1
|
Pedrosa L, Mosteiro A, Reyes L, Amaro S, Menéndez-Girón S, Rivera MC, Domínguez CJ, Planas AM, Torné R, Rodríguez-Hernández A. Profiling Tight Junction Protein Expression in Brain Vascular Malformations. Int J Mol Sci 2025; 26:4558. [PMID: 40429705 PMCID: PMC12111537 DOI: 10.3390/ijms26104558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/17/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Recent studies suggest that blood-brain barrier (BBB) disruption plays a key role in the clinical course and bleeding risk of brain arteriovenous malformations (bAVMs). The tight junctions (TJs) are complex endothelial transmembrane proteins with a significant physical contribution to BBB disruption. In this study, we hypothesized that bAVMs display a different TJ pattern than other vascular malformations and normal brain tissue. We studied the expression of claudin-5 and occludin as essential factors for functional TJs. Human specimens of surgically resected cavernomas (CCMs) (n = 9), bAVMs (n = 17), and perilesional brain parenchyma (6 from CCMs and 16 from bAVM patients) were analyzed via immunofluorescence staining, transmission electron microscopy (TEM), and Western blot tests. Compared to perilesional parenchyma, bAVMs showed a significant decrease in TJ protein expression, and these alterations were more apparent in ruptured bAVMs than in unruptured bAVMs or CCMs. TEM images provided evidence of disrupted connectivity between endothelial cells of bAVMs. This is the first clinical investigation that studies the expression of TJs in human bAVMs and their surrounding parenchyma. Despite the limitations of the sample size, we found significant differences in the expression and composition of TJs in bAVMs when compared to surrounding parenchyma and other vascular lesions such as CCMs. These results add further evidence to the role of BBB disruption in the clinical course of bAVM. A deeper understanding of these mechanisms may lead to the development of new therapeutic targets and management strategies for bAVMs.
Collapse
Affiliation(s)
- Leire Pedrosa
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (L.P.); (S.A.); (A.M.P.)
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Alejandra Mosteiro
- Department of Neurosurgery, Institute of Neuroscience, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (A.M.); (L.R.)
| | - Luis Reyes
- Department of Neurosurgery, Institute of Neuroscience, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (A.M.); (L.R.)
| | - Sergio Amaro
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (L.P.); (S.A.); (A.M.P.)
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Comprehensive Stroke Center, Institute of Neuroscience, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Sebastián Menéndez-Girón
- Department of Neurosurgery, Germans Trias i Pujol University Hospital, Carretera de Canyet, s/n, 08916 Badalona, Spain; (S.M.-G.); (M.C.R.); (C.J.D.)
| | - Mateo Cortés Rivera
- Department of Neurosurgery, Germans Trias i Pujol University Hospital, Carretera de Canyet, s/n, 08916 Badalona, Spain; (S.M.-G.); (M.C.R.); (C.J.D.)
| | - Carlos J. Domínguez
- Department of Neurosurgery, Germans Trias i Pujol University Hospital, Carretera de Canyet, s/n, 08916 Badalona, Spain; (S.M.-G.); (M.C.R.); (C.J.D.)
| | - Anna M. Planas
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (L.P.); (S.A.); (A.M.P.)
- Department of Neuroscience and Experimental Therapeutics, Institute for Biomedical Research of Barcelona (IIBB), Spanish Research Council (CSIC), 08036 Barcelona, Spain
| | - Ramon Torné
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (L.P.); (S.A.); (A.M.P.)
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Department of Neurosurgery, Institute of Neuroscience, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (A.M.); (L.R.)
| | - Ana Rodríguez-Hernández
- Department of Neurosurgery, Germans Trias i Pujol University Hospital, Carretera de Canyet, s/n, 08916 Badalona, Spain; (S.M.-G.); (M.C.R.); (C.J.D.)
| |
Collapse
|
2
|
Sun Y, Xu H, Zhu Y, Rao Y, Fan X, Wang Z, Gu H, Yue X, Zhao X, Su L, Cai R. Single-cell and spatial transcriptomic analyses reveal transcriptional cell lineage heterogeneity in extracranial arteriovenous malformation. J Dermatol Sci 2025; 118:66-75. [PMID: 40118698 DOI: 10.1016/j.jdermsci.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Extracranial arteriovenous malformations (eAVMs) are rare congenital vascular anomalies consisting of abnormal artery-vein bypass with no intervening capillary network, and can lead to disability and death. The critical genetic determination factors and key transcriptional pathways of the eAVMs genesis process are still unclear. OBJECTIVE To generate an overview of the molecular information within eAVMs at the single-cell level. METHODS We performed single-cell RNA sequencing (scRNA-seq) on nine samples of eAVMs receiving a confirmatory histopathologic evaluation from a board-certified dermatopathologist and two nonlesional tissue sample controls. 10x Visium spatial transcriptomics (ST) was performed on one eAVM to spatially localize heterogeneous cells and profile the gene expression dynamics of the cells in their morphological context. The scRNA-seq and ST data were integrated and analyzed to further query for spatially restricted mapping of intrapopulation heterogeneous cells. RESULTS We identified different cell states of endothelial cells (ECs), perivascular cells and immune cells in eAVMs, uncovered the presence of MAFB+ nidus ECs, characterized mesenchymal activation in ECs, and identified transcriptional variation within perivascular cells and the presence of smooth muscle-like pericytes in eAVMs. Dysregulated cell to cell interactions among ECs, perivascular cells and immune cells that are associated with eAVMs, including those involving MDK, VEGF, ANGPT, SEMA3 and GALECTIN-9 were cataloged. Together, our results depicted the heterogeneity underlying cell function and interaction of eAVMs at a single-cell resolution. CONCLUSION We present a comprehensive picture of the cell-resolution atlas that describes the transcriptomic heterogeneity underlying cell function and interaction in eAVMs.
Collapse
Affiliation(s)
- Yi Sun
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyang Xu
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanze Zhu
- School of Automation, Northwestern Polytechnical University, Xi 'an, China
| | - Yamin Rao
- Department of Pathology, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xindong Fan
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenfeng Wang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Gu
- Department of Burn and Plastic Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaojie Yue
- Department of Burn and Plastic Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiong Zhao
- Department of Burn and Plastic Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Lixin Su
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ren Cai
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Jimenez-Macias JL, Vaughn-Beaucaire P, Bharati A, Xu Z, Forrest M, Hong J, Sun M, Schmidt A, Clark J, Hawkins W, Mercado N, Real J, Huntington K, Zdioruk M, Nowicki MO, Cho CF, Wu B, Li W, Logan T, Manz KE, Pennell KD, Fedeles BI, Bertone P, Punsoni M, Brodsky AS, Lawler SE. Modulation of blood-tumor barrier transcriptional programs improves intratumoral drug delivery and potentiates chemotherapy in GBM. SCIENCE ADVANCES 2025; 11:eadr1481. [PMID: 40009687 PMCID: PMC11864199 DOI: 10.1126/sciadv.adr1481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
Efficient drug delivery to glioblastoma (GBM) is a major obstacle as the blood-brain barrier (BBB) and the blood-tumor barrier (BTB) prevent passage of the majority of chemotherapies into the brain. Here, we identified a transcriptional 12-gene signature associated with the BTB in GBM. We identified CDH5 as a core molecule in this set and confirmed its expression in GBM vasculature using transcriptomics and immunostaining of patient specimens. The indirubin-derivative, 6-bromoindirubin acetoxime (BIA), down-regulates CDH5 and other BTB signature genes, causing endothelial barrier disruption in vitro and in murine GBM xenograft models. Treatment with BIA increased intratumoral cisplatin accumulation and potentiated DNA damage by targeting DNA repair pathways. Last, using an injectable BIA nanoparticle formulation, PPRX-1701, we significantly improved cisplatin efficacy in murine GBM. Our work reveals potential targets of the BTB and the bifunctional properties of BIA as a BTB modulator and a potentiator of chemotherapy, supporting its further development.
Collapse
Affiliation(s)
- Jorge L. Jimenez-Macias
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Philippa Vaughn-Beaucaire
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ayush Bharati
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Zheyun Xu
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Megan Forrest
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Jason Hong
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Michael Sun
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Andrea Schmidt
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Jasmine Clark
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - William Hawkins
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Noe Mercado
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Jacqueline Real
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Kelsey Huntington
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Mykola Zdioruk
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michal O. Nowicki
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Choi-Fong Cho
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02115, USA
| | - Bin Wu
- Cytodigm Inc, Natick, MA 01760, USA
| | - Weiyi Li
- Phosphorex Inc, Hopkinton, MA 01748, USA
| | | | | | - Kurt D. Pennell
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Bogdan I. Fedeles
- Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Paul Bertone
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
- Department of Medicine, Brown University, Providence, RI 02903, USA
| | - Michael Punsoni
- Brown University Health, Warren Alpert Medical School, Providence, RI 02903, USA
| | - Alexander S. Brodsky
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Sean E. Lawler
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| |
Collapse
|
4
|
Ricciardelli AR, Genet G, Genet N, McClugage ST, Kan PT, Hirschi KK, Fish JE, Wythe JD. From bench to bedside: murine models of inherited and sporadic brain arteriovenous malformations. Angiogenesis 2025; 28:15. [PMID: 39899215 PMCID: PMC11790818 DOI: 10.1007/s10456-024-09953-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/06/2024] [Indexed: 02/04/2025]
Abstract
Brain arteriovenous malformations are abnormal vascular structures in which an artery shunts high pressure blood directly to a vein without an intervening capillary bed. These lesions become highly remodeled over time and are prone to rupture. Historically, brain arteriovenous malformations have been challenging to treat, using primarily surgical approaches. Over the past few decades, the genetic causes of these malformations have been uncovered. These can be divided into (1) familial forms, such as loss of function mutations in TGF-β (BMP9/10) components in hereditary hemorrhagic telangiectasia, or (2) sporadic forms, resulting from somatic gain of function mutations in genes involved in the RAS-MAPK signaling pathway. Leveraging these genetic discoveries, preclinical mouse models have been developed to uncover the mechanisms underlying abnormal vessel formation, and thus revealing potential therapeutic targets. Impressively, initial preclinical studies suggest that pharmacological treatments disrupting these aberrant pathways may ameliorate the abnormal pathologic vessel remodeling and inflammatory and hemorrhagic nature of these high-flow vascular anomalies. Intriguingly, these studies also suggest uncontrolled angiogenic signaling may be a major driver in bAVM pathogenesis. This comprehensive review describes the genetics underlying both inherited and sporadic bAVM and details the state of the field regarding murine models of bAVM, highlighting emerging therapeutic targets that may transform our approach to treating these devastating lesions.
Collapse
Affiliation(s)
| | - Gael Genet
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Nafiisha Genet
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Samuel T McClugage
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA
- Division of Pediatric Neurosurgery, Texas Children's Hospital, Houston, TX, USA
| | - Peter T Kan
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX, 77598, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Developmental Genomics Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Joshua D Wythe
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Developmental Genomics Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Brain, Immunology, and Glia Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
5
|
Mehdi SJ, Zhang H, Sun RW, Richter GT, Strub GM. Mural Cells Initiate Endothelial-to-Mesenchymal Transition in Adjacent Endothelial Cells in Extracranial AVMs. Cells 2024; 13:2122. [PMID: 39768212 PMCID: PMC11727354 DOI: 10.3390/cells13242122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Extracranial arteriovenous malformations (eAVMs) are complex vascular lesions characterized by anomalous arteriovenous connections, vascular instability, and disruptions in endothelial cell (EC)-to-mural cell (MC) interactions. This study sought to determine whether eAVM-MCs could induce endothelial-to-mesenchymal transition (EndMT), a process known to disrupt vascular integrity, in the eAVM microenvironment. eAVM and paired control tissues were analyzed using RT-PCR for EC (CD31, CD34, and CDH5) and EndMT-specific markers (SNAI1, SNAI2, ACTA2/α-SMA, N-cadherin/CDH2, VIM). Immunohistochemistry (IHC) was also performed to analyze MC- (PDGFR-β and α-SMA), EC (CD31, CD34, and CDH5), and EndMT-specific markers (CDH2 and SNAI1) in sequential paraffin-embedded sections of eAVM patient biopsies and in adjacent normal tissue biopsies from the same patients. Furthermore, eAVM-MCs and MCs from normal paired tissues (NMCs) were then isolated from fresh human surgical samples using flow cytometry and co-cultured with normal human umbilical vein vascular endothelial cells (HUVECs), followed by analysis of CD31 by immunofluorescence. RT-PCR analysis did not show a significant difference in the expression of EC markers between eAVM tissues and controls, whereas expression of EndMT-specific markers was upregulated in eAVM tissues compared to controls. IHC of eAVMs and paired control tissues demonstrated regions of significant perivascular eAVM-MC expansion (PDGFR-β+, and α-SMA+) surrounding dilated, morphologically abnormal vessels. These regions contained endothelium undergoing EndMT as evidenced by loss of CD31, CD34, and CDH5 expression and upregulation of the EndMT-associated genes CDH2 and SNAI1. Isolated eAVM-MCs induced loss of CD31 in HUVECs when grown in co-culture, while NMCs did not. This study suggests that the eAVM endothelium surrounded by expanded eAVM-MCs undergoes EndMT, potentially leading to the formation of dilated and fragile vessels, and implicates the eAVM-MCs in EndMT initiation and eAVM pathology.
Collapse
Affiliation(s)
- Syed J. Mehdi
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Haihong Zhang
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Ravi W. Sun
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Gresham T. Richter
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Graham M. Strub
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| |
Collapse
|
6
|
Tu T, Zhang S, Li J, Jiang C, Ren J, Zhang S, Meng X, Peng H, Xing D, Zhang H, Hong T, Yu J. Inhibition of Angiopoietin-2 rescues sporadic brain arteriovenous malformations by reducing pericyte loss. Angiogenesis 2024; 28:3. [PMID: 39636449 DOI: 10.1007/s10456-024-09957-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/11/2024] [Indexed: 12/07/2024]
Abstract
Brain arteriovenous malformations (bAVMs) are a major cause of hemorrhagic stroke in children and young adults. These lesions are thought to result from somatic KRAS/BRAF mutations in brain endothelial cells (bECs). In this study, we introduce a new bAVM model by inducing a brain endothelial-specific BrafV600E mutation using the Slc1o1c1(BAC)-CreER driver line. The pathological characteristics of this model resemble human bAVMs, including dilated and hyperpermeable vessels, as well as parenchymal hemorrhage. We observed that these lesions showed a typical reduction in pericyte coverage and disruption of the pericyte-endothelial cell connection. Additionally, we found that ANGPT2 levels were significantly increased in the endothelium of bAVM lesions, which may be a critical factor in the pericyte deficits of the malformed vessels. Treatment with an ANGPT2 neutralizing antibody confirmed that blocking ANGPT2 can restore pericyte density in bAVM lesions, improve pericyte coverage around microvessels, enhance tight junction protein coverage related to endothelial cells, and normalize endothelial barrier function. In summary, our findings suggest that increased ANGPT2 expression in endothelial cells with the BrafV600E mutation is a key factor in pericyte deficiencies in bAVMs, highlighting the potential effectiveness of anti-ANGPT2 therapy in treating bAVMs.
Collapse
Affiliation(s)
- Tianqi Tu
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Shikun Zhang
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Jingwei Li
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Chendan Jiang
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Jian Ren
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Shiju Zhang
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Xiaosheng Meng
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China
| | - Hao Peng
- Department of Neurosurgery in Hainan General Hospital, Hainan Medical University, Hainan, China
- Department of neurosurgery, The second people's hospital of hainan province, Hainan, China
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China.
| | - Tao Hong
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China.
| | - Jiaxing Yu
- Department of Neurosurgery, Xuanwu Hospital, International Neuroscience Institute, Capital Medical University, Beijing, 100053, China.
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St, Beijing, 100053, China.
| |
Collapse
|
7
|
He Q, Huo R, Sun Y, Zheng Z, Xu H, Zhao S, Ni Y, Yu Q, Jiao Y, Zhang W, Zhao J, Cao Y. Cerebral vascular malformations: pathogenesis and therapy. MedComm (Beijing) 2024; 5:e70027. [PMID: 39654683 PMCID: PMC11625509 DOI: 10.1002/mco2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Cerebral vascular malformations (CVMs), particularly cerebral cavernous malformations and cerebral arteriovenous malformations, pose significant neurological challenges due to their complex etiologies and clinical implications. Traditionally viewed as congenital conditions with structural abnormalities, CVMs have been treated primarily through resection, embolization, and stereotactic radiosurgery. While these approaches offer some efficacy, they often pose risks to neurological integrity due to their invasive nature. Advances in next-generation sequencing, particularly high-depth whole-exome sequencing and bioinformatics, have facilitated the identification of gene variants from neurosurgically resected CVMs samples. These advancements have deepened our understanding of CVM pathogenesis. Somatic mutations in key mechanistic pathways have been identified as causative factors, leading to a paradigm shift in CVM treatment. Additionally, recent progress in noninvasive and minimally invasive techniques, including gene imaging genomics, liquid biopsy, or endovascular biopsies (endovascular sampling of blood vessel lumens), has enabled the identification of gene variants associated with CVMs. These methods, in conjunction with clinical data, offer potential for early detection, dynamic monitoring, and targeted therapies that could be used as monotherapy or adjuncts to surgery. This review highlights advancements in CVM pathogenesis and precision therapies, outlining the future potential of precision medicine in CVM management.
Collapse
Affiliation(s)
- Qiheng He
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Ran Huo
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yingfan Sun
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Zhiyao Zheng
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Research Unit of Accurate DiagnosisTreatment, and Translational Medicine of Brain Tumors Chinese Academy of Medical Sciences and Peking Union Medical College Beijing ChinaBeijingChina
- Department of Neurosurgery Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical College Beijing ChinaBeijingChina
| | - Hongyuan Xu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Shaozhi Zhao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yang Ni
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Qifeng Yu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yuming Jiao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Wenqian Zhang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Jizong Zhao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yong Cao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijingChina
- Collaborative Innovation CenterBeijing Institute of Brain DisordersBeijingChina
| |
Collapse
|
8
|
Saito S, Nakamura Y, Miyashita S, Sato T, Hoshina K, Okada M, Hasegawa H, Oishi M, Fujii Y, Körbelin J, Kubota Y, Tainaka K, Natsumeda M, Ueno M. CRISPR/CasRx suppresses KRAS-induced brain arteriovenous malformation developed in postnatal brain endothelial cells in mice. JCI Insight 2024; 9:e179729. [PMID: 39576014 PMCID: PMC11601911 DOI: 10.1172/jci.insight.179729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/02/2024] [Indexed: 11/29/2024] Open
Abstract
Brain arteriovenous malformations (bAVMs) are anomalies forming vascular tangles connecting the arteries and veins, which cause hemorrhagic stroke in young adults. Current surgical approaches are highly invasive, and alternative therapeutic methods are warranted. Recent genetic studies identified KRAS mutations in endothelial cells of bAVMs; however, the underlying process leading to malformation in the postnatal stage remains unknown. Here we established a mouse model of bAVM developing during the early postnatal stage. Among 4 methods tested, mutant KRAS specifically introduced in brain endothelial cells by brain endothelial cell-directed adeno-associated virus (AAV) and endothelial cell-specific Cdh5-CreERT2 mice successfully induced bAVMs in the postnatal period. Mutant KRAS led to the development of multiple vascular tangles and hemorrhage in the brain with increased MAPK/ERK signaling and growth in endothelial cells. Three-dimensional analyses in cleared tissue revealed dilated vascular networks connecting arteries and veins, similar to human bAVMs. Single-cell RNA-Seq revealed dysregulated gene expressions in endothelial cells and multiple cell types involved in the pathological process. Finally, we employed CRISPR/CasRx to knock down mutant KRAS expression, which efficiently suppressed bAVM development. The present model reveals pathological processes that lead to postnatal bAVMs and demonstrates the efficacy of therapeutic strategies with CRISPR/CasRx.
Collapse
Affiliation(s)
- Shoji Saito
- Department of Neurosurgery and
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yuka Nakamura
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Satoshi Miyashita
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tokiharu Sato
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kana Hoshina
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | | | | | | | | | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | | | - Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
9
|
Nasim S, Bichsel C, Pinto A, Alexandrescu S, Kozakewich H, Bischoff J. Similarities and differences between brain and skin GNAQ p.R183Q driven capillary malformations. Angiogenesis 2024; 27:931-941. [PMID: 39343803 DOI: 10.1007/s10456-024-09950-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024]
Abstract
Capillary malformations (CM) are congenital vascular irregularities of capillary and venous blood vessels that appear in the skin, leptomeninges of the brain, and the choroid of the eye in the disorder known as Sturge Weber Syndrome (SWS). More common are non-syndromic CM found only in the skin, without brain or ocular involvement. A somatic activating mutation in GNAQ (p.R183Q) is found in ~ 90% of syndromic and non-syndromic CM specimens and is present in CD31pos endothelial cells isolated from brain and skin CM specimens. Endothelial expression of the GNAQ p.R183Q variant is sufficient to form CM-like vessels in mice. Given the distinct features and functions of blood vessels in the brain versus the skin, we examined the features of CM vessels in both tissues to gain insights into the pathogenesis of CM. Herein, we present morphologic characteristics of CM observed in specimens from brain and skin. The GNAQ p.R183Q variant allelic frequency in each specimen was determined by droplet digital PCR. Sections were stained for endothelial cells, tight junctions, mural cells, and macrophages to assess the endothelium as well as perivascular constituents. CM blood vessels in brain and skin were enlarged, exhibited fibrin leakage and reduced zona occludin-1 and claudin-5, and were surrounded by MRC1pos/LYVE1pos macrophages. In contrast, the CMs from brain and skin differ in endothelial sprouting activity and localization of mural cells. These characteristics might be helpful in the development of targeted and/or tissue specific therapies to prevent or reverse non-syndromic and syndromic CM.
Collapse
Affiliation(s)
- Sana Nasim
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Colette Bichsel
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- CSEM SA, Hegenheimermattweg 167 A, 4123, Allschwil, Switzerland
| | - Anna Pinto
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Sanda Alexandrescu
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Harry Kozakewich
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Joyce Bischoff
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
10
|
Jeong JY, Bafor AE, Freeman BH, Chen PR, Park ES, Kim E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines 2024; 12:1795. [PMID: 39200259 PMCID: PMC11351371 DOI: 10.3390/biomedicines12081795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Brain arteriovenous malformations (bAVMs) substantially increase the risk for intracerebral hemorrhage (ICH), which is associated with significant morbidity and mortality. However, the treatment options for bAVMs are severely limited, primarily relying on invasive methods that carry their own risks for intraoperative hemorrhage or even death. Currently, there are no pharmaceutical agents shown to treat this condition, primarily due to a poor understanding of bAVM pathophysiology. For the last decade, bAVM research has made significant advances, including the identification of novel genetic mutations and relevant signaling in bAVM development. However, bAVM pathophysiology is still largely unclear. Further investigation is required to understand the detailed cellular and molecular mechanisms involved, which will enable the development of safer and more effective treatment options. Endothelial cells (ECs), the cells that line the vascular lumen, are integral to the pathogenesis of bAVMs. Understanding the fundamental role of ECs in pathological conditions is crucial to unraveling bAVM pathophysiology. This review focuses on the current knowledge of bAVM-relevant signaling pathways and dysfunctions in ECs, particularly the endothelial-to-mesenchymal transition (EndMT).
Collapse
Affiliation(s)
| | | | | | | | | | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.Y.J.); (A.E.B.); (B.H.F.); (P.R.C.); (E.S.P.)
| |
Collapse
|
11
|
Nasim S, Bichsel C, Pinto A, Alexandrescu S, Kozakewich H, Bischoff J. Similarities and differences between brain and skin GNAQ p.R183Q driven capillary malformations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599711. [PMID: 38948880 PMCID: PMC11213000 DOI: 10.1101/2024.06.19.599711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Capillary malformations (CM) are congenital vascular irregularities of capillary and venous blood vessels that appear in the skin, leptomeninges of the brain, and the choroid of the eye in the disorder known as Sturge Weber Syndrome (SWS). More common are non-syndromic CM found only in the skin, without brain or ocular involvement. A somatic activating mutation in GNAQ (p.R183Q) is found in ~90% of syndromic and non-syndromic CM specimens and is present in CD31pos endothelial cells isolated from brain and skin CM specimens. Endothelial expression of the GNAQ p.R183Q variant is sufficient to form CM-like vessels in mice. Given the distinct features and functions of blood vessels in the brain versus the skin, we examined the features of CM vessels in both tissues to gain insights into the pathogenesis of CM. Herein, we present morphologic characteristics of CM observed in specimen from brain and skin. The GNAQ p.R183Q variant allelic frequency in each specimen was determined by droplet digital PCR. Sections were stained for endothelial cells, tight junctions, mural cells, and macrophages to assess the endothelium as well as perivascular constituents. CM blood vessels in brain and skin were enlarged, exhibited fibrin leakage and reduced zona occludin-1, and were surrounded by MRC1pos/LYVE1pos macrophages. In contrast, the CMs from brain and skin differ in endothelial sprouting activity and localization of mural cells. These characteristics might be helpful in the development of targeted and/or tissue specific therapies to prevent or reverse non-syndromic and syndromic CM.
Collapse
Affiliation(s)
- Sana Nasim
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Colette Bichsel
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Anna Pinto
- Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Sanda Alexandrescu
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Harry Kozakewich
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Joyce Bischoff
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
12
|
Mansur A, Radovanovic I. Defining the Role of Oral Pathway Inhibitors as Targeted Therapeutics in Arteriovenous Malformation Care. Biomedicines 2024; 12:1289. [PMID: 38927496 PMCID: PMC11201820 DOI: 10.3390/biomedicines12061289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Arteriovenous malformations (AVMs) are vascular malformations that are prone to rupturing and can cause significant morbidity and mortality in relatively young patients. Conventional treatment options such as surgery and endovascular therapy often are insufficient for cure. There is a growing body of knowledge on the genetic and molecular underpinnings of AVM development and maintenance, making the future of precision medicine a real possibility for AVM management. Here, we review the pathophysiology of AVM development across various cell types, with a focus on current and potential druggable targets and their therapeutic potentials in both sporadic and familial AVM populations.
Collapse
Affiliation(s)
- Ann Mansur
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, School of Graduate Studies, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ivan Radovanovic
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, School of Graduate Studies, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada
| |
Collapse
|
13
|
Ma L, Zhu X, Tang C, Pan P, Yadav A, Liang R, Press K, Nelson J, Su H. CNS resident macrophages enhance dysfunctional angiogenesis and circulating monocytes infiltration in brain arteriovenous malformation. J Cereb Blood Flow Metab 2024; 44:925-937. [PMID: 38415628 PMCID: PMC11318399 DOI: 10.1177/0271678x241236008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/08/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024]
Abstract
Myeloid immune cells are abundant in both ruptured and unruptured brain arteriovenous malformations (bAVMs). The role of central nervous system (CNS) resident and circulating monocyte-derived macrophages in bAVM pathogenesis has not been fully understood. We hypothesize that CNS resident macrophages enhance bAVM development and hemorrhage. RNA sequencing using cultured endothelial cells (ECs) and mouse bAVM samples revealed that downregulation of two bAVM causative genes, activin-like kinase 1 (ALK1) or endoglin, increased inflammation and innate immune signaling. To understand the role of CNS resident macrophages in bAVM development and hemorrhage, we administrated a colony-stimulating factor 1 receptor inhibitor to bAVM mice with brain focal Alk1 deletion. Transient depletion of CNS resident macrophages at an early stage of bAVM development mitigated the phenotype severity of bAVM, including a prolonged inhibition of angiogenesis, dysplastic vasculature formation, and infiltration of CNS resident and circulating monocyte-derived macrophages during bAVM development. Transient depletion of CNS resident macrophages increased EC tight junction protein expression, reduced the number of dysplasia vessels and severe hemorrhage in established bAVMs. Thus, EC AVM causative gene mutation can activate CNS resident macrophages promoting bAVM progression. CNS resident macrophage could be a therapeutic target to mitigate the development and severity of bAVMs.
Collapse
Affiliation(s)
- Li Ma
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| | - Xiaonan Zhu
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| | - Chaoliang Tang
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| | - Peipei Pan
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| | - Alka Yadav
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| | - Rich Liang
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| | - Kelly Press
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| | - Jeffrey Nelson
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| | - Hua Su
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| |
Collapse
|
14
|
Ota T. An Updated Review on the Pathogenesis of Brain Arteriovenous Malformations and Its Therapeutic Targets. JOURNAL OF NEUROENDOVASCULAR THERAPY 2024; 19:2024-0008. [PMID: 39958460 PMCID: PMC11826344 DOI: 10.5797/jnet.ra.2024-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/24/2024] [Indexed: 02/18/2025]
Abstract
Brain arteriovenous malformations (bAVMs) are associated with a high risk of intracerebral hemorrhage, which causes severe complications in patients. Although the genetic factors leading to hereditary bAVMs have been extensively investigated, their pathogenesis are still under study. This review examines updated data on the molecular and genetic aspects of bAVMs, the architecture of microvasculature, the roles of angiogenic factors, and signaling pathways. The compiled information may help us understand the pathogenesis of both sporadic and hereditary bAVMs and develop appropriate preemptive treatment approaches.
Collapse
Affiliation(s)
- Takahiro Ota
- Department of Neurosurgery, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| |
Collapse
|
15
|
Koester SW, Hoglund BK, Ciobanu-Caraus O, Hartke JN, Pacult MA, Winkler EA, Catapano JS, Lawton MT. Hematologic and Inflammatory Predictors of Outcome in Patients with Brain Arteriovenous Malformations. World Neurosurg 2024; 185:e342-e350. [PMID: 38340796 DOI: 10.1016/j.wneu.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVE This study investigated the prognostic value of admission blood counts for arteriovenous malformation (AVM) outcomes and compared admission blood counts for patients with ruptured and unruptured AVMs. METHODS A retrospective analysis of patients who underwent surgical treatment for a ruptured cerebral AVM between February 1, 2014, and March 31, 2020, was conducted. The primary outcome was poor neurologic outcome, defined as a modified Rankin Scale score ≥2 in patients with unruptured AVMs or >2 in patients with ruptured AVMs. RESULTS Of 235 included patients, 80 (34%) had ruptured AVMs. At admission, patients with ruptured AVMs had a significantly lower mean (SD) hemoglobin level (12.78 [2.07] g/dL vs. 13.71 [1.60] g/dL, P < 0.001), hematocrit (38.1% [5.9%] vs. 40.7% [4.6%], P < 0.001), lymphocyte count (16% [11%] vs. 26% [10%], P < 0.001), and absolute lymphocyte count (1.41 [0.72] × 103/μL vs. 1.79 [0.68] × 103/μL, P < 0.001), and they had a significantly higher mean (SD) white blood cell count (10.4 [3.8] × 103/μL vs. 7.6 [2.3] × 103/μL, P < 0.001), absolute neutrophil count (7.8 [3.8] × 103/μL vs. 5.0 [2.5] × 103/μL, P < 0.001), and neutrophil count (74% [14%] vs. 64% [13%], P < 0.001). Among patients with unruptured AVMs, white blood cell count ≥6.4 × 103/μL and absolute neutrophil count ≥3.4 × 103/μL were associated with a favorable neurologic outcome, whereas hemoglobin level ≥13.4 g/dL was associated with an unfavorable outcome. Among patients with ruptured AVMs, hypertension was associated with a 3-fold increase in odds of a poor neurologic outcome. CONCLUSIONS Patients with ruptured and unruptured AVMs present with characteristic profiles of hematologic and inflammatory parameters evident in their admission blood work.
Collapse
Affiliation(s)
- Stefan W Koester
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Brandon K Hoglund
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Olga Ciobanu-Caraus
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Joelle N Hartke
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Mark A Pacult
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Ethan A Winkler
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Joshua S Catapano
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Michael T Lawton
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA.
| |
Collapse
|
16
|
Wang S, Deng X, Wu Y, Wu Y, Zhou S, Yang J, Huang Y. Understanding the pathogenesis of brain arteriovenous malformation: genetic variations, epigenetics, signaling pathways, and immune inflammation. Hum Genet 2023; 142:1633-1649. [PMID: 37768356 DOI: 10.1007/s00439-023-02605-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/10/2023] [Indexed: 09/29/2023]
Abstract
Brain arteriovenous malformation (BAVM) is a rare but serious cerebrovascular disease whose pathogenesis has not been fully elucidated. Studies have found that epigenetic regulation, genetic variation and their signaling pathways, immune inflammation, may be the cause of BAVM the main reason. This review comprehensively analyzes the key pathways and inflammatory factors related to BAVMs, and explores their interplay with epigenetic regulation and genetics. Studies have found that epigenetic regulation such as DNA methylation, non-coding RNAs and m6A RNA modification can regulate endothelial cell proliferation, apoptosis, migration and damage repair of vascular malformations through different target gene pathways. Gene defects such as KRAS, ACVRL1 and EPHB4 lead to a disordered vascular environment, which may promote abnormal proliferation of blood vessels through ERK, NOTCH, mTOR, Wnt and other pathways. PDGF-B and PDGFR-β were responsible for the recruitment of vascular adventitial cells and smooth muscle cells in the extracellular matrix environment of blood vessels, and played an important role in the pathological process of BAVM. Recent single-cell sequencing data revealed the diversity of various cell types within BAVM, as well as the heterogeneous expression of vascular-associated antigens, while neutrophils, macrophages and cytokines such as IL-6, IL-1, TNF-α, and IL-17A in BAVM tissue were significantly increased. Currently, there are no specific drugs targeting BAVMs, and biomarkers for BAVM formation, bleeding, and recurrence are lacking clinically. Therefore, further studies on molecular biological mechanisms will help to gain insight into the pathogenesis of BAVM and develop potential therapeutic strategies.
Collapse
Affiliation(s)
- Shiyi Wang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China
| | - Xinpeng Deng
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China
| | - Yuefei Wu
- Department of Neurology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China
| | - Yiwen Wu
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China
| | - Shengjun Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China
| | - Jianhong Yang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China.
| | - Yi Huang
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China.
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, 315010, Zhejiang, China.
| |
Collapse
|
17
|
Tanzadehpanah H, Modaghegh MHS, Mahaki H. Key biomarkers in cerebral arteriovenous malformations: Updated review. J Gene Med 2023; 25:e3559. [PMID: 37380428 DOI: 10.1002/jgm.3559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/02/2023] [Accepted: 06/08/2023] [Indexed: 06/30/2023] Open
Abstract
The formation of vascular networks consisting of arteries, capillaries, and veins is vital in embryogenesis. It is also crucial in adulthood for the formation of a functional vasculature. Cerebral arteriovenous malformations (CAVMs) are linked with a remarkable risk of intracerebral hemorrhage because arterial blood is directly shunted into the veins before the arterial blood pressure is dissipated. The underlying mechanisms responsible for arteriovenous malformation (AVM) growth, progression, and rupture are not fully known, yet the critical role of inflammation in AVM pathogenesis has been noted. The proinflammatory cytokines are upregulated in CAVM, which stimulates overexpression of cell adhesion molecules in endothelial cells (ECs), leading to improved leukocyte recruitment. It is well-known that metalloproteinase-9 secretion by leukocytes disrupts CAVM walls resulting in rupture. Moreover, inflammation alters the angioarchitecture of CAVMs by upregulating angiogenic factors impacting the apoptosis, migration, and proliferation of ECs. A better understanding of the molecular signature of CAVM might allow us to identify biomarkers predicting this complication, acting as a goal for further investigations that may be potentially targeted in gene therapy. The present review is focused on the numerous studies conducted on the molecular signature of CAVM and the associated hemorrhage. The association of numerous molecular signatures with a higher risk of CAVM rupture is shown through inducing proinflammatory mediators, as well as growth factors signaling, Ras-mitogen-activated protein kinase-extracellular signal-regulated kinase, and NOTCH pathways, which are accompanied by cellular level inflammation and endothelial alterations resulting in vascular wall instability. According to the studies, it is assumed that matrix metalloproteinase, interleukin-6, and vascular endothelial growth factor are the biomarkers most associated with CAVM and the rate of hemorrhage, as well as diagnostic methods, with respect to enhancing the patient-specific risk estimation and improving treatment choices.
Collapse
Affiliation(s)
- Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hanie Mahaki
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Ricciardelli AR, Robledo A, Fish JE, Kan PT, Harris TH, Wythe JD. The Role and Therapeutic Implications of Inflammation in the Pathogenesis of Brain Arteriovenous Malformations. Biomedicines 2023; 11:2876. [PMID: 38001877 PMCID: PMC10669898 DOI: 10.3390/biomedicines11112876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/26/2023] Open
Abstract
Brain arteriovenous malformations (bAVMs) are focal vascular lesions composed of abnormal vascular channels without an intervening capillary network. As a result, high-pressure arterial blood shunts directly into the venous outflow system. These high-flow, low-resistance shunts are composed of dilated, tortuous, and fragile vessels, which are prone to rupture. BAVMs are a leading cause of hemorrhagic stroke in children and young adults. Current treatments for bAVMs are limited to surgery, embolization, and radiosurgery, although even these options are not viable for ~20% of AVM patients due to excessive risk. Critically, inflammation has been suggested to contribute to lesion progression. Here we summarize the current literature discussing the role of the immune system in bAVM pathogenesis and lesion progression, as well as the potential for targeting inflammation to prevent bAVM rupture and intracranial hemorrhage. We conclude by proposing that a dysfunctional endothelium, which harbors the somatic mutations that have been shown to give rise to sporadic bAVMs, may drive disease development and progression by altering the immune status of the brain.
Collapse
Affiliation(s)
- Ashley R. Ricciardelli
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ariadna Robledo
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX 77555, USA; (A.R.)
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada;
- Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Peter T. Kan
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX 77555, USA; (A.R.)
| | - Tajie H. Harris
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA 22903, USA;
- Brain, Immunology, and Glia (BIG) Center, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Joshua D. Wythe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA 22903, USA;
- Brain, Immunology, and Glia (BIG) Center, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| |
Collapse
|
19
|
Nakisli S, Lagares A, Nielsen CM, Cuervo H. Pericytes and vascular smooth muscle cells in central nervous system arteriovenous malformations. Front Physiol 2023; 14:1210563. [PMID: 37601628 PMCID: PMC10437819 DOI: 10.3389/fphys.2023.1210563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/29/2023] [Indexed: 08/22/2023] Open
Abstract
Previously considered passive support cells, mural cells-pericytes and vascular smooth muscle cells-have started to garner more attention in disease research, as more subclassifications, based on morphology, gene expression, and function, have been discovered. Central nervous system (CNS) arteriovenous malformations (AVMs) represent a neurovascular disorder in which mural cells have been shown to be affected, both in animal models and in human patients. To study consequences to mural cells in the context of AVMs, various animal models have been developed to mimic and predict human AVM pathologies. A key takeaway from recently published work is that AVMs and mural cells are heterogeneous in their molecular, cellular, and functional characteristics. In this review, we summarize the observed perturbations to mural cells in human CNS AVM samples and CNS AVM animal models, and we discuss various potential mechanisms relating mural cell pathologies to AVMs.
Collapse
Affiliation(s)
- Sera Nakisli
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Neuroscience Program, Ohio University, Athens, OH, United States
| | - Alfonso Lagares
- Department of Neurosurgery, University Hospital 12 de Octubre, Madrid, Spain
- Department of Surgery, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Imas12, Madrid, Spain
| | - Corinne M. Nielsen
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Neuroscience Program, Ohio University, Athens, OH, United States
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, United States
| | - Henar Cuervo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (F.S.P), Madrid, Spain
| |
Collapse
|
20
|
Ma L, Zhu X, Tang C, Pan P, Yadav A, Liang R, Press K, Su H. CNS resident macrophages enhance dysfunctional angiogenesis and circulating monocytes infiltration in brain arteriovenous malformation. RESEARCH SQUARE 2023:rs.3.rs-2899768. [PMID: 37214790 PMCID: PMC10197785 DOI: 10.21203/rs.3.rs-2899768/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Myeloid immune cells present abundantly in both ruptured and unruptured brain arteriovenous malformations (bAVMs). The role of central nervous system (CNS) resident and circulating monocyte-derived macrophages in bAVM pathogenesis has not been fully understood. RNA sequencing using cultured cells and bAVM samples revealed that downregulation of activin-like kinase 1 (ALK1) or endoglin (two bAVM causative genes) increased pro-angiogenic, endothelial inflammation and innate immune signaling, which provided endogenous underpinnings of the active inflammation in bAVM. To further understand the role of CNS resident macrophages in bAVM development and hemorrhage, we administrated a colony-stimulating factor 1 receptor (CSF1R) inhibitor to bAVM mice with endothelial Alk1 deletion. Transient depletion of CNS resident macrophages at early stage of bAVM development remarkably mitigated the subsequent phenotype severity of bAVM. This therapeutic effect exhibited a prolonged inhibition of angiogenesis, dysplastic vasculature formation, and infiltration of CNS resident and circulating monocyte-derived macrophages during bAVM development. Transient depletion of CNS resident macrophages also reduced the dysplasia vessels and improved the integrity of endothelial tight junctions in established bAVMs. Administration of CSF1R inhibitor also prevented severe hemorrhage of bAVMs. Thus, endothelial AVM causative gene mutation can activate CNS resident macrophages promoting bAVM progression. CNS resident macrophages could be specific targets to mitigate the development and severity of bAVMs.
Collapse
Affiliation(s)
- Li Ma
- University of California, San Francisco
| | | | | | | | | | | | | | - Hua Su
- University of California, San Francisco
| |
Collapse
|
21
|
Wiseman JA, Dragunow M, I-H Park T. Cell Type-Specific Nuclei Markers: The Need for Human Brain Research to Go Nuclear. Neuroscientist 2023; 29:41-61. [PMID: 34459315 DOI: 10.1177/10738584211037351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Identifying and interrogating cell type-specific populations within the heterogeneous milieu of the human brain is paramount to resolving the processes of normal brain homeostasis and the pathogenesis of neurological disorders. While brain cell type-specific markers are well established, most are localized on cellular membranes or within the cytoplasm, with limited literature describing those found in the nucleus. Due to the complex cytoarchitecture of the human brain, immunohistochemical studies require well-defined cell-specific nuclear markers for more precise and efficient quantification of the cellular populations. Furthermore, efficient nuclear markers are required for cell type-specific purification and transcriptomic interrogation of archived human brain tissue through nuclei isolation-based RNA sequencing. To sate the growing demand for robust cell type-specific nuclear markers, we thought it prudent to comprehensively review the current literature to identify and consolidate a novel series of robust cell type-specific nuclear markers that can assist researchers across a range of neuroscientific disciplines. The following review article collates and discusses several key and prospective cell type-specific nuclei markers for each of the major human brain cell types; it then concludes by discussing the potential applications of cell type-specific nuclear workflows and the power of nuclear-based neuroscientific research.
Collapse
Affiliation(s)
- James A Wiseman
- Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Hugh Green Biobank, The Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Thomas I-H Park
- Department of Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Quintin S, Figg JW, Mehkri Y, Hanna CO, Woolridge MG, Lucke-Wold B. Arteriovenous Malformations: An Update on Models and Therapeutic Targets. JOURNAL OF NEUROSCIENCE AND NEUROLOGICAL SURGERY 2023; 13:250. [PMID: 36846724 PMCID: PMC9956274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Arteriovenous malformations (AVMs) are an anomaly of the vascular system where feeding arteries are directly connected to the venous drainage network. While AVMs can arise anywhere in the body and have been described in most tissues, brain AVMs are of significant concern because of the risk of hemorrhage which carries significant morbidity and mortality. The prevalence of AVM's and the mechanisms underlying their formation are not well understood. For this reason, patients who undergo treatment for symptomatic AVM's remain at increased risk of subsequent bleeds and adverse outcomes. The cerebrovascular network is delicate and novel animal models continue to provide insight into its dynamics in the context of AVM's. As the molecular players in the formation of familial and sporadic AVM's are better understood, novel therapeutic approaches have been developed to mitigate their associated risks. Here we discuss the current literature surrounding AVM's including the development of models and therapeutic targets which are currently being investigated.
Collapse
Affiliation(s)
- Stephan Quintin
- College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - John W Figg
- Department of Neurosurgery, University of Florida, Gainesville, Florida 32610, USA
| | - Yusuf Mehkri
- College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Chadwin O Hanna
- College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | | | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, Florida 32610, USA
| |
Collapse
|
23
|
State of the Art in the Role of Endovascular Embolization in the Management of Brain Arteriovenous Malformations-A Systematic Review. J Clin Med 2022; 11:jcm11237208. [PMID: 36498782 PMCID: PMC9739246 DOI: 10.3390/jcm11237208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
As a significant cause of intracerebral hemorrhages, seizures, and neurological decline, brain arteriovenous malformations (bAVMs) are a rare group of complex vascular lesions with devastating implications for patients' quality of life. Although the concerted effort of the scientific community has improved our understanding of bAVM biology, the exact mechanism continues to be elucidated. Furthermore, to this day, due to the high heterogeneity of bAVMs as well as the lack of objective data brought by the lack of evaluative and comparative studies, there is no clear consensus on the treatment of this life-threatening and dynamic disease. As a consequence, patients often fall short of obtaining the optimal treatment. Endovascular embolization is an inherent part of multidisciplinary bAVM management that can be used in various clinical scenarios, each with different objectives. Well-trained neuro-interventional centers are proficient at curing bAVMs that are smaller than 3 cm; are located superficially in noneloquent areas; and have fewer, larger, and less tortuous feeding arteries. The transvenous approach is an emerging effective and safe technique that potentially offers a chance to cure previously untreatable bAVMs. This review provides the state of the art in all aspects of endovascular embolization in the management of bAVMs.
Collapse
|
24
|
Drapé E, Anquetil T, Larrivée B, Dubrac A. Brain arteriovenous malformation in hereditary hemorrhagic telangiectasia: Recent advances in cellular and molecular mechanisms. Front Hum Neurosci 2022; 16:1006115. [PMID: 36504622 PMCID: PMC9729275 DOI: 10.3389/fnhum.2022.1006115] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/27/2022] [Indexed: 11/25/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a genetic disorder characterized by vessel dilatation, such as telangiectasia in skin and mucosa and arteriovenous malformations (AVM) in internal organs such as the gastrointestinal tract, lungs, and brain. AVMs are fragile and tortuous vascular anomalies that directly connect arteries and veins, bypassing healthy capillaries. Mutations in transforming growth factor β (TGFβ) signaling pathway components, such as ENG (ENDOGLIN), ACVRL1 (ALK1), and SMAD4 (SMAD4) genes, account for most of HHT cases. 10-20% of HHT patients develop brain AVMs (bAVMs), which can lead to vessel wall rupture and intracranial hemorrhages. Though the main mutations are known, mechanisms leading to AVM formation are unclear, partially due to lack of animal models. Recent mouse models allowed significant advances in our understanding of AVMs. Endothelial-specific deletion of either Acvrl1, Eng or Smad4 is sufficient to induce AVMs, identifying endothelial cells (ECs) as primary targets of BMP signaling to promote vascular integrity. Loss of ALK1/ENG/SMAD4 signaling is associated with NOTCH signaling defects and abnormal arteriovenous EC differentiation. Moreover, cumulative evidence suggests that AVMs originate from venous ECs with defective flow-migration coupling and excessive proliferation. Mutant ECs show an increase of PI3K/AKT signaling and inhibitors of this signaling pathway rescue AVMs in HHT mouse models, revealing new therapeutic avenues. In this review, we will summarize recent advances and current knowledge of mechanisms controlling the pathogenesis of bAVMs, and discuss unresolved questions.
Collapse
Affiliation(s)
- Elise Drapé
- Centre de Recherche, CHU St. Justine, Montréal, QC, Canada,Département de Pharmacologie et de Physiologie, Université de Montréal, Montréal, QC, Canada
| | - Typhaine Anquetil
- Centre de Recherche, CHU St. Justine, Montréal, QC, Canada,Département De Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC, Canada
| | - Bruno Larrivée
- Département d’Ophtalmologie, Université de Montréal, Montréal, QC, Canada,Centre De Recherche, Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada,*Correspondence: Bruno Larrivée,
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montréal, QC, Canada,Département De Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC, Canada,Département d’Ophtalmologie, Université de Montréal, Montréal, QC, Canada,Alexandre Dubrac,
| |
Collapse
|
25
|
Eisa-Beygi S, Burrows PE, Link BA. Endothelial cilia dysfunction in pathogenesis of hereditary hemorrhagic telangiectasia. Front Cell Dev Biol 2022; 10:1037453. [PMID: 36438574 PMCID: PMC9686338 DOI: 10.3389/fcell.2022.1037453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/21/2022] [Indexed: 09/09/2023] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is associated with defective capillary network, leading to dilated superficial vessels and arteriovenous malformations (AVMs) in which arteries connect directly to the veins. Loss or haploinsufficiency of components of TGF-β signaling, ALK1, ENG, SMAD4, and BMP9, have been implicated in the pathogenesis AVMs. Emerging evidence suggests that the inability of endothelial cells to detect, transduce and respond to blood flow, during early development, is an underpinning of AVM pathogenesis. Therefore, components of endothelial flow detection may be instrumental in potentiating TGF-β signaling in perfused blood vessels. Here, we argue that endothelial cilium, a microtubule-based and flow-sensitive organelle, serves as a signaling hub by coupling early flow detection with potentiation of the canonical TGF-β signaling in nascent endothelial cells. Emerging evidence from animal models suggest a role for primary cilia in mediating vascular development. We reason, on recent observations, that endothelial cilia are crucial for vascular development and that embryonic loss of endothelial cilia will curtail TGF-β signaling, leading to associated defects in arteriovenous development and impaired vascular stability. Loss or dysfunction of endothelial primary cilia may be implicated in the genesis of AVMs due, in part, to inhibition of ALK1/SMAD4 signaling. We speculate that AVMs constitute part of the increasing spectrum of ciliopathy-associated vascular defects.
Collapse
Affiliation(s)
- Shahram Eisa-Beygi
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Patricia E. Burrows
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Brian A. Link
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
26
|
Van Trigt WK, Kelly KM, Hughes CCW. GNAQ mutations drive port wine birthmark-associated Sturge-Weber syndrome: A review of pathobiology, therapies, and current models. Front Hum Neurosci 2022; 16:1006027. [PMID: 36405075 PMCID: PMC9670321 DOI: 10.3389/fnhum.2022.1006027] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Port-wine birthmarks (PWBs) are caused by somatic, mosaic mutations in the G protein guanine nucleotide binding protein alpha subunit q (GNAQ) and are characterized by the formation of dilated, dysfunctional blood vessels in the dermis, eyes, and/or brain. Cutaneous PWBs can be treated by current dermatologic therapy, like laser intervention, to lighten the lesions and diminish nodules that occur in the lesion. Involvement of the eyes and/or brain can result in serious complications and this variation is termed Sturge-Weber syndrome (SWS). Some of the biggest hurdles preventing development of new therapeutics are unanswered questions regarding disease biology and lack of models for drug screening. In this review, we discuss the current understanding of GNAQ signaling, the standard of care for patients, overlap with other GNAQ-associated or phenotypically similar diseases, as well as deficiencies in current in vivo and in vitro vascular malformation models.
Collapse
Affiliation(s)
- William K. Van Trigt
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Kristen M. Kelly
- Department of Dermatology, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Christopher C. W. Hughes
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
27
|
Vetiska S, Wälchli T, Radovanovic I, Berhouma M. Molecular and genetic mechanisms in brain arteriovenous malformations: new insights and future perspectives. Neurosurg Rev 2022; 45:3573-3593. [PMID: 36219361 DOI: 10.1007/s10143-022-01883-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/30/2022] [Accepted: 10/05/2022] [Indexed: 10/17/2022]
Abstract
Brain arteriovenous malformations (bAVMs) are rare vascular lesions made of shunts between cerebral arteries and veins without the interposition of a capillary bed. The majority of bAVMs are asymptomatic, but some may be revealed by seizures and potentially life-threatening brain hemorrhage. The management of unruptured bAVMs remains a matter of debate. Significant progress in the understanding of their pathogenesis has been made during the last decade, particularly using genome sequencing and biomolecular analysis. Herein, we comprehensively review the recent molecular and genetic advances in the study of bAVMs that not only allow a better understanding of the genesis and growth of bAVMs, but also open new insights in medical treatment perspectives.
Collapse
Affiliation(s)
- Sandra Vetiska
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada
| | - Thomas Wälchli
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada.,Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, and Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Ivan Radovanovic
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada.,Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Moncef Berhouma
- Department of Neurosurgery, University Hospital of Dijon Bourgogne, Dijon, France. .,CREATIS Lab, CNRS UMR 5220, INSERM U1294, Lyon 1, University, Lyon, France.
| |
Collapse
|
28
|
Arthur HM, Roman BL. An update on preclinical models of hereditary haemorrhagic telangiectasia: Insights into disease mechanisms. Front Med (Lausanne) 2022; 9:973964. [PMID: 36250069 PMCID: PMC9556665 DOI: 10.3389/fmed.2022.973964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Endoglin (ENG) is expressed on the surface of endothelial cells (ECs) where it efficiently binds circulating BMP9 and BMP10 ligands to initiate activin A receptor like type 1 (ALK1) protein signalling to protect the vascular architecture. Patients heterozygous for ENG or ALK1 mutations develop the vascular disorder known as hereditary haemorrhagic telangiectasia (HHT). Many patients with this disorder suffer from anaemia, and are also at increased risk of stroke and high output heart failure. Recent work using animal models of HHT has revealed new insights into cellular and molecular mechanisms causing this disease. Loss of the ENG (HHT1) or ALK1 (HHT2) gene in ECs leads to aberrant arteriovenous connections or malformations (AVMs) in developing blood vessels. Similar phenotypes develop following combined EC specific loss of SMAD1 and 5, or EC loss of SMAD4. Taken together these data point to the essential role of the BMP9/10-ENG-ALK1-SMAD1/5-SMAD4 pathway in protecting the vasculature from AVMs. Altered directional migration of ECs in response to shear stress and increased EC proliferation are now recognised as critical factors driving AVM formation. Disruption of the ENG/ALK1 signalling pathway also affects EC responses to vascular endothelial growth factor (VEGF) and crosstalk between ECs and vascular smooth muscle cells. It is striking that the vascular lesions in HHT are both localised and tissue specific. Increasing evidence points to the importance of a second genetic hit to generate biallelic mutations, and the sporadic nature of such somatic mutations would explain the localised formation of vascular lesions. In addition, different pro-angiogenic drivers of AVM formation are likely to be at play during the patient’s life course. For example, inflammation is a key driver of vessel remodelling in postnatal life, and may turn out to be an important driver of HHT disease. The current wealth of preclinical models of HHT has led to increased understanding of AVM development and revealed new therapeutic approaches to treat AVMs, and form the topic of this review.
Collapse
Affiliation(s)
- Helen M. Arthur
- Biosciences Institute, Centre for Life, University of Newcastle, Newcastle, United Kingdom
- *Correspondence: Helen M. Arthur,
| | - Beth L. Roman
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
29
|
Shabani Z, Schuerger J, Su H. Cellular loci involved in the development of brain arteriovenous malformations. Front Hum Neurosci 2022; 16:968369. [PMID: 36211120 PMCID: PMC9532630 DOI: 10.3389/fnhum.2022.968369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Brain arteriovenous malformations (bAVMs) are abnormal vessels that are prone to rupture, causing life-threatening intracranial bleeding. The mechanism of bAVM formation is poorly understood. Nevertheless, animal studies revealed that gene mutation in endothelial cells (ECs) and angiogenic stimulation are necessary for bAVM initiation. Evidence collected through analyzing bAVM specimens of human and mouse models indicate that cells other than ECs also are involved in bAVM pathogenesis. Both human and mouse bAVMs vessels showed lower mural cell-coverage, suggesting a role of pericytes and vascular smooth muscle cells (vSMCs) in bAVM pathogenesis. Perivascular astrocytes also are important in maintaining cerebral vascular function and take part in bAVM development. Furthermore, higher inflammatory cytokines in bAVM tissue and blood demonstrate the contribution of inflammatory cells in bAVM progression, and rupture. The goal of this paper is to provide our current understanding of the roles of different cellular loci in bAVM pathogenesis.
Collapse
Affiliation(s)
- Zahra Shabani
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Joana Schuerger
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Hua Su
- Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Hua Su, ; orcid.org/0000-0003-1566-9877
| |
Collapse
|
30
|
Sato Y, Falcone-Juengert J, Tominaga T, Su H, Liu J. Remodeling of the Neurovascular Unit Following Cerebral Ischemia and Hemorrhage. Cells 2022; 11:2823. [PMID: 36139398 PMCID: PMC9496956 DOI: 10.3390/cells11182823] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/18/2022] [Accepted: 08/30/2022] [Indexed: 11/23/2022] Open
Abstract
Formulated as a group effort of the stroke community, the transforming concept of the neurovascular unit (NVU) depicts the structural and functional relationship between brain cells and the vascular structure. Composed of both neural and vascular elements, the NVU forms the blood-brain barrier that regulates cerebral blood flow to meet the oxygen demand of the brain in normal physiology and maintain brain homeostasis. Conversely, the dysregulation and dysfunction of the NVU is an essential pathological feature that underlies neurological disorders spanning from chronic neurodegeneration to acute cerebrovascular events such as ischemic stroke and cerebral hemorrhage, which were the focus of this review. We also discussed how common vascular risk factors of stroke predispose the NVU to pathological changes. We synthesized existing literature and first provided an overview of the basic structure and function of NVU, followed by knowledge of how these components remodel in response to ischemic stroke and brain hemorrhage. A greater understanding of the NVU dysfunction and remodeling will enable the design of targeted therapies and provide a valuable foundation for relevant research in this area.
Collapse
Affiliation(s)
- Yoshimichi Sato
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Jaime Falcone-Juengert
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
| | - Teiji Tominaga
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Hua Su
- Department of Anesthesia, UCSF, San Francisco, CA 94143, USA
- Center for Cerebrovascular Research, UCSF, San Francisco, CA 94143, USA
| | - Jialing Liu
- Department of Neurological Surgery, UCSF, San Francisco, CA 94158, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA 94158, USA
| |
Collapse
|
31
|
Selhorst S, Nakisli S, Kandalai S, Adhicary S, Nielsen CM. Pathological pericyte expansion and impaired endothelial cell-pericyte communication in endothelial Rbpj deficient brain arteriovenous malformation. Front Hum Neurosci 2022; 16:974033. [PMID: 36147294 PMCID: PMC9485665 DOI: 10.3389/fnhum.2022.974033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/15/2022] [Indexed: 11/27/2022] Open
Abstract
Pericytes, like vascular smooth muscle cells, are perivascular cells closely associated with blood vessels throughout the body. Pericytes are necessary for vascular development and homeostasis, with particularly critical roles in the brain, where they are involved in regulating cerebral blood flow and establishing the blood-brain barrier. A role for pericytes during neurovascular disease pathogenesis is less clear—while some studies associate decreased pericyte coverage with select neurovascular diseases, others suggest increased pericyte infiltration in response to hypoxia or traumatic brain injury. Here, we used an endothelial loss-of-function Recombination signal binding protein for immunoglobulin kappa J region (Rbpj)/Notch mediated mouse model of brain arteriovenous malformation (AVM) to investigate effects on pericytes during neurovascular disease pathogenesis. We tested the hypothesis that pericyte expansion, via morphological changes, and Platelet-derived growth factor B/Platelet-derived growth factor receptor β (Pdgf-B/Pdgfrβ)-dependent endothelial cell-pericyte communication are affected, during the pathogenesis of Rbpj mediated brain AVM in mice. Our data show that pericyte coverage of vascular endothelium expanded pathologically, to maintain coverage of vascular abnormalities in brain and retina, following endothelial deletion of Rbpj. In Rbpj-mutant brain, pericyte expansion was likely attributed to cytoplasmic process extension and not to increased pericyte proliferation. Despite expanding overall area of vessel coverage, pericytes from Rbpj-mutant brains showed decreased expression of Pdgfrβ, Neural (N)-cadherin, and cluster of differentiation (CD)146, as compared to controls, which likely affected Pdgf-B/Pdgfrβ-dependent communication and appositional associations between endothelial cells and pericytes in Rbpj-mutant brain microvessels. By contrast, and perhaps by compensatory mechanism, endothelial cells showed increased expression of N-cadherin. Our data identify cellular and molecular effects on brain pericytes, following endothelial deletion of Rbpj, and suggest pericytes as potential therapeutic targets for Rbpj/Notch related brain AVM.
Collapse
Affiliation(s)
- Samantha Selhorst
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Honors Tutorial College, Ohio University, Athens, OH, United States
| | - Sera Nakisli
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Neuroscience Program, Ohio University, Athens, OH, United States
| | - Shruthi Kandalai
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Honors Tutorial College, Ohio University, Athens, OH, United States
| | - Subhodip Adhicary
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Translational Biomedical Sciences Program, Ohio University, Athens, OH, United States
| | - Corinne M. Nielsen
- Department of Biological Sciences, Ohio University, Athens, OH, United States
- Neuroscience Program, Ohio University, Athens, OH, United States
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, United States
- *Correspondence: Corinne M. Nielsen,
| |
Collapse
|
32
|
Osorno-Cruz C, Hasanpour Z, Peart R, Dodd W, Laurent D, Aghili-Mehrizi S, Lucke-Wold B, Chalouhi N. Venous Outflow for Brain Arteriovenous Malformations: Overview and Treatment Implications. INTERNATIONAL JOURNAL OF NEUROBIOLOGY 2022; 4:151. [PMID: 36081858 PMCID: PMC9450881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Introduction Recent evidence has demonstrated a close relationship between the cerebral venous and lymphatic systems. Venous congestion has been implicated in a host of neurologic disorders, with relevance for vascular etiologies. Objective The authors aim to review the literature as it pertains to brain arteriovenous malformations' (BAVMs) venous hemodynamics and glymphatic pathways, as well as the implications of BAVM treatment. Results BAVMs offer a unique challenge, with sudden alteration in flow dynamics leading to increased hemorrhage risk and difficult challenges post-treatment. Conclusion Recent progress in the understanding of CNS fluid dynamics and glymphatic pathways have revealed important implications for BAVM pathology and treatment. As imaging techniques and treatment modalities advance, there is a need to further investigate this relationship as it relates to therapeutic options and post-treatment sequalae.
Collapse
Affiliation(s)
- C Osorno-Cruz
- Department of Neurosurgery, University of Iowa, Iowa City
| | - Z Hasanpour
- Department of Neurosurgery, University of Florida, Gainesville
| | - R Peart
- Department of Neurosurgery, University of Florida, Gainesville
| | - W Dodd
- Department of Neurosurgery, University of Florida, Gainesville
| | - D Laurent
- Department of Neurosurgery, University of Florida, Gainesville
| | | | - B Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville
| | - N Chalouhi
- Department of Neurosurgery, University of Florida, Gainesville
| |
Collapse
|
33
|
Han C, Lang MJ, Nguyen CL, Luna Melendez E, Mehta S, Turner GH, Lawton MT, Oh SP. Novel experimental model of brain arteriovenous malformations using conditional Alk1 gene deletion in transgenic mice. J Neurosurg 2022; 137:163-174. [PMID: 34740197 DOI: 10.3171/2021.6.jns21717] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/16/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Hereditary hemorrhagic telangiectasia is the only condition associated with multiple inherited brain arteriovenous malformations (AVMs). Therefore, a mouse model was developed with a genetics-based approach that conditionally deleted the causative activin receptor-like kinase 1 (Acvrl1 or Alk1) gene. Radiographic and histopathological findings were correlated, and AVM stability and hemorrhagic behavior over time were examined. METHODS Alk1-floxed mice were crossed with deleter mice to generate offspring in which both copies of the Alk1 gene were deleted by Tagln-Cre to form brain AVMs in the mice. AVMs were characterized using MRI, MRA, and DSA. Brain AVMs were characterized histopathologically with latex dye perfusion, immunofluorescence, and Prussian blue staining. RESULTS Brains of 55 Tagln-Cre+;Alk12f/2f mutant mice were categorized into three groups: no detectable vascular lesions (group 1; 23 of 55, 42%), arteriovenous fistulas (AVFs) with no nidus (group 2; 10 of 55, 18%), and nidal AVMs (group 3; 22 of 55, 40%). Microhemorrhage was observed on MRI or MRA in 11 AVMs (50%). AVMs had the angiographic hallmarks of early nidus opacification, a tangle of arteries and dilated draining veins, and rapid shunting of blood flow. Latex dye perfusion confirmed arteriovenous shunting in all AVMs and AVFs. Microhemorrhages were detected adjacent to AVFs and AVMs, visualized by iron deposition, Prussian blue staining, and macrophage infiltration using CD68 immunostaining. Brain AVMs were stable on serial MRI and MRA in group 3 mice (mean age at initial imaging 2.9 months; mean age at last imaging 9.5 months). CONCLUSIONS Approximately 40% of transgenic mice satisfied the requirements of a stable experimental AVM model by replicating nidal anatomy, arteriovenous hemodynamics, and microhemorrhagic behavior. Transgenic mice with AVFs had a recognizable phenotype of hereditary hemorrhagic telangiectasia but were less suitable for experimental modeling. AVM pathogenesis can be understood as the combination of conditional Alk1 gene deletion during embryogenesis and angiogenesis that is hyperactive in developing and newborn mice, which translates to a congenital origin in most patients but an acquired condition in patients with a confluence of genetic and angiogenic events later in life. This study offers a novel experimental brain AVM model for future studies of AVM pathophysiology, growth, rupture, and therapeutic regression.
Collapse
Affiliation(s)
- Chul Han
- 1Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix
| | | | - Candice L Nguyen
- 1Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix
| | - Ernesto Luna Melendez
- 3Ivy Brain Tumor Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Shwetal Mehta
- 3Ivy Brain Tumor Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Gregory H Turner
- 4Neuroimaging, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix; and
| | - Michael T Lawton
- 1Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix
- Departments of2Neurosurgery and
| | - S Paul Oh
- 1Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix
| |
Collapse
|
34
|
Winkler EA, Pacult MA, Catapano JS, Scherschinski L, Srinivasan VM, Graffeo CS, Oh SP, Lawton MT. Emerging pathogenic mechanisms in human brain arteriovenous malformations: a contemporary review in the multiomics era. Neurosurg Focus 2022; 53:E2. [DOI: 10.3171/2022.4.focus2291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/18/2022] [Indexed: 11/06/2022]
Abstract
A variety of pathogenic mechanisms have been described in the formation, maturation, and rupture of brain arteriovenous malformations (bAVMs). While the understanding of bAVMs has largely been formulated based on animal models of rare hereditary diseases in which AVMs form, a new era of “omics” has permitted large-scale examinations of contributory genetic variations in human sporadic bAVMs. New findings regarding the pathogenesis of bAVMs implicate changes to endothelial and mural cells that result in increased angiogenesis, proinflammatory recruitment, and breakdown of vascular barrier properties that may result in hemorrhage; a greater diversity of cell populations that compose the bAVM microenvironment may also be implicated and complicate traditional models. Genomic sequencing of human bAVMs has uncovered inherited, de novo, and somatic activating mutations, such as KRAS, which contribute to the pathogenesis of bAVMs. New droplet-based, single-cell sequencing technologies have generated atlases of cell-specific molecular derangements. Herein, the authors review emerging genomic and transcriptomic findings underlying pathologic cell transformations in bAVMs derived from human tissues. The application of multiple sequencing modalities to bAVM tissues is a natural next step for researchers, although the potential therapeutic benefits or clinical applications remain unknown.
Collapse
Affiliation(s)
- Ethan A. Winkler
- Department of Neurological Surgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix; and
| | - Mark A. Pacult
- Department of Neurological Surgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix; and
| | - Joshua S. Catapano
- Department of Neurological Surgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix; and
| | - Lea Scherschinski
- Department of Neurological Surgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix; and
| | - Visish M. Srinivasan
- Department of Neurological Surgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix; and
| | - Christopher S. Graffeo
- Department of Neurological Surgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix; and
| | - S. Paul Oh
- Department of Neurological Surgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix; and
- Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona
| | - Michael T. Lawton
- Department of Neurological Surgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix; and
| |
Collapse
|
35
|
Kilari S, Wang Y, Singh A, Graham RP, Iyer V, Thompson SM, Torbenson MS, Mukhopadhyay D, Misra S. Neuropilin-1 deficiency in vascular smooth muscle cells is associated with hereditary hemorrhagic telangiectasia arteriovenous malformations. JCI Insight 2022; 7:155565. [PMID: 35380991 PMCID: PMC9090252 DOI: 10.1172/jci.insight.155565] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/30/2022] [Indexed: 11/25/2022] Open
Abstract
Patients with hereditary hemorrhagic telangiectasia (HHT) have arteriovenous malformations (AVMs) with genetic mutations involving the activin-A receptor like type 1 (ACVRL1 or ALK1) and endoglin (ENG). Recent studies have shown that Neuropilin-1 (NRP-1) inhibits ALK1. We investigated the expression of NRP-1 in livers of patients with HHT and found that there was a significant reduction in NRP-1 in perivascular smooth muscle cells (SMCs). We used Nrp1SM22KO mice (Nrp1 was ablated in SMCs) and found hemorrhage, increased immune cell infiltration with a decrease in SMCs, and pericyte lining in lungs and liver in adult mice. Histologic examination revealed lung arteriovenous fistulas (AVFs) with enlarged liver vessels. Evaluation of the retina vessels at P5 from Nrp1SM22KO mice demonstrated dilated capillaries with a reduction of pericytes. In inflow artery of surgical AVFs from the Nrp1SM22KO versus WT mice, there was a significant decrease in Tgfb1, Eng, and Alk1 expression and phosphorylated SMAD1/5/8 (pSMAD1/5/8), with an increase in apoptosis. TGF-β1–stimulated aortic SMCs from Nrp1SM22KO versus WT mice have decreased pSMAD1/5/8 and increased apoptosis. Coimmunoprecipitation experiments revealed that NRP-1 interacts with ALK1 and ENG in SMCs. In summary, NRP-1 deletion in SMCs leads to reduced ALK1, ENG, and pSMAD1/5/8 signaling and reduced cell death associated with AVM formation.
Collapse
Affiliation(s)
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States of America
| | - Avishek Singh
- Department of Radiology, Mayo Clinic, Rochester, United States of America
| | - Rondell P Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, United States of America
| | - Vivek Iyer
- Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, United States of America
| | - Scott M Thompson
- Department of Radiology, Mayo Clinic, Rochester, United States of America
| | - Michael S Torbenson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, United States of America
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States of America
| | - Sanjay Misra
- Department of Radiology, Mayo Clinic, Rochester, United States of America
| |
Collapse
|
36
|
Winkler EA, Kim CN, Ross JM, Garcia JH, Gil E, Oh I, Chen LQ, Wu D, Catapano JS, Raygor K, Narsinh K, Kim H, Weinsheimer S, Cooke DL, Walcott BP, Lawton MT, Gupta N, Zlokovic BV, Chang EF, Abla AA, Lim DA, Nowakowski TJ. A single-cell atlas of the normal and malformed human brain vasculature. Science 2022; 375:eabi7377. [PMID: 35084939 PMCID: PMC8995178 DOI: 10.1126/science.abi7377] [Citation(s) in RCA: 188] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cerebrovascular diseases are a leading cause of death and neurologic disability. Further understanding of disease mechanisms and therapeutic strategies requires a deeper knowledge of cerebrovascular cells in humans. We profiled transcriptomes of 181,388 cells to define a cell atlas of the adult human cerebrovasculature, including endothelial cell molecular signatures with arteriovenous segmentation and expanded perivascular cell diversity. By leveraging this reference, we investigated cellular and molecular perturbations in brain arteriovenous malformations, which are a leading cause of stroke in young people, and identified pathologic endothelial transformations with abnormal vascular patterning and the ontology of vascularly derived inflammation. We illustrate the interplay between vascular and immune cells that contributes to brain hemorrhage and catalog opportunities for targeting angiogenic and inflammatory programs in vascular malformations.
Collapse
Affiliation(s)
- Ethan A Winkler
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Chang N Kim
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Jayden M Ross
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Joseph H Garcia
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Eugene Gil
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Irene Oh
- Rebus Biosystems, Santa Clara, CA, USA
| | | | - David Wu
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Joshua S Catapano
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kunal Raygor
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Kazim Narsinh
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Helen Kim
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Shantel Weinsheimer
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Daniel L Cooke
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Brian P Walcott
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, IL, USA
| | - Michael T Lawton
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Nalin Gupta
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Edward F Chang
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Adib A Abla
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
37
|
Sheth KN, Anderson CD, Biffi A, Dlamini N, Falcone GJ, Fox CK, Fullerton HJ, Greenberg SM, Hemphill JC, Kim A, Kim H, Ko NU, Roland JL, Sansing LH, van Veluw SJ, Rosand J. Maximizing Brain Health After Hemorrhagic Stroke: Bugher Foundation Centers of Excellence. Stroke 2022; 53:1020-1029. [PMID: 35109678 PMCID: PMC8885885 DOI: 10.1161/strokeaha.121.036197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Kevin N. Sheth
- Department of Neurology, Yale School of Medicine, New Haven, CT
| | - Christopher D. Anderson
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA,Broad Institute, Cambridge, MA,Division of Stroke and Cerebrovascular Diseases, Brigham and Women’s Hospital, Boston, MA
| | - Alessandro Biffi
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA,Division of Neuropsychiatry, Massachusetts General Hospital, Boston, MA,Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Nomazulu Dlamini
- Division of Neurology, The Hospital for Sick Children, Toronto, Canada
| | | | - Christine K. Fox
- Department of Neurology, University of California at San Francisco, San Francisco, CA
| | - Heather J. Fullerton
- Department of Neurology, University of California at San Francisco, San Francisco, CA
| | | | - J. Claude Hemphill
- Department of Neurology, University of California at San Francisco, San Francisco, CA
| | - Anthony Kim
- Department of Neurology, University of California at San Francisco, San Francisco, CA
| | - Helen Kim
- Department of Anesthesia, University of California at San Francisco, San Francisco, CA
| | - Nerissa U. Ko
- Department of Neurology, University of California at San Francisco, San Francisco, CA
| | - Jarod L Roland
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA
| | | | | | - Jonathan Rosand
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA,Broad Institute, Cambridge, MA,Department of Neurology, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
38
|
Genetics and Vascular Biology of Brain Vascular Malformations. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00012-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
39
|
Pan P, Weinsheimer S, Cooke D, Winkler E, Abla A, Kim H, Su H. Review of treatment and therapeutic targets in brain arteriovenous malformation. J Cereb Blood Flow Metab 2021; 41:3141-3156. [PMID: 34162280 PMCID: PMC8669284 DOI: 10.1177/0271678x211026771] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/23/2022]
Abstract
Brain arteriovenous malformations (bAVM) are an important cause of intracranial hemorrhage (ICH), especially in younger patients. The pathogenesis of bAVM are largely unknown. Current understanding of bAVM etiology is based on studying genetic syndromes, animal models, and surgically resected specimens from patients. The identification of activating somatic mutations in the Kirsten rat sarcoma viral oncogene homologue (KRAS) gene and other mitogen-activated protein kinase (MAPK) pathway genes has opened up new avenues for bAVM study, leading to a paradigm shift to search for somatic, de novo mutations in sporadic bAVMs instead of focusing on inherited genetic mutations. Through the development of new models and understanding of pathways involved in maintaining normal vascular structure and functions, promising therapeutic targets have been identified and safety and efficacy studies are underway in animal models and in patients. The goal of this paper is to provide a thorough review or current diagnostic and treatment tools, known genes and key pathways involved in bAVM pathogenesis to summarize current treatment options and potential therapeutic targets uncovered by recent discoveries.
Collapse
Affiliation(s)
- Peipei Pan
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, USA
| | - Shantel Weinsheimer
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, USA
| | - Daniel Cooke
- Department of Radiology, University of California, San Francisco, USA
| | - Ethan Winkler
- Department of Neurosurgery, University of California, San Francisco, USA
| | - Adib Abla
- Department of Neurosurgery, University of California, San Francisco, USA
| | - Helen Kim
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, USA
| | - Hua Su
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, USA
| |
Collapse
|
40
|
Ando K, Ishii T, Fukuhara S. Zebrafish Vascular Mural Cell Biology: Recent Advances, Development, and Functions. Life (Basel) 2021; 11:1041. [PMID: 34685412 PMCID: PMC8537713 DOI: 10.3390/life11101041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
Recruitment of mural cells to the vascular wall is essential for forming the vasculature as well as maintaining proper vascular functions. In recent years, zebrafish genetic tools for mural cell biology have improved substantially. Fluorescently labeled zebrafish mural cell reporter lines enable us to study, with higher spatiotemporal resolution than ever, the processes of mural cell development from their progenitors. Furthermore, recent phenotypic analysis of platelet-derived growth factor beta mutant zebrafish revealed well-conserved organotypic mural cell development and functions in vertebrates with the unique features of zebrafish. However, comprehensive reviews of zebrafish mural cells are lacking. Therefore, herein, we highlight recent advances in zebrafish mural cell tools. We also summarize the fundamental features of zebrafish mural cell development, especially at early stages, and functions.
Collapse
Affiliation(s)
- Koji Ando
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Tokyo 113 8602, Japan; (T.I.); (S.F.)
| | | | | |
Collapse
|
41
|
Abstract
Brain arteriovenous malformation (bAVM) is the most common cause of intracranial hemorrhage (ICH), particularly in young patients. However, the exact cause of bAVM bleeding and rupture is not yet fully understood. In bAVMs, blood bypasses the entire capillary bed and directly flows from arteries to veins. The vessel walls in bAVMs have structural defects, which impair vascular integrity. Mural cells are essential structural and functional components of blood vessels and play a critical role in maintaining vascular integrity. Changes in mural cell number and coverage have been implicated in bAVMs. In this review, we discussed the roles of mural cells in bAVM pathogenesis. We focused on 1) the recent advances in human and animal studies of bAVMs; 2) the importance of mural cells in vascular integrity; 3) the regulatory signaling pathways that regulate mural cell function. More specifically, the platelet-derived growth factor-B (PDGF-B)/PDGF receptor-β (PDGFR-β), EphrinB2/EphB4, and angiopoietins/tie2 signaling pathways that regulate mural cell-recruitment during vascular remodeling were discussed in detail.
Collapse
|
42
|
Gao S, Nelson J, Weinsheimer S, Winkler EA, Rutledge C, Abla AA, Gupta N, Shieh JT, Cooke DL, Hetts SW, Tihan T, Hess CP, Ko N, Walcott BP, McCulloch CE, Lawton MT, Su H, Pawlikowska L, Kim H. Somatic mosaicism in the MAPK pathway in sporadic brain arteriovenous malformation and association with phenotype. J Neurosurg 2021; 136:148-155. [PMID: 34214981 DOI: 10.3171/2020.11.jns202031] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/16/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Sporadic brain arteriovenous malformation (BAVM) is a tangled vascular lesion characterized by direct artery-to-vein connections that can cause life-threatening intracerebral hemorrhage (ICH). Recently, somatic mutations in KRAS have been reported in sporadic BAVM, and mutations in other mitogen-activated protein kinase (MAPK) signaling pathway genes have been identified in other vascular malformations. The objectives of this study were to systematically evaluate somatic mutations in MAPK pathway genes in patients with sporadic BAVM lesions and to evaluate the association of somatic mutations with phenotypes of sporadic BAVM severity. METHODS The authors performed whole-exome sequencing on paired lesion and blood DNA samples from 14 patients with sporadic BAVM, and 295 genes in the MAPK signaling pathway were evaluated to identify genes with somatic mutations in multiple patients with BAVM. Digital droplet polymerase chain reaction was used to validate KRAS G12V and G12D mutations and to assay an additional 56 BAVM samples. RESULTS The authors identified a total of 24 candidate BAVM-associated somatic variants in 11 MAPK pathway genes. The previously identified KRAS G12V and G12D mutations were the only recurrent mutations. Overall, somatic KRAS G12V was present in 14.5% of BAVM lesions and G12D was present in 31.9%. The authors did not detect a significant association between the presence or allelic burden of KRAS mutation and three BAVM phenotypes: lesion size (maximum diameter), age at diagnosis, and age at ICH. CONCLUSIONS The authors confirmed the high prevalence of somatic KRAS mutations in sporadic BAVM lesions and identified several candidate somatic variants in other MAPK pathway genes. These somatic variants may contribute to understanding of the etiology of sporadic BAVM and the clinical characteristics of patients with this condition.
Collapse
Affiliation(s)
- Sen Gao
- Departments of1Anesthesia and Perioperative Care.,2Center for Cerebrovascular Research, and
| | - Jeffrey Nelson
- Departments of1Anesthesia and Perioperative Care.,2Center for Cerebrovascular Research, and
| | - Shantel Weinsheimer
- Departments of1Anesthesia and Perioperative Care.,2Center for Cerebrovascular Research, and.,4Institute for Human Genetics, University of California, San Francisco, California
| | | | | | | | | | - Joseph T Shieh
- 4Institute for Human Genetics, University of California, San Francisco, California.,11Pediatrics, and
| | | | | | | | | | | | - Brian P Walcott
- 3Neurological Surgery.,8NorthShore University Health System, Evanston, Illinois; and
| | | | - Michael T Lawton
- 10Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona
| | - Hua Su
- Departments of1Anesthesia and Perioperative Care.,2Center for Cerebrovascular Research, and
| | - Ludmila Pawlikowska
- Departments of1Anesthesia and Perioperative Care.,2Center for Cerebrovascular Research, and.,4Institute for Human Genetics, University of California, San Francisco, California
| | - Helen Kim
- Departments of1Anesthesia and Perioperative Care.,2Center for Cerebrovascular Research, and.,4Institute for Human Genetics, University of California, San Francisco, California
| |
Collapse
|
43
|
Involvement of Microglia in the Pathophysiology of Intracranial Aneurysms and Vascular Malformations-A Short Overview. Int J Mol Sci 2021; 22:ijms22116141. [PMID: 34200256 PMCID: PMC8201350 DOI: 10.3390/ijms22116141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/19/2022] Open
Abstract
Aneurysms and vascular malformations of the brain represent an important source of intracranial hemorrhage and subsequent mortality and morbidity. We are only beginning to discern the involvement of microglia, the resident immune cell of the central nervous system, in these pathologies and their outcomes. Recent evidence suggests that activated proinflammatory microglia are implicated in the expansion of brain injury following subarachnoid hemorrhage (SAH) in both the acute and chronic phases, being also a main actor in vasospasm, considerably the most severe complication of SAH. On the other hand, anti-inflammatory microglia may be involved in the resolution of cerebral injury and hemorrhage. These immune cells have also been observed in high numbers in brain arteriovenous malformations (bAVM) and cerebral cavernomas (CCM), although their roles in these lesions are currently incompletely ascertained. The following review aims to shed a light on the most significant findings related to microglia and their roles in intracranial aneurysms and vascular malformations, as well as possibly establish the course for future research.
Collapse
|
44
|
Park ES, Kim S, Huang S, Yoo JY, Körbelin J, Lee TJ, Kaur B, Dash PK, Chen PR, Kim E. Selective Endothelial Hyperactivation of Oncogenic KRAS Induces Brain Arteriovenous Malformations in Mice. Ann Neurol 2021; 89:926-941. [PMID: 33675084 DOI: 10.1002/ana.26059] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Brain arteriovenous malformations (bAVMs) are a leading cause of hemorrhagic stroke and neurological deficits in children and young adults, however, no pharmacological intervention is available to treat these patients. Although more than 95% of bAVMs are sporadic without family history, the pathogenesis of sporadic bAVMs is largely unknown, which may account for the lack of therapeutic options. KRAS mutations are frequently observed in cancer, and a recent unprecedented finding of these mutations in human sporadic bAVMs offers a new direction in the bAVM research. Using a novel adeno-associated virus targeting brain endothelium (AAV-BR1), the current study tested if endothelial KRASG12V mutation induces sporadic bAVMs in mice. METHODS Five-week-old mice were systemically injected with either AAV-BR1-GFP or -KRASG12V . At 8 weeks after the AAV injection, bAVM formation and characteristics were addressed by histological and molecular analyses. The effect of MEK/ERK inhibition on KRASG12V -induced bAVMs was determined by treatment of trametinib, a US Food and Drug Administration (FDA)-approved MEK/ERK inhibitor. RESULTS The viral-mediated KRASG12V overexpression induced bAVMs, which were composed of a tangled nidus mirroring the distinctive morphology of human bAVMs. The bAVMs were accompanied by focal angiogenesis, intracerebral hemorrhages, altered vascular constituents, neuroinflammation, and impaired sensory/cognitive/motor functions. Finally, we confirmed that bAVM growth was inhibited by trametinib treatment. INTERPRETATION Our innovative approach using AAV-BR1 confirms that KRAS mutations promote bAVM development via the MEK/ERK pathway, and provides a novel preclinical mouse model of bAVMs which will be useful to develop a therapeutic strategy for patients with bAVM. ANN NEUROL 2021;89:926-941.
Collapse
Affiliation(s)
- Eun S Park
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Sehee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Shuning Huang
- Department of Diagnostic and Interventional Imaging, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Ji Young Yoo
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Jakob Körbelin
- II. Department of Internal Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tae Jin Lee
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Balveen Kaur
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Peng R Chen
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
45
|
Giarretta I, Sturiale CL, Gatto I, Pacioni S, Gaetani E, Porfidia A, Puca A, Palucci I, Tondi P, Olivi A, Pallini R, Pola R. Sonic hedgehog is expressed in human brain arteriovenous malformations and induces arteriovenous malformations in vivo. J Cereb Blood Flow Metab 2021; 41:324-335. [PMID: 32169015 PMCID: PMC8369994 DOI: 10.1177/0271678x20912405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abnormalities in arterial versus venous endothelial cell identity and dysregulation of angiogenesis are deemed important in the pathophysiology of brain arteriovenous malformations (AVMs). The Sonic hedgehog (Shh) pathway is crucial for both angiogenesis and arterial versus venous differentiation of endothelial cells, through its dual role on the vascular endothelial growth factor/Notch signaling and the nuclear orphan receptor COUP-TFII. In this study, we show that Shh, Gli1 (the main transcription factor of the Shh pathway), and COUP-TFII (a target of the non-canonical Shh pathway) are aberrantly expressed in human brain AVMs. We also show that implantation of pellets containing Shh in the cornea of Efnb2/LacZ mice induces growth of distinct arteries and veins, interconnected by complex sets of arteriovenous shunts, without an interposed capillary bed, as seen in AVMs. We also demonstrate that injection in the rat brain of a plasmid containing the human Shh gene induces the growth of tangles of tortuous and dilated vessels, in part positive and in part negative for the arterial marker αSMA, with direct connections between αSMA-positive and -negative vessels. In summary, we show that the Shh pathway is active in human brain AVMs and that Shh-induced angiogenesis has characteristics reminiscent of those seen in AVMs in humans.
Collapse
Affiliation(s)
- Igor Giarretta
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carmelo L Sturiale
- Division of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ilaria Gatto
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simone Pacioni
- Division of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Gaetani
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Angelo Porfidia
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alfredo Puca
- Division of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ivana Palucci
- Istitute of Microbiology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paolo Tondi
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Olivi
- Division of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Pallini
- Division of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Pola
- Department of Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy.,Division of Cardiovascular Research, St. Elizabeth's Medical Center, Boston, MA, USA
| |
Collapse
|
46
|
Snodgrass RO, Chico TJA, Arthur HM. Hereditary Haemorrhagic Telangiectasia, an Inherited Vascular Disorder in Need of Improved Evidence-Based Pharmaceutical Interventions. Genes (Basel) 2021; 12:174. [PMID: 33513792 PMCID: PMC7911152 DOI: 10.3390/genes12020174] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 12/15/2022] Open
Abstract
Hereditary haemorrhagic telangiectasia (HHT) is characterised by arteriovenous malformations (AVMs). These vascular abnormalities form when arteries and veins directly connect, bypassing the local capillary system. Large AVMs may occur in the lungs, liver and brain, increasing the risk of morbidity and mortality. Smaller AVMs, known as telangiectases, are prevalent on the skin and mucosal lining of the nose, mouth and gastrointestinal tract and are prone to haemorrhage. HHT is primarily associated with a reduction in endoglin (ENG) or ACVRL1 activity due to loss-of-function mutations. ENG and ACVRL1 transmembrane receptors are expressed on endothelial cells (ECs) and bind to circulating ligands BMP9 and BMP10 with high affinity. Ligand binding to the receptor complex leads to activation of the SMAD1/5/8 signalling pathway to regulate downstream gene expression. Various genetic animal models demonstrate that disruption of this pathway in ECs results in AVMs. The vascular abnormalities underlying AVM formation result from abnormal EC responses to angiogenic and haemodynamic cues, and include increased proliferation, reduced migration against the direction of blood flow and an increased EC footprint. There is growing evidence that targeting VEGF signalling has beneficial outcomes in HHT patients and in animal models of this disease. The anti-VEGF inhibitor bevacizumab reduces epistaxis and has a normalising effect on high cardiac output in HHT patients with hepatic AVMs. Blocking VEGF signalling also reduces vascular malformations in mouse models of HHT1 and HHT2. However, VEGF signalling is complex and drives numerous downstream pathways, and it is not yet clear which pathway (or combination of pathways) is critical to target. This review will consider the recent evidence gained from HHT clinical and preclinical studies that are increasing our understanding of HHT pathobiology and informing therapeutic strategies.
Collapse
Affiliation(s)
- Ryan O. Snodgrass
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, University of Sheffield, Sheffield S10 2RX, UK; (R.O.S.); (T.J.A.C.)
| | - Timothy J. A. Chico
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, University of Sheffield, Sheffield S10 2RX, UK; (R.O.S.); (T.J.A.C.)
| | - Helen M. Arthur
- Biosciences Institute, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| |
Collapse
|
47
|
Abstract
The complex development of the brain vascular system can be broken down by embryonic stages and anatomic locations, which are tightly regulated by different factors and pathways in time and spatially. The adult brain is relatively quiescent in angiogenesis. However, under disease conditions, such as trauma, stroke, or tumor, angiogenesis can be activated in the adult brain. Disruption of any of the factors or pathways may lead to malformed vessel development. In this chapter, we will discuss factors and pathways involved in normal brain vasculogenesis and vascular maturation, and the pathogenesis of several brain vascular malformations.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, United States
| | - Sonali S Shaligram
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California San Francisco, San Francisco, CA, United States
| | - Hua Su
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
48
|
Nadeem T, Bogue W, Bigit B, Cuervo H. Deficiency of Notch signaling in pericytes results in arteriovenous malformations. JCI Insight 2020; 5:125940. [PMID: 33148887 PMCID: PMC7710269 DOI: 10.1172/jci.insight.125940] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 09/24/2020] [Indexed: 01/08/2023] Open
Abstract
Arteriovenous malformations (AVMs) are high-flow lesions directly connecting arteries and veins. In the brain, AVM rupture can cause seizures, stroke, and death. Patients with AVMs exhibit reduced coverage of the vessels by pericytes, the mural cells of microvascular capillaries; however, the mechanism underlying this pericyte reduction and its association with AVM pathogenesis remains unknown. Notch signaling has been proposed to regulate critical pericyte functions. We hypothesized that Notch signaling in pericytes is crucial to maintain pericyte homeostasis and prevent AVM formation. We inhibited Notch signaling specifically in perivascular cells and analyzed the vasculature of these mice. The retinal vessels of mice with deficient perivascular Notch signaling developed severe AVMs, together with a significant reduction in pericytes and vascular smooth muscle cells (vSMC) in the arteries, while vSMCs were increased in the veins. Vascular malformations and pericyte loss were also observed in the forebrain of embryonic mice deficient for perivascular Notch signaling. Moreover, the loss of Notch signaling in pericytes downregulated Pdgfrb levels and increased pericyte apoptosis, pointing to a critical role for Notch in pericyte survival. Overall, our findings reveal a mechanism of AVM formation and highlight the Notch signaling pathway as an essential mediator in this process.
Collapse
|
49
|
Li Y, Shang Q, Li P, Yang Z, Yang J, Shi J, Ge S, Wang Y, Fan X, Jia R. BMP9 attenuates occurrence of venous malformation by maintaining endothelial quiescence and strengthening vessel walls via SMAD1/5/ID1/α-SMA pathway. J Mol Cell Cardiol 2020; 147:92-107. [PMID: 32730768 DOI: 10.1016/j.yjmcc.2020.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/30/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022]
Abstract
Venous malformation (VM) is a type of vascular morphogenic defect in humans with an incidence of 1%. Although gene mutation is considered as the most common cause of VM, the pathogenesis of those without gene mutation remains to be elucidated. Here, we aimed to explore the relation of bone morphogenetic protein 9 (BMP9) and development of VM. At first, we found serum and tissue BMP9 expression in VM patients was significantly lower than that in healthy subjects, detected via enzyme-linked immunosorbent assay. Next, with wound healing assay, transwell assay and tube formation assay, we discovered BMP9 could inhibit migration and enhance tube formation activity of human umbilical vein endothelial cells (HUVECs) via receptor activin receptor-like kinase 1 (ALK1). Besides, BMP9 improved the expression of structural proteins alpha-smooth muscle actin (α-SMA) and Desmin in human umbilical vein smooth muscle cells (HUVSMCs) via activation of the SMAD1/5-ID1 pathway, determined by RNA-based next-generation sequencing, qPCR, immunofluorescence and western blotting. Intriguingly, this effect could be blocked by receptor ALK1 inhibitor, SMAD1/5 inhibitor and siRNAs targeting ID1, verifying the BMP9/ALK1/SMAD1/5/ID1/α-SMA pathway. Meanwhile, knocking out BMP9 in C57BL/6 mice embryo led to α-SMA scarcity in walls of lung and mesenteric vessels, as well as walls of small trachea. BMP9-/- zebrafish also exhibited abnormal vascular maturity, indicating a critical role of BMP9 in vascular maturity and remodeling. Finally, a VM mice model revealed that BMP9 might have therapeutic effect in VM progression. Our study discovered that BMP9 might inhibit the occurrence of VM by strengthening the vessel wall and maintaining endothelium quiescence. These findings provide promising evidences of new therapeutic targets that might be used for the management of VM.
Collapse
Affiliation(s)
- Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Qingfeng Shang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Peng Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Zhi Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Jie Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Jiahao Shi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Yefei Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China.
| |
Collapse
|
50
|
Anderson KE, Bellio TA, Aniskovich E, Adams SL, Blusztajn JK, Delalle I. The Expression of Activin Receptor-Like Kinase 1 (ACVRL1/ALK1) in Hippocampal Arterioles Declines During Progression of Alzheimer's Disease. Cereb Cortex Commun 2020; 1:tgaa031. [PMID: 32974611 PMCID: PMC7497413 DOI: 10.1093/texcom/tgaa031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 12/18/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) in Alzheimer’s disease (AD)—deposition of beta amyloid
(Aβ) within the walls of cerebral blood vessels—typically accompanies Aβ buildup in brain
parenchyma and causes abnormalities in vessel structure and function. We recently
demonstrated that the immunoreactivity of activin receptor-like kinase 1 (ALK1), the type
I receptor for circulating BMP9/BMP10 (bone morphogenetic protein) signaling proteins, is
reduced in advanced, but not early stages of AD in CA3 pyramidal neurons. Here we
characterize vascular expression of ALK1 in the context of progressive AD pathology
accompanied by amyloid angiopathy in postmortem hippocampi using immunohistochemical
methods. Hippocampal arteriolar wall ALK1 signal intensity was 35% lower in AD patients
(Braak and Braak Stages IV and V [BBIV-V]; clinical dementia rating [CDR1-2]) as compared
with subjects with early AD pathologic changes but either cognitively intact or with
minimal cognitive impairment (BBIII; CDR0-0.5). The intensity of Aβ signal in arteriolar
walls was similar in all analyzed cases. These data suggest that, as demonstrated
previously for specific neuronal populations, ALK1 expression in blood vessels is also
vulnerable to the AD pathophysiologic process, perhaps related to CAA. However, cortical
arterioles may remain responsive to the ALK1 ligands, such as BMP9 and BMP10 in early and
moderate AD.
Collapse
Affiliation(s)
- Kelley E Anderson
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Thomas A Bellio
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Emily Aniskovich
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Stephanie L Adams
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jan Krzysztof Blusztajn
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA.,Department of Pathology and Laboratory Medicine, Lifespan Academic Medical Center, Warren Alpert Medical School of Brown University, Providence 02903 RI, USA
| |
Collapse
|