Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2016

Open Access 01.12.2016 | Research

Assessment of neuroinflammation in a mouse model of obesity and β-amyloidosis using PET

verfasst von: Anna M. Barron, Masaki Tokunaga, Ming-Rong Zhang, Bin Ji, Tetsuya Suhara, Makoto Higuchi

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2016

Abstract

Background

Obesity has been identified as a risk factor for cognitive decline and Alzheimer’s disease (AD). The aim of this study was to investigate the effect of obesity on neuroinflammation and cerebral glucose metabolism using PET in a mouse model of β-amyloidosis and determine the relationship between these PET imaging biomarkers, pathogenic changes, and functional outcomes.

Methods

Three-month-old C57BL/J6 mice were fed either a standard (control group) or high-fat diet (obese group) for 3 months and intracerebroventricularly infused with vehicle or human beta amyloid 1-42 (Aβ42). We assessed obesity-induced abnormalities in peripheral metabolic indices including adiposity, fasting glucose, and glucose tolerance. Brain glucose metabolism was assessed by 18F-FDG PET, and glial activation was assessed using the translocator protein (TSPO) ligand 11C-PBR-28. TSPO expression was confirmed by immunohistochemistry of brain sections obtained from scanned mice. The association between inflammatory state and 11C-PBR-28 PET signals was characterized by examination of the cytokine expression profile in both the serum and hippocampus by antibody array. Learning and memory performance was assessed in the object recognition task, and anxiety-related behavior was assessed in the elevated plus maze.

Results

Obesity combined with Aβ infusion promoted neuroinflammation and cerebral hypermetabolism, and these signals were significant predictors of learning and memory performance in the object recognition task. In vivo TSPO signals were associated with inflammatory markers including CXCL1, CXCL2, CXCL12, CCL3, CCL5, TIMP-1, G-CSF, sICAM-1, and IL-1ra.

Conclusions

In vivo cerebral metabolism and TSPO signals indicate that obesity can accelerate amyloid-induced inflammation and associated cognitive decline.
Abkürzungen
Beta amyloid
AD
Alzheimer’s disease
AIC
Akaike information criterion
ANOVA
Analysis of Variance
C5a
Complement component 5
CCL
Chemokine (C–C motif) ligand
CXCL
Chemokine (C–X–C motif) ligand
EPM
Elevated plus maze
FDG
Fluoro-2-deoxy-d-glucose
G-CSF
Granulocyte colony-stimulating factor
GFAP
Glial fibrillary acidic protein
GM-CSF
Granulocyte macrophage colony-stimulating factor
HDL
High-density lipoprotein
IBA-1
Ionized calcium-binding adaptor molecule 1
ICAM-1
Intracellular adhesion molecule 1
IFN-γ
Interferon gamma
IL
Interleukin
M-CSF
Macrophage colony-stimulating factor
MCI
Mild cognitive impairment
MRI
Magnetic resonance imaging
PBS
Phosphate-buffered saline
PET
Positron emission tomography
SEM
Standard error of mean
SUV
Standardized uptake value
TIMP-1
Tissue inhibitor of metalloproteinase 1
TNF-α
Tumor necrosis factor alpha
TSPO
Translocator protein
TREM-1
Triggering receptor expressed on myeloid cells 1
VOI
Volume of interest
%ID/g
Percentage of injected dose per tissue volume

Background

Obesity is a global health issue owing to high intake of refined carbohydrates and saturated fat and increasingly sedentary lifestyles [1]. Growing evidence suggests that obesity at midlife increases the risk of cognitive decline and Alzheimer’s disease (AD) in later life [24]. AD pathogenesis, which begins decades prior to the onset of symptoms [5, 6], is characterized by the accumulation of the toxic beta amyloid (Aβ) peptides, formation of neurofibrillary tangles, chronic inflammation, and neuronal loss [5, 7]. Numerous imaging modalities including structural and functional magnetic resonance imaging (MRI) and positron emission tomography (PET) assessment of cerebral glucose metabolism and amyloid burden have proven diagnostically useful in mild cognitive impairment (MCI) and AD [8]. However, the relationship between imaging biomarkers and early midlife pathogenic changes leading to AD, such as obesity, has remained elusive. Since obesity-driven inflammation and metabolic impairment have been identified as important mechanisms accelerating AD pathogenesis and functional impairment [913], they may represent potential early-stage imaging markers of cognitive decline and AD risk.
Neuroinflammation has been identified as a contributor to both the initiation and progression of AD. Accumulation of soluble Aβ is suggested to induce inflammatory processes in AD, activating glial cells and potentiating the production of proinflammatory cytokines [14]. In obesity, chronic systemic inflammation triggers neuroinflammation, thereby exacerbating Aβ-induced degenerative cascades and AD progression [15]. The mitochondrial translocator protein (TSPO) is markedly upregulated in activated glia in inflammatory disease including AD, believed to play a critical role in mediating glial responses to AD pathogenesis. In vitro studies using transgenic mouse models of AD indicated that TSPO expression is linked with neuronal toxicity and death [16, 17]. 11C-PBR-28 is a second-generation TSPO ligand with high affinity and penetration of the blood-brain barrier compared to the prototypic first-generation TSPO ligand, 11C-PK-11195 [18]. Previous studies have demonstrated the usefulness of 11C-PBR-28 in subjects with AD [19] and multiple sclerosis [20] and in animal models of aging [21] and cerebral ischemia [22]. However, the detection of early-stage inflammatory changes associated with obesity-related cognitive decline and AD risk has not been investigated using TSPO ligands.
Cerebral glucose metabolism measured by 18F-fluoro-2-deoxy-d-glucose-PET (FDG PET) is widely used in AD as an index of neuronal loss [23]. Although cerebral metabolic changes prior to the development of neuronal loss in AD remains poorly understood, emerging evidence suggests that hypometabolism may be preceded by a hypermetabolic phase [2427]. It is unclear if this early hypermetabolic stage reflects neuronal hyperactivity or inflammatory changes or if the hyperactivity is a cause (e.g., activity-driven Aβ deposition) or result of AD pathogenesis (e.g., Aβ-induced seizure activity or inflammation) [26, 27]. Transgenic mouse models of amyloidosis which do not develop neuronal loss may be useful tools for investigating metabolic changes associated early stages of Aβ pathogenesis [2831]. However, use of 18F-FDG PET in transgenic animal models may be hampered by the overexpression of amyloid precursor protein (APP) and non-Aβ fragments of APP, which are known to modify synaptic and metabolic activity [2831]. To circumvent this, here, we have used Aβ-infusion to investigate interactions between Aβ- and obesity-driven inflammatory and metabolic abnormalities in vivo using 11C-PBR-28 and 18F-FDG PET, respectively, and determine the relationship between these imaging biomarkers, underlying pathogenic changes and functional outcomes.

Methods

Animals and treatments

Three-month-old male C57Bl/J mice were purchased (Clear, Japan) and maintained at the National Institute of Radiological Sciences vivarium facilities with food and water available ad libitum. Beginning at 3 months of age, mice were randomly assigned to groups (n = 6/group) and maintained on a standard diet (10 % kCal fat; Research Diets D12450B Harlan Teklad, Indianapolis, IN) or a high-calorie, high-fat diet (60 % kCal fat; Research Diets D12492, Harlan Teklad) for a period of 3 months. Body weights were monitored weekly, and fasting blood glucose was monitored fortnightly.
After 1 month of feeding, mice were anesthetized with isoflurane (3 % v/v for induction, 1.5 % v/v for maintenance), positioned in a stereotaxic apparatus and 0.9 % saline applied to the eyes. The scalp was shaved and cut, the skull exposed, and adhering tissue was removed with acetone. A cannula (Brain Infusion Kit 3, Alzet) was implanted in the left ventricle at the following coordinates: +1.0 medial/lateral, −0.3 anterior/posterior, −2.5 dorsal/ventral. The cannula was fixed to the skull using dental cement and connected to a mini-osmotic pump (Model 1002, Alzet) that was filled with either vehicle (250 μg/mL high-density lipoprotein (HDL) in 4 mM HEPES with 2.5 % DMSO) or 120-μM oligomeric Aβ-42 [32]. Oligomeric Aβ-42 was prepared by solubilizing synthetic human Aβ-42 (Peptide Institute) to 1 mM in hexafluoroisopropanol, then drying under vacuum in a SpeedVac. The peptide film was then resuspended in DMSO to 5 mM and diluted in 4 mM HEPES containing 250 μg/mL HDL (Millipore) to a final concentration of 120 μM. Pumps were partially coated with paraffin to adjust the infusion rate to 3 μL/day for 1 month, then the filled pumps were incubated in sterile phosphate-buffered saline (PBS) at 37 °C for 40 h prior to implantation under the dorsal skin on the back. The incision site on the scalp was closed with suture, and mice were administered buprenorphrine (0.05 mg/kg i.p., Henry Schein Inc.) post-operatively for analgesia. One spontaneous death occurred in the 8 weeks post-surgery treatment duration (obese + Aβ group).
All experimentation was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health and was approved by the Institutional Animal Care and Use Committee of the National Institutes for Quantum and Radiological Science and Technology, Japan.

Glucose measurements

Fasting blood glucose was assessed using a Nipro Freestyle Glucometer (Nipro Diagnostics, Florida, USA) from the whole blood collected via the tail vein while the mouse was under isofluorane general anesthesia. Mice were fasted overnight for 16 h prior to sample collection. Mice were fasted at baseline (time 0), 1, 2 months and sacrificed for assessment of blood glucose levels. Mice were additionally fasted overnight at 2.5 months for the 18F-FDG PET scans and again for 2 days later for the glucose tolerance test. For the glucose tolerance test, baseline glucose levels were measured, then fasted mice were injected with 2 mg glucose/g body weight (i.p.), and blood glucose was measured from the whole blood collected via the tail vein 30, 60, and 120 min after injection.

In vivo PET imaging

TSPO signals were assessed by PET using 11C-PBR-28, which was prepared according to previously published methods [33]. The specific activity of the end product was 80.7 ± 14.7 GBq/μmol and the radiochemical purity exceeded 95 %. 18F-FDG was purchased from Nihon Medi-Physics Co. LTD (Tokyo, Japan). Mice were fasted prior to 18F-FDG PET scans, and blood glucose levels were assessed at the completion of scan.
Mice were anesthetized with 1.5 % (v/v) isoflurane and administered 11C-PBR-28 (27.9 ± 0.9 MBq) or 18F-FDG (17.6 ± 0.2 MBq) via a tail vein catheter while in a microPET Focus 220 animal scanner (Siemens Medical Solutions). Simultaneous with radioligand injection, 3D list mode data acquisitions were started and lasted 60 min. Summation images were reconstructed with maximum a posteriori reconstruction (40–60 min for 11C-PBR-28 and 20–60 min for 18F-FDG), and dynamic images were reconstructed with filtered back-projection using a 0.5-mm Hanning filter, which were used for quantification of standardized uptake values (SUVs) and percentage of injected dose per tissue volume (%ID/g). Volumes of interest (VOIs) were placed at the infusion site and in the hippocampal and cerebellar regions with reference to the MRI template (Fig. 1). Then, dynamic PET images were merged to the MRI template, and the tracer uptake in each VOI was estimated as %ID/g for 11C-PBR-28 (40–60 min) and SUV for 18F-FDG (20–60 min). PET image analysis described above was performed with PMOD image analysis software (version 3.4; PMOD Technologies, Inc., Zurich, Switzerland).

Behavioral assessment

Short-term working memory was assessed in the object recognition task. Mice were placed in the centre of an open field (50 × 50 × 50 cm) and allowed to freely explore for 3 min (habituation trial). Following a 2-h interval, mice were again placed in the centre of the maze between two identical objects and allowed to explore freely for 6 min (training trial). The frequency of the left and right object explorations were scored to test for spatial bias. After a 24-h inter-trial interval, one object was replaced with a novel object and the mouse was allowed to freely explore for 10 min (probe trial). The duration of novel (D n) and familiar (D f) object explorations were recorded and used to calculate the percent recognition (D n/(D n + D f)).
Anxiety-related behavior was assessed in the elevated plus maze (EPM). Mice were placed in the center of a plus-shaped maze (25 cm × 8 cm) with two open arms and two closed arms elevated 60 cm from the ground. The closed arms were surrounded by 24-cm walls. Behavior was recorded in a 5-min period via overhead digital video and analyzed using SMART Automated Recording (Panlab). The total duration spent in the open arm was calculated as an index of anxiety-related behavior.

Tissue collection and preparation

Mice were fasted for 16 h prior to sacrifice then deeply anesthetized (50 mg sodium pentobarbital/kg body weight, i.p.). Blood was collected via cardiac puncture and fasting blood glucose assessed. Serum was collected from the remaining blood and stored at −80 °C. Mice were then intracardially perfused with ice-cold, sterile saline, and one hemibrain was submersion fixed in 4 % paraformaldehyde/0.1 M PBS (pH 7.4) for 24 h for immunohistochemistry. The hippocampus was rapidly dissected from the remaining hemibrain and snap frozen on liquid nitrogen. Retroperitoneal and subcutaneous fat pads were collected at necropsy and weighed as an indicator of visceral adiposity and body composition.

Immunohistochemistry

Fixed hemibrains were cryoprotected in 30 % sucrose, frozen, and sectioned in the horizontal plane at 20 μM using a cryostat. Sections were soaked in PBS for 5 min, then immunolabeled with antibodies directed against TSPO (rabbit monoclonal, 1:1000; Abcam), glial fibrillary acidic protein (GFAP; rat monoclonal, 1:1000 dilution, 2.2B10, Zymed), or anti-ionized calcium-binding adaptor molecule-1 (IBA-1; rabbit polyclonal, 1:1000 dilution, WAKO). Non-specific staining was blocked in blocking reagent (PerkinElmer) for 1 h at room temperature, then immunolabeled overnight at 4 °C with primary antibodies. Sections were then washed three times in PBS for 5 min and then incubated with anti-rabbit or anti-rat IgG biotin (1:1000) for 1 h at room temperature. Immunoreactivity was visualized using tetramethylrhodamine-labeled tyramide signal amplification (PerkinElmer). Sections were mounted with VectaSheild (Vector Laboratories), and coverslips were sealed with enamel. Photomicrographs were captured with a fluorescence microscope/digital camera (BZ-X700, Keyence) at ×20 magnification and then thresholded at a predetermined, constant value using NIH Image 1.61 to create a binary image identifying positive and negative immunolabeling and the intensity assessed.

Cytokine antibody microarray

Mouse cytokine antibody arrays (cytokine array panel A, Proteome Profiler, R&D systems, Minneapolis, USA) were used to analyze cytokine expression profiles in serum and hippocampal samples according to the manufacturer’s instructions. Frozen hippocampus samples were homogenized by bead cell disrupter (Micro Smash) in PBS containing complete ULTRA protease inhibitors (Roche). A digital imaging system (Chemidoc, Bio-Rad) was used to detect chemiluminescent signals which were analyzed using ImageJ software (NIH). Measured cytokines included the following: complement component 5 (C5a), chemokine (C–C motif) ligand 1 (CCL1), CCL2, CCL3, CCL4, CCL5, CCL-11, CCL12, CCL17, chemokine (C–X–C motif) ligand 1 (CXCL1), CXCL2, CXCL9, CXCL10, CXCL11, CXCL12, CXCL13, granulocyte colony-stimulating factor (G-CSF), granulocyte macrophage colony-stimulating factor (GM-CSF), intracellular adhesion molecule 1 (ICAM-1), interferon gamma (IFN-γ), interleukin-1α (IL-1α), IL-1β, IL-1ra, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-10, IL-12p70, IL-13, IL-16, IL-17, IL-23, IL-27, macrophage colony-stimulating factor (M-CSF), tissue inhibitor of metalloproteinase 1 (TIMP-1), tumor necrosis factor alpha (TNF-α), and triggering receptor expressed on myeloid cells 1 (TREM-1).

Statistical analysis

Parametric data was analyzed by one-way analysis of variance (ANOVA) or linear mixed model analysis using the Statistical Package for Social Sciences (SPSS: version 11.5; SPSS Inc., IL, USA). Data were analyzed using the information-theoretic approach, using the Akaike information criterion (AIC) model selection process and linear regression with Bonferroni adjustment for multiple analyses. All data are presented as mean ± standard error of the mean (SEM). Significance was set at a threshold of p < 0.05.

Results

In this study, we investigated obesity-driven inflammatory and metabolic abnormalities in an Aβ-infusion model of β-amyloidosis using PET. Obesity and hyperglycemia were evident in the high-fat fed mice after 1 month of feeding, accompanied by significantly increased abdominal and subcutaneous fat deposition and impaired glucose tolerance at sacrifice (Fig. 2bd). Mice were infused with either vehicle or oligomeric human Aβ42 1 month after feeding initiation, once obesity was already evident in high-fat fed groups (Fig. 2a). In vivo TSPO signals were assessed at baseline, 2, and 3 months after feeding initiation via 11C-PBR-28 PET, while cerebral metabolism was assessed 2.5 months after feeding initiation via 18F-FDG PET (Fig. 2a). Learning and memory and anxiety-related behaviors were assessed 3 months after feeding initiation, prior to the final 11C-PBR-28 PET scan (Fig. 2a).

Longitudinal in vivo PET imaging of obesity and Aβ-triggered inflammation using 11C-PBR-28

To examine the effect of obesity and β-amyloidosis on TSPO inflammatory signals, 11C-PBR-28 uptake was compared at baseline, 2-, and 3-month treatment time points (Fig. 3). Obesity can alter the whole body distribution of 11C-PBR-28; therefore, the brain uptake expressed as %ID/g may result in an underestimation of TSPO signals in obese groups (Fig. 3a). Aβ significantly increased 11C-PBR-28 signals at the infusion site and extending into the dorsal hippocampus in both control and obese groups at the 2-month treatment time point (F = 10.94; p = 0.001; Fig. 3b–e). At 3 months, increased 11C-PBR-28 signals persisted in obese but not in control mice infused with Aβ (F = 3.80, p = 0.03; Fig. 3b, e).
In vivo hippocampal TSPO signals were confirmed in tissues by immunohistochemistry (Fig. 4). Significantly increased TSPO immunoreactivity was observed in the hippocampus of Aβ-infused obese mice (F = 4.51, p = 0.007; Fig. 4b), and TSPO immunoreactivity correlated with in vivo hippocampal 11C-PBR-28 signals (R = 0.74, P < 0.001; Fig. 4c).

Obesity and Aβ synergistically increase hippocampal gliosis

To assess whether 11C-PBR-28 PET signals at 3 months were associated with gliosis, IBA-1 and GFAP levels were assessed by immunohistochemistry in the hippocampus (Fig. 5a). Obesity combined with Aβ infusion significantly increased IBA-1 immunoreactivity relative to vehicle-treated control mice (F = 2.96, p = 0.05; Fig. 5b) and increased GFAP immunoreactivity compared to all other groups (F = 5.13, p = 0.004; Fig. 5c). Hippocampal 11C-PBR-28 PET signals significantly correlated with hippocampal IBA-1 (r = 0.45, p = 0.05; Fig. 5d) and GFAP immunoreactivity (r = 0.62, p = 0.005; Fig. 5e).
To characterize the association between inflammatory state and 11C-PBR-28 PET signals, the cytokine expression profile in both the serum and hippocampus was evaluated by an antibody array (Fig. 5f). Correlation and multiple regression analyses were conducted to examine the relationship between 11C-PBR-28 PET signals (%ID/g) in the hippocampus measured at the final time point and potential inflammatory predictors (Table 1). Serum levels of M-CSF and IL-2 and hippocampal levels of CCL5, CCL3, IL-1ra, CCL2, CXCL9, and TIMP-1 significantly positively correlated with 11C-PBR-28 PET signals, while hippocampal CCL17 and IL-6 significantly negatively correlated. Hippocampal 11C-PBR-28 PET signals were significantly predicted by both serum and hippocampal CXCL12 and CXCL13; serum IL-13, IL-1β, CXCL1, CXCL2, CCL3, CCL5, and TIMP-1; and hippocampal G-CSF, IL-1ra, IL-6, and ICAM-1 as assessed by multiple linear regression analysis, with the predictor variables explaining greater than 99 % of 11C-PBR-28 PET signal variability (F = 8014.77, p < 0.001, R 2 = 0.999; Table 1).
Table 1
Multivariate linear regression analysis of serum and hippocampal cytokines associated with in vivo hippocampal 11C-PBR-28 uptake assessed by PET
  
Cytokine
11C-PBR-28 correlation
Multiple regression weights
b
β
Serum
Predictors
TIMP-1
0.075
0.0011**
2.3454
CXCL1
−0.129
−0.0008**
−1.9194
CXCL2
0.25
0.0040**
1.6419
CCL3
−0.057
−0.0026**
−1.5286
CCL5
0.038
0.0023**
0.8512
IL-1β
0.053
−0.0012**
−0.6740
IL-13
0.281
0.0002**
0.5435
CXCL12
−0.099
0.0002**
0.3181
CXCL13
0.137
−0.0003**
−0.2792
Correlates
M-CSF
0.650**
  
IL-2
0.537*
  
Hippocampus
Predictors
CXCL13
0.045
−0.0024**
−2.923
G-CSF
0.069
0.0074**
2.159
IL-1ra
0.641*
0.0072**
1.550
IL-6
−0.531*
−0.0001**
−0.518
CXCL12
0.389
−0.0008**
−0.625
sICAM-1
−0.245
−0.0008*
−0.474
Correlates
CCL17
−0.838***
  
CCL3
0.691**
  
CCL2
0.637*
  
CXCL9
0.634*
  
TIMP-1
0.522*
  
CCL5
0.553*
  
Abbreviations: b unstandardized coefficients, β standardized coefficients, TIMP-1 tissue inhibitor of metalloproteinases, M-CSF macrophage colony-stimulating factor, IL-2 interleukin 2, G-CSF granulocyte colony-stimulating factor, IL-ra interleukin 1 receptor antagonist, IL-6 interleukin 6, ICAM-1 intracellular adhesion molecule 1
*p < 0.05; **p < 0.01; ***p < 0.001

PET imaging of obesity and Aβ-induced effects on cerebral metabolism using 18F-FDG PET

To examine the effect of obesity and β-amyloidosis on cerebral metabolism, 18F-FDG uptake was examined at the 2.5-month treatment time point (Fig. 6). A significant increase in 18F-FDG uptake in the hippocampus but not the cerebellum was observed in Aβ-infused obese mice (hippocampus: F = 3.99; p = 0.02; cerebellum: F = 2.23, p = 0.12; Fig. 6). Hippocampal 18F-FDG SUVs were not significantly associated with markers of adiposity or glucose metabolism (data not shown).

11C-PBR-28 and 18F-FDG PET signals predict learning and memory impairment

At the 3-month treatment time point, the effect of obesity and Aβ infusion on anxiety-related behavior and learning and memory performance was assessed in the EPM and the object recognition tasks, respectively. Obesity significantly increased in anxiety-related behavior, resulting in increased latency to enter the open arm compared to the control mice, while obesity combined with Aβ-infusion increased anxiety-related behavior compared to both vehicle and Aβ-infused control groups (F = 11.54, p < 0.001; Fig. 7a). Treatment did not affect locomotor activity, with no significant effect of treatment on total distance moved in the EPM (F = 0.11, p = 0.96). Although a significant effect of treatment was observed on object recognition performance (F = 10.32, p = 0.02), no significant group differences were observed by post hoc analysis, with the effect of obesity combined with Aβ infusion approaching significance (p = 0.06; Fig. 7b). Interestingly, 18F-FDG SUVs significantly correlated with anxiety-related behavior in the elevated plus maze (r = 0.48, p = 0.02) and negatively correlated with learning and memory in the object recognition task (r = −0.46, p = 0.02; Fig. 7c). Hippocampal 18F-FDG and 11C-PBR-28 PET signals at 3 months significantly predicted learning and memory performance but not anxiety-related behavior as assessed by multiple linear regression analysis, with the predictor variables explaining approximately 40 % of the variability in object recognition performance (F = 6.06, p = 0.009, R 2 = 0.39). Both 18F-FDG (B = −19.55, β = −0.62, p = 0.005) and 11C-PBR-28 (B = −17.59, β = −0.45, p = 0.03) significantly contributed to the model.

Discussion

Using 11C-PBR-28-PET, we detected elevated TSPO associated with activated astrocytes and microglia resulting from Aβ-induced inflammation. The Aβ-induced inflammatory response was transient, with TSPO signals no longer significantly elevated at the completion of the experiment. However, the magnitude and duration of the Aβ-induced inflammatory response was exacerbated by obesity, even though obesity alone did not increase TSPO inflammatory signals. Previous studies using TSPO as a marker of inflammation in transgenic mouse models of β-amyloidosis have also reported elevated TSPO signals detected by PET, although only in aged mice at late stages of neuropathology [17, 34, 35]. What remains unclear is to what extent TSPO expression represents deleterious versus trophic glial responses. While many studies have focused on TSPO as a marker of deleterious gliosis, reporting good correlations between microglial markers and TSPO PET signals [36], some evidence suggests that TSPO-positive glial activation may be associated with trophic glial functions including Aβ clearance [37]. To address this, in the current study, the systemic and hippocampal cytokine profile was examined to determine which specific inflammatory factors were associated with in vivo TSPO signals. Serum levels of numerous obesity-linked chemokines promoting adipocyte proliferation (TIMP-1 [38]) and adipose macrophage infiltration (CXCL1 [39, 40], CXCL2 [41], CXCL12 [42], CCL3 [43], and CCL5 [44]) were found to be significant predictors of in vivo hippocampal TSPO signals. Interestingly, in the hippocampus, levels of G-CSF [45, 46], ICAM-1 [47], and CXCL12 [48] and IL-1ra [49], which have been identified as inflammatory factors important in the regulation of amyloid clearance, significantly predicted in vivo hippocampal TSPO signals. Identification of the TPSO-associated inflammatory profile is necessary in order to understand the specific glial functions associated with TSPO signals, which is essential for the validation of TSPO as an effective biomarker of β-amyloidosis in AD.
In contrast to clinical observations in advanced AD, in the current study, β-amyloidosis and cognitive impairment were associated with increased cerebral metabolism as assessed by 18F-FDG PET. Numerous other studies have also associated hypermetabolism with early stages of amyloid pathology in transgenic mouse models of β-amyloidosis [2831]. Luo et al. reported hypermetabolism in young Tg2576 mice, prior to the development of plaques [29]. Previous studies have suggested hypermetabolism may result from increased gliosis in the absence of neuronal loss; however, we observed poor overlap between regions of hyperactivity and TSPO signals and no association between cerebral metabolism and markers of gliosis. Our findings suggest that hypermetabolic changes cannot be explained by gliosis alone. Multiple metabolic disturbances like altered glucose uptake and glycolytic metabolism has been mechanistically implicated in β-amyloidosis [50] and Aβ-induced astrocytic and neuronal hyperactivity and seizure activity [5154]. In very young AD mutation carriers, approximately 25 years before the expected onset of symptoms, region-specific hypermetabolism is observed prior to the development of hypometabolism [24]. Likewise, amyloid deposition has been found to be associated with hypermetabolism in normal aging and MCI [2527]. Functional MRI (fMRI) studies have also demonstrated increased brain activation in normal controls with amyloid deposition [55], in MCI [5660] and in subjects at genetic risk for AD [6163]. Cerebral hypermetabolism may be induced in early stages of β-amyloidosis, reflecting Aβ-induced pathogenic mechanisms including glial and neuronal hyperactivity, preceding neuronal loss.
In contrast to our previous findings [13], in the current study, obesity alone did not result in a significant deficit in learning and memory performance in the object recognition task. The lack of any deficit in obese mice in the current study is likely a consequence of poor performance of vehicle-treated control mice, which exhibited around 60 % recognition. From our previous studies, we expect the performance of control mice to be around 75 % in this task. We hypothesize that the reduced performance observed in control mice in the current study may be the consequence of repeated interventions including the pump and cannula implantation surgery and imaging experiments, with the stress on the procedures potentially impairing cognitive performance. This reduced the overall sensitivity of the object recognition test, making it difficult to detect impairment resulting from the experimental manipulations.
Interestingly, we did not observe any significant effect of obesity alone on markers of neuroinflammation, cerebral metabolism, or cognitive function. Findings from previous studies are mixed, with some studies reporting increased cortical or hippocampal markers of inflammation in high-fat fed, obese mice [6466]; others report modest region-specific effects or no detectable effect on the hippocampus [67, 68]. These differences may reflect differences in the severity of metabolic symptoms between mouse strains, diet composition (e.g., cholesterol and caloric and macronutrient content), and duration of treatment exposure. However, this may not be detectable by PET due to methodological issues.
In the current study, obesity induced some limitations including not only logistical and image quality issues due to scatter and attenuation but also in validation of analysis models where large differences in body weight affect radioligand distribution throughout the body. SUV has been widely validated as an outcome measure for 18F-FDG uptake adjusting for differences in body weight. However, hyperglycemia is associated with reduced cerebral 18F-FDG uptake due to increased competition to cross the blood-brain barrier, likely resulting in an underestimation of cerebral metabolism in obese groups [6971]. In the case of 11C-PBR-28, systemic inflammation and increased TSPO binding in peripheral tissues of obese animals [72] may also have significant impacts on brain uptake. These factors are likely contributors to the reduction in in vivo brain 11C-PBR-28 signals observed in obese mice; however, good agreement was observed between in vivo TSPO signals and immunoreactivity, validating the %ID/g for quantifying TSPO in living brains.

Conclusions

Obesity combined with Aβ infusion was found to increase both TSPO-PET signals and cerebral glucose metabolism. These signals can predict learning and memory performance. In vivo TSPO signals were confirmed by ex vivo analyses and correlated with inflammatory markers of hippocampal gliosis and serum levels of cytokine and chemokines. In contrast, since hypermetabolic regions did not directly correspond to regions of increased TSPO signals, it may reflect early pathogenic effects of Aβ toxicity on neurons rather than inflammation. These findings demonstrate the potential usefulness of TSPO and cerebral glucose metabolism for the investigation of interactions between midlife lifestyle factors and Aβ amyloidosis with the aid of neuroimaging.

Acknowledgements

The authors thank M. Ono, T. Minamihisamatsu, S. Sasaki and S. Uchida for technical support and assistance (National Institute of Radiological Sciences, NIRS), A. S. J. Wyatt for assistance with statistical analyses (University of Tokyo), and S. Chie for critical feedback on the manuscript (NIRS).

Funding

This work is supported by grants from Grants-in-Aid for Core Research for Evolutional Science and Technology (14533254), the Brain Mapping by Integrated Neurotechnologies for Disease Studies (Brain/MINDS; 15653129), the Research and Development Grants for Dementia (16768966) to M. H. from the Japan Agency for Medical Research and Development, and the Japan Advanced Molecular Imaging Program and Scientific Research on Innovative Areas (“Brain Environment” 23111009 to M. H.) from the Ministry of Education, Culture, Sports, Science and Technology, Japan.

Availability of data and materials

There are no data, software, databases, or application/tool apart from that reported in the study. Data supporting the conclusions are presented in the current manuscript.

Authors’ contributions

Experiments were designed and conceived by AMB, MT, and MH. Data was collected by AMB, MT, MRZ, and BJ. Data was interpreted by all authors. The manuscript was written by AMB, ST, and MH, and all authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
All experimentation was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health and was approved by the Institutional Animal Care and Use Committee of the National Institutes for Quantum and Radiological Science and Technology, Japan. Consent to participate is not applicable.
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
1.
Zurück zum Zitat Organization WH: Obesity: preventing and managing the global epidemic. In WHO Technical Report Series. 2000; p. 252. Organization WH: Obesity: preventing and managing the global epidemic. In WHO Technical Report Series. 2000; p. 252.
2.
Zurück zum Zitat Xu WL, Atti AR, Gatz M, Pedersen NL, Johansson B, Fratiglioni L. Midlife overweight and obesity increase late-life dementia risk: a population-based twin study. Neurology. 2011;76:1568.CrossRefPubMedPubMedCentral Xu WL, Atti AR, Gatz M, Pedersen NL, Johansson B, Fratiglioni L. Midlife overweight and obesity increase late-life dementia risk: a population-based twin study. Neurology. 2011;76:1568.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Loef M, Walach H. Midlife obesity and dementia: meta-analysis and adjusted forecast of dementia prevalence in the United States and China. Obesity. 2013;21:E51.CrossRefPubMed Loef M, Walach H. Midlife obesity and dementia: meta-analysis and adjusted forecast of dementia prevalence in the United States and China. Obesity. 2013;21:E51.CrossRefPubMed
4.
Zurück zum Zitat Smith E, Hay P, Campbell L, Trollor JN. A review of the association between obesity and cognitive function across the lifespan: implications for novel approaches to prevention and treatment. Obesity Rev. 2011;12:740.CrossRef Smith E, Hay P, Campbell L, Trollor JN. A review of the association between obesity and cognitive function across the lifespan: implications for novel approaches to prevention and treatment. Obesity Rev. 2011;12:740.CrossRef
5.
Zurück zum Zitat Thal DR, Rub U, Orantes M, Braak H. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58:1791.CrossRefPubMed Thal DR, Rub U, Orantes M, Braak H. Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology. 2002;58:1791.CrossRefPubMed
6.
Zurück zum Zitat Ritchie K, Ritchie CW, Yaffe K, Skoog I, Scarmeas N. Is late-onset Alzheimer’s disease really a disease of midlife? Alzheimers Dement. 2015;1:122. Ritchie K, Ritchie CW, Yaffe K, Skoog I, Scarmeas N. Is late-onset Alzheimer’s disease really a disease of midlife? Alzheimers Dement. 2015;1:122.
8.
Zurück zum Zitat Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Green RC, Harvey D, Jack CR, Jagust W, Liu E, et al. The Alzheimer’s Disease Neuroimaging Initiative: a review of papers published since its inception. Alzheimers Dement. 2012;8:S1.CrossRefPubMed Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Green RC, Harvey D, Jack CR, Jagust W, Liu E, et al. The Alzheimer’s Disease Neuroimaging Initiative: a review of papers published since its inception. Alzheimers Dement. 2012;8:S1.CrossRefPubMed
9.
Zurück zum Zitat Ho L, Qin W, Pompl PN, Xiang Z, Wang J, Zhao Z, Peng Y, Cambareri G, Rocher A, Mobbs CV, et al. Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s disease. FASEB J. 2004. doi:10.1096/fj.03-0978fje. Ho L, Qin W, Pompl PN, Xiang Z, Wang J, Zhao Z, Peng Y, Cambareri G, Rocher A, Mobbs CV, et al. Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s disease. FASEB J. 2004. doi:10.​1096/​fj.​03-0978fje.
10.
Zurück zum Zitat Julien C, Tremblay C, Phivilay A, Berthiaume L, Émond V, Julien P, Calon F. High-fat diet aggravates amyloid-beta and tau pathologies in the 3xTg-AD mouse model. Neurobiol Aging. 2010;31:1516.CrossRefPubMed Julien C, Tremblay C, Phivilay A, Berthiaume L, Émond V, Julien P, Calon F. High-fat diet aggravates amyloid-beta and tau pathologies in the 3xTg-AD mouse model. Neurobiol Aging. 2010;31:1516.CrossRefPubMed
11.
Zurück zum Zitat Li L, Cao D, Garber DW, Kim H, Fukuchi K-I. Association of aortic atherosclerosis with cerebral β-amyloidosis and learning deficits in a mouse model of Alzheimer’s disease. Am J Pathol. 2003;163:2155.CrossRefPubMedPubMedCentral Li L, Cao D, Garber DW, Kim H, Fukuchi K-I. Association of aortic atherosclerosis with cerebral β-amyloidosis and learning deficits in a mouse model of Alzheimer’s disease. Am J Pathol. 2003;163:2155.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Levin-Allerhand JA, Lominska CE, Smith JD. Increased amyloid-β levels in APPSWE transgenic mice treated chronically with a physiological high-fat high-cholesterol diet. J Nutr Health Aging. 2002;6:315.PubMed Levin-Allerhand JA, Lominska CE, Smith JD. Increased amyloid-β levels in APPSWE transgenic mice treated chronically with a physiological high-fat high-cholesterol diet. J Nutr Health Aging. 2002;6:315.PubMed
13.
Zurück zum Zitat Barron AM, Rosario ER, Elteriefi R, Pike CJ. Sex-specific effects of high fat diet on indices of metabolic syndrome in 3xTg-AD mice: implications for Alzheimer’s disease. PLoS One. 2013;8:e78554.CrossRefPubMedPubMedCentral Barron AM, Rosario ER, Elteriefi R, Pike CJ. Sex-specific effects of high fat diet on indices of metabolic syndrome in 3xTg-AD mice: implications for Alzheimer’s disease. PLoS One. 2013;8:e78554.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Heppner FL, Ransohoff RM, Becher B. Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci. 2015;16:358.CrossRefPubMed Heppner FL, Ransohoff RM, Becher B. Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci. 2015;16:358.CrossRefPubMed
15.
Zurück zum Zitat Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388.CrossRefPubMed Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, Jacobs AH, Wyss-Coray T, Vitorica J, Ransohoff RM, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388.CrossRefPubMed
16.
Zurück zum Zitat Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, Maeda J, Suhara T, Trojanowski JQ, Lee VM. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53:337.CrossRefPubMed Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, Maeda J, Suhara T, Trojanowski JQ, Lee VM. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53:337.CrossRefPubMed
17.
Zurück zum Zitat Ji B, Maeda J, Sawada M, Ono M, Okauchi T, Inaji M, Zhang M-R, Suzuki K, Ando K, Staufenbiel M, et al. Imaging of peripheral benzodiazepine receptor expression as biomarkers of detrimental versus beneficial glial responses in mouse models of Alzheimer’s and other CNS pathologies. J Neurosci. 2008;28:12255.CrossRefPubMedPubMedCentral Ji B, Maeda J, Sawada M, Ono M, Okauchi T, Inaji M, Zhang M-R, Suzuki K, Ando K, Staufenbiel M, et al. Imaging of peripheral benzodiazepine receptor expression as biomarkers of detrimental versus beneficial glial responses in mouse models of Alzheimer’s and other CNS pathologies. J Neurosci. 2008;28:12255.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Briard E, Zoghbi SS, Imaizumi M, Gourley JP, Shetty HU, Hong J, Cropley V, Fujita M, Innis RB, Pike VW. Synthesis and evaluation in monkey of two sensitive 11C-labeled aryloxyanilide ligands for imaging brain peripheral benzodiazepine receptors in vivo. J Med Chem. 2008;51:17.CrossRefPubMed Briard E, Zoghbi SS, Imaizumi M, Gourley JP, Shetty HU, Hong J, Cropley V, Fujita M, Innis RB, Pike VW. Synthesis and evaluation in monkey of two sensitive 11C-labeled aryloxyanilide ligands for imaging brain peripheral benzodiazepine receptors in vivo. J Med Chem. 2008;51:17.CrossRefPubMed
19.
Zurück zum Zitat Kreisl W, Lyoo CH, Wei M, Snow J, Jenko K, Morse C, Zoghbi S, Pike V, Turner R, Innis R. Serial imaging with [11C]PBR28 suggests increased neuroinflammation during the progression of Alzheimer’s disease. Neurology. 2015;84:6. Kreisl W, Lyoo CH, Wei M, Snow J, Jenko K, Morse C, Zoghbi S, Pike V, Turner R, Innis R. Serial imaging with [11C]PBR28 suggests increased neuroinflammation during the progression of Alzheimer’s disease. Neurology. 2015;84:6.
20.
Zurück zum Zitat Park E, Gallezot J-D, Delgadillo A, Liu S, Planeta B, Lin S-F, O’Connor K, Lim K, Lee J-Y, Chastre A, et al. 11C-PBR28 imaging in multiple sclerosis patients and healthy controls: test-retest reproducibility and focal visualization of active white matter areas. Eur J Nucl Med Mol Imaging. 2015;42:1081.CrossRefPubMed Park E, Gallezot J-D, Delgadillo A, Liu S, Planeta B, Lin S-F, O’Connor K, Lim K, Lee J-Y, Chastre A, et al. 11C-PBR28 imaging in multiple sclerosis patients and healthy controls: test-retest reproducibility and focal visualization of active white matter areas. Eur J Nucl Med Mol Imaging. 2015;42:1081.CrossRefPubMed
21.
Zurück zum Zitat Walker MD, Dinelle K, Kornelsen R, Lee NV, Miao Q, Adam M, Takhar C, Mak E, Schulzer M, Farrer MJ, Sossi V. [11C]PBR28 PET imaging is sensitive to neuroinflammation in the aged rat. J Cereb Blood Flow Metab. 2015;35:1331.CrossRefPubMedPubMedCentral Walker MD, Dinelle K, Kornelsen R, Lee NV, Miao Q, Adam M, Takhar C, Mak E, Schulzer M, Farrer MJ, Sossi V. [11C]PBR28 PET imaging is sensitive to neuroinflammation in the aged rat. J Cereb Blood Flow Metab. 2015;35:1331.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Imaizumi M, Kim HJ, Zoghbi SS, Briard E, Hong J, Musachio JL, Ruetzler C, Chuang DM, Pike VW, Innis RB, Fujita M. PET imaging with [11C]PBR28 can localize and quantify upregulated peripheral benzodiazepine receptors associated with cerebral ischemia in rat. Neurosci Lett. 2007;411:200.CrossRefPubMed Imaizumi M, Kim HJ, Zoghbi SS, Briard E, Hong J, Musachio JL, Ruetzler C, Chuang DM, Pike VW, Innis RB, Fujita M. PET imaging with [11C]PBR28 can localize and quantify upregulated peripheral benzodiazepine receptors associated with cerebral ischemia in rat. Neurosci Lett. 2007;411:200.CrossRefPubMed
23.
Zurück zum Zitat Rocher AB, Chapon F, Blaizot X, Baron JC, Chavoix C. Resting-state brain glucose utilization as measured by PET is directly related to regional synaptophysin levels: a study in baboons. NeuroImage. 2003;20:1894.CrossRefPubMed Rocher AB, Chapon F, Blaizot X, Baron JC, Chavoix C. Resting-state brain glucose utilization as measured by PET is directly related to regional synaptophysin levels: a study in baboons. NeuroImage. 2003;20:1894.CrossRefPubMed
24.
Zurück zum Zitat Benzinger TL, Blazey T, Jack Jr CR, Koeppe RA, Su Y, Xiong C, Raichle ME, Snyder AZ, Ances BM, Bateman RJ, et al. Regional variability of imaging biomarkers in autosomal dominant Alzheimer’s disease. PNAS. 2013;110:E4502.CrossRefPubMedPubMedCentral Benzinger TL, Blazey T, Jack Jr CR, Koeppe RA, Su Y, Xiong C, Raichle ME, Snyder AZ, Ances BM, Bateman RJ, et al. Regional variability of imaging biomarkers in autosomal dominant Alzheimer’s disease. PNAS. 2013;110:E4502.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Oh H, Habeck C, Madison C, Jagust W. Covarying alterations in Aβ deposition, glucose metabolism, and gray matter volume in cognitively normal elderly. Human Brain Map. 2014;35:297.CrossRef Oh H, Habeck C, Madison C, Jagust W. Covarying alterations in Aβ deposition, glucose metabolism, and gray matter volume in cognitively normal elderly. Human Brain Map. 2014;35:297.CrossRef
26.
Zurück zum Zitat Ashraf A, Fan Z, Brooks DJ, Edison P. Cortical hypermetabolism in MCI subjects: a compensatory mechanism? Eur J Nucl Med Mol Imaging. 2015;42:447.CrossRefPubMed Ashraf A, Fan Z, Brooks DJ, Edison P. Cortical hypermetabolism in MCI subjects: a compensatory mechanism? Eur J Nucl Med Mol Imaging. 2015;42:447.CrossRefPubMed
27.
Zurück zum Zitat Cohen AD, Price JC, Weissfeld LA, James J, Rosario BL, Bi W, Nebes RD, Saxton JA, Snitz BE, Aizenstein HA, et al. Basal cerebral metabolism may modulate the cognitive effects of Aβ in mild cognitive impairment: an example of brain reserve. J Neurosci. 2009;29:14770.CrossRefPubMedPubMedCentral Cohen AD, Price JC, Weissfeld LA, James J, Rosario BL, Bi W, Nebes RD, Saxton JA, Snitz BE, Aizenstein HA, et al. Basal cerebral metabolism may modulate the cognitive effects of Aβ in mild cognitive impairment: an example of brain reserve. J Neurosci. 2009;29:14770.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Sancheti H, Patil I, Kanamori K, Diaz Brinton R, Zhang W, Lin A-L, Cadenas E. Hypermetabolic state in the 7-month-old triple transgenic mouse model of Alzheimer’s disease and the effect of lipoic acid: a 13C-NMR study. J Cereb Blood Flow Metab. 2014;34:1749.CrossRefPubMedPubMedCentral Sancheti H, Patil I, Kanamori K, Diaz Brinton R, Zhang W, Lin A-L, Cadenas E. Hypermetabolic state in the 7-month-old triple transgenic mouse model of Alzheimer’s disease and the effect of lipoic acid: a 13C-NMR study. J Cereb Blood Flow Metab. 2014;34:1749.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Luo F, Rustay NR, Ebert U, Hradil VP, Cole TB, Llano DA, Mudd SR, Zhang Y, Fox GB, Day M. Characterization of 7- and 19-month-old Tg2576 mice using multimodal in vivo imaging: limitations as a translatable model of Alzheimer’s disease. Neurobiol Aging. 2012;33:933.CrossRefPubMed Luo F, Rustay NR, Ebert U, Hradil VP, Cole TB, Llano DA, Mudd SR, Zhang Y, Fox GB, Day M. Characterization of 7- and 19-month-old Tg2576 mice using multimodal in vivo imaging: limitations as a translatable model of Alzheimer’s disease. Neurobiol Aging. 2012;33:933.CrossRefPubMed
30.
Zurück zum Zitat Rojas S, Herance JR, Gispert JD, Abad S, Torrent E, Jimenez X, Pareto D, Perpina U, Sarroca S, Rodriguez E, et al. In vivo evaluation of amyloid deposition and brain glucose metabolism of 5XFAD mice using positron emission tomography. Neurobiol Aging. 2013;34:1790.CrossRefPubMed Rojas S, Herance JR, Gispert JD, Abad S, Torrent E, Jimenez X, Pareto D, Perpina U, Sarroca S, Rodriguez E, et al. In vivo evaluation of amyloid deposition and brain glucose metabolism of 5XFAD mice using positron emission tomography. Neurobiol Aging. 2013;34:1790.CrossRefPubMed
31.
Zurück zum Zitat Poisnel G, Herard AS, El Tannir El Tayara N, Bourrin E, Volk A, Kober F, Delatour B, Delzescaux T, Debeir T, Rooney T, et al. Increased regional cerebral glucose uptake in an APP/PS1 model of Alzheimer’s disease. Neurobiol Aging. 2012;33:1995.CrossRefPubMed Poisnel G, Herard AS, El Tannir El Tayara N, Bourrin E, Volk A, Kober F, Delatour B, Delzescaux T, Debeir T, Rooney T, et al. Increased regional cerebral glucose uptake in an APP/PS1 model of Alzheimer’s disease. Neurobiol Aging. 2012;33:1995.CrossRefPubMed
32.
Zurück zum Zitat Craft JM, Watterson DM, Frautschy SA, Van Eldik LJ. Aminopyridazines inhibit beta-amyloid-induced glial activation and neuronal damage in vivo. Neurobiol Aging. 2004;25:1283.CrossRefPubMed Craft JM, Watterson DM, Frautschy SA, Van Eldik LJ. Aminopyridazines inhibit beta-amyloid-induced glial activation and neuronal damage in vivo. Neurobiol Aging. 2004;25:1283.CrossRefPubMed
33.
Zurück zum Zitat Wang M, Yoder KK, Gao M, Mock BH, Xu XM, Saykin AJ, Hutchins GD, Zheng QH. Fully automated synthesis and initial PET evaluation of [11C]PBR28. Bioorg Med Chem Lett. 2009;19:5636.CrossRefPubMedPubMedCentral Wang M, Yoder KK, Gao M, Mock BH, Xu XM, Saykin AJ, Hutchins GD, Zheng QH. Fully automated synthesis and initial PET evaluation of [11C]PBR28. Bioorg Med Chem Lett. 2009;19:5636.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Maeda J, Zhang MR, Okauchi T, Ji B, Ono M, Hattori S, Kumata K, Iwata N, Saido TC, Trojanowski JQ, et al. In vivo positron emission tomographic imaging of glial responses to amyloid-beta and tau pathologies in mouse models of Alzheimer’s disease and related disorders. J Neurosci. 2011;31:4720.CrossRefPubMedPubMedCentral Maeda J, Zhang MR, Okauchi T, Ji B, Ono M, Hattori S, Kumata K, Iwata N, Saido TC, Trojanowski JQ, et al. In vivo positron emission tomographic imaging of glial responses to amyloid-beta and tau pathologies in mouse models of Alzheimer’s disease and related disorders. J Neurosci. 2011;31:4720.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat James ML, Belichenko NP, Nguyen TV, Andrews LE, Ding Z, Liu H, Bodapati D, Arksey N, Shen B, Cheng Z, et al. PET imaging of translocator protein (18 kDa) in a mouse model of Alzheimer’s disease using N-(2,5-dimethoxybenzyl)-2-18 F-fluoro-N-(2-phenoxyphenyl) acetamide. J Nucl Med. 2015;56:311.CrossRefPubMedPubMedCentral James ML, Belichenko NP, Nguyen TV, Andrews LE, Ding Z, Liu H, Bodapati D, Arksey N, Shen B, Cheng Z, et al. PET imaging of translocator protein (18 kDa) in a mouse model of Alzheimer’s disease using N-(2,5-dimethoxybenzyl)-2-18 F-fluoro-N-(2-phenoxyphenyl) acetamide. J Nucl Med. 2015;56:311.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Venneti S, Lopresti BJ, Wang G, Hamilton RL, Mathis CA, Klunk WE, Apte UM, Wiley CA. PK11195 labels activated microglia in Alzheimer’s disease and in vivo in a mouse model using PET. Neurobiol Aging. 2009;30:1217.CrossRefPubMed Venneti S, Lopresti BJ, Wang G, Hamilton RL, Mathis CA, Klunk WE, Apte UM, Wiley CA. PK11195 labels activated microglia in Alzheimer’s disease and in vivo in a mouse model using PET. Neurobiol Aging. 2009;30:1217.CrossRefPubMed
37.
Zurück zum Zitat Maeda J, Ji B, Irie T, Tomiyama T, Maruyama M, Okauchi T, Staufenbiel M, Iwata N, Ono M, Saido TC, et al. Longitudinal, quantitative assessment of amyloid, neuroinflammation, and anti-amyloid treatment in a living mouse model of Alzheimer’s disease enabled by positron emission tomography. J Neurosci. 2007;27:10957.CrossRefPubMed Maeda J, Ji B, Irie T, Tomiyama T, Maruyama M, Okauchi T, Staufenbiel M, Iwata N, Ono M, Saido TC, et al. Longitudinal, quantitative assessment of amyloid, neuroinflammation, and anti-amyloid treatment in a living mouse model of Alzheimer’s disease enabled by positron emission tomography. J Neurosci. 2007;27:10957.CrossRefPubMed
38.
Zurück zum Zitat Meissburger B, Stachorski L, Roder E, Rudofsky G, Wolfrum C. Tissue inhibitor of matrix metalloproteinase 1 (TIMP1) controls adipogenesis in obesity in mice and in humans. Diabetologia. 2011;54:1468.CrossRefPubMed Meissburger B, Stachorski L, Roder E, Rudofsky G, Wolfrum C. Tissue inhibitor of matrix metalloproteinase 1 (TIMP1) controls adipogenesis in obesity in mice and in humans. Diabetologia. 2011;54:1468.CrossRefPubMed
39.
Zurück zum Zitat Neels JG, Badeanlou L, Hester KD, Samad F. Keratinocyte-derived chemokine in obesity: expression, regulation, and role in adipose macrophage infiltration and glucose homeostasis. J Biol Chem. 2009;284:20692.CrossRefPubMedPubMedCentral Neels JG, Badeanlou L, Hester KD, Samad F. Keratinocyte-derived chemokine in obesity: expression, regulation, and role in adipose macrophage infiltration and glucose homeostasis. J Biol Chem. 2009;284:20692.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Nunemaker CS, Chung HG, Verrilli GM, Corbin KL, Upadhye A, Sharma PR. Increased serum CXCL1 and CXCL5 are linked to obesity, hyperglycemia, and impaired islet function. J Endocrinol. 2014;222:267.CrossRefPubMedPubMedCentral Nunemaker CS, Chung HG, Verrilli GM, Corbin KL, Upadhye A, Sharma PR. Increased serum CXCL1 and CXCL5 are linked to obesity, hyperglycemia, and impaired islet function. J Endocrinol. 2014;222:267.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Rouault C, Pellegrinelli V, Schilch R, Cotillard A, Poitou C, Tordjman J, Sell H, Clement K, Lacasa D. Roles of chemokine ligand-2 (CXCL2) and neutrophils in influencing endothelial cell function and inflammation of human adipose tissue. Endocrinol. 2013;154:1069.CrossRef Rouault C, Pellegrinelli V, Schilch R, Cotillard A, Poitou C, Tordjman J, Sell H, Clement K, Lacasa D. Roles of chemokine ligand-2 (CXCL2) and neutrophils in influencing endothelial cell function and inflammation of human adipose tissue. Endocrinol. 2013;154:1069.CrossRef
42.
Zurück zum Zitat Kim D, Kim J, Yoon JH, Ghim J, Yea K, Song P, Park S, Lee A, Hong CP, Jang MS, et al. CXCL12 secreted from adipose tissue recruits macrophages and induces insulin resistance in mice. Diabetologia. 2014;57:1456.CrossRefPubMed Kim D, Kim J, Yoon JH, Ghim J, Yea K, Song P, Park S, Lee A, Hong CP, Jang MS, et al. CXCL12 secreted from adipose tissue recruits macrophages and induces insulin resistance in mice. Diabetologia. 2014;57:1456.CrossRefPubMed
43.
Zurück zum Zitat Huber J, Kiefer F, Zeyda M, Ludvik B, Silberhumer G, Prager G, Stulnig TM. CCL2 and CCL3 chemokine gene expression and their receptors in human visceral and subcutaneous adipose tissue is associated with obesity and insulin resistance. Exp Clin Endocrinol Diabetes. 2007;115:3215.CrossRef Huber J, Kiefer F, Zeyda M, Ludvik B, Silberhumer G, Prager G, Stulnig TM. CCL2 and CCL3 chemokine gene expression and their receptors in human visceral and subcutaneous adipose tissue is associated with obesity and insulin resistance. Exp Clin Endocrinol Diabetes. 2007;115:3215.CrossRef
44.
Zurück zum Zitat Keophiphath M, Rouault C, Divoux A, Clement K, Lacasa D. CCL5 promotes macrophage recruitment and survival in human adipose tissue. Arterioscler Thromb Vasc Biol. 2010;30:39.CrossRefPubMed Keophiphath M, Rouault C, Divoux A, Clement K, Lacasa D. CCL5 promotes macrophage recruitment and survival in human adipose tissue. Arterioscler Thromb Vasc Biol. 2010;30:39.CrossRefPubMed
45.
Zurück zum Zitat Doi Y, Takeuchi H, Mizoguchi H, Fukumoto K, Horiuchi H, Jin S, Kawanokuchi J, Parajuli B, Sonobe Y, Mizuno T, Suzumura A. Granulocyte-colony stimulating factor attenuates oligomeric amyloid beta neurotoxicity by activation of neprilysin. PLoS One. 2014;9:e103458.CrossRefPubMedCentral Doi Y, Takeuchi H, Mizoguchi H, Fukumoto K, Horiuchi H, Jin S, Kawanokuchi J, Parajuli B, Sonobe Y, Mizuno T, Suzumura A. Granulocyte-colony stimulating factor attenuates oligomeric amyloid beta neurotoxicity by activation of neprilysin. PLoS One. 2014;9:e103458.CrossRefPubMedCentral
46.
Zurück zum Zitat Tsai K-J, Tsai Y-C, Shen C-KJ. G-CSF rescues the memory impairment of animal models of Alzheimer’s disease. J Experimental Med. 2007;204:1273.CrossRef Tsai K-J, Tsai Y-C, Shen C-KJ. G-CSF rescues the memory impairment of animal models of Alzheimer’s disease. J Experimental Med. 2007;204:1273.CrossRef
47.
Zurück zum Zitat Kim JY, Kim DH, Kim JH, Lee D, Jeon HB, Kwon SJ, Kim SM, Yoo YJ, Lee EH, Choi SJ, et al. Soluble intracellular adhesion molecule-1 secreted by human umbilical cord blood-derived mesenchymal stem cell reduces amyloid-beta plaques. Cell Death Differ. 2012;19:680.CrossRefPubMed Kim JY, Kim DH, Kim JH, Lee D, Jeon HB, Kwon SJ, Kim SM, Yoo YJ, Lee EH, Choi SJ, et al. Soluble intracellular adhesion molecule-1 secreted by human umbilical cord blood-derived mesenchymal stem cell reduces amyloid-beta plaques. Cell Death Differ. 2012;19:680.CrossRefPubMed
48.
Zurück zum Zitat Wang Q, Xu Y, Chen JC, Qin YY, Liu M, Liu Y, Xie MJ, Yu ZY, Zhu Z, Wang W. Stromal cell-derived factor 1alpha decreases beta-amyloid deposition in Alzheimer’s disease mouse model. Brain Res. 2012;1459:15.CrossRefPubMed Wang Q, Xu Y, Chen JC, Qin YY, Liu M, Liu Y, Xie MJ, Yu ZY, Zhu Z, Wang W. Stromal cell-derived factor 1alpha decreases beta-amyloid deposition in Alzheimer’s disease mouse model. Brain Res. 2012;1459:15.CrossRefPubMed
49.
Zurück zum Zitat Craft JM, Watterson DM, Hirsch E, Van Eldik LJ. Interleukin 1 receptor antagonist knockout mice show enhanced microglial activation and neuronal damage induced by intracerebroventricular infusion of human beta-amyloid. J Neuroinflammation. 2005;2:15.CrossRefPubMedPubMedCentral Craft JM, Watterson DM, Hirsch E, Van Eldik LJ. Interleukin 1 receptor antagonist knockout mice show enhanced microglial activation and neuronal damage induced by intracerebroventricular infusion of human beta-amyloid. J Neuroinflammation. 2005;2:15.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Vlassenko AG, Vaishnavi SN, Couture L, Sacco D, Shannon BJ, Mach RH, Morris JC, Raichle ME, Mintun MA. Spatial correlation between brain aerobic glycolysis and amyloid-β (Aβ) deposition. PNAS. 2010;107:17763.CrossRefPubMedPubMedCentral Vlassenko AG, Vaishnavi SN, Couture L, Sacco D, Shannon BJ, Mach RH, Morris JC, Raichle ME, Mintun MA. Spatial correlation between brain aerobic glycolysis and amyloid-β (Aβ) deposition. PNAS. 2010;107:17763.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice. Science. 2009;323:1211.CrossRefPubMedPubMedCentral Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice. Science. 2009;323:1211.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Busche MA, Eichhoff G, Adelsberger H, Abramowski D, Wiederhold K-H, Haass C, Staufenbiel M, Konnerth A, Garaschuk O. Clusters of hyperactive neurons near amyloid plaques in a mouse model of Alzheimer’s disease. Science. 2008;321:1686.CrossRefPubMed Busche MA, Eichhoff G, Adelsberger H, Abramowski D, Wiederhold K-H, Haass C, Staufenbiel M, Konnerth A, Garaschuk O. Clusters of hyperactive neurons near amyloid plaques in a mouse model of Alzheimer’s disease. Science. 2008;321:1686.CrossRefPubMed
53.
Zurück zum Zitat Busche MA, Chen X, Henning HA, Reichwald J, Staufenbiel M, Sakmann B, Konnerth A. Critical role of soluble amyloid-β for early hippocampal hyperactivity in a mouse model of Alzheimer’s disease. PNAS. 2012;109:8740.CrossRefPubMedPubMedCentral Busche MA, Chen X, Henning HA, Reichwald J, Staufenbiel M, Sakmann B, Konnerth A. Critical role of soluble amyloid-β for early hippocampal hyperactivity in a mouse model of Alzheimer’s disease. PNAS. 2012;109:8740.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Palop JJ, Chin J, Roberson ED, Wang J, Thwin MT, Bien-Ly N, Yoo J, Ho KO, Yu GQ, Kreitzer A, et al. Aberrant excitatory neuronal activity and compensatory remodeling of inhibitory hippocampal circuits in mouse models of Alzheimer’s disease. Neuron. 2007;55:697.CrossRefPubMed Palop JJ, Chin J, Roberson ED, Wang J, Thwin MT, Bien-Ly N, Yoo J, Ho KO, Yu GQ, Kreitzer A, et al. Aberrant excitatory neuronal activity and compensatory remodeling of inhibitory hippocampal circuits in mouse models of Alzheimer’s disease. Neuron. 2007;55:697.CrossRefPubMed
55.
Zurück zum Zitat Sperling RA, LaViolette PS, O’Keefe K, O’Brien J, Rentz DM, Pihlajamaki M, Marshall G, Hyman BT, Selkoe DJ, Hedden T, et al. Amyloid deposition is associated with impaired default network function in older persons without dementia. Neuron. 2009;63:178.CrossRefPubMedPubMedCentral Sperling RA, LaViolette PS, O’Keefe K, O’Brien J, Rentz DM, Pihlajamaki M, Marshall G, Hyman BT, Selkoe DJ, Hedden T, et al. Amyloid deposition is associated with impaired default network function in older persons without dementia. Neuron. 2009;63:178.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Dickerson BC, Salat DH, Greve DN, Chua EF, Rand-Giovannetti E, Rentz DM, Bertram L, Mullin K, Tanzi RE, Blacker D, et al. Increased hippocampal activation in mild cognitive impairment compared to normal aging and AD. Neurology. 2005;65:404.CrossRefPubMedPubMedCentral Dickerson BC, Salat DH, Greve DN, Chua EF, Rand-Giovannetti E, Rentz DM, Bertram L, Mullin K, Tanzi RE, Blacker D, et al. Increased hippocampal activation in mild cognitive impairment compared to normal aging and AD. Neurology. 2005;65:404.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Celone KA, Calhoun VD, Dickerson BC, Atri A, Chua EF, Miller SL, DePeau K, Rentz DM, Selkoe DJ, Blacker D, et al. Alterations in memory networks in mild cognitive impairment and Alzheimer’s disease: an independent component analysis. J Neurosci. 2006;26:10222.CrossRefPubMed Celone KA, Calhoun VD, Dickerson BC, Atri A, Chua EF, Miller SL, DePeau K, Rentz DM, Selkoe DJ, Blacker D, et al. Alterations in memory networks in mild cognitive impairment and Alzheimer’s disease: an independent component analysis. J Neurosci. 2006;26:10222.CrossRefPubMed
58.
Zurück zum Zitat Kircher TT, Weis S, Freymann K, Erb M, Jessen F, Grodd W, Heun R, Leube DT. Hippocampal activation in patients with mild cognitive impairment is necessary for successful memory encoding. JNNP. 2007;78:812. Kircher TT, Weis S, Freymann K, Erb M, Jessen F, Grodd W, Heun R, Leube DT. Hippocampal activation in patients with mild cognitive impairment is necessary for successful memory encoding. JNNP. 2007;78:812.
59.
Zurück zum Zitat Miller SL, Fenstermacher E, Bates J, Blacker D, Sperling RA, Dickerson BC. Hippocampal activation in adults with mild cognitive impairment predicts subsequent cognitive decline. JNNP. 2008;79:630. Miller SL, Fenstermacher E, Bates J, Blacker D, Sperling RA, Dickerson BC. Hippocampal activation in adults with mild cognitive impairment predicts subsequent cognitive decline. JNNP. 2008;79:630.
60.
Zurück zum Zitat Yassa MA, Stark SM, Bakker A, Albert MS, Gallagher M, Stark CE. High-resolution structural and functional MRI of hippocampal CA3 and dentate gyrus in patients with amnestic mild cognitive impairment. NeuroImage. 2010;51:1242.CrossRefPubMedPubMedCentral Yassa MA, Stark SM, Bakker A, Albert MS, Gallagher M, Stark CE. High-resolution structural and functional MRI of hippocampal CA3 and dentate gyrus in patients with amnestic mild cognitive impairment. NeuroImage. 2010;51:1242.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Bondi MW, Houston WS, Eyler LT, Brown GG. fMRI evidence of compensatory mechanisms in older adults at genetic risk for Alzheimer disease. Neurology. 2005;64:501.CrossRefPubMedPubMedCentral Bondi MW, Houston WS, Eyler LT, Brown GG. fMRI evidence of compensatory mechanisms in older adults at genetic risk for Alzheimer disease. Neurology. 2005;64:501.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Bookheimer SY, Strojwas MH, Cohen MS, Saunders AM, Pericak-Vance MA, Mazziotta JC, Small GW. Patterns of brain activation in people at risk for Alzheimer’s disease. NEJM. 2000;343:450.CrossRefPubMedPubMedCentral Bookheimer SY, Strojwas MH, Cohen MS, Saunders AM, Pericak-Vance MA, Mazziotta JC, Small GW. Patterns of brain activation in people at risk for Alzheimer’s disease. NEJM. 2000;343:450.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Bassett SS, Yousem DM, Cristinzio C, Kusevic I, Yassa MA, Caffo BS, Zeger SL. Familial risk for Alzheimer’s disease alters fMRI activation patterns. Brain. 2006;129:1229.CrossRefPubMedPubMedCentral Bassett SS, Yousem DM, Cristinzio C, Kusevic I, Yassa MA, Caffo BS, Zeger SL. Familial risk for Alzheimer’s disease alters fMRI activation patterns. Brain. 2006;129:1229.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Koga S, Kojima A, Kuwabara S, Yoshiyama Y. Immunohistochemical analysis of tau phosphorylation and astroglial activation with enhanced leptin receptor expression in diet-induced obesity mouse hippocampus. Neurosci Lett. 2014;571:11.CrossRefPubMed Koga S, Kojima A, Kuwabara S, Yoshiyama Y. Immunohistochemical analysis of tau phosphorylation and astroglial activation with enhanced leptin receptor expression in diet-induced obesity mouse hippocampus. Neurosci Lett. 2014;571:11.CrossRefPubMed
65.
Zurück zum Zitat Jayaraman A, Lent-Schochet D, Pike CJ. Diet-induced obesity and low testosterone increase neuroinflammation and impair neural function. J Neuroinflam. 2014;11:162.CrossRef Jayaraman A, Lent-Schochet D, Pike CJ. Diet-induced obesity and low testosterone increase neuroinflammation and impair neural function. J Neuroinflam. 2014;11:162.CrossRef
66.
Zurück zum Zitat Puig KL, Floden AM, Adhikari R, Golovko MY, Combs CK. Amyloid precursor protein and proinflammatory changes are regulated in brain and adipose tissue in a murine model of high fat diet-induced obesity. PLoS One. 2012;7:e30378.CrossRefPubMedPubMedCentral Puig KL, Floden AM, Adhikari R, Golovko MY, Combs CK. Amyloid precursor protein and proinflammatory changes are regulated in brain and adipose tissue in a murine model of high fat diet-induced obesity. PLoS One. 2012;7:e30378.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Zhang L, Dasuri K, Fernandez-Kim S-O, Bruce-Keller AJ, Freeman LR, Pepping JK, Beckett TL, Murphy MP, Keller JN. Prolonged diet induced obesity has minimal effects towards brain pathology in mouse model of cerebral amyloid angiopathy: Implications for studying obesity–brain interactions in mice. BBA-Mol Basis Dis. 2013;1832:1456.CrossRef Zhang L, Dasuri K, Fernandez-Kim S-O, Bruce-Keller AJ, Freeman LR, Pepping JK, Beckett TL, Murphy MP, Keller JN. Prolonged diet induced obesity has minimal effects towards brain pathology in mouse model of cerebral amyloid angiopathy: Implications for studying obesity–brain interactions in mice. BBA-Mol Basis Dis. 2013;1832:1456.CrossRef
68.
Zurück zum Zitat Buckman LB, Thompson MM, Moreno HN, Ellacott KL. Regional astrogliosis in the mouse hypothalamus in response to obesity. J Comp Neurol. 2013;521:1322.CrossRefPubMedPubMedCentral Buckman LB, Thompson MM, Moreno HN, Ellacott KL. Regional astrogliosis in the mouse hypothalamus in response to obesity. J Comp Neurol. 2013;521:1322.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Ishizu K, Nishizawa S, Yonekura Y, Sadato N, Magata Y, Tamaki N, Tsuchida T, Okazawa H, Miyatake S, Ishikawa M, et al. Effects of hyperglycemia on FDG uptake in human brain and glioma. J Nucl Med. 1994;35:1104.PubMed Ishizu K, Nishizawa S, Yonekura Y, Sadato N, Magata Y, Tamaki N, Tsuchida T, Okazawa H, Miyatake S, Ishikawa M, et al. Effects of hyperglycemia on FDG uptake in human brain and glioma. J Nucl Med. 1994;35:1104.PubMed
70.
Zurück zum Zitat Yamada K, Endo S, Fukuda H, Abe Y, Yoshioka S, Itoh M, Kubota K, Hatazawa J, Satoh T, Matsuzawa T, et al. Experimental studies on myocardial glucose metabolism of rats with 18 F-2-fluoro-2-deoxy-D-glucose. Eur J Nucl Med. 1985;10:341.CrossRefPubMed Yamada K, Endo S, Fukuda H, Abe Y, Yoshioka S, Itoh M, Kubota K, Hatazawa J, Satoh T, Matsuzawa T, et al. Experimental studies on myocardial glucose metabolism of rats with 18 F-2-fluoro-2-deoxy-D-glucose. Eur J Nucl Med. 1985;10:341.CrossRefPubMed
71.
Zurück zum Zitat Wahl RL, Henry CA, Ethier SP. Serum glucose: effects on tumor and normal tissue accumulation of 2-[F-18]-fluoro-2-deoxy-D-glucose in rodents with mammary carcinoma. Radiology. 1992;183:643.CrossRefPubMed Wahl RL, Henry CA, Ethier SP. Serum glucose: effects on tumor and normal tissue accumulation of 2-[F-18]-fluoro-2-deoxy-D-glucose in rodents with mammary carcinoma. Radiology. 1992;183:643.CrossRefPubMed
72.
Zurück zum Zitat Giannaccini G, Betti L, Palego L, Pirone A, Schmid L, Lanza M, Fabbrini L, Pelosini C, Maffei M, Santini F, et al. Serotonin transporter (SERT) and translocator protein (TSPO) expression in the obese ob/ob mouse. BMC Neurosci. 2011;12:18.CrossRefPubMedPubMedCentral Giannaccini G, Betti L, Palego L, Pirone A, Schmid L, Lanza M, Fabbrini L, Pelosini C, Maffei M, Santini F, et al. Serotonin transporter (SERT) and translocator protein (TSPO) expression in the obese ob/ob mouse. BMC Neurosci. 2011;12:18.CrossRefPubMedPubMedCentral
Metadaten
Titel
Assessment of neuroinflammation in a mouse model of obesity and β-amyloidosis using PET
verfasst von
Anna M. Barron
Masaki Tokunaga
Ming-Rong Zhang
Bin Ji
Tetsuya Suhara
Makoto Higuchi
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2016
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0700-x

Weitere Artikel der Ausgabe 1/2016

Journal of Neuroinflammation 1/2016 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.