1
|
Shin S, Kim YW, Sheen SH, Park SQ, Jin SC, Jeon JP, Lee JY, Lee BC, Lim YW, Kim GO, Kwon YU, Lee YR, Han SY, Oh JS. Epidemiology and Functional Outcome of Acute Stroke Patients in Korea Using Nationwide data. J Korean Neurosurg Soc 2025; 68:159-176. [PMID: 39313854 PMCID: PMC11924632 DOI: 10.3340/jkns.2024.0118] [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: 07/14/2024] [Accepted: 09/21/2024] [Indexed: 09/25/2024] Open
Abstract
OBJECTIVE Korea's healthcare system and policy promotes early, actively stroke treatment to improve prognosis. This study represents stroke epidemiology and outcomes in Korea. METHODS This study investigated data from the Acute Stroke Assessment Registry. The registry collects data from over 220 hospitals nationwide, focusing on quality stroke service management. Data analysis included patient demographics, stroke severity assessment, and discharge prognosis measurement using standardized scales. RESULTS Eighty-six thousand five hundred sixty-eight acute stroke patients were collected with demographic and clinical characteristics during 18 months from 2016, 2018, and between 2020 to 2021, focusing on acute subarachnoid hemorrhage (SAH), acute intracerebral hemorrhage (ICH), and acute ischemic stroke. Of these 86568 patients, 8.3% was SAH, 16.3% ICH, and 74.9% ischemic stroke. Trends showed decreasing SAH and increasing ICH cases over the years. 68.3% stroke patients had the clear onset time. 49.6% stroke patients arrived within 4.5 hours of symptom onset, with more patients treated at general hospitals. Good functional outcomes at discharge was obtained with 58.3% of acute stroke patients, 55.9% of SAH patients, 34.6% of ICH patients, and 63.8% of ischemic stroke patients. CONCLUSION The results showed that ischemic stroke was the most common subtype, followed by ICH and SAH. Prognosis differed among subtypes, with favorable outcomes more common in ischemic stroke and SAH compared to ICH.
Collapse
Affiliation(s)
- Seungmin Shin
- Department of Neurosurgery, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Young Woo Kim
- Department of Neurosurgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Hun Sheen
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University, Seongnam, Korea
| | - Sukh Que Park
- Department of Neurosurgery, Soonchunhyang University Seoul Hospital, College of Medicine, Soonchunhyang University, Seoul, Korea
| | - Sung-Chul Jin
- Department of Neurosurgery, Inje University Haeundae Paik Hospital, Busan, Korea
| | - Jin Pyeong Jeon
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon, Korea
| | - Ji Young Lee
- Department of Neurosurgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Boung Chul Lee
- Health Insurance Review & Assessment Service (HIRA), Wonju, Korea
| | - Young Wha Lim
- Health Insurance Review & Assessment Service (HIRA), Wonju, Korea
| | - Gui Ok Kim
- Health Insurance Review & Assessment Service (HIRA), Wonju, Korea
| | - Youg Uk Kwon
- Healthcare Review and Assessment Committee (HCRAC), Seoul, Korea
| | - Yu Ra Lee
- Cardio-Cerebrovascular Disease Assessment Division, Quality Assessment Administration Department, Healthcare Review and Assessment Committee (HCRAC), Seoul, Korea
| | - So Young Han
- Quality Assessment Management Division, Quality Assessment Department , Healthcare Review and Assessment Committee (HCRAC), Seoul, Korea
| | - Jae Sang Oh
- Department of Neurosurgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
2
|
Jabarkheel R, Li L, Frankfurter M, Zhang DY, Gajjar A, Muhammad N, Srinivasan VM, Burkhardt JK, Kahn M. Untangling sporadic brain arteriovenous malformations: towards targeting the KRAS/MAPK pathway. Front Surg 2024; 11:1504612. [PMID: 39687326 PMCID: PMC11646853 DOI: 10.3389/fsurg.2024.1504612] [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: 10/01/2024] [Accepted: 10/30/2024] [Indexed: 12/18/2024] Open
Abstract
Brain arteriovenous malformations (AVMs) are vascular lesions characterized by abnormal connections between parenchymal arteries and veins, bypassing a capillary bed, and forming a nidus. Brain AVMs are consequential as they are prone to rupture and associated with significant morbidity. They can broadly be subdivided into hereditary vs. sporadic lesions with sporadic brain AVMs representing the majority of all brain AVMs. However, little had been known about the pathogenesis of sporadic brain AVMs until the landmark discovery in 2018 that the majority of sporadic brain AVMs carry somatic activating mutations of the oncogene, Kirsten rat sarcoma viral oncogene homologue (KRAS), in their endothelial cells. Here, we review the history of brain AVMs, their treatments, and recent advances in uncovering the pathogenesis of sporadic brain AVMs. We specifically focus on the latest studies suggesting that pharmacologically targeting the KRAS/MEK pathway may be a potentially efficacious treatment for sporadic brain AVMs.
Collapse
Affiliation(s)
- Rashad Jabarkheel
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Lun Li
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Maxwell Frankfurter
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Y. Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Avi Gajjar
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Najib Muhammad
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Visish M. Srinivasan
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jan-Karl Burkhardt
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Mark Kahn
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
3
|
Das S, Kasher P, Waqar M, Parry-Jones A, Patel H. Reporting of angiographic studies in patients diagnosed with a cerebral arteriovenous malformation: a systematic review. F1000Res 2024; 12:1252. [PMID: 39931157 PMCID: PMC11809685 DOI: 10.12688/f1000research.139256.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 02/13/2025] Open
Abstract
A cerebral arteriovenous malformation (cAVM) is an abnormal tangle of cerebral blood vessels. The consensus document by the Joint Writing Group (JWG) highlighted which cAVM features should be recorded. Subsequent publications have reported cAVM angioarchitecture, but it is unknown if all followed the JWG recommendations. The aim of this systematic review was to describe use of the JWG guidelines. A database search, using the PRISMA checklist, was performed. We describe the proportion of publications that used JWG reporting standards, which standards were used, whether the definitions used differed from the JWG, or if any additional angiographic features were reported. Out of 4306 articles identified, 105 were selected, and a further 114 from other sources. Thirty-three studies (33/219; 15%) specifically referred to using JWG standards. Since the JWG publication, few studies have used their standards to report cAVMs. This implies that the angioarchitecture of cAVMs are not routinely fully described.
Collapse
Affiliation(s)
- Suparna Das
- The University of Manchester, Manchester, England, UK
| | - Paul Kasher
- The University of Manchester, Manchester, England, UK
| | - Mueez Waqar
- The University of Manchester, Manchester, England, UK
| | | | - Hiren Patel
- The University of Manchester, Manchester, England, UK
| |
Collapse
|
4
|
Tanaka M. Basic Knowledge and Overview of Brain AVMs. JOURNAL OF NEUROENDOVASCULAR THERAPY 2024; 19:2024-0037. [PMID: 40018280 PMCID: PMC11864997 DOI: 10.5797/jnet.ra.2024-0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/11/2024] [Indexed: 03/01/2025]
Abstract
Brain arteriovenous malformations (AVMs) are intricate networks of blood vessels in which arteries connect directly to veins, bypassing the capillary system. This aberration can lead to serious neurological manifestations, including seizures, headaches, and hemorrhagic strokes. The embryonic development of AVMs implicates possible disruptions in arteriovenous differentiation during angiogenesis, improper regression of the primary capillary plexus, or the retention of fetal vasculature as contributing factors. Additionally, genetic mutations and environmental influences during pregnancy may facilitate AVM formation, with identified mutations in genes such as endoglin, activin receptor-like kinase 1, SMAD family member 4, and RAS p21 protein activator 1 disrupting vascular development. Such mutations are associated with conditions like hereditary hemorrhagic telangiectasia and capillary malformation-arteriovenous malformation syndrome, thus highlighting the essential role of genetic counseling in AVM management. This review underscores the importance of a deep comprehension of the embryological and genetic foundations of AVMs to refine diagnostic, therapeutic, and prognostic approaches. The paper advocates for advanced research on intervention strategies and emphasizes the significance of a genetics-focused approach in the clinical management of AVMs.
Collapse
Affiliation(s)
- Michihiro Tanaka
- Department of Neurosurgery and Neuroendovascular Surgery, Kameda Medical Center, Kamogawa, Chiba, Japan
| |
Collapse
|
5
|
Astudillo Potes MD, Bauman MMJ, Shoushtari A, Winter BM, Singh R, Rahmani R, Catapano J, Lawton MT. Elucidating the pathogenesis behind arteriovenous malformations of the central nervous system: a bibliometric analysis. Neurosurg Rev 2024; 47:133. [PMID: 38556597 DOI: 10.1007/s10143-024-02367-3] [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: 11/18/2023] [Revised: 02/20/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024]
Abstract
Arteriovenous malformations (AVMs) are vascular malformations of the central nervous system (CNS) with potential for significant consequences. The exact pathophysiologic mechanism of AVM formation is not fully understood. This study aims to evaluate bibliometric parameters and citations of the literature of AVMs to provide an overview of how the field has evolved. We performed an electronic search on Web of Science to identify the top 100 published and indexed articles with the highest number of citations discussing the pathogenesis of AVMs. This study yielded 1863 articles, of which the top 100 were selected based on the highest total citation count. These articles included 24% basic science, 46% clinical, and 30% review articles. The most-cited article was a clinical article from 2003, and the most recent was published in 2022. The median number of authors was 6, with the highest being 46 for a clinical article. The top 5 journals were identified, with the highest impact factor being 20.1. 13 countries were identified, with the US contributing the most articles (approximately 70%). Regarding genes of investigation, VEGF was one of the early genes investigated, while more interested in RAS/MAPK has been garnered since 2015. There is a growing interest in AVM genomics and pathogenesis research. While progress has been made in understanding clinical aspects and risk factors, the exact pathophysiological mechanisms and genetic basis of AVM formation remain incompletely understood. Further investigation of key genes in AVM pathogenesis can allow identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Maria D Astudillo Potes
- Mayo Clinic Alix School of Medicine, Rochester, Minnesota, USA
- Department of Neurological Surgery, Rochester, Minnesota, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Megan M J Bauman
- Mayo Clinic Alix School of Medicine, Rochester, Minnesota, USA
- Department of Neurological Surgery, Rochester, Minnesota, USA
| | - Ali Shoushtari
- Department of Neurological Surgery, Rochester, Minnesota, USA
| | - Bailey M Winter
- Mayo Clinic Alix School of Medicine, Rochester, Minnesota, USA
- Department of Neurological Surgery, Rochester, Minnesota, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Rohin Singh
- Department of Neurosurgery, University of Rochester, Rochester, NY, USA.
| | - Redi Rahmani
- Department of Neurosurgery, University of Rochester, Rochester, NY, USA
- Barrow Neurological Institute, Phoenix, AZ, USA
| | | | | |
Collapse
|
6
|
Lauzier DC, Srienc AI, Vellimana AK, Dacey Jr RG, Zipfel GJ. Peripheral macrophages in the development and progression of structural cerebrovascular pathologies. J Cereb Blood Flow Metab 2024; 44:169-191. [PMID: 38000039 PMCID: PMC10993883 DOI: 10.1177/0271678x231217001] [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: 07/05/2023] [Revised: 09/10/2023] [Accepted: 09/15/2023] [Indexed: 11/26/2023]
Abstract
The human cerebrovascular system is responsible for maintaining neural function through oxygenation, nutrient supply, filtration of toxins, and additional specialized tasks. While the cerebrovascular system has resilience imparted by elaborate redundant collateral circulation from supportive tertiary structures, it is not infallible, and is susceptible to developing structural vascular abnormalities. The causes of this class of structural cerebrovascular diseases can be broadly categorized as 1) intrinsic developmental diseases resulting from genetic or other underlying aberrations (arteriovenous malformations and cavernous malformations) or 2) extrinsic acquired diseases that cause compensatory mechanisms to drive vascular remodeling (aneurysms and arteriovenous fistulae). Cerebrovascular diseases of both types pose significant risks to patients, in some cases leading to death or disability. The drivers of such diseases are extensive, yet inflammation is intimately tied to all of their progressions. Central to this inflammatory hypothesis is the role of peripheral macrophages; targeting this critical cell type may lead to diagnostic and therapeutic advancement in this area. Here, we comprehensively review the role that peripheral macrophages play in cerebrovascular pathogenesis, provide a schema through which macrophage behavior can be understood in cerebrovascular pathologies, and describe emerging diagnostic and therapeutic avenues in this area.
Collapse
Affiliation(s)
- David C Lauzier
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anja I Srienc
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ananth K Vellimana
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ralph G Dacey Jr
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory J Zipfel
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
7
|
González EO, Runge S, Mantziaris G, Ironside N, Sheehan JP. Stereotactic radiosurgery for brain arteriovenous malformations in patients with hereditary hemorrhagic telangiectasia. Acta Neurochir (Wien) 2024; 166:21. [PMID: 38231447 PMCID: PMC10794397 DOI: 10.1007/s00701-024-05923-4] [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: 11/21/2023] [Accepted: 12/14/2023] [Indexed: 01/18/2024]
Abstract
OBJECTIVE Brain arteriovenous malformations (AVMs) in patients with hereditary hemorrhagic telangiectasia (HHT) present different characteristics from sporadic AVMs, and they have lower initial bleeding rates. Conservative management is usually preferred for the treatment of these lesions. In this case study, we present the largest series of HHT patients treated with stereotactic radiosurgery to date. METHODS We identified eight patients with HHT and 14 AVMs. We retrospectively collected clinical, radiographic, and treatment characteristics of the patients and each AVM. RESULTS Most patients in our sample presented with small AVMs. The median volume of these AVMs was 0.22 cm3 (IQR 0.08-0.59). Three out of eight patients presented with initial intracerebral hemorrhage (ICH). The majority of lesions had low (12/14) Spetzler-Martin grades (I-II). Median maximum and margin doses used for treatment were 36.2 (IQR 35.25-44.4) and 20 (IQR 18-22.5) Gy, respectively. The overall obliteration rate after SRS was 11/14, and the median time to obliteration across all 11 obliterated AVMs was 35.83 months (IQR, 17-39.99). Neurological status was favorable with all patients having a mRS of 0 or 1 at the last follow-up. Symptomatic radiation-induced changes (RIC) after SRS were low (7.1%), and there were no permanent RIC. CONCLUSIONS Patients with HHT who present with multiple brain AVMs are generally well served by SRS. Obliteration can be achieved in the majority of HHT patients and with a low complication rate. In the current study, initial hemorrhage rates prior to SRS were noticeable which supports the decision to treat these AVMs. Future studies are needed to better address the role of SRS for HHT patients harboring ruptured and unruptured AVMs.
Collapse
Affiliation(s)
- Eduardo Orrego González
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Sean Runge
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Georgios Mantziaris
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Natasha Ironside
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Jason P Sheehan
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA, 22908, USA.
| |
Collapse
|
8
|
Zhao S, Mekbib KY, van der Ent MA, Allington G, Prendergast A, Chau JE, Smith H, Shohfi J, Ocken J, Duran D, Furey CG, Hao LT, Duy PQ, Reeves BC, Zhang J, Nelson-Williams C, Chen D, Li B, Nottoli T, Bai S, Rolle M, Zeng X, Dong W, Fu PY, Wang YC, Mane S, Piwowarczyk P, Fehnel KP, See AP, Iskandar BJ, Aagaard-Kienitz B, Moyer QJ, Dennis E, Kiziltug E, Kundishora AJ, DeSpenza T, Greenberg ABW, Kidanemariam SM, Hale AT, Johnston JM, Jackson EM, Storm PB, Lang SS, Butler WE, Carter BS, Chapman P, Stapleton CJ, Patel AB, Rodesch G, Smajda S, Berenstein A, Barak T, Erson-Omay EZ, Zhao H, Moreno-De-Luca A, Proctor MR, Smith ER, Orbach DB, Alper SL, Nicoli S, Boggon TJ, Lifton RP, Gunel M, King PD, Jin SC, Kahle KT. Mutation of key signaling regulators of cerebrovascular development in vein of Galen malformations. Nat Commun 2023; 14:7452. [PMID: 37978175 PMCID: PMC10656524 DOI: 10.1038/s41467-023-43062-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 10/30/2023] [Indexed: 11/19/2023] Open
Abstract
To elucidate the pathogenesis of vein of Galen malformations (VOGMs), the most common and most severe of congenital brain arteriovenous malformations, we performed an integrated analysis of 310 VOGM proband-family exomes and 336,326 human cerebrovasculature single-cell transcriptomes. We found the Ras suppressor p120 RasGAP (RASA1) harbored a genome-wide significant burden of loss-of-function de novo variants (2042.5-fold, p = 4.79 x 10-7). Rare, damaging transmitted variants were enriched in Ephrin receptor-B4 (EPHB4) (17.5-fold, p = 1.22 x 10-5), which cooperates with p120 RasGAP to regulate vascular development. Additional probands had damaging variants in ACVRL1, NOTCH1, ITGB1, and PTPN11. ACVRL1 variants were also identified in a multi-generational VOGM pedigree. Integrative genomic analysis defined developing endothelial cells as a likely spatio-temporal locus of VOGM pathophysiology. Mice expressing a VOGM-specific EPHB4 kinase-domain missense variant (Phe867Leu) exhibited disrupted developmental angiogenesis and impaired hierarchical development of arterial-capillary-venous networks, but only in the presence of a "second-hit" allele. These results illuminate human arterio-venous development and VOGM pathobiology and have implications for patients and their families.
Collapse
Affiliation(s)
- Shujuan Zhao
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kedous Y Mekbib
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Martijn A van der Ent
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Garrett Allington
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Andrew Prendergast
- Yale Zebrafish Research Core, Yale School of Medicine, New Haven, CT, USA
| | - Jocelyn E Chau
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT, USA
| | - Hannah Smith
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - John Shohfi
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Jack Ocken
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Daniel Duran
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS, USA
| | - Charuta G Furey
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
- Ivy Brain Tumor Center, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Le Thi Hao
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Phan Q Duy
- Department of Neurosurgery, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Benjamin C Reeves
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Junhui Zhang
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Di Chen
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Boyang Li
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Timothy Nottoli
- Yale Genome Editing Center, Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Suxia Bai
- Yale Genome Editing Center, Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Myron Rolle
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xue Zeng
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Po-Ying Fu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Yung-Chun Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Shrikant Mane
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Paulina Piwowarczyk
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Katie Pricola Fehnel
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alfred Pokmeng See
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bermans J Iskandar
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Beverly Aagaard-Kienitz
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Quentin J Moyer
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Evan Dennis
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emre Kiziltug
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Adam J Kundishora
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Tyrone DeSpenza
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Ana B W Greenberg
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Andrew T Hale
- Department of Neurosurgery, University of Alabama School of Medicine, Birmingham, AL, USA
| | - James M Johnston
- Department of Neurosurgery, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Eric M Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Phillip B Storm
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shih-Shan Lang
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - William E Butler
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul Chapman
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher J Stapleton
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aman B Patel
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Georges Rodesch
- Service de Neuroradiologie Diagnostique et Thérapeutique, Hôpital Foch, Suresnes, France
- Department of Interventional Neuroradiology, Hôpital Fondation A. de Rothschild, Paris, France
| | - Stanislas Smajda
- Department of Interventional Neuroradiology, Hôpital Fondation A. de Rothschild, Paris, France
| | - Alejandro Berenstein
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tanyeri Barak
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Hongyu Zhao
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Andres Moreno-De-Luca
- Department of Radiology, Autism & Developmental Medicine Institute, Genomic Medicine Institute, Geisinger, Danville, PA, USA
| | - Mark R Proctor
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edward R Smith
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Darren B Orbach
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurointerventional Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Seth L Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Stefania Nicoli
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale School of Medicine, New Haven, CT, USA
| | - Titus J Boggon
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT, USA
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Richard P Lifton
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Philip D King
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Sheng Chih Jin
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA.
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA.
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, US.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
9
|
Al-Smadi MW, Fazekas LA, Aslan S, Bernat B, Beqain A, Al-Khafaji MQM, Priksz D, Orlik B, Nemeth N. A Microsurgical Arteriovenous Malformation Model on Saphenous Vessels in the Rat. Biomedicines 2023; 11:2970. [PMID: 38001970 PMCID: PMC10669800 DOI: 10.3390/biomedicines11112970] [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: 10/12/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Arteriovenous malformation (AVM) is an anomaly of blood vessel formation. Numerous models have been established to understand the nature of AVM. These models have limitations in terms of the diameter of the vessels used and the impact on the circulatory system. Our goal was to establish an AVM model that does not cause prompt and significant hemodynamic and cardiac alterations but is feasible for follow-up of the AVM's progression. Sixteen female rats were randomly divided into sham-operated and AVM groups. In the AVM group, the saphenous vein and artery were interconnected using microsurgical techniques. The animals were followed up for 12 weeks. Anastomosis patency and the structural and hemodynamic changes of the heart were monitored. The hearts and vessels were histologically analyzed. During the follow-up period, shunts remained unobstructed. Systolic, diastolic, mean arterial pressure, and heart rate values slightly and non-significantly decreased in the AVM group. Echocardiogram results indicated minor systolic function impact, with slight and insignificant changes in aortic pressure and blood velocity, and minimal left ventricular wall enlargement. The small-caliber saphenous AVM model does not cause acute hemodynamic changes. Moderate but progressive alterations and venous dilatation confirmed AVM-like features. The model seems to be suitable for studying further the progression, enlargement, or destabilization of AVM.
Collapse
Affiliation(s)
- Mohammad Walid Al-Smadi
- Department of Operative Techniques and Surgical Research, Faculty of Medicine, University of Debrecen, Moricz Zsigmond ut 22, 4032 Debrecen, Hungary; (M.W.A.-S.); (L.A.F.); (S.A.); (A.B.); (M.Q.M.A.-K.)
- Kalman Laki Doctoral School, University of Debrecen, 4032 Debrecen, Hungary
| | - Laszlo Adam Fazekas
- Department of Operative Techniques and Surgical Research, Faculty of Medicine, University of Debrecen, Moricz Zsigmond ut 22, 4032 Debrecen, Hungary; (M.W.A.-S.); (L.A.F.); (S.A.); (A.B.); (M.Q.M.A.-K.)
| | - Siran Aslan
- Department of Operative Techniques and Surgical Research, Faculty of Medicine, University of Debrecen, Moricz Zsigmond ut 22, 4032 Debrecen, Hungary; (M.W.A.-S.); (L.A.F.); (S.A.); (A.B.); (M.Q.M.A.-K.)
| | - Brigitta Bernat
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (B.B.); (D.P.)
| | - Anas Beqain
- Department of Operative Techniques and Surgical Research, Faculty of Medicine, University of Debrecen, Moricz Zsigmond ut 22, 4032 Debrecen, Hungary; (M.W.A.-S.); (L.A.F.); (S.A.); (A.B.); (M.Q.M.A.-K.)
| | - Mustafa Qais Muhsin Al-Khafaji
- Department of Operative Techniques and Surgical Research, Faculty of Medicine, University of Debrecen, Moricz Zsigmond ut 22, 4032 Debrecen, Hungary; (M.W.A.-S.); (L.A.F.); (S.A.); (A.B.); (M.Q.M.A.-K.)
| | - Daniel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary; (B.B.); (D.P.)
| | - Brigitta Orlik
- Department of Pathology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, 4032 Debrecen, Hungary;
| | - Norbert Nemeth
- Department of Operative Techniques and Surgical Research, Faculty of Medicine, University of Debrecen, Moricz Zsigmond ut 22, 4032 Debrecen, Hungary; (M.W.A.-S.); (L.A.F.); (S.A.); (A.B.); (M.Q.M.A.-K.)
| |
Collapse
|
10
|
Lauzier DC, Chiang SN, Moran CJ. Etiologies of Brain Arteriovenous Malformation Recurrence: A Focus on Pediatric Disease. Pediatr Neurol 2023; 148:94-100. [PMID: 37690270 DOI: 10.1016/j.pediatrneurol.2023.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/18/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023]
Abstract
Pediatric brain arteriovenous malformations are a major cause of morbidity and mortality, with the harmful effects of this disease compounded by the additional disability-years experienced by children with ruptured or other symptomatic arteriovenous malformations. In addition to the risks shared with their adult counterparts, pediatric patients frequently experience recurrence following radiographic cure, which presents an additional source of morbidity and mortality. Therefore, there is a need to synthesize potential mechanisms contributing to the elevated recurrence risk in the pediatric population and discuss how these translate to practical considerations for managing these patients.
Collapse
Affiliation(s)
- David C Lauzier
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri.
| | - Sarah N Chiang
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Christopher J Moran
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
11
|
Zhao S, Mekbib KY, van der Ent MA, Allington G, Prendergast A, Chau JE, Smith H, Shohfi J, Ocken J, Duran D, Furey CG, Le HT, Duy PQ, Reeves BC, Zhang J, Nelson-Williams C, Chen D, Li B, Nottoli T, Bai S, Rolle M, Zeng X, Dong W, Fu PY, Wang YC, Mane S, Piwowarczyk P, Fehnel KP, See AP, Iskandar BJ, Aagaard-Kienitz B, Kundishora AJ, DeSpenza T, Greenberg ABW, Kidanemariam SM, Hale AT, Johnston JM, Jackson EM, Storm PB, Lang SS, Butler WE, Carter BS, Chapman P, Stapleton CJ, Patel AB, Rodesch G, Smajda S, Berenstein A, Barak T, Erson-Omay EZ, Zhao H, Moreno-De-Luca A, Proctor MR, Smith ER, Orbach DB, Alper SL, Nicoli S, Boggon TJ, Lifton RP, Gunel M, King PD, Jin SC, Kahle KT. Genetic dysregulation of an endothelial Ras signaling network in vein of Galen malformations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.18.532837. [PMID: 36993588 PMCID: PMC10055230 DOI: 10.1101/2023.03.18.532837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
To elucidate the pathogenesis of vein of Galen malformations (VOGMs), the most common and severe congenital brain arteriovenous malformation, we performed an integrated analysis of 310 VOGM proband-family exomes and 336,326 human cerebrovasculature single-cell transcriptomes. We found the Ras suppressor p120 RasGAP ( RASA1 ) harbored a genome-wide significant burden of loss-of-function de novo variants (p=4.79×10 -7 ). Rare, damaging transmitted variants were enriched in Ephrin receptor-B4 ( EPHB4 ) (p=1.22×10 -5 ), which cooperates with p120 RasGAP to limit Ras activation. Other probands had pathogenic variants in ACVRL1 , NOTCH1 , ITGB1 , and PTPN11 . ACVRL1 variants were also identified in a multi-generational VOGM pedigree. Integrative genomics defined developing endothelial cells as a key spatio-temporal locus of VOGM pathophysiology. Mice expressing a VOGM-specific EPHB4 kinase-domain missense variant exhibited constitutive endothelial Ras/ERK/MAPK activation and impaired hierarchical development of angiogenesis-regulated arterial-capillary-venous networks, but only when carrying a "second-hit" allele. These results illuminate human arterio-venous development and VOGM pathobiology and have clinical implications.
Collapse
|
12
|
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]
|
13
|
Kolarich AR, Solomon AJ, Bailey C, Latif MA, Rowan NR, Galiatsatos P, Weiss CR. Imaging Manifestations and Interventional Treatments for Hereditary Hemorrhagic Telangiectasia. Radiographics 2021; 41:2157-2175. [PMID: 34723698 DOI: 10.1148/rg.2021210100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Hemorrhagic hereditary telangiectasia (HHT) is a rare autosomal dominant disorder that causes multisystem vascular malformations including mucocutaneous telangiectasias and arteriovenous malformations (AVMs). Clinical and genetic screening of patients with signs, symptoms, or a family history suggestive of HHT is recommended to confirm the diagnosis on the basis of the Curaçao criteria and prevent associated complications. Patients with HHT frequently have epistaxis and gastrointestinal bleeding from telangiectasias. Pulmonary AVMs are common right-to-left shunts between pulmonary arteries and veins that can result in dyspnea and exercise intolerance, heart failure, migraine headaches, stroke or transient ischemic attacks, brain abscesses, or in rare cases, pulmonary hemorrhage. Primary neurologic complications from cerebral AVMs, which can take on many forms, are less common but particularly severe complications of HHT. Multimodality imaging, including transthoracic echocardiography, Doppler US, CT, and MRI, is used in the screening and initial characterization of vascular lesions in patients with HHT. Diagnostic angiography is an important tool in characterization of and interventional treatments for HHT, particularly those in the lungs and central nervous system. A multidisciplinary approach to early diagnosis, treatment, imaging, and surveillance at high-volume HHT Centers of Excellence is recommended. Although a variety of idiopathic, traumatic, or genetic conditions can result in similar clinical and imaging features, the Curaçao criteria are particularly useful for the proper diagnosis of HHT. Imaging and treatment options are reviewed, with a focus on screening, diagnosis, and posttreatment findings, with the use of updated international guidelines. Online supplemental material is available for this article. ©RSNA, 2021.
Collapse
Affiliation(s)
- Andrew R Kolarich
- From the Russell H. Morgan Department of Radiology and Radiological Science (A.R.K., A.J.S., C.B., M.A.L., C.R.W.), Department of Otolarygology-Head and Neck Surgery (N.R.R.), and Department of Medicine, Division of Pulmonology (P.G.), The Johns Hopkins University School of Medicine, 1800 Orleans St, Sheikh Zayed Tower, Baltimore, MD 21287
| | - Alex J Solomon
- From the Russell H. Morgan Department of Radiology and Radiological Science (A.R.K., A.J.S., C.B., M.A.L., C.R.W.), Department of Otolarygology-Head and Neck Surgery (N.R.R.), and Department of Medicine, Division of Pulmonology (P.G.), The Johns Hopkins University School of Medicine, 1800 Orleans St, Sheikh Zayed Tower, Baltimore, MD 21287
| | - Christopher Bailey
- From the Russell H. Morgan Department of Radiology and Radiological Science (A.R.K., A.J.S., C.B., M.A.L., C.R.W.), Department of Otolarygology-Head and Neck Surgery (N.R.R.), and Department of Medicine, Division of Pulmonology (P.G.), The Johns Hopkins University School of Medicine, 1800 Orleans St, Sheikh Zayed Tower, Baltimore, MD 21287
| | - Muhammad Aamir Latif
- From the Russell H. Morgan Department of Radiology and Radiological Science (A.R.K., A.J.S., C.B., M.A.L., C.R.W.), Department of Otolarygology-Head and Neck Surgery (N.R.R.), and Department of Medicine, Division of Pulmonology (P.G.), The Johns Hopkins University School of Medicine, 1800 Orleans St, Sheikh Zayed Tower, Baltimore, MD 21287
| | - Nicholas R Rowan
- From the Russell H. Morgan Department of Radiology and Radiological Science (A.R.K., A.J.S., C.B., M.A.L., C.R.W.), Department of Otolarygology-Head and Neck Surgery (N.R.R.), and Department of Medicine, Division of Pulmonology (P.G.), The Johns Hopkins University School of Medicine, 1800 Orleans St, Sheikh Zayed Tower, Baltimore, MD 21287
| | - Panagis Galiatsatos
- From the Russell H. Morgan Department of Radiology and Radiological Science (A.R.K., A.J.S., C.B., M.A.L., C.R.W.), Department of Otolarygology-Head and Neck Surgery (N.R.R.), and Department of Medicine, Division of Pulmonology (P.G.), The Johns Hopkins University School of Medicine, 1800 Orleans St, Sheikh Zayed Tower, Baltimore, MD 21287
| | - Clifford R Weiss
- From the Russell H. Morgan Department of Radiology and Radiological Science (A.R.K., A.J.S., C.B., M.A.L., C.R.W.), Department of Otolarygology-Head and Neck Surgery (N.R.R.), and Department of Medicine, Division of Pulmonology (P.G.), The Johns Hopkins University School of Medicine, 1800 Orleans St, Sheikh Zayed Tower, Baltimore, MD 21287
| |
Collapse
|
14
|
Song Y, Kwon B, Al-Abdulwahhab AH, Nam YK, Ahn Y, Jeong SY, Seo EJ, Lee JK, Suh DC. Rare Neurovascular Diseases in Korea: Classification and Related Genetic Variants. Korean J Radiol 2021; 22:1379-1396. [PMID: 34047503 PMCID: PMC8316781 DOI: 10.3348/kjr.2020.1171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/07/2020] [Accepted: 01/23/2021] [Indexed: 01/19/2023] Open
Abstract
Rare neurovascular diseases (RNVDs) have not been well-recognized in Korea. They involve the central nervous system and greatly affect the patients' lives. However, these diseases are difficult to diagnose and treat due to their rarity and incurability. We established a list of RNVDs by referring to the previous literature and databases worldwide to better understand the diseases and their current management status. We categorized 68 RNVDs based on their pathophysiology and clinical manifestations and estimated the prevalence of each disease in Korea. Recent advances in genetic, molecular, and developmental research have enabled further understanding of these RNVDs. Herein, we review each disease, while considering its classification based on updated pathologic mechanisms, and discuss the management status of RNVD in Korea.
Collapse
Affiliation(s)
- Yunsun Song
- Division of Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Boseong Kwon
- Division of Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Abdulrahman Hamed Al-Abdulwahhab
- Department of Diagnostic and Interventional Radiology, Imam Abdulrahman Bin Faisal University, King Fahd Hospital of the University, Al-Khobar City, Eastern Province, Saudi Arabia
| | - Yeo Kyoung Nam
- Division of Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yura Ahn
- Division of Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - So Yeong Jeong
- Division of Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eul Ju Seo
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong Keuk Lee
- Asan Institute of Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dae Chul Suh
- Division of Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
Florian IA, Timiș TL, Ungureanu G, Florian IS, Bălașa A, Berindan-Neagoe I. Deciphering the vascular labyrinth: role of microRNAs and candidate gene SNPs in brain AVM development - literature review. Neurol Res 2020; 42:1043-1054. [PMID: 32723034 DOI: 10.1080/01616412.2020.1796380] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: Brain arteriovenous malformations (AVMs) are a relatively infrequent vascular pathology of unknown etiology that, despite their rarity, cause the highest number of hemorrhagic strokes under the age of 30 years. They pose a challenge to all forms of treatment due to their variable morphology, location, size, and, last but not least, evolving nature. MicroRNAs (miRNAs) are non-coding RNA strands that may suppress the expression of target genes by binding completely or partially to their complementary sequences. Single nucleotide polymorphisms (SNPs), as the name implies, are variations in a single nucleotide in the DNA, usually found in the non-coding segments. Although the majority of SNPs are harmless, some located in the proximity of candidate genes may result in altered expression or function of these genes and cause diseases or affect how different pathologies react to treatment. The roles miRNAs and certain SNPs play in the development and growth of AVMs are currently uncertain, yet progress in deciphering the minutiae of this pathology is already visible. Methods and Results: We performed an electronic Medline (PubMed, PubMed Central) and Google Academic exploration using permutations of the terms: "arteriovenous malformations," "single nucleotide polymorphisms," "microRNA," "non-coding RNA," and "genetic mutations." The findings were then divided into two categories, namely the miRNAs and the candidate gene SNPs associated with AVMs respectively. 6 miRNAs and 12 candidate gene SNPs were identified and discussed. Conclusions: The following literature review focuses on the discoveries made in ascertaining the different implications of miRNAs and candidate gene SNPs in the formation and evolution of brain AVMs, as well as highlighting the possible directions of future research and biological treatment. Abbreviations: ACVRL1/ALK1: activin receptor-like kinase 1; Akt: protein kinase B; ANGPTL4: angiopoietin-like 4; ANRIL: antisense noncoding RNA in the INK4 locus; AVM: arteriovenous malformation; AVM-BEC: arteriovenous malformation brain endothelial cell; BRCA1: breast cancer type 1 susceptibility protein; CCS: case-control study; CDKN2A/B: cyclin-dependent kinase inhibitor 2A/B; CLTC: clathrin heavy chain; DNA: deoxyribonucleic acid; ERK: extracellular signal-regulated kinase; GPR124: probable G-protein coupled receptor 124; GWAS: genome-wide association study; HHT: hereditary hemorrhagic telangiectasia; HIF1A: hypoxia-inducible factor 1A; IA: intracranial aneurysm; ICH: intracranial hemorrhage; Id-1: inhibitor of DNA-binding protein A; IL-17: interleukin 17; MAP4K3: mitogen-activated protein kinase kinase kinase kinase 3; miRNA: microRNA; MMP: matrix metalloproteinase; NFkB: nuclear factor kappa-light-chain of activated B cells; NOTCH: neurogenic locus notch homolog; p38MAPK: p38 mitogen-activated protein kinase; PI3K: phosphoinositide 3-kinase; RBBP8: retinoblastoma-binding protein 8; RNA: ribonucleic acid; SNAI1: Snail Family Transcriptional Repressor 1; SNP: single nucleotide polymorphism; SOX-17: SRY-related HMG-box; TGF-β: transformation growth factor β; TGFR: transformation growth factor receptor; TIMP-4, tissue inhibitor of metalloproteinase 4; TSP-1: thrombospondin-1; UTR: untranslated region; VEGF: Vascular Endothelial Growth Factor; VSMC: vascular smooth muscle cell; Wnt1: Wnt family member 1.
Collapse
Affiliation(s)
- Ioan Alexandru Florian
- Clinic of Neurosurgery, Cluj County Emergency Clinical Hospital , Cluj-Napoca, Romania.,Department of Neurosurgery, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca, Romania
| | - Teodora Larisa Timiș
- Department of Physiology, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca, Romania
| | - Gheorghe Ungureanu
- Clinic of Neurosurgery, Cluj County Emergency Clinical Hospital , Cluj-Napoca, Romania.,Department of Neurosurgery, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca, Romania
| | - Ioan Stefan Florian
- Clinic of Neurosurgery, Cluj County Emergency Clinical Hospital , Cluj-Napoca, Romania.,Department of Neurosurgery, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca, Romania
| | - Adrian Bălașa
- Clinic of Neurosurgery, Tîrgu Mureș County Clinical Emergency Hospital , Tîrgu Mureș, Romania.,Department of Neurosurgery, Tîrgu Mureș University of Medicine, Pharmacy, Science and Technology , Tîrgu Mureș, Romania
| | - Ioana Berindan-Neagoe
- The Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy , Cluj-Napoca, Romania.,Functional Genomics and Experimental Pathology Department, The Oncology Institute "Prof. Dr. Ion Chiricuta" , Cluj-Napoca, Romania
| |
Collapse
|
16
|
Liu Y, Wu X, Nie S, Zhou S, Xiao S, Gao X, Lin Z, Sun J, Huang Y. Methylation of Phospholipase A2 Group VII Gene Is Associated with Brain Arteriovenous Malformations in Han Chinese Populations. J Mol Neurosci 2020; 70:1056-1063. [DOI: 10.1007/s12031-020-01508-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 02/19/2020] [Indexed: 11/29/2022]
|
17
|
Lopez-Rivera V, Sheriff FG, Sandberg DI, Blackburn S, Dannenbaum M, Sheth SA, Day AL, Chen PR. De novo thalamic arteriovenous malformation in a boy with a brainstem cavernous malformation. J Clin Neurosci 2020; 76:226-228. [PMID: 32331948 DOI: 10.1016/j.jocn.2020.04.073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 04/14/2020] [Indexed: 11/16/2022]
Abstract
Brain arteriovenous malformations (bAVMs) have long been considered to be congenital, developing between the third and eighth weeks of embryogenesis. However, cases reporting their de novo formation suggest that these lesions can develop after birth and have challenged this concept. We present a case of a 6-year-old boy with a history of a brainstem cavernous malformation diagnosed after birth who later developed a de novo bAVM. The de novo occurrence of this bAVM distant to the site of the cavernous malformation and a prior negative catheter angiography contributes to the uncertainty of the dynamics and pathophysiology of bAVMs.
Collapse
Affiliation(s)
- Victor Lopez-Rivera
- Departments of Neurology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Faheem G Sheriff
- Departments of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - David I Sandberg
- Departments of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States; Departments of Division of Pediatric Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Spiros Blackburn
- Departments of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Mark Dannenbaum
- Departments of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sunil A Sheth
- Departments of Neurology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Arthur L Day
- Departments of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Peng Roc Chen
- Departments of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, United States.
| |
Collapse
|
18
|
Rozhchenko LV. [Molecular mechanisms of growth and relapse of cerebral arteriovenous malformations]. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2020; 84:94-100. [PMID: 32207748 DOI: 10.17116/neiro20208401194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cerebral AVMs are not static congenital formations, they may grow, recur, and even appear de novo after complete resection, embolization, or radiosurgery. The author analyzes modern literature on the molecular mechanisms of AVM growth. The AVM intranidal vessels are exposed to abnormally high blood flows, which leads to the activation of molecular pathways in endothelial cells, causing proliferation and remodeling of AVM vessels. The existence of cerebral AVM is determined by more than 860 genes, the most important among them are the genetic mutations (SNPs) of VEGF, TGF-β, IL-6, MMP, ANG, ENG. The possible causes of AVM relapse after removal or total embolization are described, as well as the mechanisms of stimulation of angiogenesis after partial embolization: hemodynamic changes in AVM, aseptic inflammation in response to embolizate and the local regional hypoxia inside the AVM. In response to this, growth factors are expressed in the endothelium that further stimulate angiogenesis in AVM. Understanding the complex molecular biology of AVMs is critical to identifying and predicting their behavior, developing new treatments that improve the results of endovascular and surgical treatment.
Collapse
Affiliation(s)
- L V Rozhchenko
- A.L. Polenov Russian Neurosurgical Research Institute - branch of V.A. Almazov National Medical Research Center, St. Petersburg, Russia
| |
Collapse
|
19
|
Prasad K, Kumar P, Misra S, Kumar A, Faruq M, Vivekanandhan S, Srivastava A. The Relationship Between Tumor Necrosis Factor-Alpha (-308G/A, +488G/A, -857C/T, and -1031T/C) Gene Polymorphisms and Risk of Intracerebral Hemorrhage in the North Indian Population: A Hospital-Based Case-Control Study. Neurol India 2020; 68:78-83. [DOI: 10.4103/0028-3886.279665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
20
|
Anbarasen L, Lim J, Rajandram R, Mun KS, Sia SF. Expression of osteopontin, matrix metalloproteinase-2 and -9 proteins in vascular instability in brain arteriovenous malformation. PeerJ 2019; 7:e7058. [PMID: 31275742 PMCID: PMC6596408 DOI: 10.7717/peerj.7058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 05/02/2019] [Indexed: 12/21/2022] Open
Abstract
Background Matrix metalloproteinase (MMP)-2 and -9 are Osteopontin (OPN) dependent molecules implicated in the destabilization of blood vessels. OPN and MMPs have been studied in brain arteriovenous malformation (BAVM) patients’ tissues and blood samples before intervention. In this study, we compared the serum level of these markers before and after treatment, as well as assessed their protein expressions in BAVM tissues to evaluate their roles in this disease. Methodology Serum samples from six BAVM patients and three control subjects were analyzed using enzyme-linked immunoabsorbent assay (ELISA) for OPN. A total of 10 BAVM patients and five control subjects were analyzed using Multiplex ELISA for MMPs. A total of 16 BAVM tissue samples and two normal brain tissue samples were analyzed using immunohistochemistry. Result MMP-2 and -9 were significantly higher in the serum of BAVM patients before and after treatment than in control patients. There were no significant differences of OPN and MMP-9 serum level in BAVM patients before and after treatment. MMP-2 showed a significant elevation after the treatment. Expression of OPN, MMP-2 and -9 proteins were seen in endothelial cells, perivascular cells and brain parenchyma of BAVM tissues. Conclusion Findings revealed that the level of MMP-2 and -9 in the serum correlated well with the expression in BAVM tissues in several cases. Knockdown studies will be required to determine the relationships and mechanisms of action of these markers in the near future. In addition, studies will be required to investigate the expression of these markers’ potential applications as primary medical therapy targets for BAVM patients.
Collapse
Affiliation(s)
- Lalita Anbarasen
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Jasmine Lim
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Retnagowri Rajandram
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Kein Seong Mun
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Sheau Fung Sia
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Persekutuan Kuala Lumpur, Malaysia
| |
Collapse
|
21
|
Intradural spinal cord arteriovenous shunts in the pediatric population: natural history, endovascular management, and follow-up. Childs Nerv Syst 2019; 35:945-955. [PMID: 30843101 DOI: 10.1007/s00381-019-04108-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Intradural spinal cord arteriovenous shunts represent a rare entity, particularly in the pediatric population, and clinical diagnosis can be challenging. METHODS We report the analysis of clinical, angioarchitectural, procedural, and follow-up data in a population of 36 children managed by our team between 2002 and 2017. RESULTS Hemorrhage occurred in 26 children (72%). Age at onset was 9.22 ± 3.65 years. Lesions were located at the thoracic level in 16 cases, at the cervical level in 15 cases, and the thoraco-lumbar region in 5 cases. A genetic or metameric syndrome was associated in 18 children (50%). Glue embolization provided complete occlusion in 5 children, subtotal in 7, and extensive in 14 without intraprocedural complications. We observed clinical normalization in 11 children, improvement in 11 cases, and stability in 3. Four children worsened during the follow-up, and one child died. CONCLUSIONS Endovascular staged glue embolization performed in experienced centers is safe in the treatment of pediatric intradural spinal cord arteriovenous shunts. Clinical and neuroradiological follow-up is mandatory, especially for pediatric patients.
Collapse
|
22
|
Akeret K, Germans M, Sun W, Kulcsar Z, Regli L. Subarachnoid Hemorrhage Due to Flow-Related Dissection of the Posterior-Inferior Cerebellar Artery Associated with a Distal Arteriovenous Malformation. World Neurosurg 2019; 125:44-48. [PMID: 30721771 DOI: 10.1016/j.wneu.2019.01.148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Cerebral arteriovenous malformations (CAVMs) are characterized by altered hemodynamics and associated with angioanatomic changes, such as aneurysms. We encountered a patient with a CAVM-associated dissection of the medial trunk of the posterior inferior cerebellar artery (PICA) instead of an aneurysm. CASE DESCRIPTION We report the case of a 56-year-old male patient with spontaneous subarachnoid hemorrhage within the cisterna magna and fourth ventricle. Digital subtraction angiography revealed a cerebellar arteriovenous malformation located within the inferior semilunar lobule that did not anatomically match the bleeding pattern. The left PICA, serving as a primary feeding artery, showed a dissection of the proximal portion of the medial trunk with a precise anatomic association with the blood in the telovelotonsillar space. CAVM-induced hyperdynamic flow through the feeding vessel is the most plausible pathophysiologic explanation for the dissection. Complete microsurgical resection of the CAVM was performed, and 3-month follow-up digital subtraction angiography showed complete regression of the dissection, disappearance of irregularities, and significant caliber reduction of the medial PICA trunk. CONCLUSIONS This is the first report of a hyperdynamic flow-related dissection of a CAVM-associated feeding vessel. Microsurgical resection of the CAVM allowed for spontaneous resolution of the dissected area by restoration of normal rheodynamics.
Collapse
Affiliation(s)
- Kevin Akeret
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Menno Germans
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Wenhua Sun
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Zsolt Kulcsar
- Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Neuroradiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Luca Regli
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Walcott BP, Winkler EA, Rouleau GA, Lawton MT. Molecular, Cellular, and Genetic Determinants of Sporadic Brain Arteriovenous Malformations. Neurosurgery 2018; 63 Suppl 1:37-42. [PMID: 27399362 DOI: 10.1227/neu.0000000000001300] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Brian P Walcott
- Department of Neurological Surgery and.,Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, California
| | - Ethan A Winkler
- Department of Neurological Surgery and.,Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, California
| | - Guy A Rouleau
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.,Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Michael T Lawton
- Department of Neurological Surgery and.,Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, California
| |
Collapse
|
24
|
Gamboa NT, Joyce EJ, Eli I, Park MS, Taussky P, Schmidt RH, McDonald J, Whitehead KJ, Kalani MYS. Clinical presentation and treatment paradigms of brain arteriovenous malformations in patients with hereditary hemorrhagic telangiectasia. J Clin Neurosci 2018; 51:22-28. [PMID: 29483005 DOI: 10.1016/j.jocn.2018.01.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 01/08/2018] [Indexed: 11/25/2022]
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is characterized by recurrent spontaneous epistaxis, mucocutaneous telangiectases, and multisystem arteriovenous malformations (AVMs). Brain AVMs typically present at birth and are identified in approximately 10-20% of patients with HHT. A retrospective review was undertaken of all HHT patients with known single or multiple brain AVMs treated at our institution. Thirty-nine patients with brain AVM(s) were diagnosed with HHT. Most patients presented with at least one Curaçao criterion. A total of 78 brain AVMs were identified in 39 patients. Two-thirds of patients had solitary brain AVMs, whereas 33% of patients harbored at least two lesions (range: 2-16). Brain AVMs of the supratentorial cerebral hemispheres comprised 83% of all lesions, whereas infratentorial lesions accounted for only 17%. Of the 55 brain AVMs assigned Spetzler-Martin grading, the majority of patients were Grade 1 (73%), and 23% and 4% were Grades 2 and 3, respectively. Patients were treated with surgery alone (51%), embolization alone (6%), embolization followed by surgery (9%), stereotactic radiosurgery (11%), stereotactic radiosurgery followed by surgery (3%), or observation (20%). Of patients who underwent genetic analysis, 62% possessed mutations in ENG (HHT type 1), whereas 38% had mutations in ACVRL1 (HHT type 2). This robust patient cohort of brain AVMs in 39 patients with HHT advances the collective understanding of this disease's varied presentation, diagnostic workup, genetic underpinnings, and available treatment options.
Collapse
Affiliation(s)
- Nicholas T Gamboa
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Evan J Joyce
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Ilyas Eli
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Min S Park
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Philipp Taussky
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Richard H Schmidt
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, UT, United States; University of Utah Hereditary Hemorrhagic Telangiectasia Center of Excellence, Salt Lake City, UT, United States
| | - Jamie McDonald
- University of Utah Hereditary Hemorrhagic Telangiectasia Center of Excellence, Salt Lake City, UT, United States
| | - Kevin J Whitehead
- University of Utah Hereditary Hemorrhagic Telangiectasia Center of Excellence, Salt Lake City, UT, United States
| | - M Yashar S Kalani
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah School of Medicine, Salt Lake City, UT, United States; University of Utah Hereditary Hemorrhagic Telangiectasia Center of Excellence, Salt Lake City, UT, United States.
| |
Collapse
|
25
|
Association of ACVRL1 Genetic Polymorphisms with Arteriovenous Malformations: A Case-Control Study and Meta-Analysis. World Neurosurg 2017; 108:690-697. [DOI: 10.1016/j.wneu.2017.09.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/08/2017] [Accepted: 09/09/2017] [Indexed: 12/26/2022]
|
26
|
Huang J, Song J, Qu M, Wang Y, An Q, Song Y, Yan W, Wang B, Wang X, Zhang S, Chen X, Zhao B, Liu P, Xu T, Zhang Z, Greenberg DA, Wang Y, Gao P, Zhu W, Yang GY. MicroRNA-137 and microRNA-195* inhibit vasculogenesis in brain arteriovenous malformations. Ann Neurol 2017; 82:371-384. [PMID: 28802071 DOI: 10.1002/ana.25015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 08/06/2017] [Accepted: 08/08/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Brain arteriovenous malformations (AVMs) are the most common cause of nontraumatic intracerebral hemorrhage in young adults. The genesis of brain AVM remains enigmatic. We investigated microRNA (miRNA) expression and its contribution to the pathogenesis of brain AVMs. METHODS We used a large-scale miRNA analysis of 16 samples including AVMs, hemangioblastoma, and controls to identify a distinct AVM miRNA signature. AVM smooth muscle cells (AVMSMCs) were isolated and identified by flow cytometry and immunohistochemistry, and candidate miRNAs were then tested in these cells. Migration, tube formation, and CCK-8-induced proliferation assays were used to test the effect of the miRNAs on phenotypic properties of AVMSMCs. A quantitative proteomics approach was used to identify protein expression changes in AVMSMCs treated with miRNA mimics. RESULTS A distinct AVM miRNA signature comprising a large portion of lowly expressed miRNAs was identified. Among these miRNAs, miR-137 and miR-195* levels were significantly decreased in AVMs and constituent AVMSMCs. Experimentally elevating the level of these microRNAs inhibited AVMSMC migration, tube formation, and survival in vitro and the formation of vascular rings in vivo. Proteomics showed the protein expression signature of AVMSMCs and identified downstream proteins regulated by miR-137 and miR-195* that were key signaling proteins involved in vessel development. INTERPRETATION Our results indicate that miR-137 and miR-195* act as vasculogenic suppressors in AVMs by altering phenotypic properties of AVMSMCs, and that the absence of miR-137 and miR-195* expression leads to abnormal vasculogenesis. Ann Neurol 2017;82:371-384.
Collapse
Affiliation(s)
- Jun Huang
- Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jianping Song
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Meijie Qu
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qingzhu An
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Song
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Yan
- Department of Biostatistics, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bingshun Wang
- Institute of Systemic Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojin Wang
- Institute of Systemic Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Song Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xi Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Bing Zhao
- Emergency Department, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peixi Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Tongyi Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | | | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Pingjin Gao
- Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Rujijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
27
|
Delev D, Pavlova A, Grote A, Boström A, Höllig A, Schramm J, Fimmers R, Oldenburg J, Simon M. NOTCH4 gene polymorphisms as potential risk factors for brain arteriovenous malformation development and hemorrhagic presentation. J Neurosurg 2017; 126:1552-1559. [DOI: 10.3171/2016.3.jns151731] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVEArteriovenous malformations (AVMs) of the brain are a frequent and important cause of intracranial hemorrhage in young adults. Little is known about the molecular-genetic pathomechanisms underlying AVM development. Genes of the NOTCH family control the normal development of vessels and proper arteriovenous specification. Transgenic mice with constitutive expression of active NOTCH4 frequently develop AVMs. Here, the authors report a genetic association study investigating possible associations between NOTCH4 gene polymorphisms and formation and clinical presentation of AVMs.METHODSAfter PCR amplification and direct DNA sequencing or restriction digests, 10 single-nucleotide polymorphisms (SNPs) of the NOTCH4 gene were used for genotyping 153 AVM patients and 192 healthy controls (i.e., blood donors). Pertinent clinical data were available for 129 patients. Uni- and multivariate single-marker and explorative haplotype analyses were performed to identify potential genetic risk factors for AVM development and for hemorrhagic or epileptic presentation.RESULTSEleven calculated haplotypes consisting of 3–4 SNPs (most of which were located in the epidermal growth factor–like domain of the NOTCH4 gene) were observed significantly more often among AVM patients than among controls. Univariate analysis indicated that rs443198_TT and rs915895_AA genotypes both were significantly associated with hemorrhage and that an rs1109771_GG genotype was associated with epilepsy. The association between rs443198_TT and AVM bleeding remained significant in the multivariate regression analysis.CONCLUSIONSThe authors' results suggest NOTCH4 SNPs as possible genetic risk factors for the development and clinical presentation of AVMs and a role of NOTCH4 in the pathogenesis of this disease.
Collapse
Affiliation(s)
| | - Anna Pavlova
- 2Institute for Experimental Haematology and Transfusion Medicine, and
| | | | | | - Anke Höllig
- 3Department of Neurosurgery, University Hospital, RWTH Aachen University, Aachen, Germany
| | | | - Rolf Fimmers
- 4Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, University Medical Center, Bonn; and
| | | | | |
Collapse
|
28
|
Wang X, Hao Q, Zhao Y, Guo Y, Ge W. Dysregulation of cell-cell interactions in brain arteriovenous malformations: A quantitative proteomic study. Proteomics Clin Appl 2017; 11. [PMID: 28083997 DOI: 10.1002/prca.201600093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 12/08/2016] [Accepted: 01/11/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Xia Wang
- National Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; Beijing 100005 China
| | - Qiang Hao
- Department of Neurosurgery, Beijing Tiantan Hospital; Capital Medical University; Beijing 100050 China
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital; Capital Medical University; Beijing 100050 China
| | - Yi Guo
- Department of Neurosurgery; Tsinghua Changgung Hospital; Beijing 102218 China
- Department of Neurosurgery; Affiliated Hospital of Hebei University; Baoding 071000 China
| | - Wei Ge
- National Key Laboratory of Medical Molecular Biology & Department of Immunology, Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences; Beijing 100005 China
| |
Collapse
|
29
|
Shah MN, Smith SE, Dierker DL, Herbert JP, Coalson TS, Bruck BS, Zipfel GJ, Van Essen DC, Dacey RG. The relationship of cortical folding and brain arteriovenous malformations. NEUROVASCULAR IMAGING (LONDON) 2016; 2. [PMID: 28009020 PMCID: PMC5167380 DOI: 10.1186/s40809-016-0024-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background The pathogenesis of human intracranial arteriovenous malformations (AVMs) is not well understood; this study aims to quantitatively assess cortical folding in patients with these lesions. Methods Seven adult participants, 4 male and 3 female, with unruptured, surgically unresectable intracranial AVMs were prospectively enrolled in the study, with a mean age of 42.1 years and Spetzler-Martin grade range of II–IV. High-resolution brain MRI T1 and T2 sequences were obtained. After standard preprocessing, segmentation and registration techniques, three measures of cortical folding, the depth difference index (DDI), coordinate distance index (CDI) and gyrification index (GI)), were calculated for the affected and unaffected hemispheres of each subject as well as a healthy control subject set. Results Of the three metrics, CDI, DDI and GI, used for cortical folding assessment, none demonstrated significant differences between the participants and previously studied healthy adults. There was a significant negative correlation between the DDI ratio between affected and unaffected hemispheres and AVM volume (correlation coefficient r = −0.74, p = 0.04). Conclusion This study is the first to quantitatively assess human brain cortical folding in the presence of intracranial AVMs and no significant differences between AVM-affected versus unaffected hemispheres were found in a small dataset. We suggest longitudinal, larger human MRI-based cortical folding studies to assess whether AVMs are congenital lesions of vascular development or de novo, dynamic lesions.
Collapse
Affiliation(s)
- Manish N Shah
- Departments of Pediatric Surgery and Neurosurgery, McGovern Medical School at UT Health and UT MD Anderson Cancer Center, Pediatric Neurosurgery, 6431 Fannin St., MSB 5.144, Houston, TX 77030, USA
| | - Sarah E Smith
- Department of Neuroscience, Washington University, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - Donna L Dierker
- Department of Neuroscience, Washington University, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - Joseph P Herbert
- Division of Neurosurgery, University of Missouri-Columbia, One Hospital Drive, 314 McHaney Hall, Columbia, MO 65212, USA
| | - Timothy S Coalson
- Department of Neuroscience, Washington University, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - Brent S Bruck
- Department of Neurological Surgery, Washington University, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - Gregory J Zipfel
- Department of Neurological Surgery, Washington University, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - David C Van Essen
- Department of Neuroscience, Washington University, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| | - Ralph G Dacey
- Department of Neurological Surgery, Washington University, 660 S. Euclid Ave, St. Louis, MO 63110, USA
| |
Collapse
|
30
|
Nakamura M, Samii A, Lang JM, Götz F, Samii M, Krauss JK. De Novo Arteriovenous Malformation Growth Secondary to Implantation of Genetically Modified Allogeneic Mesenchymal Stem Cells in the Brain. Neurosurgery 2016; 78:E596-600. [PMID: 26382859 DOI: 10.1227/neu.0000000000001025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND IMPORTANCE Local biological drug delivery in the brain is an innovative field of medicine that developed rapidly in recent years. Our report illustrates a unique case of de novo development of a cerebral arteriovenous malformation (AVM) after implantation of genetically modified allogeneic mesenchymal stem cells in the brain. CLINICAL PRESENTATION A 50-year-old man was included in a prospective clinical study (study ID number CM GLP-1/01, 2007-004516-31) investigating a novel neuroprotective approach in stroke patients to prevent perihematomal neuronal damage. In this study, alginate microcapsules containing genetically modified allogeneic mesenchymal stem cells producing the neuroprotective glucagon-like peptide-1 (GLP-1) were implanted. Three years later, the patient presented with aphasia and a focal seizure due to a new left frontal intracerebral hemorrhage. Angiography revealed a de novo left frontal AVM. CONCLUSION The development of an AVM within a period of 3 years after implantation of the glucagon-like peptide-1-secreting mesenchymal stem cells suggests a possible relationship. This case exemplifies that further investigations are necessary to assess the safety of genetically modified cell lines for local biological drug delivery in the brain.
Collapse
Affiliation(s)
- Makoto Nakamura
- *Department of Neurosurgery, Hannover Medical University, Hannover, Germany;‡Department of Neurosurgery, International Neuroscience Institute, Hannover, Germany;§Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical University, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Brain arteriovenous malformations (bAVMs) represent a high risk of intracranial hemorrhages, which are substantial causes of morbidity and mortality of bAVMs, especially in children and young adults. Although a variety of factors leading to hemorrhages of bAVMs are investigated extensively, their pathogenesis is still not well elucidated. The author has reviewed the updated data of genetic aspects of bAVMs, especially focusing on clinical and experimental knowledge from hereditary hemorrhagic telangiectasia, which is the representative genetic disease presenting with bAVMs caused by loss-of-function in one of the two genes: endoglin and activin receptor-like kinase 1. This knowledge may allow us to infer the pathogensis of sporadic bAVMs and in the development of new medical therapies for them.
Collapse
Affiliation(s)
- Masaki Komiyama
- Department of Neuro-Intervention, Osaka City General Hospital
| |
Collapse
|
32
|
Elevated Expression of Matrix Metalloproteinase-9 not Matrix Metalloproteinase-2 Contributes to Progression of Extracranial Arteriovenous Malformation. Sci Rep 2016; 6:24378. [PMID: 27075045 PMCID: PMC4830979 DOI: 10.1038/srep24378] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 03/22/2016] [Indexed: 02/02/2023] Open
Abstract
Extracranial arteriovenous malformations (AVMs) are rare but dangerous congenital lesions arising from direct arterial-venous shunts without intervening capillaries. Progressive infiltration, expansion, and soft tissue destruction lead to bleeding, pain, debilitation and disfigurement. The pathophysiology of AVMs is not well understood. Matrix Metalloproteinases (MMPs) are thought to play an important role in pathologic processes underlying many diseases. This study investigates the expression of MMP-9 and MMP-2 in aggressive extracranial AVMs. The differential expression of MMP-9 and its regulatory factors is also examined. Herein we demonstrate that mRNA and protein expressions of MMP-9, but not MMP-2, are significantly higher in AVM tissues compared to normal tissues. The serum level of MMP-9, but not MMP-2, is also elevated in AVM patients compared to healthy controls. MMP-9/neutrophil gelatinase-associated lipocalin (NGAL) complex is also significantly increased in AVM tissues. The MMP-9/ tissue inhibitor of metalloproteases-1 (TIMP-1) complex presents as a major form detected in normal tissues. The increased and aberrant expression of MMP-9 and specific MMP-9 forms may help explain the constitutive vascular remodeling and infiltrative nature of these lesions. Specific MMP-9 inhibitors would be a promising treatment for AVMs.
Collapse
|
33
|
Kim H, Pawlikowska L, Su H, Young WL. Genetics and Vascular Biology of Angiogenesis and Vascular Malformations. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00012-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Animal Models in Studying Cerebral Arteriovenous Malformation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:178407. [PMID: 26649296 PMCID: PMC4663287 DOI: 10.1155/2015/178407] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/11/2015] [Accepted: 10/25/2015] [Indexed: 12/13/2022]
Abstract
Brain arteriovenous malformation (AVM) is an important cause of hemorrhagic stroke. The etiology is largely unknown and the therapeutics are controversial. A review of AVM-associated animal models may be helpful in order to understand the up-to-date knowledge and promote further research about the disease. We searched PubMed till December 31, 2014, with the term “arteriovenous malformation,” limiting results to animals and English language. Publications that described creations of AVM animal models or investigated AVM-related mechanisms and treatments using these models were reviewed. More than 100 articles fulfilling our inclusion criteria were identified, and from them eight different types of the original models were summarized. The backgrounds and procedures of these models, their applications, and research findings were demonstrated. Animal models are useful in studying the pathogenesis of AVM formation, growth, and rupture, as well as in developing and testing new treatments. Creations of preferable models are expected.
Collapse
|
35
|
Kremer PHC, Koeleman BPC, Pawlikowska L, Weinsheimer S, Bendjilali N, Sidney S, Zaroff JG, Rinkel GJE, van den Berg LH, Ruigrok YM, de Kort GAP, Veldink JH, Kim H, Klijn CJM. Evaluation of genetic risk loci for intracranial aneurysms in sporadic arteriovenous malformations of the brain. J Neurol Neurosurg Psychiatry 2015; 86:524-9. [PMID: 25053769 PMCID: PMC4302044 DOI: 10.1136/jnnp-2013-307276] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 07/01/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND In genome-wide association studies (GWAS) five putative risk loci are associated with intracranial aneurysm. As brain arteriovenous malformations (AVM) and intracranial aneurysms are both intracranial vascular diseases and AVMs often have associated aneurysms, we investigated whether these loci are also associated with sporadic brain AVM. METHODS We included 506 patients (168 Dutch, 338 American) and 1548 controls, all Caucasians. Controls had been recruited as part of previous GWAS. Dutch patients were genotyped by KASPar assay and US patients by Affymetrix SNP 6.0 array. Associations in each cohort were tested by univariable logistic regression modelling, with subgroup analysis in 205 American cases with aneurysm data. Meta-analysis was performed by a Mantel-Haenszel fixed-effect method. RESULTS In the Dutch cohort none of the single nucleotide polymorphisms (SNPs) were associated with AVMs. In the American cohort, genotyped SNPs near SOX-17 (OR 0.74; 95% CI 0.56-0.98), RBBP8 (OR 0.76; 95% CI 0.62-0.94) and an imputed SNP near CDKN2B-AS1 (OR 0.79; 95% CI 0.64-0.98) were significantly associated with AVM. The association with SNPs near SOX-17 and CDKN2B-AS1 but not RBBP8 were strongest in patients with AVM with associated aneurysms. In the meta-analysis we found no significant associations between allele frequencies and AVM occurrence, but rs9298506, near SOX-17 approached statistical significance (OR 0.77; 95% CI 0.57-1.03, p=0.08). CONCLUSIONS Our meta-analysis of two Caucasian cohorts did not show an association between five aneurysm-associated loci and sporadic brain AVM. Possible involvement of SOX-17 and RBBP8, genes involved in cell cycle progression, deserves further investigation.
Collapse
Affiliation(s)
- P H C Kremer
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - B P C Koeleman
- Department of Biomedical Genetics and Complex Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - L Pawlikowska
- Department of Anesthesia, Center for Cerebrovascular Research, University of California-San Francisco, San Francisco, California, USA Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA
| | - S Weinsheimer
- Department of Anesthesia, Center for Cerebrovascular Research, University of California-San Francisco, San Francisco, California, USA
| | - N Bendjilali
- Department of Anesthesia, Center for Cerebrovascular Research, University of California-San Francisco, San Francisco, California, USA
| | - S Sidney
- Division of Research, Kaiser Permanente of Northern California, Oakland, California, USA
| | - J G Zaroff
- Division of Research, Kaiser Permanente of Northern California, Oakland, California, USA
| | - G J E Rinkel
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - L H van den Berg
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Y M Ruigrok
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - G A P de Kort
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J H Veldink
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H Kim
- Department of Anesthesia, Center for Cerebrovascular Research, University of California-San Francisco, San Francisco, California, USA Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - C J M Klijn
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
36
|
|
37
|
Choquet H, Pawlikowska L, Nelson J, McCulloch CE, Akers A, Baca B, Khan Y, Hart B, Morrison L, Kim H. Polymorphisms in inflammatory and immune response genes associated with cerebral cavernous malformation type 1 severity. Cerebrovasc Dis 2014; 38:433-40. [PMID: 25472749 DOI: 10.1159/000369200] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/16/2014] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Familial cerebral cavernous malformation type 1 (CCM1) is an autosomal dominant disease caused by mutations in the Krev Interaction Trapped 1 (KRIT1/CCM1) gene, and characterized by multiple brain lesions that often result in intracerebral hemorrhage (ICH), seizures, and neurological deficits. Carriers of the same genetic mutation can present with variable symptoms and severity of disease, suggesting the influence of modifier factors. Evidence is emerging that inflammation and immune response play a role in the pathogenesis of CCM. The purpose of this study was to investigate whether common variants in inflammatory and immune response genes influence the severity of familial CCM1 disease, as manifested by ICH and greater brain lesion count. METHODS Hispanic CCM1 patients (n=188) harboring the founder Q455X 'common Hispanic mutation' (CHM) in the KRIT1 gene were analyzed at baseline. Participants were enrolled between June 2010 and March 2014 either through the Brain Vascular Malformation Consortium (BVMC) study or through the Angioma Alliance organization. Clinical assessment and cerebral susceptibility-weighted magnetic resonance imaging were performed to determine ICH as well as total and large (≥5 mm in diameter) lesion counts. Samples were genotyped on the Affymetrix Axiom Genome-Wide LAT1 Human Array. We analyzed 830 variants in 56 inflammatory and immune response genes for association with ICH and residuals of log-transformed total or large lesion count adjusted for age at enrollment and gender. Variants were analyzed individually or grouped by sub-pathways or whole pathways. RESULTS At baseline, 30.3% of CCM1-CHM subjects had ICH, with a mean ± standard deviation (SD) of 60.1±115.0 (range 0-713) for total lesions and 4.9±8.7 (range 0-104) for large lesions. The heritability estimates explained by all autosomal variants were 0.20 (SE=0.31), 0.81 (SE=0.17), and 0.48 (SE=0.19), for ICH, total lesion count, and large lesion count, respectively. TGFBR2 rs9823731 was significantly associated with ICH as well as with the total and large lesion counts (p≤0.017). Further, IL-4 rs9327638, CD14 rs778588, IL-6R rs114660934 and MSR1 rs62489577 were associated with two markers of disease severity. Finally, the whole pathway was associated with total lesion count (p=0.005) with TLR-4 rs10759930, CD14 rs778588, IL-6R rs114660934 and IGH rs57767447 mainly bearing this association. Eicosanoid signaling, extracellular pattern recognition, and immune response sub-pathways were also associated with the total lesion count. CONCLUSIONS These results suggest that polymorphisms in inflammatory and immune response pathways contribute to variability in CCM1 disease severity and might be used as predictors of disease severity. In particular, TGFBR2 rs9823731 was associated with all three markers of CCM1 disease severity tested, suggesting that TGFBR2 might be a key participant in the mechanism underlying CCM1 disease severity and phenotype variability. However, further longitudinal studies in larger sample sizes are needed to confirm these findings.
Collapse
Affiliation(s)
- Hélène Choquet
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, Calif., USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Shoemaker LD, Fuentes LF, Santiago SM, Allen BM, Cook DJ, Steinberg GK, Chang SD. Human brain arteriovenous malformations express lymphatic-associated genes. Ann Clin Transl Neurol 2014; 1:982-95. [PMID: 25574473 PMCID: PMC4284124 DOI: 10.1002/acn3.142] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/08/2014] [Accepted: 10/13/2014] [Indexed: 12/19/2022] Open
Abstract
Objective Brain arteriovenous malformations (AVMs) are devastating, hemorrhage-prone, cerebrovascular lesions characterized by well-defined feeding arteries, draining vein(s) and the absence of a capillary bed. The endothelial cells (ECs) that comprise AVMs exhibit a loss of arterial and venous specification. Given the role of the transcription factor COUP-TFII in vascular development, EC specification, and pathological angiogenesis, we examined human AVM tissue to determine if COUP-FTII may have a role in AVM disease biology. Methods We examined 40 human brain AVMs by immunohistochemistry (IHC) and qRT-PCR for the expression of COUP-TFII as well as other genes involved in venous and lymphatic development, maintenance, and signaling. We also examined proliferation and EC tube formation with human umbilical ECs (HUVEC) following COUP-TFII overexpression. Results We report that AVMs expressed COUP-TFII, SOX18, PROX1, NFATC1, FOXC2, TBX1, LYVE1, Podoplanin, and vascular endothelial growth factor (VEGF)-C, contained Ki67-positive cells and heterogeneously expressed genes involved in Hedgehog, Notch, Wnt, and VEGF signaling pathways. Overexpression of COUP-TFII alone in vitro resulted in increased EC proliferation and dilated tubes in an EC tube formation assay in HUVEC. Interpretation This suggests AVM ECs are further losing their arterial/venous specificity and acquiring a partial lymphatic molecular phenotype. There was significant correlation of gene expression with presence of clinical edema and acute hemorrhage. While the precise role of these genes in the formation, stabilization, growth and risk of hemorrhage of AVMs remains unclear, these findings have potentially important implications for patient management and treatment choice, and opens new avenues for future work on AVM disease mechanisms.
Collapse
Affiliation(s)
- Lorelei D Shoemaker
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Laurel F Fuentes
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Shauna M Santiago
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Breanna M Allen
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Douglas J Cook
- Centre for Neuroscience Studies and the Department of Surgery, Queen's University Kingston, Ontario, Canada
| | - Gary K Steinberg
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| | - Steven D Chang
- Department of Neurosurgery, Stanford Neuromolecular Innovation Program, Stanford University 300 Pasteur Drive, Stanford, California, 94305
| |
Collapse
|
39
|
BMP signaling modulation attenuates cerebral arteriovenous malformation formation in a vertebrate model. J Cereb Blood Flow Metab 2014; 34:1688-94. [PMID: 25052553 PMCID: PMC4269730 DOI: 10.1038/jcbfm.2014.134] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 06/02/2014] [Accepted: 06/30/2014] [Indexed: 12/22/2022]
Abstract
Cerebral arteriovenous malformations (AVMs) are vascular anomalies that carry a high risk of stroke and death. To test potential AVM therapies, a reverse genetics approach was used to model AVMs in zebrafish. Antisense morpholino oligonucleotides were used to knockdown activin receptor-like kinase I (alk1), which encodes a transforming growth factor (TGF)-beta family type I receptor implicated in a subset of human AVMs. Knockdown of alk1 caused a spectrum of morphologic, functional, and molecular defects that resemble those seen in humans with AVMs. It was found that losartan, an angiotensin II receptor antagonist, attenuated abnormal blood vessel morphology and systemic manifestations of high-output arteriovenous shunting in vivo. SMAD1 phosphorylation was significantly decreased in alk1 morphants compared with uninjected organisms (0.189±0.0201, 0.429±0.0164, P=0.0002). After treatment, morphant SMAD1 levels approached uninjected levels (0.326±0.0360, P=0.0355) and were significantly higher than those seen in the morphant-control group (P=0.0294). These data suggest that modulating the BMP signaling pathway with losartan, a drug in widespread clinical use in humans as an antihypertensive, may have the potential to be further evaluated as a therapeutic strategy for patients with AVMs.
Collapse
|
40
|
Morales-Valero SF, Bortolotti C, Sturiale C, Lanzino G. Are parenchymal AVMs congenital lesions? Neurosurg Focus 2014; 37:E2. [DOI: 10.3171/2014.6.focus14234] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A long-held dogma in neurosurgery is that parenchymal arteriovenous malformations (AVMs) are congenital. However, there is no strong evidence supporting this theory. An increasing number of documented cases of de novo formation of parenchymal AVMs cast doubt on their congenital nature and suggest that indeed the majority of these lesions may form after birth. Further evidence suggesting the postnatal development of parenchymal AVMs comes from the exceedingly rare diagnosis of these lesions in utero despite the widespread availability of high-resolution imaging modalities such as ultrasound and fetal MRI. The exact mechanism of AVM formation has yet to be elucidated, but most likely involves genetic susceptibility and environmental triggering factors. In this review, the authors report 2 cases of de novo AVM formation and analyze the evidence suggesting that they represent an acquired condition.
Collapse
Affiliation(s)
- Saul F. Morales-Valero
- 1Department of Neurologic Surgery, Mayo Clinic, Mayo Medical School, Rochester, Minnesota; and
| | - Carlo Bortolotti
- 2Department of Neurosurgery, IRCCS Institute of Neurological Sciences, Bellaria Hospital, Bologna, Italy
| | - Carmelo Sturiale
- 2Department of Neurosurgery, IRCCS Institute of Neurological Sciences, Bellaria Hospital, Bologna, Italy
| | - Giuseppe Lanzino
- 1Department of Neurologic Surgery, Mayo Clinic, Mayo Medical School, Rochester, Minnesota; and
| |
Collapse
|
41
|
Shen F, Degos V, Chu PL, Han Z, Westbroek EM, Choi EJ, Marchuk D, Kim H, Lawton MT, Maze M, Young WL, Su H. Endoglin deficiency impairs stroke recovery. Stroke 2014; 45:2101-6. [PMID: 24876084 DOI: 10.1161/strokeaha.114.005115] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE Endoglin deficiency causes hereditary hemorrhagic telangiectasia-1 and impairs myocardial repair. Pulmonary arteriovenous malformations in patients with hereditary hemorrhagic telangiectasia-1 are associated with a high incidence of paradoxical embolism in the cerebral circulation and ischemic brain injury. We hypothesized that endoglin deficiency impairs stroke recovery. METHODS Eng heterozygous (Eng+/-) and wild-type mice underwent permanent distal middle cerebral artery occlusion (pMCAO). Pial collateral vessels were quantified before pMCAO. Infarct/atrophic volume, vascular density, and macrophages were quantified in various days after pMCAO, and behavioral function was assessed using corner and adhesive removal tests on days 3, 15, 30, and 60 after pMCAO. The association between ENG 207G>A polymorphism and brain arteriovenous malformation rupture and surgery outcome was analyzed using logistic regression analysis in 256 ruptured and 157 unruptured patients. RESULTS After pMCAO, Eng+/- mice showed larger infarct/atrophic volumes at all time points (P<0.05) and showed worse behavior performance (P<0.05) at 15, 30, and 60 days when compared with wild-type mice. Eng+/- mice had fewer macrophages on day 3 (P=0.009) and more macrophages on day 60 (P=0.02) in the peri-infarct region. Although Eng+/- and wild-type mice had similar numbers of pial collateral vessels before pMCAO, Eng+/- mice had lower vascular density in the peri-infarct region (P=0.05) on day 60 after pMCAO. In humans, ENG 207A allele has been associated with worse outcomes after arteriovenous malformation rupture or surgery of patients with unruptured arteriovenous malformation. CONCLUSIONS Endoglin deficiency impairs brain injury recovery. Reduced angiogenesis, impaired macrophage homing, and delayed inflammation resolution could be the underlying mechanism.
Collapse
MESH Headings
- Alleles
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Behavior, Animal/physiology
- Disease Models, Animal
- Endoglin
- Humans
- Infarction, Middle Cerebral Artery/etiology
- Infarction, Middle Cerebral Artery/metabolism
- Intracellular Signaling Peptides and Proteins/deficiency
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Intracranial Arteriovenous Malformations/genetics
- Intracranial Arteriovenous Malformations/metabolism
- Intracranial Arteriovenous Malformations/surgery
- Mice
- Mice, Knockout
- Polymorphism, Genetic/genetics
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Recovery of Function/genetics
- Recovery of Function/physiology
- Time Factors
Collapse
Affiliation(s)
- Fanxia Shen
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Vincent Degos
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Pei-Lun Chu
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Zhenying Han
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Erick M Westbroek
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Eun-Jung Choi
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Douglas Marchuk
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Helen Kim
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Michael T Lawton
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Mervyn Maze
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - William L Young
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.)
| | - Hua Su
- From the Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research (F.S., V.D., Z.H., E.M.W., E.-J.C., H.K., M.M., W.L.Y., H.S.) and Departments of Neurological Surgery (M.T.L., W.L.Y.) and Neurology (W.L.Y.), University of California, San Francisco; Department of Neurology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (F.S.); Department of Anesthesia and Intensive Care, INSERM, U676, Hôpital Robert Debré, Paris, France (V.D.); and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC (P.-L.C., D.M.).
| |
Collapse
|
42
|
Developmental and pathological angiogenesis in the central nervous system. Cell Mol Life Sci 2014; 71:3489-506. [PMID: 24760128 DOI: 10.1007/s00018-014-1625-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 01/24/2023]
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing vessels, in the central nervous system (CNS) is seen both as a normal physiological response as well as a pathological step in disease progression. Formation of the blood-brain barrier (BBB) is an essential step in physiological CNS angiogenesis. The BBB is regulated by a neurovascular unit (NVU) consisting of endothelial and perivascular cells as well as vascular astrocytes. The NVU plays a critical role in preventing entry of neurotoxic substances and regulation of blood flow in the CNS. In recent years, research on numerous acquired and hereditary disorders of the CNS has increasingly emphasized the role of angiogenesis in disease pathophysiology. Here, we discuss molecular mechanisms of CNS angiogenesis during embryogenesis as well as various pathological states including brain tumor formation, ischemic stroke, arteriovenous malformations, and neurodegenerative diseases.
Collapse
|
43
|
Brain arteriovenous malformation modeling, pathogenesis, and novel therapeutic targets. Transl Stroke Res 2014; 5:316-29. [PMID: 24723256 DOI: 10.1007/s12975-014-0343-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 03/24/2014] [Accepted: 03/25/2014] [Indexed: 02/07/2023]
Abstract
Patients harboring brain arteriovenous malformation (bAVM) are at life-threatening risk of rupture and intracranial hemorrhage (ICH). The pathogenesis of bAVM has not been completely understood. Current treatment options are invasive, and ≈ 20 % of patients are not offered interventional therapy because of excessive treatment risk. There are no specific medical therapies to treat bAVMs. The lack of validated animal models has been an obstacle for testing hypotheses of bAVM pathogenesis and testing new therapies. In this review, we summarize bAVM model development and bAVM pathogenesis and potential therapeutic targets that have been identified during model development.
Collapse
|
44
|
Walcott BP, Peterson RT. Zebrafish models of cerebrovascular disease. J Cereb Blood Flow Metab 2014; 34:571-7. [PMID: 24517974 PMCID: PMC3982096 DOI: 10.1038/jcbfm.2014.27] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 12/27/2013] [Accepted: 01/07/2014] [Indexed: 12/18/2022]
Abstract
Perturbations in cerebral blood flow and abnormalities in blood vessel structure are the hallmarks of cerebrovascular disease. While there are many genetic and environmental factors that affect these entities through a heterogeneous group of disease processes, the ultimate final pathologic insult in humans is defined as a stroke, or damage to brain parenchyma. In the case of ischemic stroke, blood fails to reach its target destination whereas in hemorrhagic stroke, extravasation of blood occurs outside of the blood vessel lumen, resulting in direct damage to brain parenchyma. As these acute events can be neurologically devastating, if not fatal, development of novel therapeutics are urgently needed. The zebrafish (Danio rerio) is an attractive model for the study of cerebrovascular disease because of its morphological and physiological similarity to human cerebral vasculature, its ability to be genetically manipulated, and its fecundity allowing for large-scale, phenotype-based screens.
Collapse
Affiliation(s)
- Brian P Walcott
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Randall T Peterson
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
45
|
Navarrete MG, Hernández AD, Collado-Ortiz MA, Salinas-Lara C, Tena-Suck ML. Brain vascular lesions: a clinicopathologic, immunohistochemistry, and ultrastructural approach. Ann Diagn Pathol 2014; 18:193-8. [PMID: 24881784 DOI: 10.1016/j.anndiagpath.2014.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 01/09/2014] [Indexed: 11/19/2022]
Abstract
Brain vascular malformations are relatively common lesions that cause serious neurologic disability or death in a significant proportion of individuals bearing them. The purpose of this study was to analyze the clinicopathologic and immunohistochemistry these lesions, looking for common antibodies expressed such as CD31, CD34, CD15, factor VIII, nestin, vimentin, vascular endothelial grow factor (VEGF), vascular endothelial grow factor receptor-2 (VEGF-R2), glial fibrillar acidic protien (GFAP), and fibroblastic grow factor β (β-FGF) and ultrastructure in endothelial cells as well as in vessel walls. Fifty cases of vascular lesions were included in this study: 29 (58%) of them were arteriovenous malformations and 21 (52%) were brain cavernomas. Twenty-six (52%) patients were women and 24 (48%) men. The age range was from 13 to 68 years (mean age, 35.86 ± 15.19 years). The size of the lesions ranged between 1 and 8 cm (3 ± 1.65 cm), and parieto-occipital lesions had a bigger size. Evolution time varied from 1 month to 1 year (mean, 7.5 months). There was a significant statistical correlation between age and sex (P = -035), rupture of lesion (P = .015), brain hemorrhage (P = .033), necrosis (P = .011), hemosiderin deposit (P = .042), VEGF (P = .015), and VEGFR (P = .037), as well as localization of rupture (P = .017), loss of consciousness (P = .000), visual deficit (P = .026), hyaline vessels (P = .000), and CD31 (.009). Interactions between endothelial cells and mural cells (pericytes and vascular smooth muscle cells) in blood vessel walls have recently come into focus as central processes in the regulation of vascular formation, stabilization, remodeling, and function in brain vascular lesions. However, the molecular mechanisms that underlie the formation and growth of brain arteriovenous malformations are still poorly understood.
Collapse
Affiliation(s)
- Marisol Galván Navarrete
- General Hospital of Guadalajara Hospital civil antiguo de Guadalajara Jalisco, Guadalajara, Mexico
| | - Alma Dalia Hernández
- Laboratory of Pathology, National Institute of Rehabilitation, México City, Mexico
| | - Miguel Angel Collado-Ortiz
- Neurological Center, Department of Clinical Neurophysiology, The American British Cowdray Hospital, Mexico City, Mexico.
| | - Citlaltepetl Salinas-Lara
- Department of Neuropathology, National Institute of Neurology and Neurosurgery, México City, Mexico.
| | - Martha Lilia Tena-Suck
- Department of Neuropathology, National Institute of Neurology and Neurosurgery, México City, Mexico.
| |
Collapse
|
46
|
Choi EJ, Chen W, Jun K, Arthur HM, Young WL, Su H. Novel brain arteriovenous malformation mouse models for type 1 hereditary hemorrhagic telangiectasia. PLoS One 2014; 9:e88511. [PMID: 24520391 PMCID: PMC3919779 DOI: 10.1371/journal.pone.0088511] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 01/08/2014] [Indexed: 12/24/2022] Open
Abstract
Endoglin (ENG) is a causative gene of type 1 hereditary hemorrhagic telangiectasia (HHT1). HHT1 patients have a higher prevalence of brain arteriovenous malformation (AVM) than the general population and patients with other HHT subtypes. The pathogenesis of brain AVM in HHT1 patients is currently unknown and no specific medical therapy is available to treat patients. Proper animal models are crucial for identifying the underlying mechanisms for brain AVM development and for testing new therapies. However, creating HHT1 brain AVM models has been quite challenging because of difficulties related to deleting Eng-floxed sequence in Eng2fl/2fl mice. To create an HHT1 brain AVM mouse model, we used several Cre transgenic mouse lines to delete Eng in different cell-types in Eng2fl/2fl mice: R26CreER (all cell types after tamoxifen treatment), SM22α-Cre (smooth muscle and endothelial cell) and LysM-Cre (lysozyme M-positive macrophage). An adeno-associated viral vector expressing vascular endothelial growth factor (AAV-VEGF) was injected into the brain to induce focal angiogenesis. We found that SM22α-Cre-mediated Eng deletion in the embryo caused AVMs in the postnatal brain, spinal cord, and intestines. Induction of Eng deletion in adult mice using R26CreER plus local VEGF stimulation induced the brain AVM phenotype. In both models, Eng-null endothelial cells were detected in the brain AVM lesions, and formed mosaicism with wildtype endothelial cells. However, LysM-Cre-mediated Eng deletion in the embryo did not cause AVM in the postnatal brain even after VEGF stimulation. In this study, we report two novel HHT1 brain AVM models that mimic many phenotypes of human brain AVM and can thus be used for studying brain AVM pathogenesis and testing new therapies. Further, our data indicate that macrophage Eng deletion is insufficient and that endothelial Eng homozygous deletion is required for HHT1 brain AVM development.
Collapse
Affiliation(s)
- Eun-Jung Choi
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - Wanqiu Chen
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - Kristine Jun
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - Helen M. Arthur
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Newcastle, United Kingdom
| | - William L. Young
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
Abstract
Brain arteriovenous malformations (bAVM) are tangles of abnormal, dilated vessels that directly shunt blood between the arteries and veins. The pathogenesis of bAVM is currently unknown. Patients with hereditary hemorrhagic telangiectasia (HHT) have a higher prevalence of bAVM than the general population. Animal models are important tools for dissecting the disease etiopathogenesis and for testing new therapies. Here, we introduce a method that induces the bAVM phenotype through regional deletion of activin-like kinase 1 (Alk1, the causal gene for HHT2) and vascular endothelial growth factor (VEGF) stimulation.
Collapse
Affiliation(s)
- Wanqiu Chen
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
48
|
Guo Y, Tihan T, Kim H, Hess C, Lawton MT, Young WL, Zhao Y, Su H. Distinctive distribution of lymphocytes in unruptured and previously untreated brain arteriovenous malformation. ACTA ACUST UNITED AC 2014; 1:147-152. [PMID: 25568888 DOI: 10.4103/2347-8659.143674] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIM To test the hypothesis that lymphocyte infiltration in brain arteriovenous malformation (bAVM) is not associated with iron deposition (indicator of microhemorrhage). METHODS Sections of unruptured, previously untreated bAVM specimens (n=19) were stained immunohistochemically for T-lymphocytes (CD3+), B-lymphocytes (CD20+), plasma cells (CD138+) and macrophages (CD68+). Iron deposition was assessed by hematoxylin and eosin and Prussian blue stains. Superficial temporal arteries (STA) were used as control. RESULTS Both T lymphocytes and macrophages were present in unruptured, previously untreated bAVM specimens, whereas few B cells and plasma cells were detected. Iron deposition was detected in 8 specimens (42%; 95% confidence interval =20-67%). The samples with iron deposition tended to have more macrophages than those without (666±313 vs 478±174 cells/mm2; P=0.11). T-cells were clustered on the luminal side of the endothelial surface, on the vessel-wall, and in the perivascular regions. There was no correlation between T lymphocyte load and iron deposition (P=0.88). No macrophages and lymphocytes were detected in STA controls. CONCLUSIONS T-lymphocytes were present in bAVM specimens. Unlike macrophages, the load and location of T-lymphocytes were not associated with iron deposition, suggesting the possibility of an independent cell-mediated immunological mechanism in bAVM pathogenesis.
Collapse
Affiliation(s)
- Yi Guo
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA ; Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 071000 China
| | - Tarik Tihan
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Helen Kim
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Christopher Hess
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Michael T Lawton
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94110, USA
| | - William L Young
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA ; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94110, USA ; Department of Neurology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 410011, China
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
49
|
The ALK-1/Smad1 pathway in cardiovascular physiopathology. A new target for therapy? Biochim Biophys Acta Mol Basis Dis 2013; 1832:1492-510. [PMID: 23707512 DOI: 10.1016/j.bbadis.2013.05.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/04/2013] [Accepted: 05/13/2013] [Indexed: 01/04/2023]
Abstract
Activin receptor-like kinase-1 or ALK-1 is a type I cell surface receptor for the transforming growth factor-β (TGF-β) family of proteins. The role of ALK-1 in endothelial cells biology and in angiogenesis has been thoroughly studied by many authors. However, it has been recently suggested a possible role of ALK-1 in cardiovascular homeostasis. ALK-1 is not only expressed in endothelial cells but also in smooth muscle cells, myofibroblast, hepatic stellate cells, chondrocytes, monocytes, myoblasts, macrophages or fibroblasts, but its role in these cells have not been deeply analyzed. Due to the function of ALK-1 in these cells, this receptor plays a role in several cardiovascular diseases. Animals with ALK-1 haploinsufficiency and patients with mutations in Acvrl1 (the gene that codifies for ALK-1) develop type-2 Hereditary Hemorrhagic Telangiectasia. Moreover, ALK-1 heterozygous mice develop pulmonary hypertension. Higher levels of ALK-1 have been observed in atherosclerotic plaques, suggesting a possible protector role of this receptor. ALK-1 deficiency is also related to the development of arteriovenous malformations (AVMs). Besides, due to the ability of ALK-1 to regulate cell proliferation and migration, and to modulate extracellular matrix (ECM) protein expression in several cell types, ALK-1 has been now demonstrated to play an important role in cardiovascular remodeling. In this review, we would like to offer a complete vision of the role of ALK-1 in many process related to cardiovascular homeostasis, and the involvement of this protein in the development of cardiovascular diseases, suggesting the possibility of using the ALK-1/smad-1 pathway as a powerful therapeutic target.
Collapse
|
50
|
Allelic variation of the MMP3 promoter affects transcription activity through the transcription factor C-MYB in human brain arteriovenous malformations. PLoS One 2013; 8:e57958. [PMID: 23483952 PMCID: PMC3587415 DOI: 10.1371/journal.pone.0057958] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 01/29/2013] [Indexed: 01/29/2023] Open
Abstract
MMPs comprise a family of proteolytic enzymes that degrade pericellular substances, which may result in the destabilization of vessels and related to the development of brain arteriovenous malformations (BAVM). MMP3 is a key member of this family, overexpressed in BAVM tissues, and a single nucleotide polymorphism within MMP3, −709A>G (rs522616), is significantly associated with the risk of BAVM. In this study, we aimed to investigate the mechanism through which the polymorphism rs522616 regulates the expression of MMP3. Our results showed that −709A led to a over 2-fold higher transcriptional activity compared with the G allele (P<0.05) and this transcriptional activity can be depressed by co-transfecting cells with competitive DNA fragments containing −709A but not −709G. Bioinformatics analyses suggested that the transcription factor C-MYB might bind to the area around rs522616. Overexpressed C-MYB significantly increased the transcriptional activity of −709A compared with −709G or controls that did not overexpress c-myb (P<0.01) in HEK293 and HUVEC cells. ChIP assays indicated that C-MYB bound to the SNP region in the two cell lines and three BAVM tissue samples. Together, these data indicated that C-MYB can bind to the −709A allele of the MMP3 promoter, activate its transcription and lead to a higher expression of this gene. This novel hypothesis, supported by molecular evidence, explains how this SNP affects MMP3 promoter function and results in a risk of BAVM development.
Collapse
|