Somatic variants of MAP3K3 are sufficient to cause cerebral and spinal cord cavernous malformations

Jian Ren, Yazi Huang, Yeqing Ren, Tianqi Tu, Baoshan Qiu, Daosheng Ai, Zhanying Bi, Xue Bai, Fengzhi Li, Jun-Liszt Li, Xing-Jun Chen, Ziyan Feng, Zongpei Guo, Jianfeng Lei, An Tian, Ziwei Cui, Volkhard Lindner, Ralf H Adams, Yibo Wang, Fei Zhao, Jakob Körbelin, Wenzhi Sun, Yilong Wang, Hongqi Zhang, Tao Hong, Woo-Ping Ge, Jian Ren, Yazi Huang, Yeqing Ren, Tianqi Tu, Baoshan Qiu, Daosheng Ai, Zhanying Bi, Xue Bai, Fengzhi Li, Jun-Liszt Li, Xing-Jun Chen, Ziyan Feng, Zongpei Guo, Jianfeng Lei, An Tian, Ziwei Cui, Volkhard Lindner, Ralf H Adams, Yibo Wang, Fei Zhao, Jakob Körbelin, Wenzhi Sun, Yilong Wang, Hongqi Zhang, Tao Hong, Woo-Ping Ge

Abstract

Cerebral cavernous malformations (CCMs) and spinal cord cavernous malformations (SCCMs) are common vascular abnormalities of the CNS that can lead to seizure, haemorrhage and other neurological deficits. Approximately 85% of patients present with sporadic (versus congenital) CCMs. Somatic mutations in MAP3K3 and PIK3CA were recently reported in patients with sporadic CCM, yet it remains unknown whether MAP3K3 mutation is sufficient to induce CCMs. Here we analysed whole-exome sequencing data for patients with CCM and found that ∼40% of them have a single, specific MAP3K3 mutation [c.1323C>G (p.Ile441Met)] but not any other known mutations in CCM-related genes. We developed a mouse model of CCM with MAP3K3I441M uniquely expressed in the endothelium of the CNS. We detected pathological phenotypes similar to those found in patients with MAP3K3I441M. The combination of in vivo imaging and genetic labelling revealed that CCMs were initiated with endothelial expansion followed by disruption of the blood-brain barrier. Experiments with our MAP3K3I441M mouse model demonstrated that CCM can be alleviated by treatment with rapamycin, the mTOR inhibitor. CCM pathogenesis has usually been attributed to acquisition of two or three distinct genetic mutations involving the genes CCM1/2/3 and/or PIK3CA. However, our results demonstrate that a single genetic hit is sufficient to cause CCMs.

Keywords: MAP3K3; cavernous malformations; cerebral; somatic mutation; spinal cord; ‌endothelial cell.

Conflict of interest statement

The authors report no competing interests.

© The Author(s) 2023. Published by Oxford University Press on behalf of the Guarantors of Brain.

Figures

Figure 1
Figure 1
Formation of CCMs induced by MAP3K3I441M mutations alone in mice of different ages. (A) Schematic of injection of AAV-BR1-MAP3K3I441M-P2A-EGFP (i.e. AAV-MAP3K3I441M) through the superficial temporal vein of a mouse. Regular T2 MRI was used for CCM detection after aden-associated virus (AAV) injection. (B) Experimental design of a series of brain MRI in the mice infected with MAP3K3I441M. (C) Quantification of the number of lesions in mouse brains in the first, second, fourth and sixth weeks after AAV injection. Compared with the data from the mice before AAV injection, no obvious difference in CCM numbers was identified in the first week (n = 5). In the second (n = 5) and fourth weeks (n = 4) after AAV-MAP3K3I441M injection, the number of lesions increased significantly. The number of lesions reached a plateau in the sixth week after AAV injection (n = 3). The density of lesion in different brain regions. The number of lesions in the brainstem were significantly lower than that in other brain areas. Ob = olfactory bulb; Cx = cerebral cortex; Th = striatum, thalamus and hippocampus; Bs = brainstem; Cm = cerebellum. n.s. = not significant; *P < 0.05; ***P < 0.001; one-way ANOVA. (D) Longitudinal T2 MRI of different sections of a mouse brain at first, fourth and sixth weeks after injection of AAV-BR1-MAP3K3I441M. Numerous microlesions were detected throughout the mouse brain. Quantification of the number of lesions in the MRI of different brain regions including olfactory bulb, cerebral cortex, striatum, thalamus and hippocampus, brainstem and cerebellum. MRI were performed at the first (n = 5 mice), fourth (n = 4 mice), and sixth (n = 3 mice) weeks. n.s. = not significant; **P < 0.01; ***P < 0.001; one-way ANOVA. (E) Administration of AAV-MAP3K3I441M in mice at different ages was essential to drive CCM formation in their brains and spinal cords. Mice aged P5, P35, P150 and P330 were injected with AAV-MAP3K3I441M and T2 MRI was performed 15 days later. MRI results showed that all mice of different ages developed CCMs in 2 weeks (arrowheads). (F) Example images of MAP3K3I441M induced CCM lesions (arrowheads) in mice. AAVs were injected at P35 and brains were harvested at P50. [F(iiv)] Insets from the brain. (G) Example images of MAP3K3I441M induced spinal cord CM lesions (arrowheads). AAVs were injected at P35 and spinal cords were harvested at P50. [G(iiii)] Insets from the spinal cord.
Figure 2
Figure 2
Characteristics and morphology of MAP3K3I441M -induced CCM lesions. (A) Representative images of H&E staining of MAP3K3I441M -induced CCM lesions (arrowhead, inseti) and haemorrhage (arrowheads, insetii) in mice. (B) Characteristics of MAP3K3I441M-induced CCM lesions and the control blood vessels in mouse brains (top), and characteristics of CCM lesions from a patient with MAP3K3I441M mutations (bottom). H&E staining (lesions, arrowheads in first panel; haemorrhage, arrowheads in second panel) and iron deposits of haemosiderin (patches, arrowheads in third panel) in MAP3K3I441M-induced CCM lesions from mice or brain tissue from human patients. Dilated vascular sinusoids were lined by endothelial cells (arrowheads in first panel). Haemorrhages and haemosiderin stains surrounded the lesions (second and third panels). The images from control groups are shown in the fourth panel. (C and D) The morphology of CMs induced by MAP3K3I441M in Cdh5-CreER::Ai14tg mice. Example images of brain sections from Cdh5-CreER::Ai14tg mice injected with AAV-BR1-MAP3K3I441M-P2A-EGFP. Representative images of MAP3K3I441M-induced CM lesions with dilated lumen (top, D) or distorted lumen (bottom, D). Tdt = tdTomato signal (red) in endothelial cells from Cdh5-CreER::Ai14tg; nuclei, DAPI (blue); GFP = weak green signal from endothelial cells (green) infected by AAV-BR1-MAP3K3I441M-P2A-EGFP. (E and F) The morphology of CMs induced by MAP3K3I441M in Pdgfrb-Cre::Ai14tg mice. Example images of brain sections stained with anti-Collagen IV in Pdgfrb-Cre::Ai14tg mice 2 weeks after infection with AAV-BR1-MAP3K3I441M. Normal blood vessels in brain region (E). Representative images of MAP3K3I441M-induced CM lesions with dilated lumen (F). Blood vessels, collagen IV (green); nuclei, DAPI (blue), and tdT signal (red) from pericytes in Pdgfrb-Cre::Ai14tg mice.
Figure 3
Figure 3
In vivo imaging of the development of CCMs caused by MAP3K3I441M. (A) Schematic of time-lapse live imaging of the brain vasculature in a conscious mouse. (B) Imaging of the development of a CCM lesion (arrowheads) located in a mouse brain through a chronic cranial window. Before AAV injection (baseline, left); 20 days after AAV injection (Day 20, middle). Inset: The same region within the dashed box with FITC-dextran (green) circulating in blood vessels (right). (CE) Three-dimensions of a z-stack image taken by a two-photon laser excitation microscope. Green = FITC-dextran; red = Dsred signal in the NG2BacDsRed transgenic(tg) mouse. (FH) CCM lesions (arrowheads) with leakage (top) and without leakage (bottom) in NG2BacDsRed tg mouse (F). Percentage of CCM lesions with leakage (n = 180 lesions in total). L(+) = leakage; L(−) = with no leakage (H). (GI) A CCM lesion (arrowheads) with coverage of pericytes (top) and without coverage of pericytes (bottom). (I) The percentage of lesions with pericyte [P(+)] or without pericyte coverage [P(−)](n = 180 lesions in total). (J) The coverage of pericytes in individual CCMs mediated by MAP3K3I441M mutations from the brain of Pdgfrb-Cre::Ai14 tg. Blood vessels were stained with antibodies against Collagen IV (green). Nuclei were stained with DAPI (blue). (K and L) Diameter and volume of the CCM lesions and normal vessels; *P < 0.05; **P < 0.01; ***P < 0.001; one-way ANOVA. (MO) Characterization of blood flow in individual CCMs induced by MAP3K3I441M mutations. Positions of line scanning in normal vessels (V1, V6), parent vessels (V2, V4) and cavernous vessels of CCM lesions (V3, V5). (N) The velocity of blood cells in vessels shown in (M) measured through line scanning. Images were plotted with X-T mode (the y-axis represents time and the x-axis represents distance) in the locations of V1 to V6. (O) Summarized results of red blood cell velocities in the upstream (Up, n = 7) and downstream (Down, n = 9) of the parent vessels, in normal blood vessels (n = 15), and in cavernous vessels of CCM lesions (n = 7), one-way ANOVA and *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 4
Figure 4
Staining of endothelial cell proliferation in tissues from MAP3K3I441M mouse models and CCM human samples. (A) Cell proliferation assays (EdU) in Cdh5-CreER::Ai14tg mice 2 weeks after infected with AAV-BR1-MAP3K3I441M. EdU positive cells (green, third row) were detected in the subventricular zone (SVZ), a neurogenic region in the brain. (B) No EdU+ endothelial cells were detected in MAP3K3I441M-mutated CCMs (0.0%, n = 0/50 lesions in total) or in normal vessels (0.0%, n = 0/100 normal vessels in total). (C) H&E staining and Ki67 staining in CCMs of human with MAP3K3I441M mutations. No positive immunoreaction for Ki67 was observed. (i) Insets from H&E staining in C. (D) Ki67 staining in the ‘control’ tissue (without CCMs) of the temporal lobe from an epilepsy patient. No positive immunoreaction for Ki67 was observed. (E) Ki67 staining in the tissue of human glioma. (F) Example images of normal capillaries and MAP3K3I441M mutated CCMs in Cdh5-CreER::Ai14 mice after injection of AAV- MAP3K3I441M. DAPI (blue) and tdTomato (tdT, red) signal from endothelial cells in Cdh5-CreER::Ai14tg. (G) Comparison of the membrane area of single endothelial cell of MAP3K3I441M induced CCMs (n = 10) with that of normal capillaries (n = 10) in Cdh5-CreER::Ai14 mice after injection of AAV- MAP3K3I441M. ***P < 0.001, unpaired two-tailed Student’s t-test. (H) Schematic of the morphology of endothelial cells with MAP3K3I441M expression and without MAP3K3I441M expression.
Figure 5
Figure 5
Molecular mechanism of the induction of CCMs by MAP3K3I441M. (A) Hierarchical cluster of all samples of bulk RNA-sequencing from brain endothelial cell lines, bEnd.3. Three samples from endothelial cells with lentivirus-MAP3K3I441M infection (Mut 1–3) and five control samples with lentivirus-EGFP infection (Ctrl 1–5). (B) Downregulated (blue) and upregulated (red) genes [log2 fold-change (FC) > 1.5, adjusted P-value < 0.05] in the endothelial cells with expression of MAP3K3I441M. (C) GSEA showed the activated pathway of structural constituent of cytoskeleton in the endothelial cells with MAP3K3I441M expression. (D) Normalized counts of the top eight genes encoding actins in bulk RNA-sequencing data acquired from the control endothelial cells (n = 5) and those cells with MAP3K3I441M expression (n = 3). ***P < 0.001, unpaired two-tailed Student’s t-test. (E) The heat map showing the z-scores of normalized counts for differential genes, as measured by bulk RNA-Sequencing. Some featured genes (e.g. Traf6, Actn1, etc.) were highlighted. (F) Western blot results for demonstrating MAP3K3-mTOR interactions. Co-immunoprecipitation (IP) results showed MAP3K3 interacted with mTOR.
Figure 6
Figure 6
Blocking mTOR activity with rapamycin prevents progression of MAP3K3I441M-mediated formation of CCMs. (A) Experimental design for our strategy of the targeted therapy using mTOR inhibitor rapamycin in mice with MAP3K3I441M CCMs. (B) T2 MRI of mouse brains after treatment using rapamycin or vehicle at the second and fourth week, and visual images of the hindbrains of mice treated by rapamycin or vehicle for 4 weeks. Scale bars = 1 mm. (C) Quantification of the number of CCM lesions in mice with and without the rapamycin treatment. Vehicle group, n = 7 mice; rapamycin, n = 9 mice; *P < 0.05, unpaired two-sided Student’s t-test. (D) Quantification of the increase of CCM lesion numbers between the second and fourth week after the treatment with vehicle or rapamycin. Vehicle, n = 7 mice; rapamycin, n = 9 mice; *P < 0.05, unpaired two-sided Student’s t-test. (E) H&E staining of mouse brains after treatment using rapamycin or vehicle at the fourth week (lesions, arrowheads). (i and ii) Insets from H&E staining in E. (FH) Quantification of CCM: the density of lesions (F), the diameter of lesions (G), and the area of a single lesion (H) with H&E staining at the fourth week in vehicle or rapamycin-treat CCM mice. ***P < 0.001, unpaired two-tailed Student’s t-test. (I) Autophagy marker, p62 staining of the brain sections from mice treated with rapamycin or vehicle for 4 weeks. (J) The number of p62 bodies was significantly decreased in the rapamycin-treated group. *P < 0.05, unpaired two-tailed Student’s t-test. (K) Experimental design for our strategy of the targeted therapy using mTOR inhibitor rapamycin in aged mice (18–24 months) with MAP3K3I441M CCMs. (L) Quantification of the number of CCM lesions in aged mice in vehicle or rapamycin-treated groups. Vehicle group, n = 8 mice; rapamycin, n = 4 mice; **P < 0.01, unpaired two-tailed Student’s t-test.

References

    1. Vernooij MW, Ikram MA, Tanghe HL, et al. . Incidental findings on brain MRI in the general population. N Engl J Med. 2007;357:1821–1828.
    1. Morris Z, Whiteley WN, Longstreth WT Jr, et al. . Incidental findings on brain magnetic resonance imaging: Systematic review and meta-analysis. BMJ. 2009;339:b3016.
    1. Batra S, Lin D, Recinos PF, Zhang J, Rigamonti D. Cavernous malformations: Natural history, diagnosis and treatment. Nat Rev Neurol. 2009;5:659–670.
    1. Chohan MO, Marchio S, Morrison LA, et al. . Emerging pharmacologic targets in cerebral cavernous malformation and potential strategies to Alter the natural history of a difficult disease: A review. JAMA Neurol. 2019;76:492–500.
    1. Laberge-le Couteulx S, Jung HH, Labauge P, et al. . Truncating mutations in CCM1, encoding KRIT1, cause hereditary cavernous angiomas. Nat Genet. 1999;23:189–193.
    1. Liquori CL, Berg MJ, Siegel AM, et al. . Mutations in a gene encoding a novel protein containing a phosphotyrosine-binding domain cause type 2 cerebral cavernous malformations. Am J Hum Genet. 2003;73:1459–1464.
    1. Bergametti F, Denier C, Labauge P, et al. . Mutations within the programmed cell death 10 gene cause cerebral cavernous malformations. Am J Hum Genet. 2005;76:42–51.
    1. Cox EM, Bambakidis NC, Cohen ML. Pathology of cavernous malformations. Handb Clin Neurol. 2017;143:267–277.
    1. Zafar A, Quadri SA, Farooqui M, et al. . Familial cerebral cavernous malformations. Stroke. 2019;50:1294–1301.
    1. Flemming KD, Graff-Radford J, Aakre J, et al. . Population-Based prevalence of cerebral cavernous malformations in older adults: Mayo clinic study of aging. JAMA Neurol. 2017;74:801–805.
    1. Hong T, Xiao X, Ren J, et al. . Somatic MAP3K3 and PIK3CA mutations in sporadic cerebral and spinal cord cavernous malformations. Brain. 2021;144:2648–2658.
    1. Weng J, Yang Y, Song D, et al. . Somatic MAP3K3 mutation defines a subclass of cerebral cavernous malformation. Am J Hum Genet. 2021;108:942–950.
    1. Peyre M, Miyagishima D, Bielle F, et al. . Somatic PIK3CA mutations in sporadic cerebral cavernous malformations. N Engl J Med. 2021;385:996–1004.
    1. Snellings DA, Girard R, Lightle R, et al. . Developmental venous anomalies are a genetic primer for cerebral cavernous malformations. Nat Cardiovasc Res. 2022;1:246–252.
    1. Ren AA, Snellings DA, Su YS, et al. . PIK3CA And CCM mutations fuel cavernomas through a cancer-like mechanism. Nature. 2021;594:271–276.
    1. Akers AL, Johnson E, Steinberg GK, Zabramski JM, Marchuk DA. Biallelic somatic and germline mutations in Cerebral Cavernous Malformations (CCMs): Evidence for a two-hit mechanism of CCM pathogenesis. Hum Mol Genet. 2009;18:919–930.
    1. Zhou HJ, Qin L, Jiang Q, et al. . Caveolae-mediated tie2 signaling contributes to CCM pathogenesis in a brain endothelial cell-specific Pdcd10-deficient mouse model. Nat Commun. 2021;12:504.
    1. Zhou Z, Tang AT, Wong WY, et al. . Cerebral cavernous malformations arise from endothelial gain of MEKK3-KLF2/4 signalling. Nature. 2016;532:122–126.
    1. Maddaluno L, Rudini N, Cuttano R, et al. . EndMT contributes to the onset and progression of cerebral cavernous malformations. Nature. 2013;498:492–496.
    1. Tang AT, Choi JP, Kotzin JJ, et al. . Endothelial TLR4 and the microbiome drive cerebral cavernous malformations. Nature. 2017;545:305–310.
    1. Korbelin J, Dogbevia G, Michelfelder S, et al. . A brain microvasculature endothelial cell-specific viral vector with the potential to treat neurovascular and neurological diseases. EMBO Mol Med. 2016;8:609–625.
    1. Jia JM, Peng C, Wang Y, Zheng J, Ge WP. Control of occlusion of middle cerebral artery in perinatal and neonatal mice with magnetic force. Mol Brain. 2018;11:47.
    1. Jia JM, Chowdary PD, Gao X, et al. . Control of cerebral ischemia with magnetic nanoparticles. Nat Methods. 2017;14:160–166.
    1. Hong T, Yan Y, Li J, et al. . High prevalence of KRAS/BRAF somatic mutations in brain and spinal cord arteriovenous malformations. Brain. 2019;142:23–34.
    1. Fisher OS, Deng H, Liu D, et al. . Structure and vascular function of MEKK3-cerebral cavernous malformations 2 complex. Nat Commun. 2015;6:7937.
    1. Cuttano R, Rudini N, Bravi L, et al. . KLF4 Is a key determinant in the development and progression of cerebral cavernous malformations. EMBO Mol Med. 2016;8:6–24.
    1. Castro M, Lavina B, Ando K, et al. . CDC42 Deletion elicits cerebral vascular malformations via increased MEKK3-dependent KLF4 expression. Circ Res. 2019;124:1240–1252.
    1. Zhu X, Bergles DE, Nishiyama A. NG2 Cells generate both oligodendrocytes and gray matter astrocytes. Development. 2008;135:145–157.
    1. Hill RA, Tong L, Yuan P, Murikinati S, Gupta S, Grutzendler J. Regional blood flow in the normal and ischemic brain is controlled by arteriolar smooth muscle cell contractility and not by capillary pericytes. Neuron. 2015;87:95–110.
    1. Whitehead KJ, Chan AC, Navankasattusas S, et al. . The cerebral cavernous malformation signaling pathway promotes vascular integrity via rho GTPases. Nat Med. 2009;15:177–184.
    1. Jenny Zhou H, Qin L, Zhang H, et al. . Endothelial exocytosis of angiopoietin-2 resulting from CCM3 deficiency contributes to cerebral cavernous malformation. Nat Med. 2016;22:1033–1042.
    1. Girard R, Fam MD, Zeineddine HA, et al. . Vascular permeability and iron deposition biomarkers in longitudinal follow-up of cerebral cavernous malformations. J Neurosurg. 2017;127:102–110.
    1. Madisen L, Zwingman TA, Sunkin SM, et al. . A robust and high-throughput cre reporting and characterization system for the whole mouse brain. Nat Neurosci. 2010;13:133–140.
    1. Chan-Ling T, Page MP, Gardiner T, Baxter L, Rosinova E, Hughes S. Desmin ensheathment ratio as an indicator of vessel stability: Evidence in normal development and in retinopathy of prematurity. Am J Pathol. 2004;165:1301–1313.
    1. Armulik A, Genove G, Mae M, et al. . Pericytes regulate the blood-brain barrier. Nature. 2010;468:557–561.
    1. Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature. 2010;468:562–566.
    1. Cuttler AS, LeClair RJ, Stohn JP, et al. . Characterization of pdgfrb-cre transgenic mice reveals reduction of ROSA26 reporter activity in remodeling arteries. Genesis. 2011;49:673–680.
    1. Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy fights disease through cellular self-digestion. Nature. 2008;451:1069–1075.
    1. Linares JF, Duran A, Reina-Campos M, et al. . Amino acid activation of mTORC1 by a PB1-domain-driven kinase Complex cascade. Cell Rep. 2015;12:1339–1352.
    1. Castillo SD, Tzouanacou E, Zaw-Thin M, et al. . Somatic activating mutations in Pik3ca cause sporadic venous malformations in mice and humans. Sci Transl Med. 2016;8:332ra43.
    1. Boscolo E, Limaye N, Huang L, et al. . Rapamycin improves TIE2-mutated venous malformation in murine model and human subjects. J Clin Invest. 2015;125:3491–3504.
    1. Fish JE, Flores Suarez CP, Boudreau E, et al. . Somatic gain of KRAS function in the endothelium is sufficient to cause vascular malformations that require MEK but not PI3K signaling. Circ Res. 2020;127:727–743.
    1. Park ES, Kim S, Huang S, et al. . Selective endothelial hyperactivation of oncogenic KRAS induces brain arteriovenous malformations in mice. Ann Neurol. 2021;89:926–941.
    1. Ren J, Jiang N, Bian L, et al. . Natural history of spinal cord cavernous malformations: A Multicenter Cohort Study. Neurosurgery. 2022;90:390–398.
    1. Stapleton CJ, Barker FG. 2nd. Cranial cavernous malformations: Natural history and treatment. Stroke. 2018;49:1029–1035.
    1. Badhiwala JH, Farrokhyar F, Alhazzani W, et al. . Surgical outcomes and natural history of intramedullary spinal cord cavernous malformations: a single-center series and meta-analysis of individual patient data: clinic article. J Neurosurg Spine. 2014;21:662–676.
    1. Akers A, Al-Shahi Salman R, AA I, et al. . Synopsis of guidelines for the clinical management of cerebral cavernous malformations: Consensus recommendations based on systematic literature review by the angioma alliance scientific advisory board clinical experts panel. Neurosurgery. 2017;80:665–680.
    1. Snellings DA, Hong CC, Ren AA, et al. . Cerebral cavernous malformation: From mechanism to therapy. Circ Res. 2021;129:195–215.
    1. Huo R, Wang J, Sun YF, et al. . Simplex cerebral cavernous malformations with MAP3K3 mutation have distinct clinical characteristics. Front Neurol. 2022;13:946324.

Source: PubMed

3
Iratkozz fel