Articles

  • KSBNS 2024

Article

Original Article

Exp Neurobiol 2023; 32(6): 441-452

Published online December 31, 2023

https://doi.org/10.5607/en23032

© The Korean Society for Brain and Neural Sciences

Cerebral Cavernous Malformation (CCM)-like Vessel Lesion in the Aged ANKS1A-deficient Brain

Jiyeon Lee, Haeryung Lee, Miram Shin and Soochul Park*

Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea

Correspondence to: *To whom correspondence should be addressed.
TEL: 82-2-710-9330, FAX: 82-2-710-9331
e-mail: scpark@sookmyung.ac.kr
These authors contributed equally to this article.

Received: September 29, 2023; Revised: December 11, 2023; Accepted: December 15, 2023

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

In this study, we show that ANKS1A is specifically expressed in the brain endothelial cells of adult mice. ANKS1A deficiency in adult mice does not affect the differentiation, growth, or patterning of the cerebrovascular system; however, its absence significantly impacts the cerebrovascular system of the aged brain. In aged ANKS1A knock-out (KO) brains, vessel lesions exhibiting cerebral cavernous malformations (CCMs) are observed. In addition, CCM-like lesions show localized peripheral blood leakage into the brain. The CCM-like lesions reveal immune cells infiltrating the parenchyma. The CCM-like lesions also contain significantly fewer astrocyte endfeets and tight junctions, indicating that the integrity of the BBB has been partially compromised. CCM-like lesions display increased fibronectin expression in blood vessels, which is also confirmed in cultured endothelial cells deficient for ANKS1A. Therefore, we hypothesize that ANKS1A may play a role in maintaining or stabilizing healthy blood vessels in the brain during aging.


Keywords: ANKS1A, Cerebral cavernous malformation, Blood-brain barrier, Neurodegenerative disease

Cerebral Cavernous Malformations (CCMs) refer to abnormal blood vessels with thin walls that are tightly packed in the brain [1, 2]. The appearance of CCMs is similar to that of a small mulberry, as a result of abnormal cell divisions in the vascular endothelium [3]. Approximately 500 people out of every 100,000 are affected by this disease (0.5%) [4], with approximately 20% of cases being familial, resulting from the loss of function of one of the CCM1/KRIT1 [5], CCM2 [6], or CCM3/PDCD10 [7] genes. In the remaining 80% of patients, the disease occurs sporadic, and the genes responsible for it have not been well identified. Moreover, CCM disease is associated with brain aging and is prevalent in the elderly [8]. This type of vascular malformation is characterized by hemorrhages within brain tissue, suggesting that damage to the blood-brain barrier may play a significant role in the development of these conditions.

In light of the inherited nature of CCM and the discovery of disease-related genes, there has been a growing interest in investigating their functional roles. Each of the CCM1, CCM2, and CCM3 genes encodes a protein that is structurally distinct and does not share any sequence homology [9]. The gene products of each gene are multi-domain adaptor proteins, which participate in numerous signaling pathways and interact with numerous binding molecules [10]. The CCM1, CCM2, and CCM3 proteins form a heterotrimeric complex [11], the CCM signaling complex, which plays an important role in the development and pathogenesis of CCM. Recent studies suggest that CCM proteins play an important role in many cellular events, including cell polarity [12], cytoskeletal reorganization [13], cell proliferation [14], cellular adhesion [15], and migration, impacting angiogenesis [16], cell–cell junction integrity [17], vascular permeability [18], and apoptosis, whether they form part of the ternary complex or not.

Interestingly, the ankyrin repeat and sterile alpha motif domain-containing protein 1B (ANKS1B) was identified as a novel binding partner of CCM1 [19], a major causative gene for CCM disease. The CCM1 protein is characterized by three NPxY/F motifs and three ankyrin repeats, and it is abundantly expressed in vascular endothelial cells. Silencing of ANKS1B, however, did not have any significant effect on the proliferation, migration, or sprouting of angiogenesis in primary human endothelial cells. While silencing of ANKS1B expression disrupted endothelial barrier functions, resulting in increased permeability, the role of ANKS1B in CCM1-mediated vascular pathogenesis remains unclear [19].

ANKS1 family of proteins consists of two members, ANKS1A and ANKS1B, and they contain six ankyrin repeats at the N-terminus, two SAM domains, as well as a PTB domain at the C-terminus [20]. In this study, we show that ANKS1A plays a role in stabilizing and regulating the permeability of vascular endothelial cells in the aged brain. The findings of this study may provide a new paradigm for understanding the pathogenesis of CCM and other vascular diseases of the brain.

Mice

ANKS1A+/-(LacZ) gene trap mice have been described previously [21, 22]. ES cells with a gene trap vector insertion (pGT0lxr) in the ANKS1A gene locus (cell line ID CF0537) were obtained from the Mutant Mouse Regional Resource Center. The gene trap insertion site was determined through sequence analysis. These ES cells were microinjected into C57BL/6 blastocysts and subsequently transferred to foster mother (ICR) females to create chimeric animals. Chimeric males were bred with 129/SvJ females, and the presence of the transgene in F1 agouti pups was assessed using PCR analysis of tail genomic DNA with specific primers designed to detect mutant and wild-type ANKS1A: 5’-TGAAGGCACATGACCCTGAG-3’ (F1), 5’-ATGTCATAGCTGTTTCCTGT-3’(F2), and 5’-ACAGCGTTTGCATCTTGCTG-3’(R1). The experiments on mice were conducted according to the institutional research guidelines of the Sookmyung Women’s University and the protocol approved by the Institutional Animal Care and Use Committee (IACUC) of the university (protocol approval: SMWU-IACUC-2104-002-1). All mice had ab libitum access to standard laboratory diet and water.

X-gal staining and immunostaining analysis

For X-gal staining, the brain sections were fixed with 1% paraformaldehyde (PFA), 0.2% glutaraldehyde, 1% deoxycholate (Sigma Aldrich, St. Louis, U.S.A), and 10% NP-40 in phosphate-buffered saline (PBS) for 10 min; the sections were then washed 3 times for 10 min and then incubated with the staining buffer (K4Fe(CN)6, K3Fe(CN)6, MgCl2, X-gal) at 37°C. After 15 h, the reaction was stopped by washing each slide three times with PBS.

For immunostaining analysis of the mouse brain cryosections, the sections were washed with PBS three times for 10 min at RT. Then, they were incubated in PBS containing 0.3% Triton X-100 for 30 min and treated with primary antibody diluted in PBS containing 0.3% Triton X-100 and 3% BSA for overnight at 4°C. After the overnight incubation, the brain sections were washed, and treated with secondary antibodies for 2 hours at RT. The sections were then three times washed with PBS containing 0.3% Triton X-100 and mounted with Vectorshiled mounting medium. Primary antibodies were as follows: Collagen IV (goat, 1:100, 1340-01, Southern Biotech), Hemoglobin (rabbit, 1:100, ABIN1078132, Antibodies online), CD68 (rabbit, 1:100, ab125212, Abcam), Aquaporin 4 (rabbit, 1:100, AQP-004, Alomone labs), CD45 (rabbit, 1:100, ab10558, Abcam), Fibronectin (rabbit, 1:100, ab23750, Abcam), Sca-1 (rat, 1:100, ab51317, Abcam). Secondary antibodies were as follows: Donkey anti-rabbit IgG Alexa488 (1:500, A21296, Thermo Fisher Scientific, Waltham, MA USA), Donkey anti-rat IgG Alexa488 (1:500, A21208, Thermo Fisher Scientific), Donkey anti-goat IgG Alexa488 (1:500, A11055, Thermo Fisher Scientific), Donkey anti-goat IgG Alexa568 (1:500, A11057, Thermo Fisher Scientific).

Image analysis

Image data were collected using an LSM700 (Carl Zeiss Microscopy) with a Plan-Apochromat ×20/0.8 M27 objective lens of an Axio Observer camera (Zeiss). The images were taken at 0.5~1 μm z-stack intervals over a 5~10 μm thickness. The fluorophore excitations were with 488, 555, and 639 nm laser wavelengths. All images were processed by the ZEN Black software.

Cerebral blood vessel network was analyzed using the AngioTool 0.6a software (NIH) [23]. For producing the vessel skeletonized images for Col IV staining, various parameters (e.g., vessel diameter) were optimized such that only the true vessels were labelled. The skeletonized images were further used to calculate length, total area, branching points and lacunarity of microvessels in each microscopic field.

Evans blue extravasation assay

2% Evans blue (Sigma Aldrich, St. Louis, U.S.A) dissolved in saline was intraperitoneally injected into each mouse (200 μl) and they were sacrificed after 24 hours [24]. For qualitative assessment of Evans blue extravasation, the brains were fixed in 4% paraformaladehyde and cut 40 μm-thick cryosection. Brain sections were washed with PBS three times for 10 min at RT. Then, sections were mounted with Vectorshield mounting medium and observed them under ZEISS Axio Zoom.V16 fluorescence microscopy (Zeiss, Jena, Germany).

Primary mouse brain endothelial cell culture

Primary brain endothelial cells were obtained from 2 months old mice using published protocol [25, 26]. The removal of the meninges by rolling the brains on filter papers. Subsequently, the brains were dissociated using a tissue grinder (with 10 brains processed in one tissue grinder). The resulting homogenates were then subjected to centrifugation (2580 g for 7 minutes at 4°C), and the resulting cell pellets were re-suspended in a 20% BSA solution, followed by thorough vortexing. The precipitates were digested for 60 minutes at 37°C in DMEM containing Collagenase/Dispase (100 mg/ml, Roche), DNase I (4 ug/ml, Roche), and TLCK (Nα-Tosyl-L-lysine chloromethyl ketone hydrochloride) (0.147 ug/ml, Sigma). After the digestion, the precipitates were again centrifuged (2580 g for 7 minutes at 4°C) and washed with PBS. The isolated mouse brain endothelial cells were then seeded onto 24-well plates coated with type IV collagen. These cells were cultured in high-glucose DMEM supplemented with 20% PDS (Plasma-derived bovine serum, FirstLink), penicillin/streptomycin, heparin (750 U), endothelial cell growth supplement (Sciencell), and 8 μg/ml puromycin (Sigma). After seeding, the cells underwent a two-day puromycin treatment, and subculturing was performed for 7 days.

Quantification

Immunofluorescence analysis was performed on images obtained from confocal microscopy. By using the Zen Blue software (Zeiss, Jena, Germany), the staining for neuroinflammation and Endothelial-mesenchymal transition (Endo-MT) markers was analyzed and quantified. Briefly, we used the mean signal intensity per field. Quantification was conducted on three animals per each group.

Statistics

Statistical analysis was performed on data from three or four independent experimental replicates using GraphPad Prism 9.0 (San Diego, CA, USA). Error bars in the graphs represent the mean±SD of the data. Statistical significance test was via the unpaired student’s t test for two samples in all the figures with the following p-value designations in the plots: *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001.

Data availability

All other data are available from the corresponding author upon reasonable request.

The specific expression of ANKS1A in the brain endothelial cells

To determine the brain region where ANKS1A is specifically expressed, we used ANKS1A+/-(LacZ) mice in which the LacZ reporter was inserted into the ANKS1A gene locus: the targeted allele expresses the LacZ reporter instead of ANKS1A. Strikingly, whole mount X-gal staining of the dissected brains of adult mice at P45 revealed that ANKS1A was predominantly expressed in the brain vessels (Fig. 1A). Using Collagen IV, a marker specific for brain vessels, brain sections were stained to determine whether LacZ expression is restricted to brain vessels. Consequently, we observed that LacZ expression co-localized with Collagen IV staining. These results indicate that ANKS1A is expressed specifically in cerebrovascular cells (Fig. 1B).

In order to investigate whether the ANKS1A deficiency impairs the development of cerebrovascular patterning, brain sections were stained with Collagen IV and then analyzed for the indices that represent cerebrovascular patterning (Fig. 1C). Accordingly, we found that the density of vessels, the average vessel length, the degree of vessel branching, and the gap between neighboring vessels were not significantly different between wild type and KO groups (Fig. 1D). The results suggest that ANKS1A deficiency does not affect the overall development of cerebrovascular networks in the adult brain.

CCM-like vessel lesions in the aged ANKS1A KO brain

The ANKS1A KO mice display normal behavior, sexual reproduction, and healthy lifespan, similar to the WT littermates. However, we hypothesized that aged brains of ANKS1A KO mice would exhibit some defects in the cerebrovascular system. To accomplish this, the mice were allowed to grow for over 16 months and then their brains were carefully examined. A striking finding was that ANKS1A KO mice exhibited abnormal clusters of closely packed vessels whereas their WT littermates did not. This small mulberry-shaped blood vessel is known as a cerebral cavernous malformation (CCM) [3]. As an example of a symptom associated with CCM is bleeding surrounding the lesion [27]. In accordance with this hypothesis, we found a significant amount of hemoglobin in the CCM-like lesion of the neocortex (Fig. 2A) but also in the vessels near the brain ventricular region (Fig. 2B), suggesting brain hemorrhage in the aged ANKS1A KO brain. To further confirm the leakage from the CCM-like lesion, the brain sections were immunostained with mouse IgG-specific antibodies. Consequently, we observed that the IgG-positive signal was highly detectable only in the aged ANKS1A KO mice (Fig. 2C). Finally, we injected Evans Blue, the most widely used marker of brain barrier integrity [28], into the peritoneal cavity of aged mice and examined whether the dyes were abundantly detected around the CCM-like lesions. The intensity of Evans Blue was measured in the areas containing CCM-like lesions as well as focal cavitation found adjacent to the lesions (see Fig. 2D, right and top panel). As we expected, the intensity of Evans Blue in the aged ANKS1A KO brain was substantially higher than in the WT littermate brains (Fig. 2D). Together, our studies strongly indicate that the aged ANKS1A KO brain exhibits abnormal blood vessel integrity with bleeding.

Immune cells are focally increased around the CCM-like lesion

An analysis of high resolution confocal images revealed that the IgG-positive cells were detectable around the CCM-like lesion (Fig. 2C), suggesting peripheral blood immune cells have been infiltrated into the brain parenchymal tissue. To investigate whether peripheral immune cells infiltrate the aged ANKS1A KO brain, brain sections were stained with CD68, a marker for macrophages and other monocyte lineage cells [29]. As expected, CD68-positive cells were highly detectable in aged ANKS1A KO brains, but not WT brains (Fig. 3A). A similar experiment was conducted using CD45 antibody, which stains leukocytes [30], showing that CD45-positive cells were highly detectable around the CCM-like lesions (Fig. 3B). Together, our results suggest that lesions associated with CCM may cause local neuroinflammation in the aged ANKS1A KO brain.

CCM-like lesions are characterized by irregularly gathered vessels that swell to form a large cavity within the neighboring parenchyma (see Fig. 2D, right and top panel). We found that CCM-like lesions with focal cavities were detected in various sizes in the aged ANKS1A KO brain and that those more than 2,000 μm2 were the most common (Fig. 3D). In addition, immunostaining using Aquaporin 4 (AQ4) antibody, a marker for astrocyte endfeet [31], revealed that astrocyte endfeet in the CCM-like lesion and cavity were much less abundant in the aged ANKS1A KO brain (Fig. 3C). However, in the aged WT brain, Aquaporin 4 staining was completely matched with Collagen IV staining (Fig. 3C). Importantly, the aged ANKS1A KO brain exhibited much lower levels of ZO-1 (Fig. 3E), a tight junctional marker for brain endothelial cells [32]. The expression of ZO-1 was only affected in CCM-like lesions and focal cavitations adjacent to the lesions. However, ZO-1 expression was not affected in blood vessels away from CCM-like lesions.

CCM-like lesions in the aged ANKS1A brain exhibit increased mesenchymal markers

We further hypothesized that formation of a CCM-like lesion in ANKS1A-deficient vessels may be due to changes in the fate of brain endothelial cells with aging. To determine whether the endothelial cells in the brain are altered in the aged ANKS1A KO mice, we examined the brain tissues with various markers for mesenchymal tissue. First, we used fibronectin as a mesenchymal marker, which is known to increase in epithelial-mesenchymal transition (EMT) induced by TGF-β [33]. In the WT brain, fibronectin was barely observed. Contrary to this, fibronectin was highly detectable in the CCM-like lesions of the aged ANKS1A KO brain (Fig. 4A, 4B). We also used Sca-1, a commonly used marker for mesenchymal stem cell [14, 34], to determine whether the CCM-like lesion lost its normal endothelial properties. As a result, we found that, as opposed to vessels from WT mice, the aged ANKS1A KO brain displayed CCM-likes lesions with high levels of Sca-1 (Fig. 4C, 4D). In addition, we cultured primary brain endothelial cells from WT and ANKS1A KO brains (2 month of age) and found that fibronectin levels were higher in ANKS1A-deficient cells (Fig. 4E, 4F). More importantly, we observed that ANKS1A-deficient cells showed Ki67-positive staining, suggesting that they are more proliferative (Fig. 4G, 4H). Taken together, these results strongly indicate that during brain aging, ANKS1A-deficient brain endothelial cells gradually lose their endothelial property and become mesenchymal cells.

In this study, we found that ANKS1A is specifically expressed in the brain endothelial cells of adult mice. Even though ANKS1A deficiency did not impair the overall differentiation, growth, and patterning of the cerebrovascular system, its absence had a significant impact on the cerebrovascular system of the aged brain. First, vessel lesions that resemble cerebral cavernous malformations (CCMs) were found. Second, the CCM-like lesions also showed a local leakage of peripheral blood into the brain tissue. In addition, the CCM-like lesions revealed that immune cells had infiltrated the parenchyma. Third, the CCM-like lesions also contained much less astrocyte endfeet with reduced tight junctions, implying that the integrity of the BBB has been partially compromised. Lastly, the CCM-like lesions displayed an increased mesenchymal marker expression. In conclusion, our findings suggest that ANKS1A may play a role in maintaining a healthy blood vessel in the brain during the aging process.

In aged ANKS1A KO brains, CCM-like lesions were detected at a low frequency (3~5 per brain) whereas they were never seen in aged WT brains. Furthermore, we did not detect CCM-like lesions in the ANKS1A KO brains from 2 months of age to 12 months of age. Since ANKS1A KO mice over 16 months of age were highly susceptible to developing CCM-like lesions, brain aging is likely to play a critical role in the development of CCM-like lesions in ANKS1A KO brains. A key finding was that the brain endothelial cells in the CCM-like lesion exhibit mesenchymal characteristics. The Endo-MT transition of brain endothelial cells plays an important role in the development of dysfunctional BBB with hemorrhage [35]. Furthermore, vascular cell adhesion molecule 1 (VCAM1) is known to be upregulated in aged brain endothelial cells and facilitate vascular-immune cell interactions [36]. Together, these reports support our findings of local bleeding and immune cell infiltration in CCM-like lesions. Nevertheless, in light of the abundant expression of ANKS1A in brain endothelial cells, this focal phenotype in the brain is not well understood. One plausible hypothesis is that environmental factors combined with ANKS1A deficiency in the localized vessels trigger the development of CCM-like vessel lesions. It is therefore essential to identify the extrinsic factors that can severely disrupt the integrity of vessels together with ANKS1A deficiency. For example, animal models with a breakdown of the BBB are more susceptible to lipopolysaccharide (LPS) [37] and experimental autoimmune encephalomyelitis (EAE) [38]. It remains to be determined whether these experimental tools can disintegrate brain endothelial cells when combined with an ANKS1A deficiency.

This work was supported by grants NRF-2021R1A4A1027355, NRF-2021R1C1C2009319 from the National Research Foundation of Korea (NRF) and HU23C0017 from KHIDI and KDRC.

Fig. 1. Brain vessel patterning is not impaired by ANKS1A deficiency in adult brains. (A) Whole brain X-gal staining of ANKS1A+/-(LacZ) mice at postnatal day 45. Ob, Olfactory bulb; Cx, Cortex; Cb, Cerebellum; Ht, hypothalamus; P, Pons; M, Medulla. (scale bars, 2 mm). The white arrows indicate X-gal-stained cerebral blood vessels. (B) Coronal sections of ANKS1A+/-(LacZ) brains were subjected to X-gal staining, followed by immunohistochemical staining with Collagen IV (scale bars, 100 μm). The white dotted boxes are enlarged in the right panels to show that the Col IV-positive vessels are matched with the X-gal-stained vessels in the high resolution image (right panel scale bars, 10 μm). (C) The cerebral cortex of 2 month-old mice was stained with Collagen IV, a blood vessel marker, and then immunofluorescence images were rendered into a vascular network image using Angiotool software (scale bars, 20 μm). (D) The data in panel C were quantified for the vessel patterning indices representing the and presented as means±SD (ANKS1A+/+, N=3 mice per group, n=16 sections; ANKS1A-/-, N=3 mice per group, n=25).
Fig. 2. Brain lesions that resemble CCM are present in aged ANKS1A KO mice. (A) In the neocortex of aged ANKS1A-deficient mice, representative fluorescent images show erythrocyte leakage associated with CCM-like vessel lesions. In our experiments, we used mice that are older than 16 months of age (scale bar, 10 μm). The mean intensity of hemoglobin staining in the WT group was set to 1. The data were quantified and presented as means±SD (N=3 mice per each group, n=8 sections). Orthogonal projection images of the CCM-like lesion shown in Fig. 2A. The arrows mark erythrocytes leaking into the dilated focal cavity surrounding the CCM-like lesion (scale bars, 10 μm). (B) A similar immunohistochemical staining process was performed as described in panel A, with the exception that the images show erythrocyte leakage in the subventricular zone of the aged mice lacking ANKS1A (scale bar, 20 μm). The data were quantified and presented as means±SD (N=3 mice per each group, n=8 sections). (C) The images show immunoglobulin G-positive staining in the neocortex of aged ANKS1A-deficient mice (scale bar, 10 μm). The white arrow indicate that the IgG-positive structures are matched with the DAPI-positive cells (scale bar, 10 μm). The microscopic filed containing CCM-like lesions was used for quantification. The data were quantified and presented as means±SD (N=3 mice per each group, n=8 sections). (D) A representative fluorescence image of Evans blue extravasation in the neocortex of aged ANKS1A-deficient mice (scale bar, 20 μm). An illustration showing that focal cavitation forms in adjacent sections due to the CCM-like lesion (right and top panel). The data were quantified and presented as means±SD (N=3 per group, n=18 sections).
Fig. 3. Aged ANKS1A KO brains exhibit increased immune cells. (A) A representative image of the CCM-like vessel lesions shows CD68-positve macrophages in the neocortex of aged ANKS1A-deficient mice (scale bar, 10 μm). The data were quantified and presented as means±SD (N=3 per group, n=8 sections). (B) A representative image of the CCM-like vessel lesions shows CD45-positive leukocytes in the neocortex of aged ANKS1A-deficient mice (scale bar, 10 μm). The yellow dotted box is enlarged in the right panels to show that the CD45-positve leukocytes contain DAPI staining (scale bar, 2 μm). The data were quantified and presented as means±SD (N=3 per group, n=8 sections). (C) The white arrow indicates a dilated focal cavity due to the CCM-like lesion (scale bar, 10 μm). The intensity of Aquaphorin 4 were quantified and presented as means±SD (N=3 per group, n=8 sections). (D) The size of CCM-like vessel lesions in aged ANKS1A-deficient mice were quantified (N=3 per group, n=21 sections). (E) The ZO-1 positive endothelial tight junctions are reduced in the dilated focal cavity of aged ANKS1A-deficient brains (scale bar, 2 μm). The arrow indicates a dilated focal cavity due to the CCM-like lesion. The data were quantified and presented as means±SD (N=3 per group, ANKS1A+/+ n=7, ANKS1A-/- KO n=5 sections).
Fig. 4. The Aged ANKS1A brains show an increased mesenchymal marker expression. (A) The image shows an increase in fibronectin positive signals in the CCM-like vessel lesions (scale bar, 10 μm). (B) The data in the panel A was quantified and presented as means presented as means±SD (N=3 per group, n=8). (C) The image shows several Sca-1 positive signals can be seen in the CCM-like vessels lesions, indicating a proliferation potential of the cells (scale bar, 10 μm). (D) The data in the panel C was quantified and presented as means presented as means±SD (N=3 per group, n=8). (E) The images show an increase in fibronectin positive signals in primary brain endothelial cells derived from ANKS1A KO mice (scale bar, 10 μm). (F) The data were quantified and presented as means±SD (N=3 per group, n=10 microscopic fields for each group). (G) The images show an enhanced Ki67 positive staining in primary brain endothelial cells derived from ANKS1A KO mice (scale bar, 10 μm). (H) The data were quantified and presented as means±SD (N=3 per group, ANKS1A+/+ n=13, ANKS1A-/- n=10 microscopic fields for each group).
  1. Labauge P, Denier C, Bergametti F, Tournier-Lasserve E (2007) Genetics of cavernous angiomas. Lancet Neurol 6:237-244
    Pubmed CrossRef
  2. Snellings DA, Hong CC, Ren AA, Lopez-Ramirez MA, Girard R, Srinath A, Marchuk DA, Ginsberg MH, Awad IA, Kahn ML (2021) Cerebral cavernous malformation: from mechanism to therapy. Circ Res 129:195-215
    Pubmed KoreaMed CrossRef
  3. Maraire JN, Awad IA (1995) Intracranial cavernous malformations: lesion behavior and management strategies. Neurosurgery 37:591-605
    Pubmed CrossRef
  4. Robinson JR, Awad IA, Little JR (1991) Natural history of the cavernous angioma. J Neurosurg 75:709-714
    Pubmed CrossRef
  5. Sahoo T, Johnson EW, Thomas JW, Kuehl PM, Jones TL, Dokken CG, Touchman JW, Gallione CJ, Lee-Lin SQ, Kosofsky B, Kurth JH, Louis DN, Mettler G, Morrison L, Gil-Nagel A, Rich SS, Zabramski JM, Boguski MS, Green ED, Marchuk DA (1999) Mutations in the gene encoding KRIT1, a Krev-1/rap1a binding protein, cause cerebral cavernous malformations (CCM1). Hum Mol Genet 8:2325-2333
    Pubmed CrossRef
  6. Liquori CL, Berg MJ, Siegel AM, Huang E, Zawistowski JS, Stoffer T, Verlaan D, Balogun F, Hughes L, Leedom TP, Plummer NW, Cannella M, Maglione V, Squitieri F, Johnson EW, Rouleau GA, Ptacek L, Marchuk DA (2003) Mutations in a gene encoding a novel protein containing a phosphotyrosine-binding domain cause type 2 cerebral cavernous malformations. Am J Hum Genet 73:1459-1464
    Pubmed KoreaMed CrossRef
  7. Bergametti F, Denier C, Labauge P, Arnoult M, Boetto S, Clanet M, Coubes P, Echenne B, Ibrahim R, Irthum B, Jacquet G, Lonjon M, Moreau JJ, Neau JP, Parker F, Tremoulet M, Tournier-Lasserve E; Société Française de Neurochirurgie (2005) Mutations within the programmed cell death 10 gene cause cerebral cavernous malformations. Am J Hum Genet 76:42-51
    Pubmed KoreaMed CrossRef
  8. Al-Holou WN, O'Lynnger TM, Pandey AS, Gemmete JJ, Thompson BG, Muraszko KM, Garton HJ, Maher CO (2012) Natural history and imaging prevalence of cavernous malformations in children and young adults. J Neurosurg Pediatr 9:198-205
    Pubmed CrossRef
  9. Draheim KM, Fisher OS, Boggon TJ, Calderwood DA (2014) Cerebral cavernous malformation proteins at a glance. J Cell Sci 127(Pt 4):701-707
    Pubmed KoreaMed CrossRef
  10. Haasdijk RA, Cheng C, Maat-Kievit AJ, Duckers HJ (2012) Cerebral cavernous malformations: from molecular pathogenesis to genetic counselling and clinical management. Eur J Hum Genet 20:134-140
    Pubmed KoreaMed CrossRef
  11. Voss K, Stahl S, Schleider E, Ullrich S, Nickel J, Mueller TD, Felbor U (2007) CCM3 interacts with CCM2 indicating common pathogenesis for cerebral cavernous malformations. Neurogenetics 8:249-256
    Pubmed CrossRef
  12. Lampugnani MG, Orsenigo F, Rudini N, Maddaluno L, Boulday G, Chapon F, Dejana E (2010) CCM1 regulates vascular-lumen organization by inducing endothelial polarity. J Cell Sci 123(Pt 7):1073-1080
    Pubmed CrossRef
  13. Faurobert E, Rome C, Lisowska J, Manet-Dupé S, Boulday G, Malbouyres M, Balland M, Bouin AP, Kéramidas M, Bouvard D, Coll JL, Ruggiero F, Tournier-Lasserve E, Albiges-Rizo C (2013) CCM1-ICAP-1 complex controls β1 integrin-dependent endothelial contractility and fibronectin remodeling. J Cell Biol 202:545-561
    Pubmed KoreaMed CrossRef
  14. Maddaluno L, Rudini N, Cuttano R, Bravi L, Giampietro C, Corada M, Ferrarini L, Orsenigo F, Papa E, Boulday G, Tournier-Lasserve E, Chapon F, Richichi C, Retta SF, Lampugnani MG, Dejana E (2013) EndMT contributes to the onset and progression of cerebral cavernous malformations. Nature 498:492-496
    Pubmed CrossRef
  15. Jenny Zhou H, Qin L, Zhang H, Tang W, Ji W, He Y, Liang X, Wang Z, Yuan Q, Vortmeyer A, Toomre D, Fuh G, Yan M, Kluger MS, Wu D, Min W (2016) Endothelial exocytosis of angiopoietin-2 resulting from CCM3 deficiency contributes to cerebral cavernous malformation. Nat Med 22:1033-1042
    Pubmed KoreaMed CrossRef
  16. Wüstehube J, Bartol A, Liebler SS, Brütsch R, Zhu Y, Felbor U, Sure U, Augustin HG, Fischer A (2010) Cerebral cavernous malformation protein CCM1 inhibits sprouting angiogenesis by activating DELTA-NOTCH signaling. Proc Natl Acad Sci U S A 107:12640-12645
    Pubmed KoreaMed CrossRef
  17. Glading A, Han J, Stockton RA, Ginsberg MH (2007) KRIT-1/CCM1 is a Rap1 effector that regulates endothelial cell cell junctions. J Cell Biol 179:247-254
    Pubmed KoreaMed CrossRef
  18. Whitehead KJ, Chan AC, Navankasattusas S, Koh W, London NR, Ling J, Mayo AH, Drakos SG, Jones CA, Zhu W, Marchuk DA, Davis GE, Li DY (2009) The cerebral cavernous malformation signaling pathway promotes vascular integrity via Rho GTPases. Nat Med 15:177-184
    Pubmed KoreaMed CrossRef
  19. Herberich SE, Klose R, Moll I, Yang WJ, Wüstehube-Lausch J, Fischer A (2015) ANKS1B interacts with the cerebral cavernous malformation protein-1 and controls endothelial permeability but not sprouting angiogenesis. PLoS One 10:e0145304
    Pubmed KoreaMed CrossRef
  20. Emaduddin M, Edelmann MJ, Kessler BM, Feller SM (2008) Odin (ANKS1A) is a Src family kinase target in colorectal cancer cells. Cell Commun Signal 6:7
    Pubmed KoreaMed CrossRef
  21. Lee H, Noh H, Mun J, Gu C, Sever S, Park S (2016) Anks1a regulates COPII-mediated anterograde transport of receptor tyrosine kinases critical for tumorigenesis. Nat Commun 7:12799
    Pubmed KoreaMed CrossRef
  22. Kim J, Lee H, Kim Y, Yoo S, Park E, Park S (2010) The SAM domains of Anks family proteins are critically involved in modulating the degradation of EphA receptors. Mol Cell Biol 30:1582-1592
    Pubmed KoreaMed CrossRef
  23. Zudaire E, Gambardella L, Kurcz C, Vermeren S (2011) A computational tool for quantitative analysis of vascular networks. PLoS One 6:e27385
    Pubmed KoreaMed CrossRef
  24. Manaenko A, Chen H, Kammer J, Zhang JH, Tang J (2011) Comparison Evans Blue injection routes: Intravenous versus intraperitoneal, for measurement of blood-brain barrier in a mice hemorrhage model. J Neurosci Methods 195:206-210
    Pubmed KoreaMed CrossRef
  25. Bernard-Patrzynski F, Lécuyer MA, Puscas I, Boukhatem I, Charabati M, Bourbonnière L, Ramassamy C, Leclair G, Prat A, Roullin VG (2019) Isolation of endothelial cells, pericytes and astrocytes from mouse brain. PLoS One 14:e0226302
    Pubmed KoreaMed CrossRef
  26. Assmann JC, Müller K, Wenzel J, Walther T, Brands J, Thornton P, Allan SM, Schwaninger M (2017) Isolation and cultivation of primary brain endothelial cells from adult mice. Bio Protoc 7:e2294
    Pubmed KoreaMed CrossRef
  27. Clatterbuck RE, Eberhart CG, Crain BJ, Rigamonti D (2001) Ultrastructural and immunocytochemical evidence that an incompetent blood-brain barrier is related to the pathophysiology of cavernous malformations. J Neurol Neurosurg Psychiatry 71:188-192
    Pubmed KoreaMed CrossRef
  28. Wolman M, Klatzo I, Chui E, Wilmes F, Nishimoto K, Fujiwara K, Spatz M (1981) Evaluation of the dye-protein tracers in pathophysiology of the blood-brain barrier. Acta Neuropathol 54:55-61
    Pubmed CrossRef
  29. Betjes MG, Haks MC, Tuk CW, Beelen RH (1991) Monoclonal antibody EBM11 (anti-CD68) discriminates between dendritic cells and macrophages after short-term culture. Immunobiology 183:79-87
    Pubmed CrossRef
  30. Nakano A, Harada T, Morikawa S, Kato Y (1990) Expression of leukocyte common antigen (CD45) on various human leukemia/lymphoma cell lines. Acta Pathol Jpn 40:107-115
    Pubmed CrossRef
  31. Haj-Yasein NN, Vindedal GF, Eilert-Olsen M, Gundersen GA, Skare Ø, Laake P, Klungland A, Thorén AE, Burkhardt JM, Ottersen OP, Nagelhus EA (2011) Glial-conditional deletion of aquaporin-4 (Aqp4) reduces blood-brain water uptake and confers barrier function on perivascular astrocyte endfeet. Proc Natl Acad Sci U S A 108:17815-17820
    Pubmed KoreaMed CrossRef
  32. Stevenson BR, Siliciano JD, Mooseker MS, Goodenough DA (1986) Identification of ZO-1: a high molecular weight polypeptide associated with the tight junction (zonula occludens) in a variety of epithelia. J Cell Biol 103:755-766
    Pubmed KoreaMed CrossRef
  33. Xu J, Lamouille S, Derynck R (2009) TGF-β-induced epithelial to mesenchymal transition. Cell Res 19:156-172
    Pubmed KoreaMed CrossRef
  34. Medici D, Kalluri R (2012) Endothelial-mesenchymal transition and its contribution to the emergence of stem cell phenotype. Semin Cancer Biol 22:379-384
    Pubmed KoreaMed CrossRef
  35. Sun Z, Zhao H, Fang D, Davis CT, Shi DS, Lei K, Rich BE, Winter JM, Guo L, Sorensen LK, Pryor RJ, Zhu N, Lu S, Dickey LL, Doty DJ, Tong Z, Thomas KR, Mueller AL, Grossmann AH, Zhang B, Lane TE, Fujinami RS, Odelberg SJ, Zhu W (2022) Neuroinflammatory disease disrupts the blood-CNS barrier via crosstalk between proinflammatory and endothelial-to-mesenchymal-transition signaling. Neuron 110:3106-3120.e7
    Pubmed KoreaMed CrossRef
  36. Yousef H, Czupalla CJ, Lee D, Chen MB, Burke AN, Zera KA, Zandstra J, Berber E, Lehallier B, Mathur V, Nair RV, Bonanno LN, Yang AC, Peterson T, Hadeiba H, Merkel T, Körbelin J, Schwaninger M, Buckwalter MS, Quake SR, Butcher EC, Wyss-Coray T (2019) Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1. Nat Med 25:988-1000
    Pubmed KoreaMed CrossRef
  37. Banks WA, Gray AM, Erickson MA, Salameh TS, Damodarasamy M, Sheibani N, Meabon JS, Wing EE, Morofuji Y, Cook DG, Reed MJ (2015) Lipopolysaccharide-induced blood-brain barrier disruption: roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit. J Neuroinflammation 12:223
    Pubmed KoreaMed CrossRef
  38. Bennett J, Basivireddy J, Kollar A, Biron KE, Reickmann P, Jefferies WA, McQuaid S (2010) Blood-brain barrier disruption and enhanced vascular permeability in the multiple sclerosis model EAE. J Neuroimmunol 229:180-191
    Pubmed CrossRef