the difference between observational and randomised studies will need to be clarified in future studies.
Microglia, the resident phagocytes of the brain, plays an integral role in maintaining brain homeostasis and protecting the brain from insults by mounting an innate immune response when activated [87]. Preclinical and post-mortem studies have consistently found that activated microglia colocalises with Aβ plaque [88, 89], suggesting a close intimate relationship between microglia activation, Aβ and neuroinflammation. In AD, microglia bind to soluble Aβ oligomers and fibrils via cell surface receptors, which triggers the activation of microglia [80]. A key issue is whether this response is adaptive or maladaptive in nature. While acute microglia activation triggered by Aβ is aimed to eliminate Aβ aggregation via phagocytosis, there is an inefficient clearance of Aβ plaques [90]. Several mechanisms have been hypothesised, including ongoing formation of Aβ and positive feedback loops between inflammation and amyloid precursor protein (APP) processing which compromise the cessation of neuroinflammation. Continued exposure to Aβ, chemokines, cytokines, and inflammatory mediators leads to microglia being chronically activated at the Aβ plaque site, which further contribute to Aβ production and accumulation in a vicious cycle.
Microglia and neuroinflammation are also closely associated with tau in AD. Reactive microglia can produce inflammatory cytokines such as IL-1 which lead to an increase in tau phosphorylation in neurons [91]. This may contribute to the development of tau pathology and thus accelerate the course of disease. Furthermore, misfolded tau may also trigger microglial activation [92]. In preclinical studies, reactive microglia are found to be sufficient in driving tau pathology and contribute to the spread of pathological tau in the brain [93]. Microglia have also been shown to internalize tau protein both in vitro and in vivo. In post-mortem studies, microglia colocalise with various forms of tau in brain tissue of AD patients [94].
Given the dynamic relationship between Aβ, tau and microglia in AD, it is imperative to study the interplay between these pathophysiologies so as to further understand the sequence of events underlying the AD process. In this regard, studies that measure Aβ, tau, and neuroinflammation concurrently will be of paramount importance. The findings of these studies will further broaden the A/T/N classification of individuals to include neuroinflammation biomarkers.

Conclusion

Towards an integration of the various pathological factors leading to targeted treatments.
This expanded view of the pathological factors at play in persons with AD may be lead to therapeutic strategies targeting the most active factors at a given time in each individual. We hope that meta-analysis of current observational studies such as ADNI and others under development such as COMPASS-ND will facilitate the validation of various imaging, CSF and blood markers for each of these pathologies, as illustrated in Table 1. In other words “mixed dementia” which is most common finding in autopsy studies will be in the near future be studied based on biomarkers. This may allow for more homogeneous groups of patients to be studied in randomized clinical trials require combination therapy, as a first step towards a personalized approach to treatment of AD throughout its course.
Table 1
Study of the various known pathological factors in AD
Factor
Imaging
CSF
Blood
Potential RX
Amyloid-β load
[11C]PIB
[18F]NAV4694
[18F]florbetapir
[18F]florbetaben
[18F]flutemetamol
Amyloid-β(1–42)
APP669–711;
Amyloid-β(1–42);
Amyloid-β(1–40);
BACE inhibitors
Amyloid-β immunotherapy
Neurofibrillary tangles load
[18F]MK6240
[18F]AV1451
[11C]PBBB3
Phosphorylated tau
The association of serum phosphorylated tau with tangles is unclear
Anti-aggregation
Tau immunotherapy
Neurodegeneration
MRI
[18F]FDG
Neurofilament light chain (NFL); d neurogranin (Ng);
Visinin-like protein-1 (VILIP-1); Synaptosomal-associated protein 25 (SNAP-25);
Neuron-specific enolase (NSE); Heart fatty acid binding protein (HFABP)
Neurofilament light chain (NFL)
Neurotrophic factors
Vascular load
MRI
CSF albumin /plasma albumin ratio
Control of risk factors
Lewy Body load
NA
α-synuclein
α-synuclein
α-synuclein immunotherapy
Neuroinflammation activity
Microglial Activation:
[11C]PK11195
[11C]PBR28
[11C]DAA1106
[18F] DPA714
[11C] DPA713
[18F]GE180
Reactive astrocytes:
[11C]L-des-deprenyl
Microglial Activation:
Chitinase-3-like protein 1 (YKL-40), soluble TREM2 (sTREM2)
Cytokines:
TNF-α, IL-6, IL-1β
Chemokines: monocyte chemotactic protein 1 [MCP-1]
Microglial Activation:
Chitinase-3-like protein 1 (YKL-40)
Cytokines:
TNF-α, IL-6, IL-1β,
Chemokines: monocyte chemotactic protein 1 [MCP-1]
NSAIDS
Peroxisome proliferator-activated receptor-γ
(PPAR-γ) activators
TNF-α inhibitor

Acknowledgements

The author’s research is funded by the Canadian Consortium on Neurodegeneration in Aging, the Canadian Institutes for Health Research, and The Weston Brain Institute.

Funding

The author’s research is funded by the Canadian Consortium on Neurodegeneration in Aging, the Canadian Institutes for Health Research, and The Weston Brain Institute.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat Dubois B, Feldman H, Jacova C, DeKosky ST, Barberger-Gateau P, Cummings J, Delacourte A, Galasko D, Gauthier S, Jicha G, Meguro K, O’Brien J, Pasquier F, Robert P, Rossor M, Salloway S, Stern Y, Visser PJ, Scheltens P. Research criteria for the diagnosis of Alzheimer’s disease: revisiting of the NINCDS-ADRDA criteria. Lancet Neurol. 2007;6:734–46.CrossRefPubMed Dubois B, Feldman H, Jacova C, DeKosky ST, Barberger-Gateau P, Cummings J, Delacourte A, Galasko D, Gauthier S, Jicha G, Meguro K, O’Brien J, Pasquier F, Robert P, Rossor M, Salloway S, Stern Y, Visser PJ, Scheltens P. Research criteria for the diagnosis of Alzheimer’s disease: revisiting of the NINCDS-ADRDA criteria. Lancet Neurol. 2007;6:734–46.CrossRefPubMed
3.
Zurück zum Zitat McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, Mohs RC, Morris JC, Rossor MN, Scheltens P, Carrillo MC, Thies B, Weintraub S, Phelps CH. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:263–9.CrossRefPubMedPubMedCentral McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR, Kawas CH, Klunk WE, Koroshetz WJ, Manly JJ, Mayeux R, Mohs RC, Morris JC, Rossor MN, Scheltens P, Carrillo MC, Thies B, Weintraub S, Phelps CH. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:263–9.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, Gamst A, Holtzman DM, Jagust WJ, Petersen RC, Snyder PJ, Carrillo MC, Thies B, Phelps CH. The diagnosis of mild cognitive impairment due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:270–9.CrossRefPubMedPubMedCentral Albert MS, DeKosky ST, Dickson D, Dubois B, Feldman HH, Fox NC, Gamst A, Holtzman DM, Jagust WJ, Petersen RC, Snyder PJ, Carrillo MC, Thies B, Phelps CH. The diagnosis of mild cognitive impairment due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:270–9.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, Iwatsubo T, Jack CR, Kaye J, Montine TJ, Park DC, Reiman EM, Rowe CC, Siemers E, Stern Y, Yaffe K, Carrillo MC, Thies B, Morrison-Bogorad M, Wagster MV, Phelps CH. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging and the Alzheimer’s association workgroup. Alzheimers Dement. 2011;7:1–13. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, Iwatsubo T, Jack CR, Kaye J, Montine TJ, Park DC, Reiman EM, Rowe CC, Siemers E, Stern Y, Yaffe K, Carrillo MC, Thies B, Morrison-Bogorad M, Wagster MV, Phelps CH. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging and the Alzheimer’s association workgroup. Alzheimers Dement. 2011;7:1–13.
6.
Zurück zum Zitat Jack CR, Hampel HJ, Universities S, Cu M, Petersen RC. A/T/N: an unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology. 2016;87:539–47.CrossRefPubMedPubMedCentral Jack CR, Hampel HJ, Universities S, Cu M, Petersen RC. A/T/N: an unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology. 2016;87:539–47.CrossRefPubMedPubMedCentral
8.
13.
Zurück zum Zitat Nagy ZS, Smith MZ, Esiri MM, Barnetson L, Smith AD. Hyperhomocysteinaemia in Alzheimer’s disease and expression of cell cycle markers in the brain. J Neurol Neurosurg Psychiatry. 2000;69(4):565–6.CrossRefPubMedPubMedCentral Nagy ZS, Smith MZ, Esiri MM, Barnetson L, Smith AD. Hyperhomocysteinaemia in Alzheimer’s disease and expression of cell cycle markers in the brain. J Neurol Neurosurg Psychiatry. 2000;69(4):565–6.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Chui HC, Zheng L, Reed BR, Vinters HV, Mack WJ. 2012.Vascular risk factors and Alzheimer’s disease: are these risk factors for plaques and tangles or for concomitant vascular pathology that increases the likelihood of dementia? An evidence-based review. Alzheimers Res Ther. 4(1):1. https://doi.org/10.1186/alzrt98. Chui HC, Zheng L, Reed BR, Vinters HV, Mack WJ. 2012.Vascular risk factors and Alzheimer’s disease: are these risk factors for plaques and tangles or for concomitant vascular pathology that increases the likelihood of dementia? An evidence-based review. Alzheimers Res Ther. 4(1):1. https://​doi.​org/​10.​1186/​alzrt98.
16.
Zurück zum Zitat Richardson K, Stephan BC, Ince PG, Brayne C, Matthews FE, Esiri MM. The neuropathology of vascular disease in the Medical Research Council cognitive function and ageing study (MRC CFAS). Curr Alzheimer Res. 2012;9(6):687–96.CrossRefPubMed Richardson K, Stephan BC, Ince PG, Brayne C, Matthews FE, Esiri MM. The neuropathology of vascular disease in the Medical Research Council cognitive function and ageing study (MRC CFAS). Curr Alzheimer Res. 2012;9(6):687–96.CrossRefPubMed
20.
Zurück zum Zitat Carmona-Iragui M, Balasa M, Benejam B, Alcolea D, Fernandez S, Videla L, Sala I, Sanchez-Saudinos MB, Morenas-Rodriguez E, Ribosa-Nogue R, Illan-Gala I, Gonzalez-Ortiz S, Clarimon J, Schmitt F, Powell DK, Bosch B, Llado A, Rafii MS, Head E, Molinuevo JL, Blesa R, Videla S, Lleo A, Sanchez-Valle R, Fortea J. Cerebral amyloid angiopathy in Down syndrome and sporadic and autosomal-dominant Alzheimer’s disease. Alzheimers Dement. 2017;13(11):1251–60. https://doi.org/10.1016/j.jalz.2017.03.007.CrossRefPubMed Carmona-Iragui M, Balasa M, Benejam B, Alcolea D, Fernandez S, Videla L, Sala I, Sanchez-Saudinos MB, Morenas-Rodriguez E, Ribosa-Nogue R, Illan-Gala I, Gonzalez-Ortiz S, Clarimon J, Schmitt F, Powell DK, Bosch B, Llado A, Rafii MS, Head E, Molinuevo JL, Blesa R, Videla S, Lleo A, Sanchez-Valle R, Fortea J. Cerebral amyloid angiopathy in Down syndrome and sporadic and autosomal-dominant Alzheimer’s disease. Alzheimers Dement. 2017;13(11):1251–60. https://​doi.​org/​10.​1016/​j.​jalz.​2017.​03.​007.CrossRefPubMed
22.
Zurück zum Zitat Love S, Chalmers K, Ince P, Esiri M, Attems J, Jellinger K, Yamada M, McCarron M, Minett T, Matthews F, Greenberg S, Mann D, Kehoe PG. Development, appraisal, validation and implementation of a consensus protocol for the assessment of cerebral amyloid angiopathy in post-mortem brain tissue. Am J Neurodegenerative Dis. 2014;3(1):19–32. Love S, Chalmers K, Ince P, Esiri M, Attems J, Jellinger K, Yamada M, McCarron M, Minett T, Matthews F, Greenberg S, Mann D, Kehoe PG. Development, appraisal, validation and implementation of a consensus protocol for the assessment of cerebral amyloid angiopathy in post-mortem brain tissue. Am J Neurodegenerative Dis. 2014;3(1):19–32.
32.
Zurück zum Zitat Shang J, Yamashita T, Zhai Y, Nakano Y, Morihara R, Fukui Y, Hishikawa N, Ohta Y, Abe K. Strong impact of chronic cerebral Hypoperfusion on neurovascular unit, cerebrovascular remodeling, and neurovascular trophic coupling in Alzheimer’s disease model mouse. J Alzheimers Dis. 2016;52(1):113–26. https://doi.org/10.3233/jad-151126. CrossRefPubMed Shang J, Yamashita T, Zhai Y, Nakano Y, Morihara R, Fukui Y, Hishikawa N, Ohta Y, Abe K. Strong impact of chronic cerebral Hypoperfusion on neurovascular unit, cerebrovascular remodeling, and neurovascular trophic coupling in Alzheimer’s disease model mouse. J Alzheimers Dis. 2016;52(1):113–26. https://​doi.​org/​10.​3233/​jad-151126.​ CrossRefPubMed
33.
Zurück zum Zitat Zhai Y, Yamashita T, Nakano Y, Sun Z, Shang J, Feng T, Morihara R, Fukui Y, Ohta Y, Hishikawa N, Abe K. Chronic cerebral Hypoperfusion accelerates Alzheimer’s disease pathology with cerebrovascular remodeling in a novel mouse model. J Alzheimers Dis. 2016;53(3):893–905. https://doi.org/10.3233/jad-160345. CrossRefPubMed Zhai Y, Yamashita T, Nakano Y, Sun Z, Shang J, Feng T, Morihara R, Fukui Y, Ohta Y, Hishikawa N, Abe K. Chronic cerebral Hypoperfusion accelerates Alzheimer’s disease pathology with cerebrovascular remodeling in a novel mouse model. J Alzheimers Dis. 2016;53(3):893–905. https://​doi.​org/​10.​3233/​jad-160345.​ CrossRefPubMed
36.
38.
Zurück zum Zitat Lucas C, Parent M, Delandsheer E, Delacourte A, Fournier Y, Defossez A, Leys D. Multiple cerebral hemorrhage and amyloid angiopathy of the white matter in a case of Alzheimer’s disease. Rev Neurol. 1992;148(3):218–20.PubMed Lucas C, Parent M, Delandsheer E, Delacourte A, Fournier Y, Defossez A, Leys D. Multiple cerebral hemorrhage and amyloid angiopathy of the white matter in a case of Alzheimer’s disease. Rev Neurol. 1992;148(3):218–20.PubMed
39.
Zurück zum Zitat Mehdorn HM, Gerhard L, Muller SP, Olbrich HM. Clinical and cerebral blood flow studies in patients with intracranial hemorrhage and amyloid angiopathy typical of Alzheimer’s disease. Neurosurg Rev. 1992;15(2):111–6.CrossRefPubMed Mehdorn HM, Gerhard L, Muller SP, Olbrich HM. Clinical and cerebral blood flow studies in patients with intracranial hemorrhage and amyloid angiopathy typical of Alzheimer’s disease. Neurosurg Rev. 1992;15(2):111–6.CrossRefPubMed
40.
42.
Zurück zum Zitat Olichney JM, Hansen LA, Hofstetter CR, Grundman M, Katzman R, Thal LJ. Cerebral infarction in Alzheimer’s disease is associated with severe amyloid angiopathy and hypertension. Arch Neurol. 1995;52(7):702–8.CrossRefPubMed Olichney JM, Hansen LA, Hofstetter CR, Grundman M, Katzman R, Thal LJ. Cerebral infarction in Alzheimer’s disease is associated with severe amyloid angiopathy and hypertension. Arch Neurol. 1995;52(7):702–8.CrossRefPubMed
43.
46.
Zurück zum Zitat McKeith IG. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. J Alzheimers Dis. 2006;9(3 Suppl):417–23.CrossRefPubMed McKeith IG. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. J Alzheimers Dis. 2006;9(3 Suppl):417–23.CrossRefPubMed
47.
Zurück zum Zitat Uchikado H, Lin WL, DeLucia MW, Dickson DW. Alzheimer disease with amygdala Lewy bodies: a distinct form of alpha-synucleinopathy. J Neuropathol Exp Neurol. 2006;65(7):685–97.CrossRefPubMedPubMedCentral Uchikado H, Lin WL, DeLucia MW, Dickson DW. Alzheimer disease with amygdala Lewy bodies: a distinct form of alpha-synucleinopathy. J Neuropathol Exp Neurol. 2006;65(7):685–97.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Schneider JA, Arvanitakis Z, Leurgans SE, Bennett DA. The neuropathology of probable Alzheimer disease and mild cognitive impairment. Ann Neurol. 2009;66(2):200–8.CrossRefPubMedPubMedCentral Schneider JA, Arvanitakis Z, Leurgans SE, Bennett DA. The neuropathology of probable Alzheimer disease and mild cognitive impairment. Ann Neurol. 2009;66(2):200–8.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Lippa CF, Fujiwara H, Mann DM, Giasson B, Baba M, Schmidt ML, et al. Lewy bodies contain altered alpha-synuclein in brains of many familial Alzheimer’s disease patients with mutations in presenilin and amyloid precursor protein genes. Am J Pathol. 1998;153(5):1365–70.CrossRefPubMedPubMedCentral Lippa CF, Fujiwara H, Mann DM, Giasson B, Baba M, Schmidt ML, et al. Lewy bodies contain altered alpha-synuclein in brains of many familial Alzheimer’s disease patients with mutations in presenilin and amyloid precursor protein genes. Am J Pathol. 1998;153(5):1365–70.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Hamilton RL. Lewy bodies in Alzheimer’s disease: a neuropathological review of 145 cases using alpha-synuclein immunohistochemistry. Brain Pathol. 2000;10(3):378–84.CrossRefPubMed Hamilton RL. Lewy bodies in Alzheimer’s disease: a neuropathological review of 145 cases using alpha-synuclein immunohistochemistry. Brain Pathol. 2000;10(3):378–84.CrossRefPubMed
51.
Zurück zum Zitat Chung EJ, Babulal GM, Monsell SE, Cairns NJ, Roe CM, Morris JC. Clinical features of Alzheimer disease with and without Lewy bodies. JAMA Neurol. 2015;72(7):789–96.CrossRefPubMedPubMedCentral Chung EJ, Babulal GM, Monsell SE, Cairns NJ, Roe CM, Morris JC. Clinical features of Alzheimer disease with and without Lewy bodies. JAMA Neurol. 2015;72(7):789–96.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Brenowitz WD, Keene CD, Hawes SE, Hubbard RA, Longstreth WT Jr, Woltjer RL, et al. Alzheimer’s disease neuropathologic change, Lewy body disease, and vascular brain injury in clinic- and community-based samples. Neurobiol Aging. 2017;53:83–92.CrossRefPubMedPubMedCentral Brenowitz WD, Keene CD, Hawes SE, Hubbard RA, Longstreth WT Jr, Woltjer RL, et al. Alzheimer’s disease neuropathologic change, Lewy body disease, and vascular brain injury in clinic- and community-based samples. Neurobiol Aging. 2017;53:83–92.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat McKeith IG, Galasko D, Kosaka K, Perry EK, Dickson DW, Hansen LA, et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology. 1996;47(5):1113–24.CrossRefPubMed McKeith IG, Galasko D, Kosaka K, Perry EK, Dickson DW, Hansen LA, et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology. 1996;47(5):1113–24.CrossRefPubMed
54.
Zurück zum Zitat Kotzbauer PT, Trojanowsk JQ, Lee VM. Lewy body pathology in Alzheimer’s disease. J Mol Neurosci. 2001;17(2):225–32.CrossRefPubMed Kotzbauer PT, Trojanowsk JQ, Lee VM. Lewy body pathology in Alzheimer’s disease. J Mol Neurosci. 2001;17(2):225–32.CrossRefPubMed
55.
Zurück zum Zitat Vereecken TH, Vogels OJ, Nieuwenhuys R. Neuron loss and shrinkage in the amygdala in Alzheimer’s disease. Neurobiol Aging. 1994;15(1):45–54.CrossRefPubMed Vereecken TH, Vogels OJ, Nieuwenhuys R. Neuron loss and shrinkage in the amygdala in Alzheimer’s disease. Neurobiol Aging. 1994;15(1):45–54.CrossRefPubMed
56.
Zurück zum Zitat Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology. 1992;42(3 Pt 1):631–9.CrossRefPubMed Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology. 1992;42(3 Pt 1):631–9.CrossRefPubMed
57.
Zurück zum Zitat Zald DH. The human amygdala and the emotional evaluation of sensory stimuli. Brain Res Brain Res Rev. 2003;41(1):88–123.CrossRefPubMed Zald DH. The human amygdala and the emotional evaluation of sensory stimuli. Brain Res Brain Res Rev. 2003;41(1):88–123.CrossRefPubMed
58.
Zurück zum Zitat Clinton LK, Blurton-Jones M, Myczek K, Trojanowski JQ, LaFerla FM. Synergistic interactions between Abeta, tau, and alpha-synuclein: acceleration of neuropathology and cognitive decline. J Neurosci. 2010;30(21):7281–9.CrossRefPubMedPubMedCentral Clinton LK, Blurton-Jones M, Myczek K, Trojanowski JQ, LaFerla FM. Synergistic interactions between Abeta, tau, and alpha-synuclein: acceleration of neuropathology and cognitive decline. J Neurosci. 2010;30(21):7281–9.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Kotzbauer PT, Cairns NJ, Campbell MC, Willis AW, Racette BA, Tabbal SD, et al. Pathologic accumulation of alpha-synuclein and Abeta in Parkinson disease patients with dementia. Arch Neurol. 2012;69(10):1326–31.CrossRefPubMedPubMedCentral Kotzbauer PT, Cairns NJ, Campbell MC, Willis AW, Racette BA, Tabbal SD, et al. Pathologic accumulation of alpha-synuclein and Abeta in Parkinson disease patients with dementia. Arch Neurol. 2012;69(10):1326–31.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Swirski M, Miners JS, de Silva R, Lashley T, Ling H, Holton J, et al. Evaluating the relationship between amyloid-beta and alpha-synuclein phosphorylated at Ser129 in dementia with Lewy bodies and Parkinson’s disease. Alzheimers Res Ther. 2014;6(5–8):77.CrossRefPubMedPubMedCentral Swirski M, Miners JS, de Silva R, Lashley T, Ling H, Holton J, et al. Evaluating the relationship between amyloid-beta and alpha-synuclein phosphorylated at Ser129 in dementia with Lewy bodies and Parkinson’s disease. Alzheimers Res Ther. 2014;6(5–8):77.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Jellinger KA, Attems J. Prevalence and impact of vascular and Alzheimer pathologies in Lewy body disease. Acta Neuropathol. 2008;115(4):427–36.CrossRefPubMed Jellinger KA, Attems J. Prevalence and impact of vascular and Alzheimer pathologies in Lewy body disease. Acta Neuropathol. 2008;115(4):427–36.CrossRefPubMed
62.
Zurück zum Zitat Sonnen JA, Postupna N, Larson EB, Crane PK, Rose SE, Montine KS, et al. Pathologic correlates of dementia in individuals with Lewy body disease. Brain Pathol. 2010;20(3):654–9.CrossRefPubMedPubMedCentral Sonnen JA, Postupna N, Larson EB, Crane PK, Rose SE, Montine KS, et al. Pathologic correlates of dementia in individuals with Lewy body disease. Brain Pathol. 2010;20(3):654–9.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Olichney JM, Galasko D, Salmon DP, Hofstetter CR, Hansen LA, Katzman R, et al. Cognitive decline is faster in Lewy body variant than in Alzheimer’s disease. Neurology. 1998;51(2):351–7.CrossRefPubMed Olichney JM, Galasko D, Salmon DP, Hofstetter CR, Hansen LA, Katzman R, et al. Cognitive decline is faster in Lewy body variant than in Alzheimer’s disease. Neurology. 1998;51(2):351–7.CrossRefPubMed
64.
Zurück zum Zitat Lopez OL, Wisniewski S, Hamilton RL, Becker JT, Kaufer DI, DeKosky ST. Predictors of progression in patients with AD and Lewy bodies. Neurology. 2000;54(9):1774–9.CrossRefPubMed Lopez OL, Wisniewski S, Hamilton RL, Becker JT, Kaufer DI, DeKosky ST. Predictors of progression in patients with AD and Lewy bodies. Neurology. 2000;54(9):1774–9.CrossRefPubMed
65.
Zurück zum Zitat Tsuang D, Leverenz JB, Lopez OL, Hamilton RL, Bennett DA, Schneider JA, et al. APOE epsilon4 increases risk for dementia in pure synucleinopathies. JAMA Neurol. 2013;70(2):223–8.CrossRefPubMedPubMedCentral Tsuang D, Leverenz JB, Lopez OL, Hamilton RL, Bennett DA, Schneider JA, et al. APOE epsilon4 increases risk for dementia in pure synucleinopathies. JAMA Neurol. 2013;70(2):223–8.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Samuel W, Alford M, Hofstetter CR, Hansen L. Dementia with Lewy bodies versus pure Alzheimer disease: differences in cognition, neuropathology, cholinergic dysfunction, and synapse density. J Neuropathol Exp Neurol. 1997;56(5):499–508.CrossRefPubMed Samuel W, Alford M, Hofstetter CR, Hansen L. Dementia with Lewy bodies versus pure Alzheimer disease: differences in cognition, neuropathology, cholinergic dysfunction, and synapse density. J Neuropathol Exp Neurol. 1997;56(5):499–508.CrossRefPubMed
67.
Zurück zum Zitat Galasko D, Katzman R, Salmon DP, Hansen L. Clinical and neuropathological findings in Lewy body dementias. Brain Cogn. 1996;31(2):166–75.CrossRefPubMed Galasko D, Katzman R, Salmon DP, Hansen L. Clinical and neuropathological findings in Lewy body dementias. Brain Cogn. 1996;31(2):166–75.CrossRefPubMed
68.
Zurück zum Zitat Heyman A, Fillenbaum GG, Gearing M, Mirra SS, Welsh-Bohmer KA, Peterson B, et al. Comparison of Lewy body variant of Alzheimer’s disease with pure Alzheimer’s disease: consortium to establish a registry for Alzheimer’s disease, part XIX. Neurology. 1999;52(9):1839–44.CrossRefPubMed Heyman A, Fillenbaum GG, Gearing M, Mirra SS, Welsh-Bohmer KA, Peterson B, et al. Comparison of Lewy body variant of Alzheimer’s disease with pure Alzheimer’s disease: consortium to establish a registry for Alzheimer’s disease, part XIX. Neurology. 1999;52(9):1839–44.CrossRefPubMed
69.
Zurück zum Zitat Weiner MF, Risser RC, Cullum CM, Honig L, White C 3rd, Speciale S, et al. Alzheimer’s disease and its Lewy body variant: a clinical analysis of postmortem verified cases. Am J Psychiatry. 1996;153(10):1269–73.CrossRefPubMed Weiner MF, Risser RC, Cullum CM, Honig L, White C 3rd, Speciale S, et al. Alzheimer’s disease and its Lewy body variant: a clinical analysis of postmortem verified cases. Am J Psychiatry. 1996;153(10):1269–73.CrossRefPubMed
70.
Zurück zum Zitat Stern Y, Jacobs D, Goldman J, Gomez-Tortosa E, Hyman BT, Liu Y, et al. An investigation of clinical correlates of Lewy bodies in autopsy-proven Alzheimer disease. Arch Neurol. 2001;58(3):460–5.PubMed Stern Y, Jacobs D, Goldman J, Gomez-Tortosa E, Hyman BT, Liu Y, et al. An investigation of clinical correlates of Lewy bodies in autopsy-proven Alzheimer disease. Arch Neurol. 2001;58(3):460–5.PubMed
71.
Zurück zum Zitat Borghi R, Marchese R, Negro A, Marinelli L, Forloni G, Zaccheo D, et al. Full length alpha-synuclein is present in cerebrospinal fluid from Parkinson’s disease and normal subjects. Neurosci Lett. 2000;287(1):65–7.CrossRefPubMed Borghi R, Marchese R, Negro A, Marinelli L, Forloni G, Zaccheo D, et al. Full length alpha-synuclein is present in cerebrospinal fluid from Parkinson’s disease and normal subjects. Neurosci Lett. 2000;287(1):65–7.CrossRefPubMed
72.
Zurück zum Zitat Hall S, Ohrfelt A, Constantinescu R, Andreasson U, Surova Y, Bostrom F, et al. Accuracy of a panel of 5 cerebrospinal fluid biomarkers in the differential diagnosis of patients with dementia and/or parkinsonian disorders. Arch Neurol. 2012;69(11):1445–52.CrossRefPubMed Hall S, Ohrfelt A, Constantinescu R, Andreasson U, Surova Y, Bostrom F, et al. Accuracy of a panel of 5 cerebrospinal fluid biomarkers in the differential diagnosis of patients with dementia and/or parkinsonian disorders. Arch Neurol. 2012;69(11):1445–52.CrossRefPubMed
73.
Zurück zum Zitat Toledo JB, Korff A, Shaw LM, Trojanowski JQ, Zhang J. CSF alpha-synuclein improves diagnostic and prognostic performance of CSF tau and Abeta in Alzheimer’s disease. Acta Neuropathol. 2013;126(5):683–97.CrossRefPubMed Toledo JB, Korff A, Shaw LM, Trojanowski JQ, Zhang J. CSF alpha-synuclein improves diagnostic and prognostic performance of CSF tau and Abeta in Alzheimer’s disease. Acta Neuropathol. 2013;126(5):683–97.CrossRefPubMed
74.
Zurück zum Zitat Slaets S, Vanmechelen E, Le Bastard N, Decraemer H, Vandijck M, Martin JJ, et al. Increased CSF alpha-synuclein levels in Alzheimer’s disease: correlation with tau levels. Alzheimers Dement. 2014;10(5 Suppl):S290–8.CrossRefPubMed Slaets S, Vanmechelen E, Le Bastard N, Decraemer H, Vandijck M, Martin JJ, et al. Increased CSF alpha-synuclein levels in Alzheimer’s disease: correlation with tau levels. Alzheimers Dement. 2014;10(5 Suppl):S290–8.CrossRefPubMed
75.
Zurück zum Zitat Ohrfelt A, Grognet P, Andreasen N, Wallin A, Vanmechelen E, Blennow K, et al. Cerebrospinal fluid alpha-synuclein in neurodegenerative disorders-a marker of synapse loss? Neurosci Lett. 2009;450(3):332–5.CrossRefPubMed Ohrfelt A, Grognet P, Andreasen N, Wallin A, Vanmechelen E, Blennow K, et al. Cerebrospinal fluid alpha-synuclein in neurodegenerative disorders-a marker of synapse loss? Neurosci Lett. 2009;450(3):332–5.CrossRefPubMed
76.
Zurück zum Zitat Kapaki E, Paraskevas GP, Emmanouilidou E, Vekrellis K. The diagnostic value of CSF alpha-synuclein in the differential diagnosis of dementia with Lewy bodies vs. normal subjects and patients with Alzheimer’s disease. PLoS One. 2013;8(11):e81654.CrossRefPubMedPubMedCentral Kapaki E, Paraskevas GP, Emmanouilidou E, Vekrellis K. The diagnostic value of CSF alpha-synuclein in the differential diagnosis of dementia with Lewy bodies vs. normal subjects and patients with Alzheimer’s disease. PLoS One. 2013;8(11):e81654.CrossRefPubMedPubMedCentral
77.
Zurück zum Zitat Wennstrom M, Surova Y, Hall S, Nilsson C, Minthon L, Bostrom F, et al. Low CSF levels of both alpha-synuclein and the alpha-synuclein cleaving enzyme neurosin in patients with synucleinopathy. PLoS One. 2013;8(1):e53250.CrossRefPubMedPubMedCentral Wennstrom M, Surova Y, Hall S, Nilsson C, Minthon L, Bostrom F, et al. Low CSF levels of both alpha-synuclein and the alpha-synuclein cleaving enzyme neurosin in patients with synucleinopathy. PLoS One. 2013;8(1):e53250.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Reesink FE, Lemstra AW, van Dijk KD, Berendse HW, van de Berg WD, Klein M, et al. CSF alpha-synuclein does not discriminate dementia with Lewy bodies from Alzheimer’s disease. J Alzheimers Dis. 2010;22(1):87–95.CrossRefPubMed Reesink FE, Lemstra AW, van Dijk KD, Berendse HW, van de Berg WD, Klein M, et al. CSF alpha-synuclein does not discriminate dementia with Lewy bodies from Alzheimer’s disease. J Alzheimers Dis. 2010;22(1):87–95.CrossRefPubMed
79.
Zurück zum Zitat Heneka MT, Kummer MP, Latz E. Innate immune activation in neurodegenerative disease. Nat Rev Immunol. 2014;14:463–77.CrossRefPubMed Heneka MT, Kummer MP, Latz E. Innate immune activation in neurodegenerative disease. Nat Rev Immunol. 2014;14:463–77.CrossRefPubMed
80.
Zurück zum Zitat Heneka MT, Carson MJ, El KJ, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.CrossRefPubMedPubMedCentral Heneka MT, Carson MJ, El KJ, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Eikelenboom P, Van Exel E, Hoozemans JJM, Veerhuis R, Rozemuller AJM, Van Gool WA. Neuroinflammation - an early event in both the history and pathogenesis of Alzheimer’s disease. Neurodegener Dis. 2010;7:38–41.CrossRefPubMed Eikelenboom P, Van Exel E, Hoozemans JJM, Veerhuis R, Rozemuller AJM, Van Gool WA. Neuroinflammation - an early event in both the history and pathogenesis of Alzheimer’s disease. Neurodegener Dis. 2010;7:38–41.CrossRefPubMed
82.
Zurück zum Zitat Griciuc A, Serrano-Pozo A, Parrado AR, Lesinski AN, Asselin CN, Mullin K, et al. Alzheimer’s disease risk gene cd33 inhibits microglial uptake of amyloid beta. Neuron. 2013;78:631–43.CrossRefPubMedPubMedCentral Griciuc A, Serrano-Pozo A, Parrado AR, Lesinski AN, Asselin CN, Mullin K, et al. Alzheimer’s disease risk gene cd33 inhibits microglial uptake of amyloid beta. Neuron. 2013;78:631–43.CrossRefPubMedPubMedCentral
83.
Zurück zum Zitat Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, et al. TREM2 variants in Alzheimer’s disease. N Engl J Med. 2013;368:117–27.CrossRefPubMed Guerreiro R, Wojtas A, Bras J, Carrasquillo M, Rogaeva E, Majounie E, et al. TREM2 variants in Alzheimer’s disease. N Engl J Med. 2013;368:117–27.CrossRefPubMed
84.
Zurück zum Zitat in’ t Veld BA, Ruitenberg A, Hofman A, Launer LJ, van Duijn CM, Stijnen T, et al. Nonsteroidal antiinflammatory drugs and the risk of Alzheimer’s disease. N Engl J Med 2001; 345: 1515–1521. in’ t Veld BA, Ruitenberg A, Hofman A, Launer LJ, van Duijn CM, Stijnen T, et al. Nonsteroidal antiinflammatory drugs and the risk of Alzheimer’s disease. N Engl J Med 2001; 345: 1515–1521.
85.
Zurück zum Zitat Szekely CA, Town T, Zandi PP. NSAIDs for the chemoprevention of Alzheimer’s disease. Inflamm Pathog Chronic Dis. 2007:229–48. Szekely CA, Town T, Zandi PP. NSAIDs for the chemoprevention of Alzheimer’s disease. Inflamm Pathog Chronic Dis. 2007:229–48.
86.
Zurück zum Zitat Alzheimer’s Disease Anti-inflammatory Prevention Trial Research Group. Results of a follow-up study to the randomized Alzheimer’s disease anti-inflammatory prevention trial (ADAPT). Alzheimers Dement. 2013;9:714–23. Alzheimer’s Disease Anti-inflammatory Prevention Trial Research Group. Results of a follow-up study to the randomized Alzheimer’s disease anti-inflammatory prevention trial (ADAPT). Alzheimers Dement. 2013;9:714–23.
87.
Zurück zum Zitat Tremblay M-E, Stevens B, Sierra A, Wake H, Bessis A, Nimmerjahn A. The role of microglia in the healthy brain. J Neurosci. 2011;31:16064–9.CrossRefPubMed Tremblay M-E, Stevens B, Sierra A, Wake H, Bessis A, Nimmerjahn A. The role of microglia in the healthy brain. J Neurosci. 2011;31:16064–9.CrossRefPubMed
88.
Zurück zum Zitat Lee CYD, Landreth GE. The role of microglia in amyloid clearance from the AD brain. J Neural Transm. 2010;117:949–60.CrossRefPubMed Lee CYD, Landreth GE. The role of microglia in amyloid clearance from the AD brain. J Neural Transm. 2010;117:949–60.CrossRefPubMed
89.
Zurück zum Zitat Hopperton KE, Mohammad D, Trépanier MO, Giuliano V, Bazinet RP. Markers of microglia in post-mortem brain samples from patients with Alzheimer’s disease: a systematic review. Molecular Psychiatry. 2018;23:177–198. Hopperton KE, Mohammad D, Trépanier MO, Giuliano V, Bazinet RP. Markers of microglia in post-mortem brain samples from patients with Alzheimer’s disease: a systematic review. Molecular Psychiatry. 2018;23:177–198.
90.
Zurück zum Zitat Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, et al. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science. 2010;330:1774.CrossRefPubMedPubMedCentral Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, et al. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science. 2010;330:1774.CrossRefPubMedPubMedCentral
91.
Zurück zum Zitat Li Y, Liu L, Barger SW, Griffin WS. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci 2003; 23: 1605–1611.Available from: http://www.ncbi.nlm.nih.gov/pubmed/12629164%5Cn. http://www.jneurosci.org/content/23/5/1605.full.pdf. Li Y, Liu L, Barger SW, Griffin WS. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci 2003; 23: 1605–1611.Available from: http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​12629164%5Cn. http://www.jneurosci.org/content/23/5/1605.full.pdf.
92.
Zurück zum Zitat Zilka N, Kazmerova Z, Jadhav S, Neradil P, Madari A, Obetkova D, et al. Who fans the flames of Alzheimer’s disease brains? Misfolded tau on the crossroad of neurodegenerative and inflammatory pathways. J Neuroinflammation. 2012;9:47. Zilka N, Kazmerova Z, Jadhav S, Neradil P, Madari A, Obetkova D, et al. Who fans the flames of Alzheimer’s disease brains? Misfolded tau on the crossroad of neurodegenerative and inflammatory pathways. J Neuroinflammation. 2012;9:47.
93.
Zurück zum Zitat Maphis N, Xu G, Kokiko-Cochran ON, Jiang S, Cardona A, Ransohoff RM, et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138:1738–55.CrossRefPubMedPubMedCentral Maphis N, Xu G, Kokiko-Cochran ON, Jiang S, Cardona A, Ransohoff RM, et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138:1738–55.CrossRefPubMedPubMedCentral
94.
Zurück zum Zitat Bolós M, Llorens-Martín M, Jurado-Arjona J, Hernández F, Rábano A, Avila J. Direct evidence of internalization of tau by microglia in vitro and in vivo. J Alzheimers Dis. 2015;50:77–87.CrossRef Bolós M, Llorens-Martín M, Jurado-Arjona J, Hernández F, Rábano A, Avila J. Direct evidence of internalization of tau by microglia in vitro and in vivo. J Alzheimers Dis. 2015;50:77–87.CrossRef
Metadaten
Titel
Impact of the biological definition of Alzheimer’s disease using amyloid, tau and neurodegeneration (ATN): what about the role of vascular changes, inflammation, Lewy body pathology?
verfasst von
S. Gauthier
H. Zhang
K. P. Ng
T.A. Pascoal
P. Rosa-Neto
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Translational Neurodegeneration / Ausgabe 1/2018
Elektronische ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-018-0117-9

Weitere Artikel der Ausgabe 1/2018

Translational Neurodegeneration 1/2018 Zur Ausgabe