• Non ci sono risultati.

Brain angioarchitecture and intussusceptive microvascular growth in a murine model of Krabbe disease

N/A
N/A
Protected

Academic year: 2021

Condividi "Brain angioarchitecture and intussusceptive microvascular growth in a murine model of Krabbe disease"

Copied!
12
0
0

Testo completo

(1)

O R I G I N A L P A P E R

Brain angioarchitecture and intussusceptive microvascular

growth in a murine model of Krabbe disease

Arianna Giacomini1•Maximilian Ackermann2•Mirella Belleri1•Daniela Coltrini3• Beatrice Nico4•Domenico Ribatti4,5•Moritz A. Konerding2•Marco Presta1,3• Marco Righi6,7,8

Received: 19 May 2015 / Accepted: 13 August 2015 / Published online: 27 August 2015 Ó Springer Science+Business Media Dordrecht 2015

Abstract Defects of the angiogenic process occur in the brain of twitcher mouse, an authentic model of human Krabbe disease caused by genetic deficiency of lysosomal b-galactosylceramidase (GALC), leading to lethal neuro-logical dysfunctions and accumulation of neurotoxic psy-chosine in the central nervous system. Here, quantitative computational analysis was used to explore the alterations of brain angioarchitecture in twitcher mice. To this aim, customized ImageJ routines were used to assess calibers, amounts, lengths and spatial dispersion of CD31?vessels

in 3D volumes from the postnatal frontal cortex of twitcher animals. The results showed a decrease in CD31 immunoreactivity in twitcher brain with a marked reduc-tion in total vessel lengths coupled with increased vessel fragmentation. No significant changes were instead observed for the spatial dispersion of brain vessels throughout volumes or in vascular calibers. Notably, no CD31?vessel changes were detected in twitcher kidneys in which psychosine accumulates at very low levels, thus confirming the specificity of the effect. Microvascular corrosion casting followed by scanning electron micro-scopy morphometry confirmed the presence of significant alterations of the functional angioarchitecture of the brain cortex of twitcher mice with reduction in microvascular density, vascular branch remodeling and intussusceptive angiogenesis. Intussusceptive microvascular growth, con-firmed by histological analysis, was paralleled by alter-ations of the expression of intussusception-related genes in twitcher brain. Our data support the hypothesis that a marked decrease in vascular development concurs to the onset of neuropathological lesions in twitcher brain and suggest that neuroinflammation-driven intussusceptive responses may represent an attempt to compensate impaired sprouting angiogenesis.

Keywords Angioarchitecture Brain  Computational analysis  Corrosion casting  Neurodegenerative Krabbe disease  Intussusceptive angiogenesis

Introduction

Globoid cell leukodystrophy (GLD), or Krabbe disease, is an autosomal recessive sphingolipidosis caused by the genetic deficiency of the lysosomal hydrolase Electronic supplementary material The online version of this

article (doi:10.1007/s10456-015-9481-6) contains supplementary material, which is available to authorized users.

& Marco Presta marco.presta@unibs.it & Marco Righi

m.righi@in.cnr.it

1 Unit of Experimental Oncology and Immunology, University

of Brescia, Brescia, Italy

2 Institute of Functional and Clinical Anatomy, University

Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany

3 Unit of Histology, Department of Molecular and

Translational Medicine, University of Brescia, viale Europa 11, 25123 Brescia, Italy

4 Unit of Human Anatomy and Histology, Department of Basic

Biomedical Sciences, University of Bari, Bari, Italy

5 National Cancer Institute ‘‘Giovanni Paolo II’’, Bari, Italy 6 CNR – Institute of Neuroscience, Milan, Italy

7 Department of Medical Biotechnology and Translational

Medicine, University of Milano, Milan, Italy

8 Consiglio Nazionale delle Ricerche, Institute of

Neuroscience, Via Vanvitelli 32, 20129 Milan, Italy DOI 10.1007/s10456-015-9481-6

(2)

b-galactosylceramidase (GALC) [1]. The disease is char-acterized by degeneration of oligodendroglia and progres-sive demyelination of the peripheral and central nervous system (CNS). Clinically, GLD manifests in early infancy and results in a severe neurological dysfunction that often leads to death by 2 years of age [2–4]. GALC degrades galactosylceramide (a major component of myelin) and other terminal b-galactose-containing sphingolipids, including b-galactosylsphingosine (psychosine). Psy-chosine is a bioactive water-soluble sphingolipid with cytotoxic properties: normally undetectable; this sphingoid base accumulates at micromolar concentrations in the CNS of GLD patients and it is strongly implicated in the molecular pathogenesis of the disease [5]. At present, the only clinical treatment for GLD is bone marrow or umbilical cord blood cell transplantation for late-onset and presymptomatic patients [3, 6]. Thus, understanding the molecular pathogenesis of GLD remains a high priority from a clinical standpoint.

Brain is a highly vascularized organ, total length of blood capillaries in adult human brain being equal to approximately 6.5 9 105m [7]. Accordingly, angiogenesis plays an important role in the development of CNS and in neurological disorders [8, 9]. The tight cross-talk among glial, neuronal and endothelial cells in CNS [9] is under-lined by the capacity of angiogenic factors, including vascular endothelial growth factor-A (VEGF) and fibrob-last growth factor-2 (FGF2), to modulate neurogenesis and neuroprotection. In turn, neurotrophic factors may regulate angiogenesis. Thus, vascular alterations appear to be implicated in the pathogenesis of stroke and neurodegen-erative disorders, including Alzheimer’s disease, Parkin-son’s disease, multiple sclerosis and amyotrophic lateral sclerosis [7–10].

Recently, the effects of GALC deficiency on CNS microvascularization and angiogenesis have been investi-gated in twitcher mice, an authentic murine model of GLD harboring a naturally occurring premature stop codon in the murine GALC gene [11]. GALC deficiency, with consequent psychosine accumulation, induces significant defects in the endothelium of the postnatal brain of twitcher mice. More-over, twitcher endothelium shows a progressively reduced capacity to respond to pro-angiogenic factors. In addition, RNA interference-mediated GALC gene silencing hampers the pro-angiogenic response of human endothelial cells to VEGF. Accordingly, microvascular alterations occur in a cortical brain biopsy from a GLD patient [11]. These observations suggest that GALC deficiency affects not only the glial/neuronal compartment of the neurovascular brain unit but also its vascular moiety. Thus, given the interactions between neurogenesis and angiogenesis and the pivotal role played by neovascularization in postnatal neuroprotection, alterations in blood vessel development are likely to

contribute to the injury occurring in the nervous system of GLD patients during early infancy.

In this report, in order to get further insights about blood vessel alterations consequent to GALC deficiency, we performed a quantitative assessment of the 3D microvas-cular architecture of the postnatal brain cortex of twitcher mice by computational analysis of CD31-immunostained histological sections [12] and by microvascular corrosion casting followed by scanning electron microscopy (SEM) morphometry [13]. The results highlight significant alter-ations in the angioarchitecture of the frontal cortex of twitcher mice with a uniform, generalized reduction in microvascular density. In parallel, vascular remodeling and intussusceptive angiogenesis were observed in twitcher brain as a possible mechanism of vascularization alterna-tive to impaired sprouting angiogenesis.

Methods

Mice

Breeder twitcher heterozygous mice (C57BL/6 J; Jackson Laboratories, ME, USA) were maintained under standard housing conditions. Animal handling protocols were in accordance with Italian institutional guidelines for animal care and use. Twitcher mutation was determined by poly-merase chain reaction (PCR) on DNA extracted from clipped tails [14]. In all the experiments, littermate wild-type (wt) and homozygous (twi/twi) animals were used at postnatal days 34–36 (P34–P36).

Immunostaining of brain microvasculature

Brains from P34 to P36 wt and twi/twi mice were either fixed overnight at 4°C in 4 % paraformaldehyde (PFA) or frozen. For histological analysis of endothelial intraluminal tissue folds, sections (8-lm-thick) of PFA-fixed paraffin-embedded samples were stained with an aqueous solution of 0.1 % toluidine blue. For 3D analysis, PFA-fixed 6 % agarose-embedded samples were cut at a thickness of 100 lm using a vibratome. Free-floating sections were first blocked in 2 % BSA (bovine serum albumine) and 0.2 % Triton X-100 for 45 min at RT and then incubated over-night at 4°C with an anti-CD31 antibody (Dianova GmbH, Hamburg, Germany, EU) diluted 1:50 in PBS containing 0.2 % BSA and 0.1 % Triton X-100. After several rinses in PBS, sections were incubated overnight at 4°C with sec-ondary AlexaFluor594-conjugated antibody (Invitrogen, Carlsbad, CA, USA) diluted 1:200 in PBS containing 0.1 % Triton X-100. For double-immunofluorescent anal-ysis, sections (4-lm-thick) of frozen brains were fixed with

(3)

acetone (for 5 min at 4°C), rinsed with PBS, and blocked with 2 % BSA in PBS. Sections were first incubated for 1 h at RT with the anti-CD31 antibody and for 30 min at RT with the secondary AlexaFluor594-conjugated antibody. After several rinses in PBS, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining (Roche, Milano, Italy, EU) was performed according to the manufacturer’s instructions to highlight apoptotic cells that were detected in the green channel. In order to analyze pericyte coverage, CD31-stained sections were incubated for 1 h at RT with an anti-desmin antibody (Dako, Milano, Italy, EU). After washing with PBS, sections were incu-bated for 30 min at RT with the appropriate secondary AlexaFluor488-conjugated antibody (Invitrogen). After further rinsing in PBS, fluorescent-stained sections were mounted in a drop of anti-bleaching mounting medium (Vectashield, Vector Laboratories, Burlingame, CA, USA) and examined under an Axiovert 200 M microscope equipped with ApoTome optical sectioning device (Carl Zeiss, Oberkochen, Germany, EU).

Quantification of endothelial intraluminal tissue folds

Thirty transversally sectioned vessel profiles with a single layer of endothelial cells, either without or with a thin basement membrane, were randomly chosen for wt and twi/twi animals. Measurements were performed at 409 magnification using a computer-assisted image analysis system (AnalySIS, Olympus Italia). For each vessel, the number of connections of intraluminal tissue folds with the opposite vascular wall, expression of intussusceptive microvascular growth, was counted. They were recognized by moving the focus through the thickness of the histo-logical slide and counting only the strands completely crossing the lumen. Those appearing incomplete and cross sections of pillars within the vascular lumen were not considered. Within each sample, the number of lumen crossing endothelial folds per vessel was averaged to pro-vide a representative value of each parameter for that sample.

Image sampling, acquisition and pre-processing of CD31-immunostained samples

For each treatment, images were acquired from samples obtained from two independent experiments with two or more animals per group. At least six Z-series of vascular fields from different sections per animal group were acquired from 100-lm-thick samples using an Axiovert 200 M microscope equipped with ApoTome optical sec-tioning device (Carl Zeiss) with a 209 objective at 3.125 pixels/lm nominal resolution. In the vertical axis,

optical slices were 1 lm apart. Acquired images were scaled to 1.56 pixels/lm, cropped to 512 9 512 pixels and organized as 8-bit image stacks which were made isotropic using the ImageJ plug-in developed by Cooper (http:// rsbweb.nih.gov/ij/plugins/make-isotropic/index.html). The stacks were then converted to binary using a custom rou-tine devised to recover faint signals while discarding excessive random noise. Briefly, after a median filter, the variance of each single slice was used to calculate an integrated density value. If variance was higher than a fixed arbitrary threshold, all the signals from the slice were discarded as spurious. The resulting stack was then nor-malized, and background was subtracted (‘‘rolling ball’’ method) to obtain a ‘‘clean’’ stack. At this point, data in the stack were processed with the Renyi entropy algorithm to build a binary mask which was dilated by two cycles using standard mathematical morphology [15, 16]. This proce-dure gave a 3D binary map roughly reporting vessel positions as found in the original stack. Finally, the ‘‘clean’’ stack was filtered using the spatial map as a mask, and the recovered grayscale signal was converted to binary using the default algorithm present in ImageJ. As the very last step, each slice was filtered by size, keeping particles sized 1.6 lm2 (4 pixels) or more. This procedure and the arbitrary thresholds used were validated on a set of at least three image stacks for each experimental condition by visual inspection of a trained operator.

Voxel classification according to vessel caliber

Binary voxels were assigned to vascular components of different caliber using cross-sectional areas projected on Cartesian planes according to a previously described workflow [12]. After fill-up of hollow vessels, we classified the resulting voxels according to the area of the smallest cross section of the particle they belonged to. To this respect, we considered only cross sections defined by the three Cartesian planes passing through the voxel. This step allowed to built maps of caliber-filtered vessels in the analyzed volumes. Then, these maps were intersected with the input binary volume to obtain the final classification of input voxels.

Analyses of CD311percent volume and spatial dispersion

The endothelial percent volume of a binary sample was calculated as the ratio of all CD31? voxels to the total voxels of the volume. Then, to quantitate the spatial dis-persion, we considered the 3D fill-up of each volume using a mathematical morphology procedure we have previously validated [12]. Accordingly, we counted the number of dilation cycles needed to fill each volume up to 90 %

(4)

starting from the original distribution of CD31?voxels and following a rhombicuboctahedral expansion scheme. For each analysis, we compared only equal volumes and used the greatest amount of signal observed among all volumes to normalize the amount of input voxels. Thus, for each sample, the normalized result was obtained subtracting the number of dilation cycles needed to reach the normaliza-tion value from the overall number of cycles needed to fill 90 % of the volume.

Automatic quantification of vascular lengths

To obtain these data, we reduced filled vessels to their basic 3D skeletons using the ImageJ plug-in Skeletonize (2D/3D) (http:// imagejdocu.tudor.lu/doku.php?id=plugin:morphology:skeleto nize3d:start) developed by Arganda-Carreras and based on the decision tree algorithm created by Lee et al. [17]. The plug-in was applied recursively to each input image until no further skeletonization was observed. Then, taking advan-tage of the analytical ImageJ plugin Analyze Skeleton (2D/ 3D) (http://imagejdocu.tudor.lu/doku.php?id=plugin:analy sis:analyzeskeleton:start) developed by Arganda-Carreras et al. [18], we used a custom ImageJ S1 script to approximate vessel lengths (the script and its instructions/rationale are enclosed as Online Resource 1 and Online Resource 2, respectively). The sum of all contributes was then multiplied for the size of the voxel, to yield the total vascular length of the analyzed volume.

Automatic classification of skeletonized voxels and assessment of free ends

Quantification of skeletonized ends was performed using the ImageJ S1 script mentioned above applied to the ImageJ plug-in Analyze Skeleton (2D/3D) that provides also basic voxel classification (node, stalk or end). These preliminary results were integrated with a custom auto-matic test to state whether tentative ‘‘ends’’ and ‘‘nodes’’ should be really considered as such on the basis of their degree of connection with surrounding noncontiguous voxels. This step was performed considering a 3x3x3 voxel cube centered around each signal (black) voxel from skeletonized samples. For each cube, we counted the iso-lated groups of external black voxels connected through faces (6-based connectivity). Given that all external voxels connect with the central one, the center voxel was classi-fied as an ‘‘end’’ when we identiclassi-fied only one group of connected voxels (see the script rationale in Online Resource 2); the voxel was considered a ‘‘stalk’’ when connected to two groups and a ‘‘node’’ when connected with more than two noncontiguous voxels or groups of voxels. This approach eased the classification of end voxels involved in skeleton kinks. Script functions were integrated

with a basic 3D filter to ignore end-to-end vessels (and their ends) shorter than an arbitrary length, as calculated by the Analyze Skeleton (2D/3D) plug-in in voxel units. A folder with image files suitable for testing the S1 script can be provided by the authors on request.

Corrosion casting

P36 wt and twi/twi mice were thoracotomized under deep pentobarbital anesthesia. The left ventricle was cannulated, and the ascending aorta was entered with an olive-tipped cannula. The entire vasculature of the animal was thoroughly rinsed by perfusing with lukewarm saline (10–20 ml) and then fixed by perfusing with 2.5 % buffered glutaraldehyde (10–20 ml, 860 mosmol, pH 7.4). Finally, up to 15 ml of the polyurethane-based casting resin PU4ii (vasQtec, Zurich, Switzerland, EU) was gently perfused as a casting medium [19]. After complete polymerization in a lukewarm water bath, the brains were excised. The specimens were macerated in 5 % potassium hydroxide, rinsed, dried and mounted on stubs for scanning electron microscopy. The microvascular corrosion casts were viewed on after coating with gold in argon atmosphere with a Philips ESEM XL 30 scanning electron microscope (FEI, Eindhoven, Netherlands, EU). From all specimens, areas were recorded as stereo images using a tilt angle of 6°.

Morphometry

Pairs of stereo images with a tilt angle difference of 6° obtained using an eucentric specimen holder were used after digitization and 3D reconstruction with an image analysis program (Kontron KS 300, Kontron, Eching, Germany, EU) to calculate parameters describing the microvascular network architecture. For details of the reconstruction and calculation, see [20].

Quantitative RT-PCR (RT-qPCR) analysis

Brains from P35 wt and twi/twi mice were analyzed for the expression of the indicated genes by RT-qPCR, and data were normalized for Gapdh expression as described [21]. To this purpose, total RNA was extracted from frozen samples and contaminating DNA was digested using DNAse, following indications reported in RNeasy Micro Handbook (Qiagen, Valencia, CA, USA). Two micrograms of total RNA was retrotranscribed with MMLV reverse transcriptase (Invitrogen, Carlsbad, CA, USA) using ran-dom hexaprimers in a final 20 ll volume. Quantitative PCR was performed with a ViiATM 7 Real-Time PCR Detection System (Applied Biosystems) using a iQTM SYBR Green Supermix (Bio-Rad) according to the man-ufacturer’s instructions. The primers are listed in Supple-mentary Table S1 in Online Resource 3.

(5)

Software and statistical analyses

All the images from CD31?-stained samples were analyzed using ImageJ software v. 1.48. Vascular tree renderings were obtained with the ImageJ 3D viewer rendering tool. The ImageJ scripts already published and the ImageJ plug-in for 3D spatial dispersion can be found as supplementary information in [12] together with instructions for use (Sa_ImageJ_routines.pdf). Statistical analyses and tests were performed using the Prism software environment (GraphPad, La Jolla, CA, USA) and its basic statistical package. Student’s t test for unpaired data (two-tailed) was used to test the probability of significant differences between two groups of samples. Differences were consid-ered significant when P \ 0.05.

Results

Computational analysis of the 3D microvascular architecture of the brain cortex of twitcher mice Immunohistochemical analysis performed using the vas-cular markers FVIII/vWF and CD31 had shown a signifi-cant decrease in the density of capillary blood vessels in the brain cortex of twi/twi mice when compared to wt animals [11]. On this basis, in order to get further insights about blood vessel alterations consequent to GALC deficiency, we performed a quantitative assessment of the microvas-cular architecture of the postnatal brain cortex of twitcher mice by computational analysis of 3D reconstructions of CD31-immunostained histological sections.

To this aim, the vascularization of the frontal brain cortex of wt and twi/twi mice was quantified in a first set of experiments by analysis of Z-stacks images after CD31 immunostaining of free-floating, 100-lm-thick tissue sec-tions. We identified vessel walls by applying a custom threshold algorithm to isotropic grayscale stacks repre-senting nominal volumes of 327 9 327 9 70 lm3. Direct inspection (Fig.1a, b) and analysis of the volumes (Fig.1c) pointed out that the frontal cortex of twi/twi mice presented reduced CD31? vessels (expressed as CD31? percent volume) than wt controls. Conversely, we did not observe inhomogeneities upon analysis of vessel spatial dispersion in each volume (Fig.1d). The reduction in CD31? vessels in twitcher samples was paralleled by a significant decrease in total microvascular length per unit of tissue volume, as assessed after vessel skeletonization (Fig.1e). Also, direct inspection of twi/twi samples (Fig.1a, b) pointed out marked changes in the organization of CD31? voxels that appeared to represent a pool of fragmented vessels more than a well-organized vascular tree. In order to quantify these changes, we calculated the

number of free CD31?segment ends in each sample that were normalized for the total vascular length measured in the same sample after removal of isolated fragments shorter than 2.4–3.0 lm. The results, shown in Fig.1f, confirmed that the normalized amounts of free vascular ends were significantly increased in the cerebral tissue of twi/twi animals with respect to wt controls in both exper-iments. Finally, normalized CD31 stainings, defined as the amounts of CD31 signal per unit of vascular length, demonstrated the absence of significant differences in specific staining between twi/twi and wt samples (Fig. 1g). This indicates that the reduction in CD31?signal in twi/twi samples reflects a real paucity of blood vessels and not a reduction in their CD31 immunoreactivity. Similar results were obtained in a second independent experiment, con-firming the reproducibility of these observations (Supple-mentary Fig. S1a–e in Online Resource 3). Thus, this preliminary analysis suggested a homogeneous reduction in the vascularization of the frontal brain cortex of twitcher mice, extending previous indications obtained by standard histological techniques [11].

In order to deepen these observations, we continued sorting out CD31? voxels according to the approximate caliber of the microvessel to which they belonged [12]. To this aim, we classified image voxels considering cross-sectional surface classes ranging from 1.6 to about 95 lm2 (theoretical diameters between 1.4 and 11 lm assuming a circular cross section). In this process, we took advantage of the vascular maps of reconstructed filled microvessels to assign poorly connected voxels to their correct class (Fig.2a). Then, we quantified the differences in brain cortex vascularization by analyzing samples from twi/twi and wt mice to obtain the percent fraction of CD31?voxels in each volume for each class of vascular calibers. According to our classification, the majority of signal belonged to vessels with a diameter ranging between 4.0 and 5.5 lm for both healthy and diseased brains (Fig. 2b). However, twi/twi mice showed a marked drop in the amount of positive voxels for most class of calibers, with up to 45–50 % signal reduction for vessels with a diameter ranging between 2.8 and 5.5 lm (Table1). The statistical significance of these differences varied from class to class of calibers but was markedly significant for well-populated classes. Similar results were obtained in a second inde-pendent experiment (Supplementary Fig. S1f in Online Resource 3 and Table1).

To rule out the possibility that the observed differences in vascularization were the mere consequence of a different diffusion of the anti-CD31 antibody through the whole 100-lm-thick tissue sections, the computational analysis was repeated on the topmost 23 lm of all samples. Even under these experimental conditions, we obtained a similar distribution of signal losses for twi/twi samples in the two

(6)

independent experiments (Supplementary Table S2 in Online Resource 3).

Cytotoxic psychosine, present at high micromolar con-centrations in the CNS of patients with GLD and twitcher mice, is detectable at very low levels in GALC-deficient non-nervous tissues/organs (see [11] and references therein). On this basis, to investigate whether vascular alterations in twitcher mice were specific for cerebral tis-sue, we repeated these vascular analyses on kidney explants from wt and twi/twi animals. At variance with data from brain samples, no significant difference in the amount of CD31? vascular signals, total vascular length and normalized amounts of free vascular ends was

observed in the kidneys of control and diseased animals (Supplementary Fig. S2 in Online Resource 3).

Ultrastructural analysis of the frontal cortex vasculature of twi/twi mice had shown significant alterations of microvascular endothelial cells that were characterized by swollen glial endfeet, altered tight junctions, decrease in the expression of the tight junction-associated protein ZO-1 and increased permeability. Also, the microvasculature of brain biopsies obtained from a 2.5-year-old GLD patient showed an irregularly shaped Factor VIII? endothelium with discontinuous wrapping by smooth muscle actin cells [11]. Accordingly, the endothelium of the brain cortex of twitcher mice is characterized by the disorganization of Fig. 1 Vascular alterations in the brain of twitcher mice by

computational analysis. Z-series of vascular fields from 100-lm-thick CD31-immunostained sections of the frontal cortex from the brains of P35 wt and twitcher mice were acquired using an Axiovert 200 M microscope equipped with ApoTome optical sectioning device (Carl Zeiss) with a 920 objective at 3.125 pixels/lm nominal resolution. a Acquired images were scaled to 1.56 pixels/lm, cropped to 512 9 512 pixels and organized as 8-bit grayscale image stacks which were made isotropic using the ImageJ plug-in developed by Cooper (http://rsbweb.nih.gov/ij/plugins/make-isotropic/index.html). bThe 8-bit grayscale stacks were then converted to binary using a custom routine devised to recover faint signals while discarding excessive random noise (for more details see ‘‘Materials and meth-ods’’ section). c Quantification of vascular density expressed as per-cent ratio of CD31? voxels/total voxels (V %). d Quantification of

spatial dispersion expressed considering the number of expansion cycles needed to fill 90 % of the volume following a rhombicuboc-tahedral expansion scheme [12]. Sample normalization as described in ‘‘Materials and methods’’ section. Data are shown as classical box and whiskers graphs: the boxes extend from the 25 to the 75th per-centiles, the lines indicate the median values, and the whiskers indi-cate the range of values. e Quantification of oriented vascular lengths calculated in whole samples after vessel skeletonization (for more details see ‘‘Materials and methods’’ section). f Number of vascular ends normalized by the sum of the total oriented vascular lengths determined after vessel skeletonization. g Amounts of CD31?signal normalized by the sum of the total oriented vascular lengths deter-mined after vessel skeletonization. In each graph, data are the mean ± SD, n = 8; ***P \ 0.001; **P \ 0.01; n.s., not significant

(7)

perivascular desmin? pericytes, leading to a significant reduction in endothelial coverage (Fig.3), thus confirming that GALC deficiency results in profound alteration of the blood–brain barrier functionality.

SEM morphometry of the 3D microvascular architecture of the brain cortex of twitcher mice Microvascular corrosion casting followed by SEM mor-phometry allows a fine qualitative and quantitative char-acterization of the vascular networks restricted to patent vessels connected to the bloodstream [22]. On this basis, in order to validate the information provided by the compu-tational analysis of immunofluorescence data, microvas-cular corrosion casts of the brain cortex of P36 twi/twi mice were studied by SEM morphometry.

Morphometric analysis of corrosion casts (Fig.4) indi-cates that, when compared to wt mice, the patent microvasculature of the brain cortex of twi/twi animals shows a significant increase in the intervascular distance, a morphometric measure of vascular density, thus confirming the data obtained by computational analysis of CD31? microvasculature (see above). In addition, direct inspection of SEM images of corrosion casts indicated that twitcher microvasculature is characterized by dilated vessels with alterations in vessel diameter, vascular leakage and endothelial dehiscence (Fig.5a). Remarkably, vascular branch remodeling and intussusceptive angiogenesis, characterized by the presence of intussusceptive pillars, were observed in twitcher mice but not in control animals (Fig.5b). In keeping with these observations, microscopic analysis of coronal sections of the brain cortex of P36 twi/ twi mice stained with toluidine blue demonstrated the presence of microvessels with morphological aspects sug-gestive of intussusceptive microvascular growth in form of intraluminal tissue folds connecting the opposite vascular walls. These aspects were not recognizable in control specimens (Fig.5c). Accordingly, the average number of endothelial intraluminal tissue folds was equal to 3 ± 1 lumen-crossing pillars per vessel in twitcher mice and equal to 0 ± 0 in control animals (n = 30) (Fig. 5d). Fig. 2 Quantification of vascular CD31?signals classified by vessel

calibers. a Color-coded renderings of wt and twi/twi microvasculature after vessel classification by approximated cross section. Classes are identified by their theoretical diameters (in lm) as it follows: magenta 1.4–2.0, red 2.0–2.8, yellow 2.8–4.0, green 4.0–5.5, cyan 5.5–8, blue 8.0–11.0. b Quantification of CD31?signals for each classes of vessel

calibers from wt (closed color bars) and twi/twi samples (color-bordered open bars). Data are the mean ± SD, n = 8. Color codings as in panel a. ***P \ 0.001; **P \ 0.01; *P \ 0.05; n.s., not significant. (Color figure online)

Table 1 Percent vascular volume differences between wt and twi/twi brain cortex samples

Vessel diameter (lm) Experiment 1 Experiment 2

Loss in vol (%) Statistical significancea Loss in vol (%) Statistical significancea

1.4–2.0 8.2 n.s. -16.4 n.s. 2.0–2.8 33.4 P\ 0.05 29.5 P\ 0.05 2.8–4.0 44.5 P\ 0.001 51.2 P\ 0.01 4.0–5.5 48.7 P\ 0.001 68.5 P\ 0.001 5.5–8.0 33.5 n.s. 73.4 P\ 0.01 8.0–11.0 15.3 n.s. 64.2 n.s. n = 8 n = 6

Values are expressed as percent reduction among the different classes of vessels grouped according to their theoretical diameter in twi/twi with respect to wt samples as observed in two independent experiments

(8)

Expression of intussusceptive angiogenesis-related genes in the brain of twitcher mice

Intussusceptive angiogenesis is a morphogenetic, non-sprouting angiogenic process in which a single vessel is split into two lumens. A distinguishing anatomic feature of intussusceptive angiogenesis is the intussusceptive pillar, a transluminal tissue bridge that spans the vessel lumen. Physical expansion or growth of the pillar along the vessel axis divides the lumen resulting in vascular duplication [23]. Even though the molecular mechanisms leading to intussusceptive angiogenesis are not fully elucidated, recent observations have shown that inhibition of Notch signaling in already existing vascular beds induces exten-sive intussusceptive angiogenesis [24]. The process, char-acterized by pericyte detachment, increased vessel permeability and extravasation of mononuclear cells, occurs in parallel with downregulation of Notch-dependent Hes5 and Tie-2 gene expression and Fgf2 and Cxcr4 upregulation.

On this basis, we evaluated the expression levels of intussusception-related genes in the total brain extracts of twitcher mice at P35. RT-qPCR analysis demonstrates a significant downregulation of the Notch target gene Hes5. Also, we observed a significant upregulation of Fgf2 and Cxcr4 expression as well as of various pro-inflammatory cytokines and chemotactic chemokines, including Tumor necrosis factor-a, Interleukin-1a and Cxcl-1, paralleled by a sustained inflammatory infiltrate as indicated by the significant increase in CD45 mRNA levels (Fig.6). This occurs in the absence of significant changes in the expression levels of Notch1 and of its ligands Dll4 and Jag1 (data not shown).

Discussion

Previous observations from our laboratory had shown that GALC deficiency induces significant defects in the endothelium of the postnatal brain of twitcher mice, an authentic murine model of GLD [11]. Here, we extend these observations by showing that quantitative computational analysis of the three-dimensional CD31? microvascular architecture of the postnatal frontal cortex coupled with microvascular corrosion casting/SEM morphometry [22] highlights significant alterations in the angioarchitecture of the brain of twitcher mice with a uniform, generalized reduction in microvascular density. In parallel, vascular branch remodeling and intussusceptive angiogenesis were observed in twitcher brain as a tentative mechanism of vas-cularization alternative to impaired sprouting angiogenesis. CD31?vessel quantification was initially approached by customized ImageJ scripts developed in order to speed up Fig. 3 Immunofluorescent analysis of endothelial coverage. a Frontal

cortex microvessels of P36 wt and twi/twi mice were double-stained with anti-CD31 and anti-desmin antibodies. Scale bar: 30 lm. bQuantification of the percentage of pericyte coverage calculated as desmin? area/CD31? area ratio. Data are the mean ± SEM. ***P \ 0.001. (Color figure online)

Fig. 4 Morphometric analysis of microvascular corrosion casts. The analysis was performed on corrosion casts of the frontal cortex of P36 wt and twi/twi mice. Vessel diameters are expressed as box plots (left) and cumulative percentual frequency graphs (right). The boxes extend from the 25th to the 75th percentiles, the lines indicate the median values, and the whiskers indicate the range of values; the mean values are indicated with a dashed line. ***P \ 0.001

(9)

the analysis of significant numbers of isotropic 3D samples. The analysis provided cumulative information about functional and non-functional vessels (including frag-mented vessels apparently not connected with the blood-stream) and was integrated by SEM observations of the corrosion casts of patent vessels. The results demonstrate a remarkable reduction in CD31? vascularity in twitcher brains analyzed at an advanced stage of disease, possibly due to a decrease in the angiogenic process that drives postnatal brain vascularization [11] and/or to cytotoxic effects due to the accumulation of high levels of psy-chosine in the CNS [5] with consequent degenerative neuroinflammation [6,25]. Taken together, our current and previous observations [11] indicate that GALC deficiency may affect not only the glial/neuronal compartment of the neurovascular brain unit but also its vascular moiety. A tight cross-talk exists between angiogenesis and

neurogenesis, and neovascularization plays an important role in postnatal neuroprotection [8–10,26]. Consequently, alterations in blood vessel development may contribute greatly to the CNS damage occurring in GLD. It remains challenging to establish whether vascular alterations in GLD represent the outcome of the astrocytic/neuronal injury, with a consequent deficiency in the production of trophic angioneurins [27], or are due to a direct damage of endothelial cell functions by the loss of GALC function and psychosine accumulation. Relevant to this point, TUNEL staining of postnatal frontal cortex sections showed a significant overall increase in apoptotic events in twitcher mice when compared to control animals. How-ever, they were limited to non-endothelial CNS compo-nents, as shown by double TUNEL/CD31 staining that failed to indicate the presence of apoptotic events in brain endothelium (Supplementary Fig. S3 in Online Resource Fig. 5 Scanning electron micrographs of the frontal cortex

microvas-culature of twitcher mice. Microvascular corrosion casting/SEM analysis of the frontal cortex of P36 twi/twi mice. a The vasculature was characterized by dilated vessels and high frequency of diameter changes (black asterisks). The dehiscences in endothelium result in a leakage of vasculature (white arrows). Scale bar: 10 lm. b Example of intussusceptive site in twitcher mice characterized by vessel bifurcation reflecting structural intussusceptive branch remodeling (see magnified 1, scale bar: 10 lm) and intussusceptive pillar indicating vessel duplication (see magnified 2, scale bar: 10 lm).

Scale bar: 20 lm. c Frontal cortex microvessels of P36 wt and twi/twi mice stained with toluidine blue showing the presence of intraluminal tissue fold connecting the opposite vascular walls (black arrows) in twi/twi mice but not in wt mice. Scale bar: 100 lm. d Quantification of endothelial intraluminal tissue folds. Transversally sectioned vessel profiles with a single layer of endothelial cells were randomly chosen for wt and twi/twi animals. For each vessel, the number of connections of intraluminal tissue folds with the opposite vascular wall, expression of intussusceptive microvascular growth, was counted. Data are the mean ± SD, n = 30. *P \ 0.05

(10)

3). This is in keeping with the capacity of psychosine to induce oligodendrocyte apoptosis [28] with no effect on endothelial cell survival [11].

Additional suggestions come from the analysis of the spatial distribution of CD31? vessels. In the search for focal sites where vascular pruning and remodeling might be taking place, we did not observe marked differences between control and twitcher animals. Previous observa-tions had shown that brain neovascularization is reduced in twi/twi mice also at postnatal stages that precede the occurrence of significant signs of disease [11]. Thus, a progressive slowdown of vascular development, and mat-uration, might explain why twitcher brains showed a larger amount of fragmented vessels with no apparent continuity with the vascular tree. Further experiments will be required to depict the exact spatiotemporal contribution of reduced neovascularization and psychosine toxicity to the observed decrease in functional microvascular density in the brain of twitcher mice. Unfortunately, it must be pointed out that, to this respect, the increased vascular leakage of the GALC-deficient endothelium precludes the possibility to use bloodstream-injected molecular tracers for a direct visual-ization and quantification of patent, functional vessels [11]. Search for signs of compensatory neovascularization in twitcher brains did not reveal evident hot spots of sprouting vessels or a marked increase in microvessels with small cross sections. Indeed, previous observation had shown a progressively reduced capacity of twitcher endothelium to respond to pro-angiogenic factors [11]. In contrast, SEM and histological analyses showed increased signs of intussusceptive angiogenesis. This occurs in parallel with the modulation of genes associated with this process (e.g., Hes5, Cxcr4, Fgf2) [24] and in the presence of a sustained inflammatory response. Intussusceptive (non-sprouting)

angiogenesis is a well-characterized morphogenetic pro-cess of blood capillaries that produces two lumens from a single vessel by intravascular septation, the intussusceptive pillar being a 1- to 5-lm transluminal tissue bridge that spans the vessel lumen [29]. Intussusceptive angiogenesis is distinct from sprouting angiogenesis because it does not require cell proliferation. It might represent a general fast recovery adaption of vascularization to growth necessities with a rapid expansion of the existing microvascular net-work [23]. In inflammatory tissue, a shift is observed from sprouting angiogenesis toward intussusceptive angiogene-sis [23]. Thus, the dynamic process of intussusceptive angiogenesis by rapidly increasing capillary exchange surface area appears to be a relevant adaptive mechanism in inflammatory and ischemic tissues [23, 30,31] and is characterized by pericyte detachment, increased vessel permeability and extravasation of mononuclear cells [24]. Our data suggest that structural and architectural angioad-aptations may occur in twi/twi mice in response to GALC deficiency [6,25], intussusceptive angiogenesis represent-ing a possible mechanism of escape in the attempt to overcome defective sprouting angiogenic responses. On note, no difference in mRNA and protein levels of hypoxia-inducible factor-1a was observed in the total brain extracts of wt and twi/twi animals (A.G., unpublished observa-tions). Further experiments will be required to elucidate the role of the neuroinflammatory process and the molecular mechanism leading to intussusceptive microvascular growth following GALC deficiency.

The pathogenesis of GLD has been proposed to arise from the accumulation of psychosine, the neurotoxic lysolipid metabolite detected at high levels in the CNS of GLD patients and twitcher mice [5,32,33]. Accumulation of psychosine leads to cytotoxic effects on oligodendroglial cells, triggering apoptotic cell death in vitro and in vivo [28, 34, 35]. Given the important role played by oligo-dendrocytes in brain vascularization [36], it is conceivable that the vascular alterations observed in the brain of twitcher mice may represent the outcome of the astrocytic/ neuronal injury, with a consequent deficiency in the pro-duction of trophic angioneurins [27] and activation of inflammatory responses [6, 25]. Accordingly, at variance with the results of the computational analysis of brain cortex microvasculature, no significant alterations of the three-dimensional CD31?microvascular architecture were observed in the kidneys of twitcher mice in which psy-chosine is detectable at very low levels when compared to CNS [37]. On the other hand, endothelial cells of different origins express GALC in vitro and in vivo, and GALC deficiency in twitcher mice affects the capacity of periph-eral endothelium to respond to pro-angiogenic factors [11]. Thus, GALC loss-of-function may affect the postnatal microvasculature of different organs besides the CNS. Fig. 6 RT-qPCR analysis of intussusceptive angiogenesis-related

genes in the brain of twitcher mice. Total brain extracts from P35 wt and twi/twi mice (n = 3–7 per group) were analyzed for the expression of the indicated genes by RT-qPCR, and data were normalized for Gapdh expression. Data are the mean ± SD; **P \ 0.01

(11)

Indeed, significant vascular permeability defects occur in different visceral organs of twi/twi mice, including kidney, lung and liver [11]. Taken together, these data suggest that GALC deficiency may cause a generalized endothelial cell dysfunction in peripheral tissues, leading to increased vessel permeability, with no significant consequence on their angioarchitecture.

Lysosomal storage disorders represent one of the most frequent classes of human genetic diseases. They are characterized by the accumulation of disease-specific metabolic intermediates within lysosomes, leading to sev-ere organ damages and premature death [32]. Besides GLD, brain alterations of the microvascular angioarchi-tecture may occur also in adrenoleukostrophy, Canavan’s disease and Alexander’s disease patients [38,39]. Of note, b-glucosylpsychosine (1-b-D-glucosylsphingosine) puri-fied from human Gaucher spleen exerts an inhibitory effect on endothelial cell proliferation and motility similar to psychosine [11], and ultrastructural abnormalities occur in capillary endothelial cells in skin biopsies from patients with acute neuronopathic infantile Gaucher disease [40]. Together, these observations point to endothelial dysfunc-tion as a common factor in severe lysosomal diseases. At present, the only clinical treatment for GLD is bone mar-row or umbilical cord blood cell transplantation for late-onset and presymptomatic patients [3,6]. A better under-standing of the alterations of CNS and peripheral microvascular angioarchitecture and of endothelial cell dysfunctions may provide further insights into the patho-genesis of lysosomal storage disorders and pave the way toward more efficacious therapeutic approaches. To this respect, our observations show that quantitative vascular analysis by a simple, automatic computational approach may provide relevant information when applied to twitcher mice, an authentic model of Krabbe disease. Given the increasing availability of image analysis tools and the intimate link between vascular and neurological systems, we consider that time is ripe for a systematic analysis of vascular trees in animal models of neurodegenerative pathologies.

Acknowledgments This work is dedicated in the memory of late Prof. Moritz A. Konerding. The work was supported in part by the CNR Research Project on Aging (Regione Lombardia MbMM-con-venzione No. 18099/RCC) to the Institute of Neuroscience (Milan, Italy) and by Grants from Ministero dell’Istruzione, Universita` e Ricerca (FIRB Project RBAP11H2R9 2011), Associazione Italiana per la Ricerca sul Cancro (AIRC Grant No. 14395) to M.P., A.G. was supported by a Fondazione Italiana per la Ricerca sul Cancro Fellowship.

Compliance with ethical standards

Conflict of interest The authors declare that they have no conflict of interest.

References

1. Suzuki K, Suzuki Y (1970) Globoid cell leucodystrophy (Krabbe’s disease): deficiency of galactocerebroside beta-galac-tosidase. Proc Natl Acad Sci USA 66:302–309

2. Loonen MC, Van Diggelen OP, Janse HC, Kleijer WJ, Arts WF (1985) Late-onset globoid cell leucodystrophy (Krabbe’s dis-ease). Clinical and genetic delineation of two forms and their relation to the early-infantile form. Neuropediatrics 16:137–142 3. Wenger DA, Rafi MA, Luzi P, Datto J, Costantino-Ceccarini E

(2000) Krabbe disease: genetic aspects and progress toward therapy. Mol Genet Metab 70:1–9

4. Suzuki K (2003) Globoid cell leukodystrophy (Krabbe’s disease): update. J Child Neurol 18:595–603

5. Suzuki K (1998) Twenty five years of the ‘‘psychosine hypoth-esis’’: a personal perspective of its history and present status. Neurochem Res 23:251–259

6. Sakai N (2009) Pathogenesis of leukodystrophy for Krabbe dis-ease: molecular mechanism and clinical treatment. Brain Dev 31:485–487

7. Zlokovic BV (2005) Neurovascular mechanisms of Alzheimer’s neurodegeneration. Trends Neurosci 28:202–208

8. Greenberg DA, Jin K (2005) From angiogenesis to neuropathol-ogy. Nature 438:954–959

9. Segura I, De Smet F, Hohensinner PJ, Ruiz de Almodovar C, Carmeliet P (2009) The neurovascular link in health and disease: an update. Trends Mol Med 15:439–451

10. Zacchigna S, Lambrechts D, Carmeliet P (2008) Neurovascular signalling defects in neurodegeneration. Nat Rev Neurosci 9:169–181

11. Belleri M, Ronca R, Coltrini D, Nico B, Ribatti D, Poliani PL, Giacomini A, Alessi P, Marchesini S, Santos MB, Bongarzone ER, Presta M (2013) Inhibition of angiogenesis by beta-galac-tosylceramidase deficiency in globoid cell leukodystrophy. Brain 136:2859–2875

12. Righi M, Giacomini A, Cleris L, Carlo-Stella C (2013) (3)D [corrected] quantification of tumor vasculature in lymphoma xenografts in NOD/SCID mice allows to detect differences among vascular-targeted therapies. PLoS One 8:e59691 13. Ackermann M, Houdek JP, Gibney BC, Ysasi A, Wagner W,

Belle J, Schittny JC, Enzmann F, Tsuda A, Mentzer SJ, Kon-erding MA (2014) Sprouting and intussusceptive angiogenesis in postpneumonectomy lung growth: mechanisms of alveolar neo-vascularization. Angiogenesis 17:541–551

14. Sakai N, Inui K, Tatsumi N, Fukushima H, Nishigaki T, Taniike M, Nishimoto J, Tsukamoto H, Yanagihara I, Ozono K, Okada S (1996) Molecular cloning and expression of cDNA for murine galactocerebrosidase and mutation analysis of the twitcher mouse, a model of Krabbe’s disease. J Neurochem 66:1118–1124 15. Serra J (1982) Image analysis and mathematical morphology, vol

I. Academic Press, New York

16. Serra J (1988) Image analysis and mathematical morphology, part ii: theoretical advances, vol II. Academic Press, London 17. Lee TC, Kashyap RL, Chu CN (1994) Building skeleton models

via 3-D medial surface axis thinning algorithms. Cvgip-Graph Model Im 56:462–478

18. Arganda-Carreras I, Fernandez-Gonzalez R, Munoz-Barrutia A, Ortiz-De-Solorzano C (2010) 3D reconstruction of histological sections: application to mammary gland tissue. Microsc Res Tech 73:1019–1029

19. Konerding MA, Steinberg F, Budach V (1989) The vascular system of xenotransplanted tumors—scanning electron and light microscopic studies. Scanning Microsc 3:327–335

20. Malkusch W, Konerding MA, Klapthor B, Bruch J (1995) A simple and accurate method for 3-D measurements in

(12)

microcorrosion casts illustrated with tumour vascularization. Anal Cell Pathol 9:69–81

21. Coltrini D, Di Salle E, Ronca R, Belleri M, Testini C, Presta M (2013) Matrigel plug assay: evaluation of the angiogenic response by reverse transcription-quantitative PCR. Angiogenesis 16:469–477

22. Konerding MA (1991) Scanning electron microscopy of corro-sion casting in medicine. Scanning Microsc 5:851–865 23. Mentzer SJ, Konerding MA (2014) Intussusceptive angiogenesis:

expansion and remodeling of microvascular networks. Angio-genesis 17:499–509

24. Dimova I, Hlushchuk R, Makanya A, Styp-Rekowska B, Ceausu A, Flueckiger S, Lang S, Semela D, Le Noble F, Chatterjee S, Djonov V (2013) Inhibition of Notch signaling induces extensive intussusceptive neo-angiogenesis by recruitment of mononuclear cells. Angiogenesis 16:921–937

25. Formichi P, Radi E, Battisti C, Pasqui A, Pompella G, Lazzerini PE, Laghi-Pasini F, Leonini A, Di Stefano A, Federico A (2007) Psychosine-induced apoptosis and cytokine activation in immune peripheral cells of Krabbe patients. J Cell Physiol 212:737–743 26. Ment LR, Stewart WB, Fronc R, Seashore C, Mahooti S,

Scaramuzzino D, Madri JA (1997) Vascular endothelial growth factor mediates reactive angiogenesis in the postnatal developing brain. Brain Res Dev Brain Res 100:52–61

27. Lee HS, Han J, Bai HJ, Kim KW (2009) Brain angiogenesis in developmental and pathological processes: regulation, molecular and cellular communication at the neurovascular interface. FEBS J 276:4622–4635

28. Zaka M, Wenger DA (2004) Psychosine-induced apoptosis in a mouse oligodendrocyte progenitor cell line is mediated by cas-pase activation. Neurosci Lett 358:205–209

29. Burri PH, Hlushchuk R, Djonov V (2004) Intussusceptive angiogenesis: its emergence, its characteristics, and its signifi-cance. Dev Dyn 231:474–488

30. Ackermann M, Tsuda A, Secomb TW, Mentzer SJ, Konerding MA (2013) Intussusceptive remodeling of vascular branch angles in chemically-induced murine colitis. Microvasc Res 87:75–82

31. Wnuk M, Hlushchuk R, Tuffin G, Huynh-Do U, Djonov V (2011) The effects of PTK787/ZK222584, an inhibitor of VEGFR and PDGFRbeta pathways, on intussusceptive angiogenesis and glomerular recovery from Thy1.1 nephritis. Am J Pathol 178:1899–1912

32. Ballabio A, Gieselmann V (2009) Lysosomal disorders: from storage to cellular damage. Biochim Biophys Acta 1793:684–696 33. Igisu H, Suzuki K (1984) Progressive accumulation of toxic

metabolite in a genetic leukodystrophy. Science 224:753–755 34. Jatana M, Giri S, Singh AK (2002) Apoptotic positive cells in

Krabbe brain and induction of apoptosis in rat C6 glial cells by psychosine. Neurosci Lett 330:183–187

35. Haq E, Giri S, Singh I, Singh AK (2003) Molecular mechanism of psychosine-induced cell death in human oligodendrocyte cell line. J Neurochem 86:1428–1440

36. Yuen TJ, Silbereis JC, Griveau A, Chang SM, Daneman R, Fancy SP, Zahed H, Maltepe E, Rowitch DH (2014) Oligodendrocyte-encoded HIF function couples postnatal myelination and white matter angiogenesis. Cell 158:383–396

37. Whitfield PD, Sharp PC, Taylor R, Meikle P (2001) Quantifica-tion of galactosylsphingosine in the twitcher mouse using elec-trospray ionization-tandem mass spectrometry. J Lipid Res 42:2092–2095

38. Kondo A, Suzuki K (1993) The blood brain barrier in human leukodystrophies and allied diseases. Ultrastructural and mor-phometric studies on the capillaries in brain biopsies. Clin Neu-ropathol 12:169–174

39. Buee L, Hof PR, Bouras C, Delacourte A, Perl DP, Morrison JH, Fillit HM (1994) Pathological alterations of the cerebral microvasculature in Alzheimer’s disease and related dementing disorders. Acta Neuropathol 87:469–480

40. Hulkova H, Poupetova H, Harzer K, Mistry P, Aerts JM, Elleder M (2010) Abnormal nonstoring capillary endothelium: a novel feature of Gaucher disease. Ultrastructural study of dermal cap-illaries. J Inherit Metab Dis 33:69–78

Figura

Table 1 Percent vascular volume differences between wt and twi/twi brain cortex samples
Fig. 4 Morphometric analysis of microvascular corrosion casts. The analysis was performed on corrosion casts of the frontal cortex of P36 wt and twi/twi mice

Riferimenti

Documenti correlati

Measurement of alcohol content in wine using electronic density-meter has been recently made official by OIV (Resolution OENO 8/2000) and admitted in the Annex A of the

(a) Longitudinal evolution of streamwise turbulence intensity / behind the central twine and the adjacent mesh hole for case N5; (b) Power decay region of / behind the twine

La disprassia evolutiva viene definita come un disturbo nel quale la caratteristica principale è una grave difficoltà di coordinazione motoria presente fin dalle prime

When perception algorithm identify a specific object gives position and orientation to next step, we join these informations together with the kind of object to look for and we

Il recente dibattito sulla crescita economica endogena (Lucas 1988; Romer 1990) ha evidenziato il ruolo degli spillover di conoscenza e, più in generale, delle esternalità dinamiche

Pietro di Cristoforo Vannucci, detto il Perugino, Battaglia di Castità. contro Lascivia, Paris, Musée

GC×GC- TANDEM IONIZATION-TOFMS WITH COMBINED UNTARGETED AND TARGETED (UT) FINGERPRINTING OF VOLATILES FROM EXTRA VIRGIN OLIVE OIL: CHALLENGES FOR DATA ALIGMENT AND