Skip to main content
Erschienen in: Fluids and Barriers of the CNS 1/2015

Open Access 01.12.2015 | Research

Kaolin-induced chronic hydrocephalus accelerates amyloid deposition and vascular disease in transgenic rats expressing high levels of human APP

verfasst von: Gerald D Silverberg, Miles C Miller, Crissey L Pascale, Ilias N Caralopoulos, Yuksel Agca, Cansu Agca, Edward G Stopa

Erschienen in: Fluids and Barriers of the CNS | Ausgabe 1/2015

Abstract

Background

Normal pressure hydrocephalus (NPH) is most common in the elderly and has a high co-morbidity with Alzheimer’s disease (AD) and cerebrovascular disease (CVD). To understand the relationship between NPH, AD and CVD, we investigated how chronic hydrocephalus impacts brain amyloid-beta peptide (Aβ) accumulation and vascular pathology in an AD transgenic rodent model. Previously we showed that the altered CSF physiology produced by kaolin-hydrocephalus in older wild-type Sprague–Dawley rats increased Aβ and hyperphosphorylated Tau (Silverberg et. al. Brain Res. 2010, 1317:286–296). We postulated that hydrocephalus would similarly affect an AD rat model.

Methods

Thirty-five transgenic rats (tgAPP21) that express high levels of human APP and naturally overproduce Aβ40 were used. Six- (n = 7) and twelve-month-old (n = 9) rats had hydrocephalus induced by cisternal kaolin injection. We analyzed Aβ burden (Aβ40, Aβ42 and oligomeric Aβ) and vascular integrity (Masson trichrome and Verhoeff-Van Gieson) by immunohistochemistry and chemical staining at 10 weeks (n = 8) and 6 months (n = 5) post hydrocephalus induction. We also analyzed whether the vascular pathology seen in tgAPP21 rats, which develop amyloid angiopathy, was accelerated by hydrocephalus. Age-matched naïve and sham-operated tgAPP21 rats served as controls (n = 19).

Results

In hydrocephalic tgAPP21 rats, compared to naïve and sham-operated controls, there was increased Aβ 40 and oligomeric Aβ in hippocampal and cortical neurons at 10 weeks and 6 months post-hydrocephalus induction. No dense-core amyloid plaques were seen, but diffuse Aβ immunoreactivity was evident in neurons. Vascular pathology was accelerated by the induction of hydrocephalus compared to controls. In the six-month-old rats, subtle degenerative changes were noted in vessel walls at 10 weeks post-kaolin, whereas at six months post-kaolin and in the 12-month-old hydrocephalic rats more pronounced amyloid angiopathic changes were seen, with frequent large areas of infarction noted.

Conclusions

Kaolin-hydrocephalus can accelerate intraneuronal Aβ40 accumulation and vascular pathology in tgAPP21 rats. In addition, disrupted CSF production and reduced CSF turnover results in impaired Aβ clearance and accelerated vascular pathology in chronic hydrocephalus. The high co-morbidity seen in NPH, AD and CVD is likely not to be an age-related coincidence, but rather a convergence of pathologies related to diminished CSF clearance.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

GDS conceived the research project and wrote the first draft of the paper. EGS and MCM did the histological studies and the interpretation of the results. CLP and INC did the cisternal kaolin injections and cared for the rats until sacrificed. YA and CA provided the tgAPP21 breeding pairs. All authors contributed to the writing and editing of the manuscript and agree with its contents and conclusions.
Abkürzungen
Amyloid-beta peptide
AD
Alzheimer’s disease
APP
Amyloid precursor protein
CSF
Cerebrospinal fluid
CVD
Cerebrovascular disease
H&E
Hematoxylin and eosin
HRP
Horseradish peroxidase
IACUC
Institutional Animal Care and Use Committee
NPH
Normal pressure hydrocephalus
pTau
Hyperphosphorylated tau protein
REM
Rapid eye movement
SAS
Subarachnoid space
TBST
0.05 M Tris-buffered saline with 0.05% Tween-20
tgAPP21
Double mutant APP Sw/Ind transgenic rat line.

Background

Normal pressure hydrocephalus (NPH) is a clinically-diagnosed disease that presents with one or several symptoms and signs, including gait disturbance, incontinence and dementia [1]. The gait disturbance is described as a gait apraxia (magnetic gait), and the urinary incontinence is often nocturnal. There is an associated ventricular enlargement, usually involving all ventricles, and some enlargement of the subarachnoid space (SAS) over the convexities. There is usually intermittent elevation of the cerebrospinal fluid (CSF) pressure most often at night and often during rapid eye movement (REM) sleep [24]. Clinical diagnosis rests on clinical and radiologic findings as well as the effects of large volume CSF removal [5].
Although there is no pathological finding that confirms the clinical diagnosis of NPH on brain biopsy or at post-mortem, there is often evidence for both Alzheimer’s disease (AD) and cerebrovascular disease (CVD) with a frequency that precludes the idea that these are unrelated afflictions of the elderly [69]. Indeed, initially these findings were thought to be simply unrelated coincidental diseases of the elderly, but that is no longer tenable.
In patients with the symptoms and signs of NPH, there is a very high co-morbidity with both AD and CVD. The incidence of AD pathology in patients with NPH is much higher than expected if the two diseases were unrelated diseases of advancing age. For instance, anywhere from 25% to 75% of NPH patients will have evidence of AD pathology, depending upon the degree of dementia, on brain biopsy at the time of shunt placement or at autopsy [1012]. In aging, only 10% of subjects over 65 will have clinical or histological evidence of AD [13]. Evidence of CVD is similarly increased in NPH over normally aged subjects.
Alzheimer’s disease is characterized by amyloid plaques (neuritic or dense-core), composed of amyloid-beta peptides (Aβ) and neurofibrillary tangles made up of paired helical filaments of hyperphosphorylated tau protein (pTau). Amyloid angiopathy is also characteristic of AD brains. This vascular pathology is characterized by vascular deposition of Aβ [14, 15]. In recent years it has become accepted that the accumulation of Aβ in non-familial AD is due to an inability to clear these peptides from the brain, rather than an overproduction as seen in familial AD [1618].
In previous reports, it was shown that both Aβ and pTau protein accumulated in older (12 months) wild-type Sprague–Dawley rats, rendered hydrocephalic by intra-cisternal injection of kaolin, in concentrations significantly higher than age-matched and sham-operated controls [19, 20]. In this histological and immunohistochemical study we report the effects of induced hydrocephalus on a transgenic rat model of AD (tgAPP21): a double transgenic (Sw/Ind mutant) human amyloid precursor protein (APP) construct that expresses high levels of human APP and Aβ40 [21]. Our hypothesis was that if the CSF was not an important clearance pathway for Aβ removal from the brain, then the accumulation of Aβ would be the same between hydrocephalic and control tgAPP21 rats. We found that the hydrocephalic tgAPP21 rats accumulated Aβ40 and oligomeric Aβ, as well as manifesting evidence of vascular disease and ischemic infarction, well before age-matched and sham-operated controls. Cortical infarction was seen only in the hydrocephalic rats.

Methods

Animals

Breeding pairs of APP 21 transgenic rats (tgAPP21) were obtained from the Department of Veterinary Pathobiology at the University of Missouri. These rats express high levels of human APP and naturally overproduce Aβ40, but not Aβ42. The tgAPP21 rats were produced from inbred Fischer 344 rats that express human APP driven by the ubiquitin-C promoter. They were generated via lentiviral vector infection of the Fischer 344 zygotes [21]. Immunohistochemistry in brain showed that the human APP transgene was expressed in neurons, but not in glial cells. After quarantine, the tgAPP21 rats were allowed to breed normally. The rats were housed in the veterinary care facility of the Aldrich Laboratories at Rhode Island Hospital and had food and water ad lib. All experiments were approved by the Institutional Animal Care and Use Committee (IACUC) at Rhode Island Hospital.
Hydrocephalus was induced by cisternal injection of kaolin (aluminum silicate 0.9%). The technique has been previously published [19, 20, 22]. Thirty-five tgAPP21 rats were used in these studies. Six-month (n = 7) and twelve-month-old (n = 9) rats had hydrocephalus induced by cisternal kaolin injection. After 10 weeks or six months of hydrocephalus the rats were euthanized by intra-peritoneal pentobarbital injection (125 mg/kg). Age-matched naïve and sham-operated tgAPP21 rats served as controls (n = 19) and their brains were processed in exactly the same way. Three of the 12-month-old hydrocephalic rats and four of the controls were allowed to survive to a natural death to assess the effects of the vascular changes on brain parenchyma (see Table 1).
Table 1
Summary of experimental animals and histological findings
Duration
Group
N
Ventricular Enlargement
Histiocytic & Granulomatous Inflammation
Neuronal Aβ40
Neuronal Aβ42
Neuronal Oligomeric Aβ
Vascular Pathology
Ischemic Infarcts
6 mo. + 10 wk.
Kaolin
4
++
+++
++
-
++
++
-
6 mo. + 10 wk.
Sham
2
-
-
+
-
+
+
-
6 mo. + 10 wk.
Naïve
2
-
-
+
-
+
+
-
6 mo. + 6 mo.
Kaolin
3
+
+
++
-
+++
++
-
6 mo. + 6 mo.
Sham
2
-
-
+
-
++
+
-
6 mo. + 6 mo.
Naïve
2
-
-
+
-
++
+
-
12 mo. + 10 wk.
Kaolin
4
++
+++
++
-
+++
+++
-
12 mo. + 10 wk.
Sham
2
-
-
+
-
++
+
-
12 mo. + 10 wk.
Naïve
2
-
-
+
-
++
+
-
12 mo. + 6 mo.
Kaolin
2
+
+
+++
-
+++
+++
+
12 mo. + 6 mo.
Sham
2
-
-
+
-
++
+
-
12 mo. + 6 mo.
Naïve
1
-
-
+
-
++
+
-
12 mo. to natural end-of-life
Kaolin
3
+
+
+++
-
+++
+++
++
12 mo. to natural end-of-life
Sham
2
-
-
+
-
++
++
-
12 mo. to natural end-of-life
Naïve
2
-
-
+
-
++
++
-
The pathology and morphological changes, as observed in histochemically-stained sections, were qualitatively graded using a scale ranging from no detectable changes (−) to mild (+), moderate (++), or severe (+++) changes.
After intracardiac cannulation and perfusion with phosphate-buffered saline, the brains were removed and immersed in 4% paraformaldehyde. Following standard tissue processing and paraffin embedding procedures, coronal brain sections were serially cut at 8 μm starting from the level of the median eminence. Ventricular enlargement was measured by the Evans ratio for control rats compared to rats at 10 weeks post hydrocephalus induction. The maximum ventricular diameter on coronal section at the bregma was divided by the maximum brain diameter on the post-mortem brain sections.
We analyzed Aβ burden by immunohistochemistry (Aβ40, Aβ42 and oligomeric Aβ), and vascular integrity by histochemical staining (Masson trichrome and Verhoeff-Van Gieson) at 10 weeks (n = 8) and six months (n = 5) post hydrocephalus induction. We also analyzed whether the vascular pathology seen in tgAPP21 rats, which normally develop amyloid angiopathy, was accelerated by hydrocephalus. Age-matched naïve and sham-operated tgAPP21 rats served as controls (n = 15).

Immunohistochemistry

Eight μm-thick tissue sections (on poly-L-lysine-coated slides) were incubated in an oven at 60°C for 1 hour, and after deparaffinization and rehydration, sections were treated with hot (85°C) 10 mM citrate buffer, pH 6, for 15 minutes. Sections were washed with distilled water and then quenched with a dual endogenous enzyme-blocking reagent (Dako, Carpinteria, CA, USA; Catalog #S2003) for 10 minutes at room temperature to eliminate endogenous peroxidase activity. After washing in 0.05 M Tris-buffered saline with 0.05% Tween-20 (TBST), pH 7.6, sections were incubated overnight at 4°C with rabbit polyclonal antibodies directed against Aβ40 (Alpha Diagnostic International, San Antonio, TX, USA; Catalog #BAM401-A, diluted 1:100), Aβ42 (Alpha Diagnostic International; Catalog #BAM421-A, diluted 1:200), or oligomeric Aβ (A11; Chemicon, Temecula, CA, USA; Catalog #AB9234, diluted 1:2000). After washing the sections in TBST, a horseradish peroxidase (HRP)-labeled polymer conjugated with secondary antibodies (anti-rabbit; Dako; Catalog #K4002) was applied for 30 minutes at room temperature, in accordance with the EnVision + System for immunohistochemical staining. The tissue sections were washed in TBST and then the immunoreaction product was developed using 3,3-diaminobenzidine (Dako; Catalog #K3468) as the chromogen. Sections were dehydrated through a series of graded alcohols back to xylene, and then coverslipped and sealed using Cytoseal XYL (Richard-Allan Scientific, Kalamazoo, MI, USA; Catalog #8312-4). Primary antibody omission controls were run alongside the other samples to check for non-specific binding due to the secondary antibodies, and advanced AD human prefrontal cortical sections were run as positive controls. In place of using a counterstain on immunohistochemically-stained slides, adjacent serial sections were stained with hematoxylin and eosin (H&E) for analysis of general tissue morphology.

Immunofluorescence

Following deparaffinization and rehydration, tissue sections were treated with hot (85°C) 10 mM citrate buffer, pH 6, for 15 minutes. Sections were washed with distilled water and then quenched with a dual endogenous enzyme-blocking reagent (Dako) for 10 minutes at room temperature. After washing in TBST, sections were blocked with 5% normal goat serum (Vector Laboratories, Burlingame, CA, USA; Catalog #S-1000) for 2 hours at room temperature, and then dually incubated overnight (at 4°C) with the following primary antibodies: a mouse monoclonal antibody directed against NeuN (A60; Abcam, Cambridge, MA, USA; Catalog #ab77315, diluted 1:1000) and a rabbit polyclonal antibody directed against oligomeric Aβ (A11; Chemicon, diluted 1:2000). Sections were then washed in TBST, and the secondary antibodies were applied for 90 minutes at room temperature in the dark: AlexaFluor 488 goat anti-mouse IgG (Molecular Probes, Eugene, OR, USA; Catalog #A-11001, diluted 1:1000) and AlexaFluor 594 goat anti-rabbit IgG (Molecular Probes; Catalog #A-11012, diluted 1:1000). To eliminate possible lipofuscin autofluorescence, tissue sections were incubated in a 0.3% Sudan Black B (Sigma-Aldrich, St. Louis, MO, USA; Catalog #S-0395) solution in 70% ethanol for 20 minutes at room temperature in the dark. Sections were washed in distilled water and coverslipped using Vectashield Hard Set Mounting Medium with DAPI (Vector Laboratories; Catalog #H-1500). Primary antibody omission controls were run alongside the other samples to check for non-specific binding due to the secondary antibodies, and advanced AD human prefrontal cortical sections were run as positive controls.

Masson trichrome staining

Masson trichrome staining was carried out in accordance with well-characterized protocols [23, 24]. Briefly, tissue sections were deparaffinized and hydrated in distilled water prior to a 1-hour treatment in Bouin’s fixative (Richard-Allan Scientific; Catalog #NC9674780) at 56°C. Sections were washed in running distilled water until clear, and then stained in Weigert’s iron hematoxylin (Richard-Allan Scientific; Catalog #NC9231529) for 10 minutes. Following a 10-minute wash in running water, sections were stained in Biebrich scarlet-acid fuchsin (Richard-Allan Scientific; Catalog #NC9424144) for 2 minutes. Sections were rinsed in distilled water followed by a 10-minute differentiation in phosphomolybdic-phosphotungstic acid (Richard-Allan Scientific; Catalog #NC9443038). Aniline blue (Richard-Allan Scientific; Catalog #NC9684104) was used as a counterstain for 10 minutes, and then sections were differentiated in 1% acetic acid for 3 minutes. Sections were dehydrated through a series of graded alcohols back to xylene, and then coverslipped and sealed using Cytoseal XYL (Richard-Allan Scientific).

Verhoeff-Van Gieson staining

The Verhoeff-Van Gieson staining protocol for elastic fibers was performed using well-established protocols [24, 25]. Briefly, tissue sections were deparaffinized and hydrated to distilled water followed by a1-hour incubation in Verhoeff’s working solution (Polysciences, Warrington, PA, USA; Catalog #25089). Sections were rinsed in running water, and then differentiated in 2% ferric chloride (Sigma-Aldrich; Catalog #451649) for 2 minutes. Following a 10-minute wash in running water, sections were treated with 5% aqueous sodium thiosulfate (Sigma-Aldrich; Catalog #S7026) for 1 minute. Tissue sections were then washed in running water for 5 minutes, and counterstained with Van Gieson’s solution (Poly Scientific, Bay Shore, NY, USA; Catalog #s289) for 3 minutes. Sections were quickly dehydrated through a series of graded alcohols back to xylene, and then coverslipped and sealed using Cytoseal XYL (Richard-Allan Scientific).

Microscopy, image acquisition & qualitative grading

All immunohistochemistry and histochemically-stained slides were converted to digital images using Aperio Scan Scope (Aperio Technologies, Vista, CA, USA) as 8-bit acquisitions of color. For confocal microscopy, images were acquired with a Nikon C1si confocal microscope (Nikon Inc., Melville, NY, USA) using 488 nm and 561 nm diode lasers. Serial optical sections were performed with EZ-C1 computer software (Nikon Inc.). Z-series sections were collected at 1.5 μm with a 20× PlanApo lens and scan zoom of 2×. Each wavelength was acquired separately by invoking frame lambda, and images were processed with Elements computer software (Nikon Inc.). Pathology and morphological changes, as observed in histochemically-stained sections, were qualitatively graded using a scale ranging from no detectable changes (−), to mild (+), moderate (++), or severe (+++) changes.

Results

All histological and immunohistochemical findings are summarized in Table 1. The tgAPP21rats injected with kaolin developed hydrocephalus similar to that reported in our wild-type Sprague–Dawley rats [20]. Evans index of ventricular size in the hydrocephalic tgAPP21 rats was significantly larger than controls and was similar to what we previously reported [10 weeks 0.30 ± 0.04 compared to sham-operated controls 0.19 ± 0.02 (mean ± SD)]. Figure 1 compares the ventricular and aqueductal morphological changes observed in a typical hydrocephalic tgAPP21 rat to a typical age-matched sham-operated control.
The hydrocephalus was due to an intense histiocytic and granulomatous reaction in the SAS which was largely resolved by six months, though some small granulomas remained. Despite the resolution of the inflammation, scarring and blockage of the SAS remained (Figure 2).
Aβ immunostaining showed a marked increase in intraneuronal Aβ40 in the hippocampus and frontoparietal cortex compared to sham-operated controls (Figure 3). No amyloid plaques were observed, but diffuse cytoplasmic Aβ40 immunoreactivity was evident across multiple neuronal populations. The absence of dense-core plaques was not surprising given the absence of Aβ42 accumulation.
Although Aβ42 is more apt to self-assemble, Aβ40 can also self-assemble into oligomeric forms. Oligomeric Aβ immunoreactivity was predominately confined to neurons in both the hippocampus and frontoparietal cortex of tgAPP21 rats (Figure 4).
In hydrocephalic tgAPP21 rats, compared to controls, there was increased oligomeric Aβ immunoreactivity in addition to the increase in Aβ 40 in both hippocampal (data not shown) and cortical neurons at 10 weeks post-hydrocephalus induction in 6 and 12-month-old animals (Figure 5).
Vascular pathology was accelerated by the induction of hydrocephalus compared to controls. In the six-month-old rats, subtle degenerative changes were noted in vessel walls at 10 weeks post-kaolin, whereas in the six-month-old rat at six months post-kaolin and in the 12-month-old rat 10 weeks post-kaolin, more pronounced degenerative changes were seen with clear expansion of the Virchow-Robin space in interstitial vessels (Figure 6).
Amyloid angiopathic changes were seen by immunostaining for Aβ40. These changes were more dramatic in the 12-month-old tgAPP21 rats than in the six-month-old rats, and in both sets of hydrocephalic rats, the difference from sham-operated controls was clearly evident (Figure 7).
Seven of the tgAPP21 rats were allowed to reach their natural end of life (approximately 30 months). Three hydrocephalic rats were compared to four controls. The hydrocephalic tgAPP21 rats were found to have frequent areas of microscopic cortical infarction in their brains (Figure 8). In the non-kaolin controls, no infarcts were seen.

Discussion

Clearance of macromolecules, such as Aβ, from the brain interstitial space involves at least four different pathways: i) via in situ degradation [2630], ii) active transport across the blood–brain barrier [3136], iii) across the choroid plexus epithelium by active transport [37], and iv) via the production and turnover of the CSF. CSF turnover is defined as the number of times the CSF is renewed in 24 hours and is calculated by dividing CSF production in 24 hours by the volume of the CSF space [3842]. Normally in humans, CSF turnover occurs 4–5 times per day.
Hydrocephalus is known to disrupt normal CSF physiological functions. In both AD and in hydrocephalic patients, CSF turnover is reduced threefold [6, 43, 44]. In both wild-type rat models of NPH and in human NPH patients therefore, CSF clearance of potentially toxic solutes like Aβ is significantly reduced, resulting in the accumulation of these molecules in brain parenchyma. Several investigations in laboratory animals have described significantly decreased CSF production and turnover after kaolin hydrocephalus induction [45, 46], and is also seen in humans with NPH [44]. Resistance to CSF absorption is also increased in hydrocephalus [47, 48]. Despite eventual clearance of the inflammation produced by the kaolin, increased resistance to CSF absorption and decreased compliance remain [47, 48].
This study examined the effects of kaolin-induced hydrocephalus on amyloid accumulation and vascular pathology in a transgenic rat model of AD. The analysis was carried out by qualitative histological and immunohistochemical staining, comparing the hydrocephalic tgAPP21 brains to age-matched, sham-operated and naïve controls. We found that induced hydrocephalus accelerated Aβ accumulation in neurons and Aβ deposition in the cerebral vasculature, presumably due to decreased clearance of Aβ. Aβ immunostaining in cerebral cortex and hippocampus was increased in the hydrocephalic rats compared to controls, and amyloid angiopathic degeneration of cerebral vessels was also accelerated compared to controls. The amyloid angiopathy associated with the hydrocephalic rats appeared to cause microscopic ischemic infarcts not seen in the control animals.
It is well known that there is variability in the degree of hydrocephalus produced by intracisternal kaolin. Also in any qualitative histological and immunohistochemical study, fixation artifact must always be considered in assessing changes. Therefore, comparison to both sham-operated and naïve controls processed in exactly the same way as the hydrocephalic rats, is essential to identifying true differences from artifact. The microscopic findings in the two groups (hydrocephalic and controls) in this study were internally consistent but were strikingly different in Aβ accumulation, self-assembly into oligomeric forms and vascular pathology. Although not quantitative, the group comparisons were sufficiently different to conclude that accelerated amyloid deposition and vascular pathology occurs in tgAPP21 rats with kaolin-induced hydrocephalus.
One can argue that the tgAPP21 rat is more a model for amyloid angiopathy [49] rather than AD, in that there is no increase in Aβ42 concentrations and no amyloid plaque formation was evident. Instead we see that the predominantly expressed Aβ40 accumulates in the cerebral microvessels, reportedly localized to the basement membrane [50, 51]. However, our study was meant to explore whether the CSF plays a significant role in the clearance of macromolecular solutes from the brain interstitial space, and whether its failure in chronic hydrocephalus accelerates the accumulation of many brain metabolites. The present study suggests that this is the case in rats and likely in humans as well.

Conclusions

The results of this study underscore the importance of normal CSF physiologic functions in clearing potentially toxic macromolecules from the brain. The study shows that kaolin-induced hydrocephalus can accelerate intraneuronal Aβ accumulation and self-assembly, and accelerate vascular pathology in tgAPP21 rats. In addition, it demonstrates that disrupted CSF production and turnover results in impaired Aβ clearance from the brain and accelerates vascular pathology in chronic hydrocephalus. The high co-morbidity seen in NPH, AD and CVD is likely not an age-related coincidence, but rather a convergence of pathologies related to reduced solute clearance.

Acknowledgements

The authors would like to thank Virginia Hovanesian for aid in microscopy/image acquisition, and Paul Monfils, Catherine Chiu and Doreen P. Osgood, PhD for their technical assistance. We also thank Dr. Petra Klinge for development of the kaolin-hydrocephalus protocol and for teaching the lab how to do it. Funding for this study was provided by the Saunders Family Fund at the Neurosurgery Foundation, Rhode Island Hospital, and the Warren Alpert Medical School of Brown University.
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/​4.​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

GDS conceived the research project and wrote the first draft of the paper. EGS and MCM did the histological studies and the interpretation of the results. CLP and INC did the cisternal kaolin injections and cared for the rats until sacrificed. YA and CA provided the tgAPP21 breeding pairs. All authors contributed to the writing and editing of the manuscript and agree with its contents and conclusions.
Literatur
1.
Zurück zum Zitat Hakim S, Adams RD: The special clinical problem of symptomatic hydrocephalus with normal cerebrospinal fluid pressure. Observations on cerebrospinal fluid hydrodynamics.J Neurol Sci 1965, 2:307–27. 10.1016/0022-510X(65)90016-XCrossRefPubMed Hakim S, Adams RD: The special clinical problem of symptomatic hydrocephalus with normal cerebrospinal fluid pressure. Observations on cerebrospinal fluid hydrodynamics.J Neurol Sci 1965, 2:307–27. 10.1016/0022-510X(65)90016-XCrossRefPubMed
2.
Zurück zum Zitat Symon L, Dorsch NW: Use of long-term intracranial pressure measurement to assess hydrocephalic patients prior to shunt surgery.J Neurosurg 1975, 42:258–73. 10.3171/jns.1975.42.3.0258CrossRefPubMed Symon L, Dorsch NW: Use of long-term intracranial pressure measurement to assess hydrocephalic patients prior to shunt surgery.J Neurosurg 1975, 42:258–73. 10.3171/jns.1975.42.3.0258CrossRefPubMed
3.
Zurück zum Zitat Symon L, Dorsch NW, Stephens RJ: Pressure waves in so-called low-pressure hydrocephalus.Lancet 1977, 2:1291–2. Symon L, Dorsch NW, Stephens RJ: Pressure waves in so-called low-pressure hydrocephalus.Lancet 1977, 2:1291–2.
4.
Zurück zum Zitat Krauss JK, Droste DW, Bohus M, Regel JP, Scheremet R, Riemann D, et al.: The relation of intracranial pressure B-waves to different sleep stages in patients with suspected normal pressure hydrocephalus.Acta Neurochir (Wien) 1995, 136:195–203. 10.1007/BF01410626CrossRef Krauss JK, Droste DW, Bohus M, Regel JP, Scheremet R, Riemann D, et al.: The relation of intracranial pressure B-waves to different sleep stages in patients with suspected normal pressure hydrocephalus.Acta Neurochir (Wien) 1995, 136:195–203. 10.1007/BF01410626CrossRef
5.
Zurück zum Zitat Wikkelsø C, Hellström P, Klinge PM, Tans JT, European iNPH Multicentre Study Group: The European iNPH Multicentre Study on the predictive values of resistance to CSF outflow and the CSF Tap Test in patients with idiopathic normal pressure hydrocephalus.J Neurol Neurosurg Psychiatry 2013, 84:562–8. 10.1136/jnnp-2012-303314CrossRefPubMed Wikkelsø C, Hellström P, Klinge PM, Tans JT, European iNPH Multicentre Study Group: The European iNPH Multicentre Study on the predictive values of resistance to CSF outflow and the CSF Tap Test in patients with idiopathic normal pressure hydrocephalus.J Neurol Neurosurg Psychiatry 2013, 84:562–8. 10.1136/jnnp-2012-303314CrossRefPubMed
6.
Zurück zum Zitat Silverberg GD, Mayo M, Saul T, Rubenstein E, McGuire D: Alzheimer’s disease, normal-pressure hydrocephalus, and senescent changes in CSF circulatory physiology: a hypothesis.Lancet Neurol 2003, 2:506–11. 10.1016/S1474-4422(03)00487-3CrossRefPubMed Silverberg GD, Mayo M, Saul T, Rubenstein E, McGuire D: Alzheimer’s disease, normal-pressure hydrocephalus, and senescent changes in CSF circulatory physiology: a hypothesis.Lancet Neurol 2003, 2:506–11. 10.1016/S1474-4422(03)00487-3CrossRefPubMed
7.
Zurück zum Zitat Silverberg GD: Normal pressure hydrocephalus (NPH): ischaemia, CSF stagnation or both.Brain 2004, 127:947–8. 10.1093/brain/awh178CrossRefPubMed Silverberg GD: Normal pressure hydrocephalus (NPH): ischaemia, CSF stagnation or both.Brain 2004, 127:947–8. 10.1093/brain/awh178CrossRefPubMed
8.
Zurück zum Zitat Tullberg M, Hultin L, Ekholm S, Månsson JE, Fredman P, Wikkelsø C: White matter changes in normal pressure hydrocephalus and Binswanger disease: specificity, predictive value and correlations to axonal degeneration and demyelination.Acta Neurol Scand 2002, 105:417–26. 10.1034/j.1600-0404.2002.01189.xCrossRefPubMed Tullberg M, Hultin L, Ekholm S, Månsson JE, Fredman P, Wikkelsø C: White matter changes in normal pressure hydrocephalus and Binswanger disease: specificity, predictive value and correlations to axonal degeneration and demyelination.Acta Neurol Scand 2002, 105:417–26. 10.1034/j.1600-0404.2002.01189.xCrossRefPubMed
9.
Zurück zum Zitat Bateman GA: The pathophysiology of idiopathic normal pressure hydrocephalus: cerebral ischemia or altered venous hemodynamics?Am J Neuroradiol 2008, 29:198–203. 10.3174/ajnr.A0739CrossRefPubMed Bateman GA: The pathophysiology of idiopathic normal pressure hydrocephalus: cerebral ischemia or altered venous hemodynamics?Am J Neuroradiol 2008, 29:198–203. 10.3174/ajnr.A0739CrossRefPubMed
10.
Zurück zum Zitat Del Bigio MR, Cardoso ER, Halliday WC: Neuropathological changes in chronic adult hydrocephalus: cortical biopsies and autopsy findings.Can J Neurol Sci 1997, 24:121–6.CrossRefPubMed Del Bigio MR, Cardoso ER, Halliday WC: Neuropathological changes in chronic adult hydrocephalus: cortical biopsies and autopsy findings.Can J Neurol Sci 1997, 24:121–6.CrossRefPubMed
11.
Zurück zum Zitat Savolainen S, Paljärvi L, Vapalahti M: Prevalence of Alzheimer’s disease in patients investigated for presumed normal pressure hydrocephalus: a clinical and neuropathological study.Acta Neurochir (Wien) 1999, 141:849–53. 10.1007/s007010050386CrossRef Savolainen S, Paljärvi L, Vapalahti M: Prevalence of Alzheimer’s disease in patients investigated for presumed normal pressure hydrocephalus: a clinical and neuropathological study.Acta Neurochir (Wien) 1999, 141:849–53. 10.1007/s007010050386CrossRef
12.
Zurück zum Zitat Golomb J, Wisoff J, Miller DC, Boksay I, Kluger A, Weiner H, et al.: Alzheimer’s disease comorbidity in normal pressure hydrocephalus: prevalence and shunt response.J Neurol Neurosurg Psychiatry 2000, 68:778–81. 10.1136/jnnp.68.6.778CrossRefPubMedPubMedCentral Golomb J, Wisoff J, Miller DC, Boksay I, Kluger A, Weiner H, et al.: Alzheimer’s disease comorbidity in normal pressure hydrocephalus: prevalence and shunt response.J Neurol Neurosurg Psychiatry 2000, 68:778–81. 10.1136/jnnp.68.6.778CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Association A’s: Alzheimer’s disease facts and figures.Alzheimers Dement 2014,2014(10):e47–92. Association A’s: Alzheimer’s disease facts and figures.Alzheimers Dement 2014,2014(10):e47–92.
14.
Zurück zum Zitat Braak H, Braak E: Neuropathological stageing of Alzheimer-related changes.Acta Neuropathol 1991, 82:239–59. 10.1007/BF00308809CrossRefPubMed Braak H, Braak E: Neuropathological stageing of Alzheimer-related changes.Acta Neuropathol 1991, 82:239–59. 10.1007/BF00308809CrossRefPubMed
15.
Zurück zum Zitat The National Institute on Aging, and Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer’s Disease: Consensus recommendations for the postmortem diagnosis of Alzheimer’s disease.Neurobiol Aging 1997,18(4 suppl):S1–2. The National Institute on Aging, and Reagan Institute Working Group on Diagnostic Criteria for the Neuropathological Assessment of Alzheimer’s Disease: Consensus recommendations for the postmortem diagnosis of Alzheimer’s disease.Neurobiol Aging 1997,18(4 suppl):S1–2.
16.
Zurück zum Zitat Selkoe DJ: Toward a comprehensive theory for Alzheimer’s disease. Hypothesis: Alzheimer’s disease is caused by the cerebral accumulation and cytotoxicity of amyloid beta-protein.Ann NY Acad Sci 2000, 924:17–25.CrossRefPubMed Selkoe DJ: Toward a comprehensive theory for Alzheimer’s disease. Hypothesis: Alzheimer’s disease is caused by the cerebral accumulation and cytotoxicity of amyloid beta-protein.Ann NY Acad Sci 2000, 924:17–25.CrossRefPubMed
17.
Zurück zum Zitat Hardy J: A hundred years of Alzheimer’s disease research.Neuron 2006, 52:3–13. 10.1016/j.neuron.2006.09.016CrossRefPubMed Hardy J: A hundred years of Alzheimer’s disease research.Neuron 2006, 52:3–13. 10.1016/j.neuron.2006.09.016CrossRefPubMed
18.
Zurück zum Zitat Mawuenyega KG, Sigurdson W, Ovod V, Munselli T, Kasten T, Morris JC, et al.: Decreased Clearance of CNS β-Amyloid in Alzheimer’s Disease.Science 2010, 330:1774. 10.1126/science.1197623CrossRefPubMedPubMedCentral Mawuenyega KG, Sigurdson W, Ovod V, Munselli T, Kasten T, Morris JC, et al.: Decreased Clearance of CNS β-Amyloid in Alzheimer’s Disease.Science 2010, 330:1774. 10.1126/science.1197623CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Klinge PM, Samii A, Niesken S, Brinker T, Silverberg GD: Brain amyloid accumulation in aged rats with kaolin-induced hydrocephalus.NeuroReport 2006, 17:657–60. 10.1097/00001756-200604240-00020CrossRefPubMed Klinge PM, Samii A, Niesken S, Brinker T, Silverberg GD: Brain amyloid accumulation in aged rats with kaolin-induced hydrocephalus.NeuroReport 2006, 17:657–60. 10.1097/00001756-200604240-00020CrossRefPubMed
20.
Zurück zum Zitat Silverberg GD, Miller MC, Machan JT, Johanson CE, Caralopoulos IN, Pascale CL, et al.: Amyloid and Tau accumulate in the brains of aged hydrocephalic rats.Brain Res 2010, 1317:286–96.CrossRefPubMed Silverberg GD, Miller MC, Machan JT, Johanson CE, Caralopoulos IN, Pascale CL, et al.: Amyloid and Tau accumulate in the brains of aged hydrocephalic rats.Brain Res 2010, 1317:286–96.CrossRefPubMed
21.
Zurück zum Zitat Agca C, Fritz JJ, Walker LC, Levey AI, Chan AWS, Lah JJ, et al.: Development of transgenic rats producing human β-amyloid precursor protein as a model for Alzheimer’s disease: Transgene and endogenous APP genes are regulated tissue-specifically.BMC Neurosci 2008, 9:28. 10.1186/1471-2202-9-28CrossRefPubMedPubMedCentral Agca C, Fritz JJ, Walker LC, Levey AI, Chan AWS, Lah JJ, et al.: Development of transgenic rats producing human β-amyloid precursor protein as a model for Alzheimer’s disease: Transgene and endogenous APP genes are regulated tissue-specifically.BMC Neurosci 2008, 9:28. 10.1186/1471-2202-9-28CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Klinge PM, Samii A, Muhlendyck A, Visnyei K, Meyer GJ, Walter GF, et al.: Cerebral hypoperfusion and delayed hippocampal response after induction of adult kaolin hydrocephalus.Stroke 2003, 34:193–9. 10.1161/01.STR.0000048820.17198.15CrossRefPubMed Klinge PM, Samii A, Muhlendyck A, Visnyei K, Meyer GJ, Walter GF, et al.: Cerebral hypoperfusion and delayed hippocampal response after induction of adult kaolin hydrocephalus.Stroke 2003, 34:193–9. 10.1161/01.STR.0000048820.17198.15CrossRefPubMed
23.
Zurück zum Zitat Masson PJ: Trichrome stainings and their preliminary techniques.J Tech Meth 1929, 12:75. Masson PJ: Trichrome stainings and their preliminary techniques.J Tech Meth 1929, 12:75.
24.
Zurück zum Zitat Luna L: Manual of histological staining methods of the armed forces insitiute of pathology. 3rd edition. New York: McGraw Hill; 1968:76–95. Luna L: Manual of histological staining methods of the armed forces insitiute of pathology. 3rd edition. New York: McGraw Hill; 1968:76–95.
25.
Zurück zum Zitat Carson F: Histotechnology: a self-instructional text. 1st edition. Chicago: American Society for Clinical Pathology Press; 1990:142–9. Carson F: Histotechnology: a self-instructional text. 1st edition. Chicago: American Society for Clinical Pathology Press; 1990:142–9.
26.
Zurück zum Zitat Qiu WQ, Walsh DM, Ye Z, Vekrellis K, Zhang J, Podlisny MB, et al.: Insulin-degrading enzyme regulates extracellular levels of amyloid beta-protein by degradation.J Biol Chem 1998, 273:32730–8. 10.1074/jbc.273.49.32730CrossRefPubMed Qiu WQ, Walsh DM, Ye Z, Vekrellis K, Zhang J, Podlisny MB, et al.: Insulin-degrading enzyme regulates extracellular levels of amyloid beta-protein by degradation.J Biol Chem 1998, 273:32730–8. 10.1074/jbc.273.49.32730CrossRefPubMed
27.
Zurück zum Zitat Iwata N, Tsubuki S, Takaki Y, Watanabe K, Sekiguchi M, Hosoki E, et al.: Identification of the major Abeta1–42-degrading catabolic pathway in brain parenchyma: suppression leads to biochemical and pathological deposition.Nat Med 2000, 6:143–50. 10.1038/72237CrossRefPubMed Iwata N, Tsubuki S, Takaki Y, Watanabe K, Sekiguchi M, Hosoki E, et al.: Identification of the major Abeta1–42-degrading catabolic pathway in brain parenchyma: suppression leads to biochemical and pathological deposition.Nat Med 2000, 6:143–50. 10.1038/72237CrossRefPubMed
28.
Zurück zum Zitat Iwata N, Tsubuki S, Takaki Y, Shirotani K, Lu B, Gerard NP, et al.: Metabolic regulation of brain Abeta by neprilysin.Science 2001, 292:1550–2. 10.1126/science.1059946CrossRefPubMed Iwata N, Tsubuki S, Takaki Y, Shirotani K, Lu B, Gerard NP, et al.: Metabolic regulation of brain Abeta by neprilysin.Science 2001, 292:1550–2. 10.1126/science.1059946CrossRefPubMed
29.
Zurück zum Zitat Kanemitsu H, Tomiyama T, Mori H: Human neprilysin is capable of degrading amyloid beta peptide not only in the monomeric form but also the pathological oligomeric form.Neurosci Lett 2003, 350:113–6. 10.1016/S0304-3940(03)00898-XCrossRefPubMed Kanemitsu H, Tomiyama T, Mori H: Human neprilysin is capable of degrading amyloid beta peptide not only in the monomeric form but also the pathological oligomeric form.Neurosci Lett 2003, 350:113–6. 10.1016/S0304-3940(03)00898-XCrossRefPubMed
30.
Zurück zum Zitat Zlokovic BV, Yamada S, Holtzman D, Ghiso J, Frangione B: Clearance of amyloid beta-peptide from brain: transport or metabolism?Nat Med 2000, 6:718–9.CrossRef Zlokovic BV, Yamada S, Holtzman D, Ghiso J, Frangione B: Clearance of amyloid beta-peptide from brain: transport or metabolism?Nat Med 2000, 6:718–9.CrossRef
31.
Zurück zum Zitat Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, et al.: Clearance of Alzheimer’s amyloid-ss(1–40) peptide from brain by LDL receptor-related protein-1 at the blood–brain barrier.J Clin Invest 2000, 106:1489–99. 10.1172/JCI10498CrossRefPubMedPubMedCentral Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Frangione B, et al.: Clearance of Alzheimer’s amyloid-ss(1–40) peptide from brain by LDL receptor-related protein-1 at the blood–brain barrier.J Clin Invest 2000, 106:1489–99. 10.1172/JCI10498CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, et al.: RAGE mediates amyloid-beta peptide transport across the blood–brain barrier and accumulation in brain.Nat Med 2003, 9:907–13. 10.1038/nm890CrossRefPubMed Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, et al.: RAGE mediates amyloid-beta peptide transport across the blood–brain barrier and accumulation in brain.Nat Med 2003, 9:907–13. 10.1038/nm890CrossRefPubMed
33.
Zurück zum Zitat Deane R, Zlokovic BV: Role of the blood–brain barrier in the pathogenesis of Alzheimer’s disease.Curr Alzheimer Res 2007, 4:191–7. 10.2174/156720507780362245CrossRefPubMed Deane R, Zlokovic BV: Role of the blood–brain barrier in the pathogenesis of Alzheimer’s disease.Curr Alzheimer Res 2007, 4:191–7. 10.2174/156720507780362245CrossRefPubMed
34.
Zurück zum Zitat Zlokovic BV: Clearing amyloid through the blood–brain barrier.J Neurochem 2004, 89:807–11. 10.1111/j.1471-4159.2004.02385.xCrossRefPubMed Zlokovic BV: Clearing amyloid through the blood–brain barrier.J Neurochem 2004, 89:807–11. 10.1111/j.1471-4159.2004.02385.xCrossRefPubMed
35.
Zurück zum Zitat Silverberg GD, Miller MC, Messier AA, Majmudar S, Machan JT, Donahue JE, et al.: Amyloid deposition and influx transporter expression at the blood–brain barrier increase in normal aging.J Neuropathol Exp Neurol 2010, 69:98–108. 10.1097/NEN.0b013e3181c8ad2fCrossRefPubMed Silverberg GD, Miller MC, Messier AA, Majmudar S, Machan JT, Donahue JE, et al.: Amyloid deposition and influx transporter expression at the blood–brain barrier increase in normal aging.J Neuropathol Exp Neurol 2010, 69:98–108. 10.1097/NEN.0b013e3181c8ad2fCrossRefPubMed
36.
Zurück zum Zitat Silverberg GD, Messier AA, Miller MC, Machan JT, Majmudar S, Stopa EG, et al.: Amyloid efflux transporter expression at the blood–brain barrier declines in normal aging.J Neuropathol Exp Neurol 2010, 69:1034–43. 10.1097/NEN.0b013e3181f46e25CrossRefPubMed Silverberg GD, Messier AA, Miller MC, Machan JT, Majmudar S, Stopa EG, et al.: Amyloid efflux transporter expression at the blood–brain barrier declines in normal aging.J Neuropathol Exp Neurol 2010, 69:1034–43. 10.1097/NEN.0b013e3181f46e25CrossRefPubMed
37.
Zurück zum Zitat Pascale CL, Miller MC, Chiu C, Boylan M, Caralopoulos IN, Gonzalez L, et al.: Amyloid-beta transporter expression at the blood-cerebrospinal fluid barrier is age-dependent.Fluids Barriers CNS 2011, 8:21. 10.1186/2045-8118-8-21CrossRefPubMedPubMedCentral Pascale CL, Miller MC, Chiu C, Boylan M, Caralopoulos IN, Gonzalez L, et al.: Amyloid-beta transporter expression at the blood-cerebrospinal fluid barrier is age-dependent.Fluids Barriers CNS 2011, 8:21. 10.1186/2045-8118-8-21CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Davson H, Oldendorf WH: Symposium on membrane transport. Transport in the central nervous system.Proc R Soc Med 1967, 60:326–9.PubMedPubMedCentral Davson H, Oldendorf WH: Symposium on membrane transport. Transport in the central nervous system.Proc R Soc Med 1967, 60:326–9.PubMedPubMedCentral
39.
Zurück zum Zitat Oldendorf WH, Davson H: Brain extracellular space and the sink action of cerebrospinal fluid. Measurement of rabbit brain extracellular space using sucrose labeled with carbon 14.Arch Neurol 1967, 17:196–205. 10.1001/archneur.1967.00470260086010CrossRefPubMed Oldendorf WH, Davson H: Brain extracellular space and the sink action of cerebrospinal fluid. Measurement of rabbit brain extracellular space using sucrose labeled with carbon 14.Arch Neurol 1967, 17:196–205. 10.1001/archneur.1967.00470260086010CrossRefPubMed
40.
Zurück zum Zitat Rennels ML, Blaumanis OR, Grady PA: Rapid solute transport throughout the brain via paravascular fluid pathways.Adv Neurol 1990, 52:431–9.PubMed Rennels ML, Blaumanis OR, Grady PA: Rapid solute transport throughout the brain via paravascular fluid pathways.Adv Neurol 1990, 52:431–9.PubMed
41.
Zurück zum Zitat Johanson CE: Ventricles and Cerebrospinal fluid. In Neuroscience in Medicine. Edited by: Conn PM. Philadelphia: J.B. Lippincott Company; 1995:171–96. Johanson CE: Ventricles and Cerebrospinal fluid. In Neuroscience in Medicine. Edited by: Conn PM. Philadelphia: J.B. Lippincott Company; 1995:171–96.
42.
Zurück zum Zitat Chiu C, Miller MC, Caralopoulos IN, Worden MS, Brinker T, Gordon ZN, et al.: Temporal course of cerebrospinal fluid dynamics and amyloid accumulation in the aging rat brain from three to thirty months.Fluids Barriers CNS 2012, 9:3. 10.1186/2045-8118-9-3CrossRefPubMedPubMedCentral Chiu C, Miller MC, Caralopoulos IN, Worden MS, Brinker T, Gordon ZN, et al.: Temporal course of cerebrospinal fluid dynamics and amyloid accumulation in the aging rat brain from three to thirty months.Fluids Barriers CNS 2012, 9:3. 10.1186/2045-8118-9-3CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Silverberg GD, Heit G, Huhn S, Jaffe RA, Chang SD, Bronte-Stewart H, et al.: The cerebrospinal fluid production rate is reduced in dementia of the Alzheimer’s type.Neurology 2001, 57:1763–6. 10.1212/WNL.57.10.1763CrossRefPubMed Silverberg GD, Heit G, Huhn S, Jaffe RA, Chang SD, Bronte-Stewart H, et al.: The cerebrospinal fluid production rate is reduced in dementia of the Alzheimer’s type.Neurology 2001, 57:1763–6. 10.1212/WNL.57.10.1763CrossRefPubMed
44.
Zurück zum Zitat Silverberg GD, Huhn S, Jaffe RA, Chang SD, Saul T, Heit G, et al.: Downregulation of cerebrospinal fluid production in patients with chronic hydrocephalus.J Neurosurg 2002, 97:1271–5. 10.3171/jns.2002.97.6.1271CrossRefPubMed Silverberg GD, Huhn S, Jaffe RA, Chang SD, Saul T, Heit G, et al.: Downregulation of cerebrospinal fluid production in patients with chronic hydrocephalus.J Neurosurg 2002, 97:1271–5. 10.3171/jns.2002.97.6.1271CrossRefPubMed
45.
Zurück zum Zitat Hochwald GM, Nakamura S, Camins MB: The rat in experimental obstructive hydrocephalus.Z Kinderchir 1981, 34:403–10.PubMed Hochwald GM, Nakamura S, Camins MB: The rat in experimental obstructive hydrocephalus.Z Kinderchir 1981, 34:403–10.PubMed
46.
Zurück zum Zitat Hochwald GM, Sahar A: Effect of spinal fluid pressure on cerebrospinal fluid formation.Exp Neurol 1971, 32:30–40. 10.1016/0014-4886(71)90162-2CrossRefPubMed Hochwald GM, Sahar A: Effect of spinal fluid pressure on cerebrospinal fluid formation.Exp Neurol 1971, 32:30–40. 10.1016/0014-4886(71)90162-2CrossRefPubMed
47.
Zurück zum Zitat Brinker T, Beck H, Klinge P, Kischnik B, Oi S, Samii M: Sinusoidal intrathecal infusion for assessment of CSF dynamics in kaolin-induced hydrocephalus.Acta Neurochir (Wien) 1998, 140:1069–75. 10.1007/s007010050216CrossRef Brinker T, Beck H, Klinge P, Kischnik B, Oi S, Samii M: Sinusoidal intrathecal infusion for assessment of CSF dynamics in kaolin-induced hydrocephalus.Acta Neurochir (Wien) 1998, 140:1069–75. 10.1007/s007010050216CrossRef
48.
Zurück zum Zitat Kondziella D, Ludemann W, Brinker T, Sletvold O, Sonnewald U: Alterations in brain metabolism, CNS morphology and CSF dynamics in adult rats with kaolin-induced hydrocephalus.Brain Res 2002, 927:35–41. 10.1016/S0006-8993(01)03320-0CrossRefPubMed Kondziella D, Ludemann W, Brinker T, Sletvold O, Sonnewald U: Alterations in brain metabolism, CNS morphology and CSF dynamics in adult rats with kaolin-induced hydrocephalus.Brain Res 2002, 927:35–41. 10.1016/S0006-8993(01)03320-0CrossRefPubMed
49.
Zurück zum Zitat Rosen RF, Fritz JJ, Dooyema J, Cintron AF, Hamaguchi T, Lah JJ, et al.: Exogenous seeding of cerebral β-amyloid deposition in βAPP-transgenic rats.J Neurochem 2012, 120:660–6. 10.1111/j.1471-4159.2011.07551.xCrossRefPubMed Rosen RF, Fritz JJ, Dooyema J, Cintron AF, Hamaguchi T, Lah JJ, et al.: Exogenous seeding of cerebral β-amyloid deposition in βAPP-transgenic rats.J Neurochem 2012, 120:660–6. 10.1111/j.1471-4159.2011.07551.xCrossRefPubMed
50.
Zurück zum Zitat Weller RO, Subash M, Preston SD, Mazanti I, Carare RO: Perivascular drainage of amyloid-beta peptides from thebrain and its failure in cerebral amyloid angiopathy and Alzheimer’s disease.Brain Pathol 2008, 18:253–66.CrossRefPubMed Weller RO, Subash M, Preston SD, Mazanti I, Carare RO: Perivascular drainage of amyloid-beta peptides from thebrain and its failure in cerebral amyloid angiopathy and Alzheimer’s disease.Brain Pathol 2008, 18:253–66.CrossRefPubMed
51.
Zurück zum Zitat Carare RO, Hawkes CA, Jeffrey M, Kalaria RN, Weller RO: Cerebral amyloid angiopathy, prion angiopathy, CADASIL and the spectrum of protein elimination-failure angiopathies (PEFA) in neurodegenerative disease with a focus on therapy.Neuropathol Appl Neurobiol 2013, 39:593–611. 10.1111/nan.12042CrossRefPubMed Carare RO, Hawkes CA, Jeffrey M, Kalaria RN, Weller RO: Cerebral amyloid angiopathy, prion angiopathy, CADASIL and the spectrum of protein elimination-failure angiopathies (PEFA) in neurodegenerative disease with a focus on therapy.Neuropathol Appl Neurobiol 2013, 39:593–611. 10.1111/nan.12042CrossRefPubMed
Metadaten
Titel
Kaolin-induced chronic hydrocephalus accelerates amyloid deposition and vascular disease in transgenic rats expressing high levels of human APP
verfasst von
Gerald D Silverberg
Miles C Miller
Crissey L Pascale
Ilias N Caralopoulos
Yuksel Agca
Cansu Agca
Edward G Stopa
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Fluids and Barriers of the CNS / Ausgabe 1/2015
Elektronische ISSN: 2045-8118
DOI
https://doi.org/10.1186/2045-8118-12-2

Weitere Artikel der Ausgabe 1/2015

Fluids and Barriers of the CNS 1/2015 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

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.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.