Skip to main content
Erschienen in: Translational Stroke Research 3/2020

Open Access 30.10.2019 | Original Article

Progression and Classification of Granular Osmiophilic Material (GOM) Deposits in Functionally Characterized Human NOTCH3 Transgenic Mice

verfasst von: Gido Gravesteijn, Leon P. Munting, Maurice Overzier, Aat A. Mulder, Ingrid Hegeman, Marc Derieppe, Abraham J. Koster, Sjoerd G. van Duinen, Onno C. Meijer, Annemieke Aartsma-Rus, Louise van der Weerd, Carolina R. Jost, Arn M. J. M. van den Maagdenberg, Julie W. Rutten, Saskia A. J. Lesnik Oberstein

Erschienen in: Translational Stroke Research | Ausgabe 3/2020

Abstract

CADASIL is a NOTCH3-associated cerebral small vessel disease. A pathological ultrastructural disease hallmark is the presence of NOTCH3-protein containing deposits called granular osmiophilic material (GOM), in small arteries. How these GOM deposits develop over time and what their role is in disease progression is largely unknown. Here, we studied the progression of GOM deposits in humanized transgenic NOTCH3Arg182Cys mice, compared them to GOM deposits in patient material, and determined whether GOM deposits in mice are associated with a functional CADASIL phenotype. We found that GOM deposits are not static, but rather progress in ageing mice, both in terms of size and aspect. We devised a GOM classification system, reflecting size, morphology and electron density. Six-month-old mice showed mostly early stage GOM, whereas older mice and patient vessels showed predominantly advanced stage GOM, but also early stage GOM. Mutant mice did not develop the most severe GOM stage seen in patient material. This absence of end-stage GOM in mice was associated with an overall lack of histological vascular pathology, which may explain why the mice did not reveal functional deficits in cerebral blood flow, cognition and motor function. Taken together, our data indicate that GOM progress over time, and that new GOM deposits are continuously being formed. The GOM staging system we introduce here allows for uniform GOM deposit classification in future mouse and human studies, which may lead to more insight into a potential association between GOM stage and CADASIL disease severity, and the role of GOM in disease progression.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1007/​s12975-019-00742-7) contains supplementary material, which is available to authorized users.
Julie W. Rutten and Saskia A. J. Lesnik Oberstein shared last authorship.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Deposition of granular osmiophilic material (GOM) is the vascular pathological hallmark of CADASIL, which is the most prevalent hereditary small vessel disease [1] and is caused by missense mutations in the NOTCH3 gene [2, 3]. GOM have been shown to contain NOTCH3 ectodomain (NOTCH3ECD) and extracellular matrix proteins [46], and can be visualized ultrastructurally in the tunica media of small arteries and capillaries. These electron dense GOM deposits are located in the basement membrane of mural cells, i.e. vascular smooth muscle cells and pericytes [711]. In both manifest and pre-manifest CADASIL patients, GOM deposits are present not only in brain vessels, but also in vessels of other organs, such as the skin [1113]. Other CADASIL-associated vascular pathology includes mural cell degeneration, smooth muscle actin (SMA)-positive (neo)intima formation, fibrosis and vessel wall thickening [1419]. These vascular alterations are associated with compromised cerebrovascular reactivity (CVR) [20, 21] and reduced cerebral blood flow (CBF), and eventually lead to mid-adult onset of recurrent strokes, vascular cognitive impairment and ultimately dementia [1]. Brain MRI reveals progressive symmetrical white matter hyperintensities, lacunes, microbleeds and brain atrophy [1].
We have previously described that our humanized CADASIL transgenic NOTCH3Arg182Cys mouse model, which overexpresses human mutant NOTCH3 protein from a genomic construct, shows granular NOTCH3ECD immunostaining as early as 4 weeks of age, while GOM deposits first appear around 6 months of age [22]. However, little is known about how GOM deposits evolve over time and what their relation is to other CADASIL-associated vascular pathology and vascular dysfunction. Here, we performed a longitudinal study of GOM pathology in the transgenic NOTCH3Arg182Cys mice. In addition, we assessed cerebrovascular, motor and cognitive function in these mice.

Methods

Mice

Transgenic mice were used that harbour the human full-length NOTCH3 gene (located on a 143 kb BAC construct) in either the wild-type or the mutant (c.544C>T, p.Arg182Cys) form, generated on a C57BL/6J background [22]. Mice were bred at the animal facility of the Leiden University Medical Center and housed individually under standard conditions, i.e. a 12-h light/dark cycle with food and water available ad libitum.
Three different mouse strains were used, with various human NOTCH3 expression levels: 100% for wild-type mice (tgN3WT100), and 100% and 350% for mutant mice (tgN3MUT100 and tgN3MUT350, respectively) [22]. Non-transgenic littermates were used as additional controls. A prospective study with 6–8 mice per group was performed to study body weight and motor function at various time points (1.5, 3, 6, 12, 16 and 20 months), and cerebral hemodynamics, cognition and immunohistochemical staining was studied at 20 months. Three mice had to be sacrificed before the end of the study; one due to an eye infection (tgN3WT100, at 15 months), one due to having a wound on its back (tgN3MUT350, at 19 months) and one due to low body weight (tgN3MUT100, at 20 months). In addition to the prospective study, tgN3MUT350 mice were sacrificed at the age of 1.5 (n = 1), 3 (n = 1), 6 (n = 2), 12 (n = 2) and 20 (n = 2) months for electron microscopy (EM) studies.

Electron Microscopy

In addition to mouse brain, post-mortem brain tissue was obtained from three CADASIL patients (deceased at age 59, 66 and 69 years) for comparison with human pathology. Mouse (frontal lobe grey matter) and human (frontal lobe grey matter) brain tissue was fixed overnight at 4 °C in 1.5% glutaraldehyde and 1% paraformaldehyde (pH = 7.4). Tissue blocks of ≤ 1 mm3 were post-fixated for 90 min in 2% osmium tetroxide and 2% potassium ferrocyanide after filtrating the post-fixative through a 0.2-μm filter. After post-fixation, the tissue was washed for 30 min in MilliQ and dehydrated in a series of ethanol (70%, 80% and 90%) for 30 min each and twice for 1 h in 100% ethanol. Blocks were incubated for 10 min in propylene oxide, 2 h in propylene oxide and Epon LX-112 (1:1) and finally for 2 h in propylene oxide and epon LX-112 (1:2). Subsequently, the epon was polymerized for 48 h at 70 °C.
One-micrometre-thick sections were checked for the presence of blood vessels by light microscopy. Then, areas with high blood vessel density were selected for further analysis with EM, i.e. 80-nm sections were collected on a one hole grid and subsequently stained with uranyl acetate and lead citrate. Images were acquired with a digital camera (One View, Gatan Inc., Pleasanton, CA) mounted on a 120 kV transmission electron microscope (Tecnai T12 with a twin objective lens, Fei Inc., Hillsburough, OR). Overviews of relatively large regions on the specimen that contained abundant numbers of cross-sections of vessels were collected by stitching many individual images together (40,000–60,000 nm2) using software described earlier [23]. Stitched images were examined using Aperio ImageScope (version 10.0.35).

GOM Analysis

The number of GOM deposits was counted per vessel and expressed as counts per 100 μm vessel circumference. Vessel circumference was approximated using the formula of oval circumference (Ramanujan’s approximation for ellipse circumference = π[3(a + b) − √((3a + b) × (a + 3b))]) where a and b were defined as the major diameter (a) and minor diameter (b) for each vessel between endothelial basement membranes. GOM deposits were studied in vessels with a minor diameter < 8 μm, referred to as microvessels, as these were the most abundant in the stitched images. Twenty-three to 84 microvessels were studied per time point (1.5, 3, 6, 12 and 20 months). Width of the basement membrane was determined averaging 40 measurements in two ntg mice and two tgN3MUT350 mice each. In the human brain sample, 21 microvessels were analysed. Also, the GOM deposit area was measured using ImageJ after manually drawn region-of-interests around GOM deposits.

Immunohistochemistry

Two 5-μm coronal frontal brain sections per mouse (approximately at the height of the infundibulum) and human brain sections of frontal white matter were analysed with immunohistochemistry. NOTCH3ECD staining was performed as described before [22]. Smooth muscle actin (SMA) staining was performed after pre-treatment with trypsin for 30 min at 37 °C and washed three times for 5 min with PBS. The primary antibody (Alpha-Smooth Muscle Actin, 1:4000, goat polyclonal, NB300-978, Novus Biologicals) was incubated overnight at room temperature. The secondary antibody (Rabbit Anti-Goat IgG, biotinylated, 1:400, Jackson Immunoresearch Lab. Inc.) was incubated for 1 h at room temperature and developed with the Vectastain Elite ABC HRP Kit (PK-6100, Vectorlabs) for 30 min at room temperature. Finally, slices were stained with 0.05% 3,3′-diaminobenzidine (DAB, Sigma) supplemented with 0.0045% H2O2 for 10 min and stained with Harris’ haematoxylin solution (diluted 1:3, Merck) for 5 s. Sections were Verhoeff-Van Gieson and Periodic Schiff acid stained using the Artisan Link Pro staining machine (DAKO, Agilent), and Van Gieson stained as described previously [24]. Sections were stained with Klüver-Barrera luxol fast blue to quantify white matter vacuolization (Supplementary Methods 4).
Microscopy imaging was performed with the Keyence BZ-X710 (Keyence). Using 20 times magnification, full colour images were taken of the complete section at 1-ms capture time. Images were stitched by Keyence BZ-X Analyzer software version 1.3.0.3 to obtain one high resolution full brain image per section. The SMA positive area was determined using a Colour Threshold (Hue 0–50; Saturation 0–255; Brightness 0–175) in ImageJ, and expressed as percentage of total brain surface of the section.

Neuroimaging, Cerebrovascular Reactivity, Cognition and Motor Function

At the age of 20 months, neuroimaging (T2W, FLAIR, SWI, cerebral hemodynamics) was performed under medetomidine anaesthesia using a 7 Tesla MRI (Bruker PharmaScan), see also Supplementary Methods 1. In short, absolute cerebral blood flow (CBF) was measured using arterial spin labelling (ASL)-MRI during a 7-min baseline and a 7-min CO2 challenge, using the measured signal difference between the labelled and control images in three brain slices. Absolute CBF at baseline and absolute CBF at challenge were quantified during the last 140 s of baseline and challenge, respectively. Absolute CBF increase was calculated as well as the cerebrovascular reactivity (CVR), which was defined as the relative cerebral blood flow (CBF) increase. One non-transgenic mouse was excluded from neuroimaging analysis due to a poor hemodynamic response to anaesthesia.
A Morris Water Maze protocol was used to assess cognitive function in 20-month-old mice, starting 2 weeks before neuroimaging assessment (Supplementary Methods 2). In short, during the training phase, mice were trained to find a hidden platform in the north-west quadrant of a circular swimming pool, while at the reversal training phase, mice were retrained to find a hidden platform in the south-east quadrant. Path length of the mice was determined between release in the pool and finding the hidden platform. Motor function was determined by analysing speed on a rotarod, on a beam and during swimming in the Morris water maze (Supplementary Methods 3). Motor and cognitive mouse experiments were performed by the same experienced researcher.

Statistical Analyses

Differences between groups were analysed using one-way ANOVA analyses with Tukey’s post-hoc correction. The increase in GOM area over time, as well as the association between baseline CBF and CVR were analysed using simple linear regression. All statistical analyses were two-sided tests with threshold for statistical significance of 0.05, using the IBM SPSS Statistics version 23.0.0.2 software.

Results

Temporal GOM Assessment

GOM appearance, size and count were studied in brain microvessels of tgN3MUT350 mice at the age of 1.5, 3, 6, 12 and 20 months. GOM deposits were first observed at the age of 6 months, appearing as small, round deposits in the basement membrane of mural cells, which were sometimes only slightly more electron dense than the surrounding basement membrane (Fig. 1a). At 12 months, most GOM deposits were larger and more electron dense. At 20 months, large amorphous GOM deposits were observed, which spanned the full width of the basement membrane (Fig. 1b). Irrespective of age, GOM deposits were predominantly (85%) located on the abluminal side of mural cells, bulging out the basement membrane and thereby leaving an indentation in the adjacent mural cell. Between 6 and 20 months, GOM size increased (b = 0.0030 μm2/month, P = 0.002) (Fig. 1c), GOM count more than doubled (0.9 to 2.4 GOM/100 μm) and the percentage of GOM-positive vessels increased from 26 to 39%. Based on these observations, we categorized GOM deposits into various stages that reflect relative size, morphology, electron density and the GOM deposit-induced bulging of the basement membrane with concomitant indentation of adjacent mural cells (Fig. 2a). Stage I GOM deposits are small and slightly electron dense and induce minimal mural cell indentation. Stage II and III GOM deposits are more electron dense, induce basement membrane bulging with mural cell indentation and are within (II) or extend beyond (III) the normal width of the basement membrane (~ 150 nm). Stage IV GOM deposits extend beyond the normal width of the basement membrane and are amorphous. At 6 months of age, stage I–III GOM deposits were observed in tgN3MUT350 mice. At 12 months, the first stage IV GOM appeared. At 20 months, the majority of the GOM were at stages III and IV (Fig. 2b), but stage I GOM deposits were also observed. This suggests that new GOM deposits are continuously being formed, and that once formed, the initially small round GOM deposits progress over time into large amorphous deposits.
Next, we compared GOM in brain tissue of three CADASIL patients to GOM in 20-month-old tgN3MUT350 mice (Fig. 1d). Almost all (96%) of the analysed microvessels in the patients contained GOM deposits, whereas GOM deposits were observed in only 39% of microvessels in the mutant mice. In human brain material, like in mice, GOM deposits of all stages were observed, but stage IV GOM deposits were most frequent (Fig. 2c). In addition, the patients’ microvessels contained large confluent patches of GOM (stage V GOM) that were not observed in the mice. Of note, the electron density of GOM deposits in the patients’ microvessels was less homogeneous than of GOM in mice. GOM deposits in patient microvessels either bulged out of the basement membrane and thereby left an indentation in the adjacent mural cell, or were located further away from any recognisable mural cells within an overall thickened basement membrane. In contrast, GOM deposits in mice were always located close to the mural cell, often in indentations of the mural cell formed by the GOM deposits.

Other CADASIL-Associated Vessel Wall and Brain Parenchyma Changes

Despite large GOM deposits and extensive granular NOTCH3ECD staining at 20 months (Fig. 3a), tgN3MUT350 mice did not show basement membrane thickening (tgN3MUT350 0.145 ± 0.050 μm, ntg 0.148 ± 0.055 μm, P = 0.73) and there were no signs of mural degeneration, such as electron lucent vacuoles, large intracellular vesicles or a shrunken appearance (Fig. 1a, b). Also, there was no difference in the amount or pattern of SMA staining between mutant and wild-type mice (Fig. 3b, c). In addition, Van Gieson’s, Verhoeff-Van Gieson’s and Periodic acid-Schiff staining did not show vessel wall thickening, in contrast to our observations in vessels of the CADASIL patients (Fig. 3c). There was no difference in white matter vacuolization between wild-type and mutant mice (Supplementary Data 2).

Cerebral Blood Flow, Cerebrovascular Reactivity and Neuroimaging

Next, we assessed cerebral blood flow (CBF) dynamics in 20-month-old mice using ASL-MRI with CO2 as vasodilative stimulus (Fig. 4a). Compared to non-transgenic and wild-type mice, mutant mice (tgN3MUT100 and tgN3MUT350) did not show a significant difference in CVR, which was defined as relative CBF increase upon challenge (Fig. 4b, d), absolute CBF increase upon challenge (Fig. 4c, e) or in CBF at baseline (Fig. 4f). There was, however, a non-significant trend towards a reduced CVR and a slightly increased baseline CBF in the tgN3MUT350 mice. Differences in baseline CBF likely contributed to the observed differences in CVR, as there was an inverse correlation between baseline CBF and CVR (s = − 0.69, P < 0.001, Supplementary data 1). Separate analysis of CBF and CVR for cortex and subcortex showed similar results (Supplementary data 3, 4).
High-resolution T2W, FLAIR and SWI MRI scans of mouse brains did not show white matter hyperintensities, lacunes or microbleeds. Also, gadolinium enhancement showed the typical pattern of contrast enhancement in and around the ventricles, but no differences were observed between tgN3MUT350 and wild-type mice (Supplementary data 5).

Cognition and Motor Function

Motor function, as assessed by rotarod running, beam walk and swimming speed, did not differ between mutant and wild-type mice at any of the time points (Supplementary data 6). In the Morris water maze tests, mutant mice did not show signs of impaired memory formation. If anything, tgN3MUT350 mice performed slightly better at some of the tasks (Fig. 5).

Discussion

We investigated the development of GOM deposits over time in a transgenic mouse model of CADASIL, which overexpresses mutant human NOTCH3 protein from a large genomic construct. GOM deposits evolved with respect to size, morphology and number in microvessels of the mutant mouse brain. Here, we propose a five-stage GOM classification system to facilitate uniform analysis and description of GOM deposits and show that this staging can also be used to systematically classify GOM in vessels of CADASIL patient material.
The GOM deposits observed in aged mice (20 months) included stages I–IV, i.e. from small, circumscript deposits within the basement membrane (stage I), to large, amorphous GOM that induced bulging of the basement membrane (stage IV). As mice aged 6 months only showed stages I–III GOM, individual GOM deposits seem to increase in size and become increasingly amorphous over time, and new GOM seem to be continuously formed. This is further illustrated by the observation that GOM deposits of stage I and II are also present in post-mortem CADASIL patient brain microvessels, next to the more extensive GOM pathology, including patches of confluent GOM (stage V) [7, 25, 26].
Although many studies have reported the presence and morphology of GOM in CADASIL patients [9, 1113, 25, 2730], little is known about how GOM deposits progress over time. Brulin et al. analysed GOM in skin biopsies of CADASIL patients of different ages, and found that the number GOM deposits increase up to 50 years of age, but also found that the number of GOM seems to decrease in elderly patients [11]. The latter may either be attributed to other end-stage vessel wall changes hampering the visualization of GOM, or because GOM seem to become confluent and disintegrate over time [26]. Whether different GOM stages in skin biopsies of CADASIL patients are associated with disease severity and disease progression, therefore, remains to be determined. If so, the proposed GOM classification system may aid future efforts to monitor and predict disease progression at the individual patient level.
In our humanized CADASIL mouse model and in a rat Notch3 CADASIL mouse model, GOM deposits are observed several months after the first signs of NOTCH3ECD-positive granular immunostaining [22, 30], suggesting that NOTCH3ECD granules may act as seeds for GOM development. Since NOTCH3ECD aggregates attract extracellular matrix proteins that are components of GOM deposits, such as TIMP3 and clusterin [4, 5], there seems a direct temporal relation between NOTCH3ECD aggregates and the formation of GOM deposits. Delineating molecular differences between early- and end-stage GOM deposits may help to understand the sequence of events in CADASIL vascular pathology, and perhaps the identification of early therapeutic targets.
Whereas the CADASIL mice in our study seem to faithfully replicate early signs of disease pathology (NOTCH3ECD accumulation and GOM deposition), we did not observe other CADASIL-associated disease features which have been observed in other CADASIL mouse models, such as vessel wall thickening, changes in SMA staining, mural cell degeneration and blood-brain barrier leakage [7, 9, 14, 15, 17, 31]. In line with this, we previously showed that our mutant mice do not show brain parenchyma pathology at age 20 months, which we confirmed in this study using high-resolution T2W neuroimaging [22]. Furthermore, we extended the characterization of the mice with functional tests, which did not reveal any cognitive or motor dysfunction. Our cerebral blood flow studies showed a small, not significant, reduction in CVR in tgN3MUT350 mice, while studies in other, genetically different, CADASIL mouse models did show a reduced CVR [8, 30, 32]. It may be relevant that we used a different method to anaesthetize mice and also to measure CVR, namely an ASL-based MRI approach. Although less sensitive in detecting small CVR changes, the advantage of ASL-MRI is that it allows for absolute perfusion quantification and detection of differences in baseline CBF. In that way, we found that the slight reduction of CVR in tgN3MUT350 mice could at least partially be explained by a higher CBF at baseline. Future research is needed to determine whether the differences in CVR findings between this study and others [8, 30, 32] can be explained by differences in genetics of the mouse models, by differences in baseline perfusion states or by differences in the study set-up, including the anaesthesia protocol. Although we did not find evidence for overt functional cerebrovascular deficits in our mouse model, this humanized mouse model captures early markers of CADASIL pathology, making it suitable for therapeutic studies targeting human mutant NOTCH3ECD accumulation early in the disease course.
In summary, we show progression of GOM deposits in a humanized CADASIL mouse model. We propose a five-stage GOM classification system for uniform assessment of GOM depositions in translational research. In future pre-clinical studies of therapeutic approaches aimed at reducing or preventing NOTCH3ECD aggregation, GOM classification may serve as a valuable tool to monitor therapeutic efficiency on an ultrastructural level.

Acknowledgements

We acknowledge the financial support from the Netherlands Brain Foundation (HA2016-02-03). We thank Baudouin Denis de Senneville for his help with the MR image registration; Lydiane Hirschler and Emmanuel Barbier for their help with the pCASL sequence; Sandra van Heiningen and the LUMC animal facility for mouse breeding; and Peter Neeskens for electron microscopy sample preparation.

Compliance with Ethical Standards

Ethical Approval

All applicable international, national and/or institutional guidelines for the care and use of animals were followed. Experiments were approved by the local animal ethics committee (code 14073) and conducted in accordance with recommendations of the European Communities Council Directive (2010/63/EU) and ARRIVE guidelines. All efforts were made to minimize animal suffering. Except from using post-mortem material, this article does not contain any studies with human participants performed by any of the authors.

Conflict of interest

GG, JWR and SAJLO were supported by the Netherlands Brain Foundation (HA2016-02-03). SAJLO is co-inventor on an LUMC owned patent, not related to this work. LPM and LvdW were supported by NWO-VIDI (864.13.014). MO, AAM, IH, MD, AJK, SGvD, OCM, CRJ and AMJMvdM have nothing to disclose. AAR has nothing to disclose related to this work. For full transparency, she discloses being employed by LUMC and being co-inventor on several LUMC owned patents. As such she is entitled to a share of royalties. AAR further discloses being ad hoc consultant for PTC Therapeutics, BioMarin Pharmaceuticals Inc., Global Guidepoint, GLG consultancy, Grunenthal, Eisai, Sarepta Therapeutics and BioClinica, and having been a member of the Duchenne Network Steering Committee (BioMarin) and of the scientific advisory boards of ProQR and Philae Pharmaceuticals. Remuneration for these activities is paid to LUMC. LUMC also received speaker honoraria from PTC Therapeutics and BioMarin Pharmaceuticals.
Open Access This 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.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

Weitere Produktempfehlungen anzeigen
Anhänge

Electronic Supplementary Material

Literatur
1.
Zurück zum Zitat Chabriat H, Joutel A, Dichgans M, Tournier-Lasserve E, Bousser M-G. Cadasil. Lancet Neurol. 2009;8:643–53.CrossRef Chabriat H, Joutel A, Dichgans M, Tournier-Lasserve E, Bousser M-G. Cadasil. Lancet Neurol. 2009;8:643–53.CrossRef
2.
Zurück zum Zitat Joutel A, Vahedi K, Corpechot C, Troesch A, Chabriat H, Vayssière C, et al. Strong clustering and stereotyped nature of Notch3 mutations in CADASIL patients. Lancet. 1997;350:1511–5.CrossRef Joutel A, Vahedi K, Corpechot C, Troesch A, Chabriat H, Vayssière C, et al. Strong clustering and stereotyped nature of Notch3 mutations in CADASIL patients. Lancet. 1997;350:1511–5.CrossRef
3.
Zurück zum Zitat Joutel A, Corpechot C, Ducros A, Vahedi K, Chabriat H, Mouton P, et al. Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature. 1996;383:707–10.CrossRef Joutel A, Corpechot C, Ducros A, Vahedi K, Chabriat H, Mouton P, et al. Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature. 1996;383:707–10.CrossRef
4.
Zurück zum Zitat Ishiko A, Shimizu A, Nagata E, Takahashi K, Tabira T, Suzuki N. Notch3 ectodomain is a major component of granular osmiophilic material (GOM) in CADASIL. Acta Neuropathol. 2006;112:333–9.CrossRef Ishiko A, Shimizu A, Nagata E, Takahashi K, Tabira T, Suzuki N. Notch3 ectodomain is a major component of granular osmiophilic material (GOM) in CADASIL. Acta Neuropathol. 2006;112:333–9.CrossRef
5.
Zurück zum Zitat Monet-Leprêtre M, Haddad I, Baron-Menguy C, Fouillot-Panchal M, Riani M, Domenga-Denier V, et al. Abnormal recruitment of extracellular matrix proteins by excess Notch3 ECD: a new pathomechanism in CADASIL. Brain. 2013;136:1830–45.CrossRef Monet-Leprêtre M, Haddad I, Baron-Menguy C, Fouillot-Panchal M, Riani M, Domenga-Denier V, et al. Abnormal recruitment of extracellular matrix proteins by excess Notch3 ECD: a new pathomechanism in CADASIL. Brain. 2013;136:1830–45.CrossRef
6.
Zurück zum Zitat Arboleda-Velasquez JF, Manent J, Lee JH, Tikka S, Ospina C, Vanderburg CR, et al. Hypomorphic notch 3 alleles link notch signaling to ischemic cerebral small-vessel disease. Proc Natl Acad Sci. 2011;108:E128–35.CrossRef Arboleda-Velasquez JF, Manent J, Lee JH, Tikka S, Ospina C, Vanderburg CR, et al. Hypomorphic notch 3 alleles link notch signaling to ischemic cerebral small-vessel disease. Proc Natl Acad Sci. 2011;108:E128–35.CrossRef
7.
Zurück zum Zitat Yamamoto Y, Craggs LJL, Watanabe A, Booth T, Attems J, Low RWC, et al. Brain microvascular accumulation and distribution of the NOTCH3 ectodomain and granular osmiophilic material in CADASIL. J Neuropathol Exp Neurol. 2013;72:416–31.CrossRef Yamamoto Y, Craggs LJL, Watanabe A, Booth T, Attems J, Low RWC, et al. Brain microvascular accumulation and distribution of the NOTCH3 ectodomain and granular osmiophilic material in CADASIL. J Neuropathol Exp Neurol. 2013;72:416–31.CrossRef
8.
Zurück zum Zitat Ghosh M, Balbi M, Hellal F, Dichgans M, Lindauer U, Plesnila N. Pericytes are involved in the pathogenesis of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Ann Neurol. 2015;78:887–900.CrossRef Ghosh M, Balbi M, Hellal F, Dichgans M, Lindauer U, Plesnila N. Pericytes are involved in the pathogenesis of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Ann Neurol. 2015;78:887–900.CrossRef
9.
Zurück zum Zitat Gu X, Liu X-Y, Fagan A, Gonzalez-Toledo ME, Zhao L-R. Ultrastructural changes in cerebral capillary pericytes in aged Notch3 mutant transgenic mice. Ultrastruct Pathol. 2012;36:48–55.CrossRef Gu X, Liu X-Y, Fagan A, Gonzalez-Toledo ME, Zhao L-R. Ultrastructural changes in cerebral capillary pericytes in aged Notch3 mutant transgenic mice. Ultrastruct Pathol. 2012;36:48–55.CrossRef
10.
Zurück zum Zitat Joutel A, Andreux F, Gaulis S, Domenga V, Cecillon M, Battail N, et al. The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. J Clin Invest. 2000;105:597–605.CrossRef Joutel A, Andreux F, Gaulis S, Domenga V, Cecillon M, Battail N, et al. The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. J Clin Invest. 2000;105:597–605.CrossRef
11.
Zurück zum Zitat Brulin P, Godfraind C, Leteurtre E, Ruchoux M-M. Morphometric analysis of ultrastructural vascular changes in CADASIL: analysis of 50 skin biopsy specimens and pathogenic implications. Acta Neuropathol. 2002;104:241–8.CrossRef Brulin P, Godfraind C, Leteurtre E, Ruchoux M-M. Morphometric analysis of ultrastructural vascular changes in CADASIL: analysis of 50 skin biopsy specimens and pathogenic implications. Acta Neuropathol. 2002;104:241–8.CrossRef
12.
Zurück zum Zitat Tikka S, Mykknen K, Ruchoux MM, Bergholm R, Junna M, Pyhnen M, et al. Congruence between NOTCH3 mutations and GOM in 131 CADASIL patients. Brain. 2009;132:933–9.CrossRef Tikka S, Mykknen K, Ruchoux MM, Bergholm R, Junna M, Pyhnen M, et al. Congruence between NOTCH3 mutations and GOM in 131 CADASIL patients. Brain. 2009;132:933–9.CrossRef
13.
Zurück zum Zitat Morroni M, Marzioni D, Ragno M, Di Bella P, Cartechini E, Pianese L, et al. Role of Electron microscopy in the diagnosis of Cadasil syndrome: a study of 32 patients. PLoS One. 2013;8:6–11.CrossRef Morroni M, Marzioni D, Ragno M, Di Bella P, Cartechini E, Pianese L, et al. Role of Electron microscopy in the diagnosis of Cadasil syndrome: a study of 32 patients. PLoS One. 2013;8:6–11.CrossRef
14.
Zurück zum Zitat Ruchoux MM, Guerouaou D, Vandenhaute B, Pruvo J-PP, Vermersch P, Leys D. Systemic vascular smooth muscle cell impairment in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Acta Neuropathol. 1995;89:500–12.CrossRef Ruchoux MM, Guerouaou D, Vandenhaute B, Pruvo J-PP, Vermersch P, Leys D. Systemic vascular smooth muscle cell impairment in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Acta Neuropathol. 1995;89:500–12.CrossRef
15.
Zurück zum Zitat Kalimo H, Viitanen M, Amberla K, Juvonen V, Marttila R, Pöyhönen M, et al. CADASIL: hereditary disease of arteries causing brain infarcts and dementia. Neuropathol Appl Neurobiol. 1999;25:257–65.CrossRef Kalimo H, Viitanen M, Amberla K, Juvonen V, Marttila R, Pöyhönen M, et al. CADASIL: hereditary disease of arteries causing brain infarcts and dementia. Neuropathol Appl Neurobiol. 1999;25:257–65.CrossRef
16.
Zurück zum Zitat Miao Q, Paloneva T, Tuisku S, Roine S, Poyhonen M, Viitanen M, et al. Arterioles of the lenticular nucleus in CADASIL. Stroke. 2006;37:2242–7.CrossRef Miao Q, Paloneva T, Tuisku S, Roine S, Poyhonen M, Viitanen M, et al. Arterioles of the lenticular nucleus in CADASIL. Stroke. 2006;37:2242–7.CrossRef
17.
Zurück zum Zitat Gatti JR, Zhang X, Korcari E, Lee SJ, Greenstone N, Dean JG, et al. Redistribution of mature smooth muscle markers in brain arteries in cerebral autosomal dominant arteriopathy with subcortical infarcts and Leukoencephalopathy. Transl Stroke Res. 2019;10:160–9.CrossRef Gatti JR, Zhang X, Korcari E, Lee SJ, Greenstone N, Dean JG, et al. Redistribution of mature smooth muscle markers in brain arteries in cerebral autosomal dominant arteriopathy with subcortical infarcts and Leukoencephalopathy. Transl Stroke Res. 2019;10:160–9.CrossRef
18.
Zurück zum Zitat Yamamoto Y, Ihara M, Tham C, Low RWC, Slade JY, Moss T, et al. Neuropathological correlates of temporal pole white matter hyperintensities in CADASIL. Stroke. 2009;40:2004–11.CrossRef Yamamoto Y, Ihara M, Tham C, Low RWC, Slade JY, Moss T, et al. Neuropathological correlates of temporal pole white matter hyperintensities in CADASIL. Stroke. 2009;40:2004–11.CrossRef
19.
Zurück zum Zitat Dong H, Ding H, Young K, Blaivas M, Christensen PJ, Wang MM. Advanced intimal hyperplasia without luminal narrowing of leptomeningeal arteries in CADASIL. Stroke. 2013;44:1456–8.CrossRef Dong H, Ding H, Young K, Blaivas M, Christensen PJ, Wang MM. Advanced intimal hyperplasia without luminal narrowing of leptomeningeal arteries in CADASIL. Stroke. 2013;44:1456–8.CrossRef
20.
Zurück zum Zitat Pfefferkorn T, von Stuckrad-Barre S, Herzog J, Gasser T, Hamann GF, Dichgans M, et al. Reduced cerebrovascular CO(2) reactivity in CADASIL: a transcranial Doppler sonography study. Stroke. 2001;32:17–21.CrossRef Pfefferkorn T, von Stuckrad-Barre S, Herzog J, Gasser T, Hamann GF, Dichgans M, et al. Reduced cerebrovascular CO(2) reactivity in CADASIL: a transcranial Doppler sonography study. Stroke. 2001;32:17–21.CrossRef
21.
Zurück zum Zitat Chabriat H, Pappata S, Ostergaard L, Clark CA, Pachot-Clouard M, Vahedi K, et al. Cerebral hemodynamics in CADASIL before and after acetazolamide challenge assessed with MRI bolus tracking. Stroke. 2000;31:1904–12.CrossRef Chabriat H, Pappata S, Ostergaard L, Clark CA, Pachot-Clouard M, Vahedi K, et al. Cerebral hemodynamics in CADASIL before and after acetazolamide challenge assessed with MRI bolus tracking. Stroke. 2000;31:1904–12.CrossRef
22.
Zurück zum Zitat Rutten JW, Klever RR, Hegeman IM, Poole DS, Dauwerse HG, LAM B, et al. The NOTCH3 score: a pre-clinical CADASIL biomarker in a novel human genomic NOTCH3 transgenic mouse model with early progressive vascular NOTCH3 accumulation. Acta Neuropathol Commun. 2015;3:89.CrossRef Rutten JW, Klever RR, Hegeman IM, Poole DS, Dauwerse HG, LAM B, et al. The NOTCH3 score: a pre-clinical CADASIL biomarker in a novel human genomic NOTCH3 transgenic mouse model with early progressive vascular NOTCH3 accumulation. Acta Neuropathol Commun. 2015;3:89.CrossRef
23.
Zurück zum Zitat Faas FGA, Cristina Avramut M, van den Berg BM, Mieke Mommaas A, Koster AJ, Ravelli RBG. Virtual nanoscopy: generation of ultra-large high resolution electron microscopy maps. J Cell Biol. 2012;198:457–69.CrossRef Faas FGA, Cristina Avramut M, van den Berg BM, Mieke Mommaas A, Koster AJ, Ravelli RBG. Virtual nanoscopy: generation of ultra-large high resolution electron microscopy maps. J Cell Biol. 2012;198:457–69.CrossRef
24.
Zurück zum Zitat Dodds HM, Clark G. An improved Van Gieson. Stain Technol. 1985;60:55.CrossRef Dodds HM, Clark G. An improved Van Gieson. Stain Technol. 1985;60:55.CrossRef
25.
Zurück zum Zitat Lewandowska E, Dziewulska D, Parys M, Pasennik E. Ultrastructure of granular osmiophilic material deposits (GOM) in arterioles of CADASIL patients. Folia Neuropathol. 2011;49:174–80.PubMed Lewandowska E, Dziewulska D, Parys M, Pasennik E. Ultrastructure of granular osmiophilic material deposits (GOM) in arterioles of CADASIL patients. Folia Neuropathol. 2011;49:174–80.PubMed
26.
Zurück zum Zitat Lewandowska E, Felczak P, Buczek J, Gramza K, Rafałowska J. Blood vessel ultrastructural picture in a CADASIL patient diagnosed at an advanced age. Folia Neuropathol. 2014;4:443–51.CrossRef Lewandowska E, Felczak P, Buczek J, Gramza K, Rafałowska J. Blood vessel ultrastructural picture in a CADASIL patient diagnosed at an advanced age. Folia Neuropathol. 2014;4:443–51.CrossRef
27.
Zurück zum Zitat Ruchoux MM, Domenga V, Brulin P, Maciazek J, Limol S, Tournier-Lasserve E, et al. Transgenic mice expressing mutant Notch3 develop vascular alterations characteristic of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Am J Pathol. 2003;162:329–42.CrossRef Ruchoux MM, Domenga V, Brulin P, Maciazek J, Limol S, Tournier-Lasserve E, et al. Transgenic mice expressing mutant Notch3 develop vascular alterations characteristic of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Am J Pathol. 2003;162:329–42.CrossRef
28.
Zurück zum Zitat Monet M, Domenga V, Lemaire B, Souilhol C, Langa F, Babinet C, et al. The archetypal R90C CADASIL-NOTCH3 mutation retains NOTCH3 function in vivo. Hum Mol Genet. 2007;16:982–92.CrossRef Monet M, Domenga V, Lemaire B, Souilhol C, Langa F, Babinet C, et al. The archetypal R90C CADASIL-NOTCH3 mutation retains NOTCH3 function in vivo. Hum Mol Genet. 2007;16:982–92.CrossRef
29.
Zurück zum Zitat Monet-Lepretre M, Bardot B, Lemaire B, Domenga V, Godin O, Dichgans M, et al. Distinct phenotypic and functional features of CADASIL mutations in the Notch3 ligand binding domain. Brain. 2009;132:1601–12.CrossRef Monet-Lepretre M, Bardot B, Lemaire B, Domenga V, Godin O, Dichgans M, et al. Distinct phenotypic and functional features of CADASIL mutations in the Notch3 ligand binding domain. Brain. 2009;132:1601–12.CrossRef
30.
Zurück zum Zitat Joutel A, Monet-Leprêtre M, Gosele C, Baron-menguy C, Hammes A, Schmidt S, et al. Cerebrovascular dysfunction and microcirculation rarefaction precede white matter lesions in a mouse genetic model of cerebral ischemic small vessel disease. J Clin Invest. 2010;120:433–45.CrossRef Joutel A, Monet-Leprêtre M, Gosele C, Baron-menguy C, Hammes A, Schmidt S, et al. Cerebrovascular dysfunction and microcirculation rarefaction precede white matter lesions in a mouse genetic model of cerebral ischemic small vessel disease. J Clin Invest. 2010;120:433–45.CrossRef
31.
Zurück zum Zitat Miao Q, Paloneva T, Tuominen S, Pöyhönen M, Tuisku S, Viitanen M, et al. Fibrosis and stenosis of the long penetrating cerebral arteries: the cause of the white matter pathology in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Brain Pathol. 2006;14:358–64.CrossRef Miao Q, Paloneva T, Tuominen S, Pöyhönen M, Tuisku S, Viitanen M, et al. Fibrosis and stenosis of the long penetrating cerebral arteries: the cause of the white matter pathology in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Brain Pathol. 2006;14:358–64.CrossRef
32.
Zurück zum Zitat Lacombe P, Oligo C, Domenga V, Tournier-Lasserve E, Joutel A. Impaired cerebral vasoreactivity in a transgenic mouse model of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy arteriopathy. Stroke. 2005;36:1053–8.CrossRef Lacombe P, Oligo C, Domenga V, Tournier-Lasserve E, Joutel A. Impaired cerebral vasoreactivity in a transgenic mouse model of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy arteriopathy. Stroke. 2005;36:1053–8.CrossRef
Metadaten
Titel
Progression and Classification of Granular Osmiophilic Material (GOM) Deposits in Functionally Characterized Human NOTCH3 Transgenic Mice
verfasst von
Gido Gravesteijn
Leon P. Munting
Maurice Overzier
Aat A. Mulder
Ingrid Hegeman
Marc Derieppe
Abraham J. Koster
Sjoerd G. van Duinen
Onno C. Meijer
Annemieke Aartsma-Rus
Louise van der Weerd
Carolina R. Jost
Arn M. J. M. van den Maagdenberg
Julie W. Rutten
Saskia A. J. Lesnik Oberstein
Publikationsdatum
30.10.2019
Verlag
Springer US
Erschienen in
Translational Stroke Research / Ausgabe 3/2020
Print ISSN: 1868-4483
Elektronische ISSN: 1868-601X
DOI
https://doi.org/10.1007/s12975-019-00742-7

Weitere Artikel der Ausgabe 3/2020

Translational Stroke Research 3/2020 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Neu im Fachgebiet Neurologie

Update Neurologie

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