Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2014

Open Access 01.12.2014 | Research

RIG-1 receptor expression in the pathology of Alzheimer’s disease

verfasst von: Juan Pablo de Rivero Vaccari, Frank J Brand III, Christina Sedaghat, Deborah C Mash, W Dalton Dietrich, Robert W Keane

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2014

Abstract

Background

Neuroinflammation plays a critical role in the pathogenesis of Alzheimer’s disease (AD) and involves activation of the innate immune response via recognition of diverse stimuli by pattern recognition receptors (PRRs). The inflammatory inducers and precise innate signaling pathway contributing to AD pathology remain largely undefined.

Results

In the present study we analyzed expression levels of innate immune proteins in temporal and occipital cortices from preclinical (no cognitive impairment, NCI, N = 22) to mild cognitive impairment (MCI, N = 20) associated with AD pathology (N = 20) and AD patients (N = 23). We found that retinoic acid-inducible gene-I (RIG-1) is significantly elevated in the temporal cortex and plasma in patients with MCI. In addition, primary human astrocytes stimulated with the RIG-1 ligand 5′ppp RNA showed increased expression of amyloid precursor protein (APP) and amyloid-β (Aβ), supporting the idea that RIG-1 is involved in the pathology of MCI associated with early progression to AD.

Conclusion

These findings suggest that RIG-1 may play a critical role in incipient AD.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1742-2094-11-67) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

JPdRV designed the study, performed experiments, analyzed the data, interpreted the results and prepared the manuscript. FJB and CS performed experiments and analyzed the data. DCM, WDD and RWK reviewed and discussed the manuscript. All authors have read and approved the final version of the manuscript.
Abkürzungen
AD
Alzheimer’s disease
PRRs
pattern recognition receptors
NCI
no cognitive impairment
MCI
mild cognitive impairment
RIG-1
retinoic acid-inducible gene-I
APP
amyloid precursor protein
amyloid-β
CNS
central nervous system
RLR
RIG-I-like receptors
NLR
NOD-like receptors
CLR
C-type lectin receptors
MDA5
melanoma differentiation-associated gene 5
LGP2
laboratory of genetics and physiology 2
CDR
clinical dementia rating
CERAD
Consortium to Establish a Registry for AD
Apoe
apolipoprotein e
5′ppp dsRNA
5′ triphosphate double-stranded RNA.

Background

Alzheimer’s disease (AD) pathogenesis is associated with central nervous system (CNS) inflammatory responses [14]. Amyloid-β (Aβ) fibrils trigger inflammatory responses mediated by Toll-like receptors (TLR)4/TLR6 in the presence of CD36 [14]. Moreover, a polymorphism in the TLR4 extracellular domain has been reported to be associated with protection against late-onset AD in an Italian population [5], suggesting that a sterile inflammatory response could influence AD pathology through TLR4 signaling. In addition, TLR2 has been shown to act as a receptor for Aβ, and to trigger an inflammatory response [6]. Activation of innate immunity in the CNS appears to be a universal component of neuroinflammation. AD may be distinguished by a disease-specific mechanism for induction of inflammatory responses. In addition, distinct pathways for production of inflammation inducers in vulnerable brain regions where these processes occur are potential biomarkers of AD pathophysiology.
Infection of cells by viruses and microorganisms activates innate immune inflammatory responses. The initial sensing of infection is mediated by pattern recognition receptors, which include TLRs, RIG-I-like receptors (RLR), NOD-like receptors (NLR), and C-type lectin receptors (CLR). The RLR family is a RNA sensing system that is comprised of retinoic acid inducible gene-like-I (RIG-1), melanoma differentiation-associated gene 5 (MDA5), and laboratory of genetics and physiology 2 (LGP2). RIG-1 recognizes relatively short dsRNA (up to 1 kb) whereas MDA5 detects long dsRNA (more than 2 kb) to activate synthesis of type I IFNs, including IFN-α and IFN-β [7]. RLRs are localized in the cytoplasm and recognize the genomic RNA of dsRNA viruses and dsRNA generated as the replication intermediate of ssRNA viruses and also act as sensors of cellular damage [8]. RLRs activate downstream signaling proteins evoking type I IFN production. Type I IFNs play central roles in antiviral responses by inducing apoptotic cell death in virally infected cells, rendering cells resistant to virus infection, activating acquired immunity, and stimulating hematopoietic stem cell turnover and proliferation. In addition, type I IFNs have been implicated in the inflammatory response in AD [9].
We have shown recently that RLR signaling proteins are present in CNS neurons and glial cells, and RLR signaling stimulation resulted in astrocyte activation [10]. In addition, activation of the inflammasome, an NLR innate immune complex, contributes to age-related cognitive decline in elderly animals [11]. However, limited information is available about the role of RLRs in AD pathology or early disease progression. Since MCI is considered a transitional phase between normal aging (or cognition) and AD [1214], it is important to identify the molecular events that characterize MCI associated with AD pathology.

Methods

Patient consents and subjects demographics

The study was approved by the University of Miami Miller School of Medicine institutional review board. Written informed consent for research and brain autopsy was obtained for all subjects in this study.
Neuropathologic specimens (3 millimeters) of fresh-frozen human temporal (BA38) and occipital cortex (BA17) were obtained from the University of Miami Brain Endowment Bank™. The temporopolar cortex (BA38) was sampled from frozen tissue blocks at the level of the fundus of the temporopolar sulcus. The occipital cortex was sampled from the primary visual cortex (BA17). Postmortem specimens were selected from age-matched subjects with no cognitive impairment (NCI), MCI, and from AD patients. The diagnosis of AD was made using standard diagnostic criteria [15]. Subjects with NCI, MCI, and AD were selected based on their antemortem clinical dementia rating (CDR) score one year prior to death and postmortem pathologic evaluation for AD pathology and Braak stage. Neuropathologic diagnosis was based on NIA-Regan criteria recommendations of the Consortium to Establish a Registry for AD (CERAD) [16] and Braak staging of neurofibrillary tangles [17]. The diagnosis of MCI included assessment of normal activities of daily living, normal general cognitive function, abnormal memory for age, and no dementia [17]. MCI patients met neuropathologic criteria for possible to probable AD and Braak stages I to IV [17]. AD cases selected for this study included patients with a diagnosis of clinical dementia and definite AD on postmortem examination (Braak stages V or VI; Table 1).
Table 1
Characteristics of subjects used in the study (brain cortex)
Characteristic
NCI
MCI
AD
Number of subjects
22
20
23
Male (%)
15 (67)
4 (19)
9 (36)
Female (%)
7 (33)
16 (81)
14 (64)
Age at death
   
Median (IQR)
68 (61 to 79)
86 (70 to 91)
80 (70 to 85)
Range
59 to 95
61 to 105
60 to 88
Race
20C 1H
20C 1H
24C 1H
Brain weight
   
Median (IQR)
1,352 (1,298 to 1,505)
1,210 (1,043 to 1,398)
1,115 (950 to 1,215)
Range
1,054 to 1,570
880 to 1,840
825 to 1,250
CDR score
   
0 (%)
22 (100)
12 (60)
0 (0)
1 (%)
0 (0)
8 (40)
0 (0)
2 (%)
0 (0)
0 (0)
2 (9)
3 (%)
0 (0)
0 (0)
21 (91)
Braak score
   
0 (%)
22 (100)
0 (0)
0 (0)
I (%)
0 (0)
6 (30)
0 (0)
II (%)
0 (0)
5 (25)
0 (0)
III (%)
0 (0)
9 (45)
0 (0)
IV (%)
0 (0)
0 (0)
1 (4)
V (%)
0 (0)
0 (0)
13 (56)
VI (%)
0 (0)
0 (0)
9 (40)
AD CERAD
   
Not present (%)
22 (100)
1 (5)
0 (0)
Possible (%)
0 (0)
4 (20)
0 (0)
Probable (%)
0 (0)
5 (25)
0 (0)
Definite (%)
0 (0)
10 (50)
23 (100)
AD: Alzheimer’s disease, CDR: clinical dementia rating, CERAD: Consortium to Establish a Registry for Alzheimer’s Disease, IQR: interquartile range, MCI: mild cognitive impairment, NCI: no cognitive impairment. Race: C = Caucasian and H = Hispanic.

Plasma and serum samples

All plasma and serum samples were obtained from the University of Kentucky Alzheimer’s Disease Center Brain Bank. The samples were obtained from patients diagnosed postmortem as either age-matched controls with no cognitive impairment (NCI; Braak stage (0 to I), MCI (Braak stages II to IV), or AD (Braak stages V to VI). The section of the study included six age-matched controls (NCI; Braak stages 0 to I), seven MCI patients with possible AD, determined by pathological evidence of neurofibrillary tangles and senile plaques (Braak stages II to IV), and ten patients who met clinical diagnostic criteria for definite AD (Braak stages V to VI; Table 2).
Table 2
Characteristics of subjects used in the study (plasma and serum)
Group
Braak stage
AD (CERAD)
Age at death
Gender
Apoe
PMI (hours)
NCI
0
B = CERAD Probable
92
M
3/5
3.33
NCI
0
No
85
F
3/3
2.50
NCI
1
No
90
F
2/3
4.00
NCI
1
No
100
F
2/3
2.25
NCI
1
No
84
F
3/4
3.00
NCI
1
No
79
F
3/4
1.75
MCI
2
B = CERAD Probable
91
F
3/4
1.75
MCI
2
B = CERAD Probable
93
F
3/4
2.75
MCI
2
B = CERAD Probable
80
F
3/4
2.00
MCI
2
A = CERAD Probable
81
M
3/5
2.83
MCI
2
C = Definite AD
79
M
3/3
1.75
MCI
4
B = CERAD Probable
77
M
3/4
2.75
MCI
3
B = CERAD Probable
92
F
2/3
3.25
AD
6
C = Definite AD
78
M
3/4
3.50
AD
6
C = Definite AD
84
M
3/4
2.75
AD
6
C = Definite AD
83
F
3/3
3.50
AD
6
B = CERAD Probable
85
M
3/3
2.75
AD
6
B = CERAD Probable
80
M
3/3
2.75
AD
6
C = Definite AD
87
M
3/4
3.25
AD
6
C = Definite AD
73
M
3/3
2.00
AD
6
C = Definite AD
80
F
3/3
4.00
AD
6
C = Definite AD
83
F
3/4
2.25
AD
5
C = Definite AD
91
F
3/3
3.00
AD: Alzheimer’s disease, Apoe, apolipoprotein e; CERAD: Consortium to Establish a Registry for AD: MCI: mild cognitive impairment, NCI: no cognitive impairment, PMI: postmortem interval.

Plasma and serum immunoglobulin isolation

To prevent interference of immunoglobulin G (IgG) during immunoblot analysis of plasma and serum, IgG was isolated using a Pierce Albumin/IgG Removal kit (Thermo Scientific Waltham, MA, USA) according to manufacturer’s instructions.

Immunoblotting

Occipital and temporal cortices were homogenized in lysis buffer (20 mM Tris, pH 7.5, 150 mM NaCl, 1 mM EDTA, 1 mM EGTA, 1% Triton X-100, 2.5 mM pyrophosphate, 1 mM β-glycerophosphate) with protease inhibitor cocktail (Sigma). Twenty five micrograms of protein per sample were resolved in 10 to 20% Tris-HCl Criterion precasted gels (Bio-Rad, Hercules, CA, USA), transferred to polyvinylidene difluoride membranes (Applied Biosystems Waltham, MA, USA) and placed in blocking buffer (PBS, 0.1% Tween-20, 0.4% I-Block (Applied Biosystems Waltham, MA, USA) and then incubated for one hour with an antibody against RIG-1 (Anaspec Fremont, CA, USA) at a dilution of 1:1,000. To authenticate the presumptive bands shown in Figures 1 and 2, a RIG-1 positive control sample (Novus Biologicals Littleton, CO, USA) was used. Immunoabsorption is more appropriate to demonstrate the authenticity of the bands. Membranes were incubated for one hour with primary antibodies followed by appropriate secondary horseradish peroxidase (HRP)-linked antibodies (Cell Signaling Danvers, MA, USA). Visualization of signal was enhanced by chemiluminescence using a Phototope-HRP detection kit (Cell Signaling Danvers, MA, USA). To control for protein loading, immunoblots were stripped with Restore, Western blot stripping buffer (Pierce Rockford, IL, USA) and blotted for β-actin using monoclonal anti-β-actin antibody (1:8,000, Sigma St. Louis, MO, USA). Quantification of band density was performed using the UN-SCAN-IT gel software, and data were normalized to β-actin. For immunoblotting of serum and plasma 5 μg of protein were loaded equally across all samples used to keep data normalized.

Astrocyte culture preparation and RIG-1 stimulation

Human astrocytes were grown in culture as described in de Rivero Vaccari et al. in 2012 [10]. Primary human astrocytes (Lonza Basel, Switzerland) were grown in culture in complete Astrocyte Growth Medium (Lonza Basel, Switzerland) for seven days. RIG-1 signaling was stimulated with 5′ triphosphate double-stranded RNA (5′ppp dsRNA, Invivogen San Diego, CA, USA) as a specific ligand to stimulate RIG-1 signaling at different concentrations (2 and 4 μg/ml) for 18 hours. After stimulation, cells were harvested and immunoblotted for RIG-1 (Anaspec Fremont, CA, USA), phosphorylated IRF3 (Novus Biologicals Littleton, CO, USA), amyloid precursor protein (Abcam Cambridge, MA, USA) and amyloid-β (Epitomics Burlingame, CA, USA) expression as described.

Stimulation of human astrocytes with 3-42 amyloid-β

Human astrocytes were grown in culture for seven days and stimulated with 3-42 amyloid-β (Anaspec Fremont, CA USA) at a concentration of 0.5, 1 and 3 μM for 18 hours. Then cells were harvested and immunoblotted for expression of caspase-1 (Imgenex San Diego, CA, USA) and RIG-1 (Anaspec Fremont, CA USA) as described.

Statistical analysis

The primary outcome measures were levels of immune proteins in two brain regions. The demographic, clinical and neuropathological characteristics were used to group assignment. Association between individual protein measures and age, gender or postmortem interval were explored in multivariate analyses to ensure that the results were unchanged. Statistical comparisons between groups were made using one-way ANOVA and one-tailed Student’s t-test. The level of statistical significance was set at * P < 0.05.

Results

RIG-1 is elevated in the temporal cortex of MCI patients

The demographic and neuropathology characteristics of the cohort used in this section of the study are summarized in Table 1. The study included 22 age-matched controls (NCI), 20 MCI patients with pathologic evidence of senile plaques and neurofibrillary tangles consistent with possible or probable AD (Braak stages I to IV), and 23 patients who met clinical diagnostic criteria for AD and definite pathologic evidence (Braak V to VI). Immunoblot analysis of temporal cortical samples revealed an increase in RIG-1 expression in the MCI group when compared to the NCI and AD groups (Figure 1B). In contrast, the levels of RIG-1 in the occipital cortex were higher in the AD group than in the NCI and MCI groups (Figure 1C). Thus, these results show for the first time that RIG-1 is increased in the temporopolar cortex of MCI patients.

RIG-1 is elevated in the plasma of MCI patients

To determine the levels of RIG-1 in the plasma and serum of patients with MCI associated with AD, immunoglobulin G was isolated from serum and plasma obtained from patients corresponding to the NCI, MCI and AD groups, as described above. Figure 2 shows that RIG-1 was significantly increased in the plasma (Figure 2B) from MCI patients compared to the NCI and AD groups, whereas the levels of RIG-1 in serum (Figure 2C) did not differ among the three groups. Thus, these results show for the first time that RIG-1 is increased in the plasma of MCI patients.

3-42 Aβ increases expression of RIG-1

3-42 Aβ species have been shown to be the most prevalent form of Aβ peptides present in early and later stages of human AD amyloid pathology [18]. Since we found that levels of RIG-1 expression are elevated in the temporal cortex from MCI patients when compared to end-stage AD pathology (AD, Figures 1 and 2), we stimulated human cortical astrocytes with 3-42 Aβ for 18 hours at different concentrations (C, 0.5, 1 and 3 μM) to determine if Aβ peptide levels regulate the protein expression levels of RIG-1. Interestingly, there was a concentration dependent effect of 3-42 Aβ on the expression of RIG-1. At 0.5 μM treatment, the RIG-1 levels did not change when compared to the control/untreated group, whereas at 1 μM, the levels of RIG-1 increased, and at 3 μM, the protein levels of RIG-1 returned to basal/control levels (Figure 3). Importantly, no morphological or toxic changes were noticed in the cultured astrocytes at the concentrations of 3-42 Aβ used for 18 hours (data not shown). Thus, it appears that Aβ may be involved in regulating the levels of the RIG-1 protein.

5′ppp dsRNA activates RIG-1 signaling in primary human cortical astrocytes

5′ppp dsRNA has been shown to be a specific ligand of RIG-1 signaling activation [19]. To determine whether 5′ppp dsRNA is responsible for the activation of RIG-1 in primary human cortical astrocytes, 5′ppp dsRNA was administered to primary astrocytes in culture for 18 hours at two different concentrations (2 and 4 μg/ml). As shown in Figure 4B and 4C, RIG-1 and phospho-interferon regulatory factor 3 (P-IRF3), respectively, were significantly elevated after the administration of 4 μg/ml of 5′ppp dsRNA, thus indicating RIG-1 signaling activation.

5′ppp dsRNA increases expression of APP and Aβ in primary human cortical astrocytes

To identify if RIG-1 signaling stimulation is involved in the pathogenesis of AD, astrocytes were stimulated with the RIG-1 signaling agonist 5′ppp dsRNA (4 μg/ml) for 18 hours. Samples were then resolved by immunoblotting using antibodies against two hallmark proteins of AD, APP (Figure 4E) and Aβ (Figure 4F). Stimulation of RIG-1 with 4 μg/ml 5′ppp dsRNA, which activates RIG-1 signaling in astrocytes, resulted in a significant elevation in the expression of APP and Aβ when compared to the control group, suggesting an involvement of RIG-1 signaling in the expression of two hallmark proteins in AD pathology.

Discussion

The results of the present study demonstrate that RIG-1 is significantly elevated in the plasma and temporal cortex of MCI patients with AD pathology whereas RIG-1 is elevated in the occipital cortex of AD patients. Stimulation of RIG-1 with 5′ppp dsRNA in human cortical astrocytes resulted in increased expression of APP and Aβ. Thus, these findings suggest a potential role of the RIG-1 signaling system in incipient AD.
AD is a progressive neurodegenerative disorder characterized by impaired judgment, confusion, changes in behavior, disorientation [20], impairment of daily living, and loss of the ability to function independently [21]. AD is expected to become more prevalent as life expectancy continues to rise. It has been estimated that by 2050, the number of AD cases could double or triple to between 11 to 16 million [22]. A major limitation in finding therapeutic solutions for AD has been the lack of reliable methods for early diagnosis of this devastating disease. AD is a neurodegenerative disorder characterized by a progressive cognitive impairment as a consequence of neuronal dysfunction and ultimately the death of neurons. MCI is considered a transitional phase between normal aging and AD [1214]. The amyloid hypothesis of AD proposes that neuronal damage results from the accumulation of insoluble, hydrophobic, fibrillar peptides such as amyloid-β1-42 [2326]. These peptides activate enzymes resulting in a cascade of second messengers including prostaglandins and platelet-activating factor. Apoptosis of neurons is thought to follow as a consequence of the uncontrolled release of second messengers. It is possible that RIG-1 signaling in the temporal cortex is involved in the early events leading to AD pathology such as the accumulation of APP. On the other hand, the presence of RIG-1 in the occipital cortex of AD patients may be associated with exacerbated production of cytokines in AD patients [27] as a result of disease progression in later stages of AD when the pathology spreads throughout the cortex from the limbic to koniocortical areas.
Neuroinflammation has been considered to play a critical role in the pathogenesis of AD [2833], but the role of the innate immune response has not been thoroughly examined [34, 35]. Human neurons, in the absence of glia, have the intrinsic machinery to trigger robust inflammatory, chemoattractive, and antiviral responses [36]. The innate immune system senses microbial and viral pathogen and danger signals released from damaged or stressed cells to trigger conserved intracellular signaling pathways that drive proinflammatory responses that are critical for productive innate and adaptive immunity. Excessive inflammatory responses become deleterious adding to tissue destruction. Here we have provided evidence demonstrating that the RIG-1 is elevated in the innate immune response in disease-affected brain areas of MCI patients.
RIG-1 signaling may be activated by small self-RNA cleavage products generated by RNase L that stimulate signaling of RIG-1 [37] or by reactive oxygen species (ROS) [38]. Since damaged CNS cells release small self-nucleic acids and ROS, these molecules may play an important role in the initiation of the innate immune response in MCI [39]. Alternatively, foreign nucleic acids, the signature of invading viruses and certain bacteria, are sensed intracellularly and then stimulate RIG-1 signaling [7]. Other, yet to be identified ligands may be involved in the activation of RIG-1 signaling in MCI. Moreover, our data suggest that RIG-1 signaling activation results in increased expression of APP and Aβ, and that in addition Aβ contributes to the expression of RIG-1. It is important to consider that this study used samples from individuals in the MCI group that had a slightly greater number of females and a wider age range; thus, when interpreting these results one must take into account the effects of gender and age [40].

Conclusions

In this study, we used immunoblot analysis to determine whether RIG-1 signaling stimulation results in increased expression of Aβ and APP. In order to determine whether human cortical astrocytes respond to RIG-1 stimulation, we treated primary cortical astrocytes in culture with the specific RIG-1 ligand 5′ppp dsRNA and assayed for the expression of the RIG-1 signaling proteins RIG-1 and P-IRF3.
as well as APP and Aβ. The levels of these proteins were increased upon stimulation with the RIG-1 ligand, consistent with the hypothesis that RIG-1 signaling is involved in the pathogenesis of AD. Astrocytes have been previously implicated in the pathogenesis of AD [4144]. In addition, we have previously shown that RIG-1 signaling is involved in the activation of astrocytes [10]. Thus, our findings further support an involvement of astrocytes in AD pathology.

Acknowledgments

These studies were supported by grants from the NIH to RWK (NS059836), to WDD (NS042133) and gifts to DCM (McGowan Endowment Fund). We thank David Sequeira for help with immunohistochemistry and Dennis Dixon, MD and Jocelyn Bruce, MD for expert neuropathological consultation. Postmortem neuropathological specimens were obtained from the University of Miami Brain Endowment BankTM. Plasma and serum samples were obtained from the University of Kentucky Alzheimer’s Disease Center Brain Bank.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. 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.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

JPdRV designed the study, performed experiments, analyzed the data, interpreted the results and prepared the manuscript. FJB and CS performed experiments and analyzed the data. DCM, WDD and RWK reviewed and discussed the manuscript. All authors have read and approved the final version of the manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Vollmar P, Kullmann JS, Thilo B, Claussen MC, Rothhammer V, Jacobi H, Sellner J, Nessler S, Korn T, Hemmer B: Active immunization with amyloid-beta 1-42 impairs memory performance through TLR2/4-dependent activation of the innate immune system. J Immunol. 2010, 185: 6338-6347. 10.4049/jimmunol.1001765.CrossRefPubMed Vollmar P, Kullmann JS, Thilo B, Claussen MC, Rothhammer V, Jacobi H, Sellner J, Nessler S, Korn T, Hemmer B: Active immunization with amyloid-beta 1-42 impairs memory performance through TLR2/4-dependent activation of the innate immune system. J Immunol. 2010, 185: 6338-6347. 10.4049/jimmunol.1001765.CrossRefPubMed
2.
Zurück zum Zitat Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, Rayner KJ, Boyer L, Zhong R, Frazier WA, Lacy-Hulbert A, El Khoury J, Golenbock DT, Moore KJ: CD36 ligands promote sterile inflammation through assembly of a toll-like receptor 4 and 6 heterodimer. Nat Immunol. 2010, 11: 155-161. 10.1038/ni.1836.PubMedCentralCrossRefPubMed Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, Rayner KJ, Boyer L, Zhong R, Frazier WA, Lacy-Hulbert A, El Khoury J, Golenbock DT, Moore KJ: CD36 ligands promote sterile inflammation through assembly of a toll-like receptor 4 and 6 heterodimer. Nat Immunol. 2010, 11: 155-161. 10.1038/ni.1836.PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Tang SC, Lathia JD, Selvaraj PK, Jo DG, Mughal MR, Cheng A, Siler DA, Markesbery WR, Arumugam TV, Mattson MP: Toll-like receptor-4 mediates neuronal apoptosis induced by amyloid beta-peptide and the membrane lipid peroxidation product 4-hydroxynonenal. Exp Neurol. 2008, 213: 114-121. 10.1016/j.expneurol.2008.05.014.PubMedCentralCrossRefPubMed Tang SC, Lathia JD, Selvaraj PK, Jo DG, Mughal MR, Cheng A, Siler DA, Markesbery WR, Arumugam TV, Mattson MP: Toll-like receptor-4 mediates neuronal apoptosis induced by amyloid beta-peptide and the membrane lipid peroxidation product 4-hydroxynonenal. Exp Neurol. 2008, 213: 114-121. 10.1016/j.expneurol.2008.05.014.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Jin JJ, Kim HD, Maxwell JA, Li L, Fukuchi K: Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer’s disease. J Neuroinflammation. 2008, 5: 23-10.1186/1742-2094-5-23.PubMedCentralCrossRefPubMed Jin JJ, Kim HD, Maxwell JA, Li L, Fukuchi K: Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer’s disease. J Neuroinflammation. 2008, 5: 23-10.1186/1742-2094-5-23.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Minoretti P, Politi P, Coen E, Di Vito C, Bertona M, Bianchi M, Emanuele E: The T393C polymorphism of the GNAS1 gene is associated with deficit schizophrenia in an Italian population sample. Neurosci Lett. 2006, 397: 159-163. 10.1016/j.neulet.2005.12.028.CrossRefPubMed Minoretti P, Politi P, Coen E, Di Vito C, Bertona M, Bianchi M, Emanuele E: The T393C polymorphism of the GNAS1 gene is associated with deficit schizophrenia in an Italian population sample. Neurosci Lett. 2006, 397: 159-163. 10.1016/j.neulet.2005.12.028.CrossRefPubMed
6.
Zurück zum Zitat Liu S, Liu Y, Hao W, Wolf L, Kiliaan AJ, Penke B, Rube CE, Walter J, Heneka MT, Hartmann T, Menger MD, Fassbender K: TLR2 is a primary receptor for Alzheimer’s amyloid beta peptide to trigger neuroinflammatory activation. J Immunol. 2012, 188: 1098-1107. 10.4049/jimmunol.1101121.CrossRefPubMed Liu S, Liu Y, Hao W, Wolf L, Kiliaan AJ, Penke B, Rube CE, Walter J, Heneka MT, Hartmann T, Menger MD, Fassbender K: TLR2 is a primary receptor for Alzheimer’s amyloid beta peptide to trigger neuroinflammatory activation. J Immunol. 2012, 188: 1098-1107. 10.4049/jimmunol.1101121.CrossRefPubMed
8.
Zurück zum Zitat Takeuchi O, Akira S: Pattern recognition receptors and inflammation. Cell. 2010, 140: 805-820. 10.1016/j.cell.2010.01.022.CrossRefPubMed Takeuchi O, Akira S: Pattern recognition receptors and inflammation. Cell. 2010, 140: 805-820. 10.1016/j.cell.2010.01.022.CrossRefPubMed
9.
Zurück zum Zitat Taylor JM, Minter MR, Newman AG, Zhang M, Adlard PA, Crack PJ: Type-1 interferon signaling mediates neuro-inflammatory events in models of Alzheimer’s disease. Neurobiol Aging. 2014, 35: 1012-1023. 10.1016/j.neurobiolaging.2013.10.089.CrossRefPubMed Taylor JM, Minter MR, Newman AG, Zhang M, Adlard PA, Crack PJ: Type-1 interferon signaling mediates neuro-inflammatory events in models of Alzheimer’s disease. Neurobiol Aging. 2014, 35: 1012-1023. 10.1016/j.neurobiolaging.2013.10.089.CrossRefPubMed
10.
Zurück zum Zitat de Rivero Vaccari JP, Minkiewicz J, Wang X, De Rivero Vaccari JC, German R, Marcillo AE, Dietrich WD, Keane RW: Astrogliosis involves activation of retinoic acid-inducible gene-like signaling in the innate immune response after spinal cord injury. Glia. 2012, 60: 414-421. 10.1002/glia.22275.PubMedCentralCrossRefPubMed de Rivero Vaccari JP, Minkiewicz J, Wang X, De Rivero Vaccari JC, German R, Marcillo AE, Dietrich WD, Keane RW: Astrogliosis involves activation of retinoic acid-inducible gene-like signaling in the innate immune response after spinal cord injury. Glia. 2012, 60: 414-421. 10.1002/glia.22275.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Mawhinney LJ, de Vaccari Rivero JP, Dale GA, Keane RW, Bramlett HM: Heightened inflammasome activation is linked to age-related cognitive impairment in fischer 344 rats. BMC Neurosci. 2011, 12: 123-10.1186/1471-2202-12-123.PubMedCentralCrossRefPubMed Mawhinney LJ, de Vaccari Rivero JP, Dale GA, Keane RW, Bramlett HM: Heightened inflammasome activation is linked to age-related cognitive impairment in fischer 344 rats. BMC Neurosci. 2011, 12: 123-10.1186/1471-2202-12-123.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Morris JC, Storandt M, Miller JP, McKeel DW, Price JL, Rubin EH, Berg L: Mild cognitive impairment represents early-stage Alzheimer disease. Arch Neurol. 2001, 58: 397-405.PubMed Morris JC, Storandt M, Miller JP, McKeel DW, Price JL, Rubin EH, Berg L: Mild cognitive impairment represents early-stage Alzheimer disease. Arch Neurol. 2001, 58: 397-405.PubMed
13.
Zurück zum Zitat Mufson EJ, Binder L, Counts SE, DeKosky ST, de Toledo-Morrell L, Ginsberg SD, Ikonomovic MD, Perez SE, Scheff SW: Mild cognitive impairment: pathology and mechanisms. Acta Neuropathol. 2012, 123: 13-30. 10.1007/s00401-011-0884-1.PubMedCentralCrossRefPubMed Mufson EJ, Binder L, Counts SE, DeKosky ST, de Toledo-Morrell L, Ginsberg SD, Ikonomovic MD, Perez SE, Scheff SW: Mild cognitive impairment: pathology and mechanisms. Acta Neuropathol. 2012, 123: 13-30. 10.1007/s00401-011-0884-1.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen E: Mild cognitive impairment: clinical characterization and outcome. Arch Neurol. 1999, 56: 303-308. 10.1001/archneur.56.3.303.CrossRefPubMed Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen E: Mild cognitive impairment: clinical characterization and outcome. Arch Neurol. 1999, 56: 303-308. 10.1001/archneur.56.3.303.CrossRefPubMed
15.
Zurück zum Zitat Fleming KC, Adams AC, Petersen RC: Dementia: diagnosis and evaluation. Mayo Clin Proc. 1995, 70: 1093-1107. 10.4065/70.11.1093.CrossRefPubMed Fleming KC, Adams AC, Petersen RC: Dementia: diagnosis and evaluation. Mayo Clin Proc. 1995, 70: 1093-1107. 10.4065/70.11.1093.CrossRefPubMed
16.
Zurück zum Zitat Mirra SS, Heyman A, McKeel D, Sumi SM, Crain BJ, Brownlee LM, Vogel FS, Hughes JP, van Belle G, Berg L: The consortium to establish a registry for Alzheimer’s disease (CERAD): part II standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology. 1991, 41: 479-486. 10.1212/WNL.41.4.479.CrossRefPubMed Mirra SS, Heyman A, McKeel D, Sumi SM, Crain BJ, Brownlee LM, Vogel FS, Hughes JP, van Belle G, Berg L: The consortium to establish a registry for Alzheimer’s disease (CERAD): part II standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology. 1991, 41: 479-486. 10.1212/WNL.41.4.479.CrossRefPubMed
17.
Zurück zum Zitat Braak H, Braak E: Neuropathological staging of Alzheimer-related changes. Acta Neuropathol. 1991, 82: 239-259. 10.1007/BF00308809.CrossRefPubMed Braak H, Braak E: Neuropathological staging of Alzheimer-related changes. Acta Neuropathol. 1991, 82: 239-259. 10.1007/BF00308809.CrossRefPubMed
18.
Zurück zum Zitat Schieb H, Kratzin H, Jahn O, Mobius W, Rabe S, Staufenbiel M, Wiltfang J, Klafki HW: Beta-amyloid peptide variants in brains and cerebrospinal fluid from amyloid precursor protein (APP) transgenic mice: comparison with human Alzheimer amyloid. J Biol Chem. 2011, 286: 33747-33758. 10.1074/jbc.M111.246561.PubMedCentralCrossRefPubMed Schieb H, Kratzin H, Jahn O, Mobius W, Rabe S, Staufenbiel M, Wiltfang J, Klafki HW: Beta-amyloid peptide variants in brains and cerebrospinal fluid from amyloid precursor protein (APP) transgenic mice: comparison with human Alzheimer amyloid. J Biol Chem. 2011, 286: 33747-33758. 10.1074/jbc.M111.246561.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Hornung V, Ellegast J, Kim S, Brzozka K, Jung A, Kato H, Poeck H, Akira S, Conzelmann KK, Schlee M, Endres S, Hartmann G: 5′-triphosphate RNA is the ligand for RIG-I. Science. 2006, 314: 994-997. 10.1126/science.1132505.CrossRefPubMed Hornung V, Ellegast J, Kim S, Brzozka K, Jung A, Kato H, Poeck H, Akira S, Conzelmann KK, Schlee M, Endres S, Hartmann G: 5′-triphosphate RNA is the ligand for RIG-I. Science. 2006, 314: 994-997. 10.1126/science.1132505.CrossRefPubMed
20.
Zurück zum Zitat Thies W, Bleiler L: Alzheimer’s disease facts and figures. Alzheimers Dement. 2011, 2011 (7): 208-244. Thies W, Bleiler L: Alzheimer’s disease facts and figures. Alzheimers Dement. 2011, 2011 (7): 208-244.
21.
Zurück zum Zitat Salloway S, Mintzer J, Weiner MF, Cummings JL: Disease-modifying therapies in Alzheimer’s disease. Alzheimers Dement. 2008, 4: 65-79. 10.1016/j.jalz.2007.10.001.CrossRefPubMed Salloway S, Mintzer J, Weiner MF, Cummings JL: Disease-modifying therapies in Alzheimer’s disease. Alzheimers Dement. 2008, 4: 65-79. 10.1016/j.jalz.2007.10.001.CrossRefPubMed
22.
Zurück zum Zitat Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA: Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol. 2003, 60: 1119-1122. 10.1001/archneur.60.8.1119.CrossRefPubMed Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA: Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol. 2003, 60: 1119-1122. 10.1001/archneur.60.8.1119.CrossRefPubMed
23.
Zurück zum Zitat Lorenzo A, Yuan M, Zhang Z, Paganetti PA, Sturchler-Pierrat C, Staufenbiel M, Mautino J, Vigo FS, Sommer B, Yankner BA: Amyloid beta interacts with the amyloid precursor protein: a potential toxic mechanism in Alzheimer’s disease. Nat Neurosci. 2000, 3: 460-464. 10.1038/74833.CrossRefPubMed Lorenzo A, Yuan M, Zhang Z, Paganetti PA, Sturchler-Pierrat C, Staufenbiel M, Mautino J, Vigo FS, Sommer B, Yankner BA: Amyloid beta interacts with the amyloid precursor protein: a potential toxic mechanism in Alzheimer’s disease. Nat Neurosci. 2000, 3: 460-464. 10.1038/74833.CrossRefPubMed
24.
Zurück zum Zitat Yankner BA: New clues to Alzheimer’s disease: unraveling the roles of amyloid and tau. Nat Med. 1996, 2: 850-852. 10.1038/nm0896-850.CrossRefPubMed Yankner BA: New clues to Alzheimer’s disease: unraveling the roles of amyloid and tau. Nat Med. 1996, 2: 850-852. 10.1038/nm0896-850.CrossRefPubMed
25.
Zurück zum Zitat Yankner BA: The pathogenesis of Alzheimer’s disease: is amyloid beta-protein the beginning or the end?. Ann N Y Acad Sci. 2000, 924: 26-28.CrossRefPubMed Yankner BA: The pathogenesis of Alzheimer’s disease: is amyloid beta-protein the beginning or the end?. Ann N Y Acad Sci. 2000, 924: 26-28.CrossRefPubMed
26.
Zurück zum Zitat Yankner BA, Lu T: Amyloid beta-protein toxicity and the pathogenesis of Alzheimer disease. J Biol Chem. 2009, 284: 4755-4759. 10.1074/jbc.R800018200.PubMedCentralCrossRefPubMed Yankner BA, Lu T: Amyloid beta-protein toxicity and the pathogenesis of Alzheimer disease. J Biol Chem. 2009, 284: 4755-4759. 10.1074/jbc.R800018200.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Ricci S, Fuso A, Ippoliti F, Businaro R: Stress-induced cytokines and neuronal dysfunction in Alzheimer’s disease. J Alzheimers Dis. 2012, 28: 11-24.PubMed Ricci S, Fuso A, Ippoliti F, Businaro R: Stress-induced cytokines and neuronal dysfunction in Alzheimer’s disease. J Alzheimers Dis. 2012, 28: 11-24.PubMed
28.
Zurück zum Zitat Rogers J: Inflammation as a pathogenic mechanism in Alzheimer’s disease. Arzneimittelforschung. 1995, 45: 439-442.PubMed Rogers J: Inflammation as a pathogenic mechanism in Alzheimer’s disease. Arzneimittelforschung. 1995, 45: 439-442.PubMed
29.
Zurück zum Zitat Rogers J: The inflammatory response in Alzheimer’s disease. J Periodontol. 2008, 79: 1535-1543. 10.1902/jop.2008.080171.CrossRefPubMed Rogers J: The inflammatory response in Alzheimer’s disease. J Periodontol. 2008, 79: 1535-1543. 10.1902/jop.2008.080171.CrossRefPubMed
30.
Zurück zum Zitat Rogers J, Shen Y: A perspective on inflammation in Alzheimer’s disease. Ann N Y Acad Sci. 2000, 924: 132-135.CrossRefPubMed Rogers J, Shen Y: A perspective on inflammation in Alzheimer’s disease. Ann N Y Acad Sci. 2000, 924: 132-135.CrossRefPubMed
31.
Zurück zum Zitat Rogers JT, Leiter LM, McPhee J, Cahill CM, Zhan SS, Potter H, Nilsson LN: Translation of the Alzheimer amyloid precursor protein mRNA is up-regulated by interleukin-1 through 5′-untranslated region sequences. J Biol Chem. 1999, 274: 6421-6431. 10.1074/jbc.274.10.6421.CrossRefPubMed Rogers JT, Leiter LM, McPhee J, Cahill CM, Zhan SS, Potter H, Nilsson LN: Translation of the Alzheimer amyloid precursor protein mRNA is up-regulated by interleukin-1 through 5′-untranslated region sequences. J Biol Chem. 1999, 274: 6421-6431. 10.1074/jbc.274.10.6421.CrossRefPubMed
32.
Zurück zum Zitat Simard AR, Rivest S: Neuroprotective properties of the innate immune system and bone marrow stem cells in Alzheimer’s disease. Mol Psychiatry. 2006, 11: 327-335. 10.1038/sj.mp.4001809.CrossRefPubMed Simard AR, Rivest S: Neuroprotective properties of the innate immune system and bone marrow stem cells in Alzheimer’s disease. Mol Psychiatry. 2006, 11: 327-335. 10.1038/sj.mp.4001809.CrossRefPubMed
33.
Zurück zum Zitat Weninger SC, Yankner BA: Inflammation and Alzheimer disease: the good, the bad, and the ugly. Nat Med. 2001, 7: 527-528. 10.1038/87839.CrossRefPubMed Weninger SC, Yankner BA: Inflammation and Alzheimer disease: the good, the bad, and the ugly. Nat Med. 2001, 7: 527-528. 10.1038/87839.CrossRefPubMed
34.
Zurück zum Zitat McGeer PL, Rogers J, McGeer EG: Inflammation, anti-inflammatory agents and Alzheimer disease: the last 12 years. J Alzheimers Dis. 2006, 9: 271-276.PubMed McGeer PL, Rogers J, McGeer EG: Inflammation, anti-inflammatory agents and Alzheimer disease: the last 12 years. J Alzheimers Dis. 2006, 9: 271-276.PubMed
35.
Zurück zum Zitat Shen Y, Lue L, Yang L, Roher A, Kuo Y, Strohmeyer R, Goux WJ, Lee V, Johnson GV, Webster SD, Cooper NR, Bradt B, Rogers J: Complement activation by neurofibrillary tangles in Alzheimer’s disease. Neurosci Lett. 2001, 305: 165-168. 10.1016/S0304-3940(01)01842-0.CrossRefPubMed Shen Y, Lue L, Yang L, Roher A, Kuo Y, Strohmeyer R, Goux WJ, Lee V, Johnson GV, Webster SD, Cooper NR, Bradt B, Rogers J: Complement activation by neurofibrillary tangles in Alzheimer’s disease. Neurosci Lett. 2001, 305: 165-168. 10.1016/S0304-3940(01)01842-0.CrossRefPubMed
36.
Zurück zum Zitat Lafon M, Megret F, Lafage M, Prehaud C: The innate immune facet of brain: human neurons express TLR-3 and sense viral dsRNA. J Mol Neurosci. 2006, 29: 185-194. 10.1385/JMN:29:3:185.CrossRefPubMed Lafon M, Megret F, Lafage M, Prehaud C: The innate immune facet of brain: human neurons express TLR-3 and sense viral dsRNA. J Mol Neurosci. 2006, 29: 185-194. 10.1385/JMN:29:3:185.CrossRefPubMed
37.
Zurück zum Zitat Malathi K, Paranjape JM, Bulanova E, Shim M, Guenther-Johnson JM, Faber PW, Eling TE, Williams BR, Silverman RH: A transcriptional signaling pathway in the IFN system mediated by 2′-5′-oligoadenylate activation of RNase L. Proc Natl Acad Sci USA. 2005, 102: 14533-14538. 10.1073/pnas.0507551102.PubMedCentralCrossRefPubMed Malathi K, Paranjape JM, Bulanova E, Shim M, Guenther-Johnson JM, Faber PW, Eling TE, Williams BR, Silverman RH: A transcriptional signaling pathway in the IFN system mediated by 2′-5′-oligoadenylate activation of RNase L. Proc Natl Acad Sci USA. 2005, 102: 14533-14538. 10.1073/pnas.0507551102.PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Tal MC, Sasai M, Lee HK, Yordy B, Shadel GS, Iwasaki A: Absence of autophagy results in reactive oxygen species-dependent amplification of RLR signaling. Proc Natl Acad Sci USA. 2009, 106: 2770-2775. 10.1073/pnas.0807694106.PubMedCentralCrossRefPubMed Tal MC, Sasai M, Lee HK, Yordy B, Shadel GS, Iwasaki A: Absence of autophagy results in reactive oxygen species-dependent amplification of RLR signaling. Proc Natl Acad Sci USA. 2009, 106: 2770-2775. 10.1073/pnas.0807694106.PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Rainer TH, Lam NY: Circulating nucleic acids and critical illness. Ann N Y Acad Sci. 2006, 1075: 271-277. 10.1196/annals.1368.035.CrossRefPubMed Rainer TH, Lam NY: Circulating nucleic acids and critical illness. Ann N Y Acad Sci. 2006, 1075: 271-277. 10.1196/annals.1368.035.CrossRefPubMed
40.
Zurück zum Zitat Kitamura T, Kitamura M, Hino S, Tanaka N, Kurata K: Gender differences in clinical manifestations and outcomes among hospitalized patients with behavioral and psychological symptoms of dementia. J Clin Psychiatry. 2012, 73: 1548-1554. 10.4088/JCP.11m07614.CrossRefPubMed Kitamura T, Kitamura M, Hino S, Tanaka N, Kurata K: Gender differences in clinical manifestations and outcomes among hospitalized patients with behavioral and psychological symptoms of dementia. J Clin Psychiatry. 2012, 73: 1548-1554. 10.4088/JCP.11m07614.CrossRefPubMed
41.
Zurück zum Zitat Allaman I, Gavillet M, Belanger M, Laroche T, Viertl D, Lashuel HA, Magistretti PJ: Amyloid-beta aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability. J Neurosci. 2010, 30: 3326-3338. 10.1523/JNEUROSCI.5098-09.2010.CrossRefPubMed Allaman I, Gavillet M, Belanger M, Laroche T, Viertl D, Lashuel HA, Magistretti PJ: Amyloid-beta aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability. J Neurosci. 2010, 30: 3326-3338. 10.1523/JNEUROSCI.5098-09.2010.CrossRefPubMed
42.
Zurück zum Zitat Calvillo M, Diaz A, Limon DI, Mayoral MA, Chanez-Cardenas ME, Zenteno E, Montano LF, Guevara J, Espinosa B: Amyloid-beta induces a permanent phosphorylation of HSF-1, but a transitory and inflammation-independent overexpression of Hsp-70 in C6 astrocytoma cells. Neuropeptides. 2013, 47: 339-346. 10.1016/j.npep.2013.06.002.CrossRefPubMed Calvillo M, Diaz A, Limon DI, Mayoral MA, Chanez-Cardenas ME, Zenteno E, Montano LF, Guevara J, Espinosa B: Amyloid-beta induces a permanent phosphorylation of HSF-1, but a transitory and inflammation-independent overexpression of Hsp-70 in C6 astrocytoma cells. Neuropeptides. 2013, 47: 339-346. 10.1016/j.npep.2013.06.002.CrossRefPubMed
43.
Zurück zum Zitat Grolla AA, Sim JA, Lim D, Rodriguez JJ, Genazzani AA, Verkhratsky A: Amyloid-beta and Alzheimer’s disease type pathology differentially affects the calcium signalling toolkit in astrocytes from different brain regions. Cell Death Dis. 2013, 4: e623-10.1038/cddis.2013.145.PubMedCentralCrossRefPubMed Grolla AA, Sim JA, Lim D, Rodriguez JJ, Genazzani AA, Verkhratsky A: Amyloid-beta and Alzheimer’s disease type pathology differentially affects the calcium signalling toolkit in astrocytes from different brain regions. Cell Death Dis. 2013, 4: e623-10.1038/cddis.2013.145.PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat 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. 10.1016/S0006-8993(03)02361-8.CrossRefPubMed 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. 10.1016/S0006-8993(03)02361-8.CrossRefPubMed
Metadaten
Titel
RIG-1 receptor expression in the pathology of Alzheimer’s disease
verfasst von
Juan Pablo de Rivero Vaccari
Frank J Brand III
Christina Sedaghat
Deborah C Mash
W Dalton Dietrich
Robert W Keane
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2014
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-11-67

Weitere Artikel der Ausgabe 1/2014

Journal of Neuroinflammation 1/2014 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie