1
|
da Fontoura Galvão G, Verly G, Valença P, Domingues FS, da Silva MR, Marcondes J. Early and long-term outcome of surgical versus conservative management for intracranial cerebral cavernous malformation: Meta-analysis of reconstructed time-to-event data. Clin Neurol Neurosurg 2024; 246:108567. [PMID: 39332049 DOI: 10.1016/j.clineuro.2024.108567] [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: 07/23/2024] [Revised: 08/28/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Cerebral cavernous malformations (CCMs) present challenges in clinical management due to a lack of definitive evidence from clinical trials. Surgical intervention and observational management are commonly used, yet their efficacy and long-term outcomes remain controversial. OBJECTIVE This meta-analysis evaluates the effectiveness of surgical intervention versus conservative management in patients with symptomatic CCMs over various time frames to determine optimal treatment strategies. METHODS A systematic review and reconstructed time-to-event meta-analysis were conducted, following PRISMA guidelines. Data from selected studies comparing surgical intervention to conservative management for CCMs were analyzed using pooled patient data from Kaplan-Meier curves. New focal neurological deficit (FND) or intracranial hemorrhage (ICH) were the outcome metrics. RESULTS Four eligible studies, comprising 290 patients, were included. Surgical intervention showed 43 events over a mean time to FND/ICH of 6.372 years (95 % CI: 3.536-8.005), while observational management had 48 events with a significantly longer mean time of 10.992 years (95 % CI: 6.070-8.005). No significant difference was found at 2 years (p = 0.910), but at 5 and 10 years, surgical intervention had more events and shorter mean times (p < 0.0001). Sensitivity analysis for previously bleeding CCMs showed no significant difference in events (p = 0.131). CONCLUSION This meta-analysis suggests observational management may achieve favorable long-term outcomes for symptomatic CCMs. Despite ongoing controversies, the findings highlight the need for further research, particularly randomized controlled trials, to refine treatment strategies and optimize patient care.
Collapse
Affiliation(s)
- Gustavo da Fontoura Galvão
- Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Department of Neurosurgery, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil.
| | - Gabriel Verly
- Medical School Graduate, Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Department of Neurosurgery, Rio de Janeiro, RJ, Brazil.
| | - Pablo Valença
- Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil.
| | - Flávio Sampaio Domingues
- Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Department of Neurosurgery, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil.
| | - Marcello Reis da Silva
- Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Department of Neurosurgery, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil.
| | - Jorge Marcondes
- Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Department of Neurosurgery, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
2
|
van de Goor A, Schuermans VNE, Postma AA, Boselie TFM. In toto resection of a rare intradural extramedullary cavernous malformation on the S1 nerve root: illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2024; 8:CASE24270. [PMID: 39467311 DOI: 10.3171/case24270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/26/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Cavernous malformations (CMs) are vascular malformations that usually occur in the brain and occasionally in the spine. Most CMs are sporadic, but they can also be caused by heterozygous cerebral cavernous malformation (CCM) gene mutations. This case describes an intradural extramedullary CM in a unique location, within a fascicle of the S1 nerve root. This tumor was found to have a CCM2 mutation. An operative video demonstrates the surgical procedure used to remove this S1 intradural extramedullary CM. OBSERVATIONS A 61-year-old male with an intradural extramedullary CM, located within a fascicle of the S1 nerve root, presented with pain and neurological symptoms. Additional imaging revealed two CCMs. The S1 CM was removed in toto. Subsequently, genetic screening revealed a CCM2 mutation. LESSONS Given the rarity of a radicular CM, diagnosis can be challenging. CMs should be considered in patients with radicular pain and corresponding nerve root thickening. When a CM is suspected, imaging of the complete neuraxis should be performed. If multiple CMs are found, genetic screening is indicated. Complete resection of the CM yields good clinical results. The operative video in this case might help surgeons encountering this rare pathology to prepare for surgery. https://thejns.org/doi/10.3171/CASE24270.
Collapse
Affiliation(s)
- Ank van de Goor
- Departments of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
- Departments of Maastricht University, Maastricht, The Netherlands
| | - Valérie N E Schuermans
- Departments of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Neurosurgery, Zuyderland Medical Center, Heerlen, The Netherlands
- CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Alida A Postma
- Departments of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Sciences, Maastricht University, Maastricht, The Netherlands
| | - Toon F M Boselie
- Departments of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Neurosurgery, Zuyderland Medical Center, Heerlen, The Netherlands
- CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
3
|
Shapeti A, Barrasa-Fano J, Abdel Fattah AR, de Jong J, Sanz-Herrera JA, Pezet M, Assou S, de Vet E, Elahi SA, Ranga A, Faurobert E, Van Oosterwyck H. Force-mediated recruitment and reprogramming of healthy endothelial cells drive vascular lesion growth. Nat Commun 2024; 15:8660. [PMID: 39370485 PMCID: PMC11456588 DOI: 10.1038/s41467-024-52866-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
Force-driven cellular interactions are crucial for cancer cell invasion but remain underexplored in vascular abnormalities. Cerebral cavernous malformations (CCM), a vascular abnormality characterized by leaky vessels, involves CCM mutant cells recruiting wild-type endothelial cells to form and expand mosaic lesions. The mechanisms behind this recruitment remain poorly understood. Here, we use an in-vitro model of angiogenic invasion with traction force microscopy to reveal that hyper-angiogenic Ccm2-silenced endothelial cells enhance angiogenic invasion of neighboring wild-type cells through force and extracellular matrix-guided mechanisms. We demonstrate that mechanically hyperactive CCM2-silenced tips guide wild-type cells by transmitting pulling forces and by creating paths in the matrix, in a ROCKs-dependent manner. This is associated with reinforcement of β1 integrin and actin cytoskeleton in wild-type cells. Further, wild-type cells are reprogrammed into stalk cells and activate matrisome and DNA replication programs, thereby initiating proliferation. Our findings reveal how CCM2 mutants hijack wild-type cell functions to fuel lesion growth, providing insight into the etiology of vascular malformations. By integrating biophysical and molecular techniques, we offer tools for studying cell mechanics in tissue heterogeneity and disease progression.
Collapse
Affiliation(s)
- Apeksha Shapeti
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium.
| | - Jorge Barrasa-Fano
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
| | - Abdel Rahman Abdel Fattah
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
- CeMM The Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Janne de Jong
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
| | - José Antonio Sanz-Herrera
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBIS), Seville, Spain
| | - Mylène Pezet
- Univ. Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Said Assou
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Emilie de Vet
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
| | - Seyed Ali Elahi
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
- KU Leuven, Department of Movement Sciences, Human Movement Biomechanics Research Group, Leuven, Belgium
| | - Adrian Ranga
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
| | - Eva Faurobert
- Univ. Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, Grenoble, France.
| | - Hans Van Oosterwyck
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium.
- KU Leuven, Prometheus, Division of Skeletal Tissue Engineering, Leuven, Belgium.
| |
Collapse
|
4
|
Galvão GDF, Neumann VB, Verly G, Valença P, Cunha AM, da Silva MR, Domingues FS, de Souza JM. Clinical features, hemorrhage risk and epilepsy outcomes of familial cerebral cavernous malformation: A 20-year observational pragmatic single-center study. J Stroke Cerebrovasc Dis 2024; 33:108041. [PMID: 39332546 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024] Open
Abstract
INTRODUCTION Familial Cerebral Cavernous Malformations (fCCMs) are rare, hereditary conditions characterized by multiple central nervous system lesions. Despite their rarity, CCMs can cause significant clinical challenges when symptomatic, manifesting as seizure and symptomatic hemorrhage (CASH). Guidelines suggest neurosurgical intervention for symptomatic or previously symptomatic lesions, while conservative management is recommended for new-onset epilepsy. However, the natural history and optimal management remain unclear, necessitating further research. OBJECTIVE This study aims to provide a comprehensive analysis of the clinical features, hemorrhage risk, and epilepsy outcomes in fCCM patients over an extended follow-up period, offering a more precise estimate of CASH and epilepsy rates in this population. METHODS This retrospective longitudinal cohort study included fCCM patients enrolled from 2001 to May 2024. Data collected included demographic information, new neurological symptoms, symptomatic hemorrhages, seizures, and modified Rankin Scale (mRS) scores. Incidence rates of first symptomatic events and Kaplan-Meier survival curves were calculated, with logistic and Cox-proportional hazard regression models used to evaluate outcomes. RESULTS A total of 47 patients were included in this study, with a mean age at diagnosis of 37.51 years. At diagnosis, 68 % were symptomatic, with 30 % having CASH and 36 % experiencing seizures without CASH. During a median follow-up of 126.0 months (interquartile range, 110.5 months), 17 % had a new CASH event, 20 % had seizures without CASH, and 60 % remained asymptomatic. The bleeding rate was 1.02 % per patient-year, with new focal neurological symptoms at 2.045 per 1000 patient-years and new CASH at 10.225 per 1000 patient-years. Most patients maintained minimal or no disability (mRS 0 or 1). Presenting with epilepsy at baseline significantly increased the odds of future seizures (OR 18.13, p = 0.001). CONCLUSION This study highlights the complex presentation and progression of fCCMs, emphasizing the necessity for long-term monitoring. Baseline epilepsy is a significant predictor of future seizures, underscoring the need for individualized management strategies. Future research with larger cohorts and standardized criteria is essential to refine the understanding and management of fCCMs.
Collapse
Affiliation(s)
- Gustavo da Fontoura Galvão
- Division of Neurosurgery, Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil.
| | - Vinicius Barbosa Neumann
- Division of Neurosurgery, Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Rio de Janeiro, RJ, Brazil
| | - Gabriel Verly
- Division of Neurosurgery, Medical School Graduate, Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Rio de Janeiro, RJ, Brazil
| | - Pablo Valença
- Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil
| | - Alexandre Martins Cunha
- Division of Neurosurgery, Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Rio de Janeiro, RJ, Brazil
| | - Marcello Reis da Silva
- Division of Neurosurgery, Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil
| | - Flavio Sampaio Domingues
- Division of Neurosurgery, Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil
| | - Jorge Marcondes de Souza
- Division of Neurosurgery, Federal University of Rio de Janeiro, University Hospital Clementino Fraga Filho, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Neurosurgery Center, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
5
|
Galvão GF, Trefilio LM, Salvio AL, da Silva EV, Alves-Leon SV, Fontes-Dantas FL, de Souza JM. Genetic variants in FCGR2A, PTPN2, VDR as predictive signatures of aggressive phenotypes in cerebral cavernous malformation. Gene 2024; 933:148918. [PMID: 39236970 DOI: 10.1016/j.gene.2024.148918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVE The biological behavior of Cerebral Cavernous Malformation (CCM) is still controversial, lacking a clear-cut signature for a mechanistic explanation of lesion aggressiveness. In this study, we evaluated the predictive capacity of genetic variants concerning the aggressive behavior of CCM and their implications in biological processes. METHODS We genotyped the variants in VDRrs7975232, VDRrs731236, VDRrs11568820, PTPN2rs72872125 and FCGR2Ars1801274 genes using TaqMan Genotyping Assays in a cohort study with 103 patients, 42 of whom had close follow-up visits for 4 years, focusing on 2 main aspects of the disease: (1) symptomatic events, which included both intracranial bleeding or epilepsy, and (2) the onset of symptoms. The genotypes were correlated with the levels of several cytokines quantified in peripheral blood, measured using the x-MAP method. RESULTS We report a novel observation that the PTPN2rs72872125 CT and the VDRrs7975232 CC genotype were independently associated with an asymptomatic phenotype. Additionally, PTPN2rs72872125 CC genotype and serum level of GM-CSF could predict a diagnostic association with symptomatic phenotype in CCM patients, while the FCGR2Ars1801274 GG genotype could predict a symptomatic event during follow-up. The study also found a correlation between VDRrs731236 AA and VDRrs11568820 CC genotype to the time to the first symptomatic event. CONCLUSIONS These genetic markers could pave the way for precision medicine strategies for CCM, enhancing patient outcomes by enabling customized therapeutic approaches.
Collapse
Affiliation(s)
- Gustavo F Galvão
- Laboratório de Neurociências Translacional, Programa de Pós-Graduação em Neurologia, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, 20211-030, Brazil; Departamento de Neurocirurgia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 3938-2480, Brazil
| | - Luisa M Trefilio
- Laboratório de Neurociências Translacional, Programa de Pós-Graduação em Neurologia, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, 20211-030, Brazil; Laboratório de Neurofarmacogenetica, Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcântara Gomes, Universidade Estadual do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Andreza L Salvio
- Laboratório de Neurociências Translacional, Programa de Pós-Graduação em Neurologia, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, 20211-030, Brazil
| | - Elielson V da Silva
- Laboratório de Neurociências Translacional, Programa de Pós-Graduação em Neurologia, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, 20211-030, Brazil
| | - Soniza V Alves-Leon
- Laboratório de Neurociências Translacional, Programa de Pós-Graduação em Neurologia, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, 20211-030, Brazil; Departamento de Neurologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 3938-2480, Brazil
| | - Fabrícia L Fontes-Dantas
- Laboratório de Neurociências Translacional, Programa de Pós-Graduação em Neurologia, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, 20211-030, Brazil; Laboratório de Neurofarmacogenetica, Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcântara Gomes, Universidade Estadual do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil.
| | - Jorge M de Souza
- Laboratório de Neurociências Translacional, Programa de Pós-Graduação em Neurologia, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, 20211-030, Brazil; Departamento de Neurocirurgia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 3938-2480, Brazil.
| |
Collapse
|
6
|
Fisher DG, Hoch MR, Gorick CM, Huchthausen C, Breza VR, Sharifi KA, Tvrdik P, Miller GW, Price RJ. Focused Ultrasound Impels the Delivery and Penetration of Model Therapeutics into Cerebral Cavernous Malformations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609060. [PMID: 39253521 PMCID: PMC11383029 DOI: 10.1101/2024.08.27.609060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
BACKGROUND Cerebral cavernous malformations (CCMs) are vascular neoplasms in the brain that can cause debilitating symptoms. Current treatments pose significant risks to some patients, motivating the development of new nonsurgical options. We recently discovered that focused ultrasound-mediated blood-brain barrier opening (FUS) arrests CCM formation and growth. Here, we build on this discovery and assess the ability of FUS to deliver model therapeutics into CCMs. METHODS Quantitative T1 mapping MRI sequences were used with 1 kDa (MultiHance; MH) and 17 kDa (GadoSpin D; GDS) contrast agents to assess the FUS-mediated delivery and penetration of model small molecule drugs and biologics, respectively, into CCMs of Krit1 mutant mice. RESULTS FUS elevated the rate of MH delivery to both the lesion core (4.6-fold) and perilesional space (6.7-fold). Total MH delivery more than doubled in the lesion core and tripled in the perilesional space when FUS was applied immediately prior to MH injection. For the model biologic drug (i.e. GDS), FUS was of greater relative benefit, resulting in 21.7-fold and 3.8-fold delivery increases to the intralesional and perilesional spaces, respectively. CONCLUSIONS FUS is capable of impelling the delivery and penetration of therapeutics into the complex and disorganized CCM microenvironment. Benefits to small molecule drug delivery are more evident in the perilesional space, while benefits to biologic delivery are more evident in CCM cores. These findings, when combined with ability of FUS alone to control CCMs, highlight the potential of FUS to serve as a powerful non-invasive therapeutic platform for CCM.
Collapse
Affiliation(s)
- Delaney G Fisher
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Matthew R Hoch
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Catherine M Gorick
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | | | - Victoria R Breza
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Khadijeh A Sharifi
- Department of Neuroscience, University of Virginia, Charlottesville, VA
- Department of Neurosurgery, University of Virginia Health System, Charlottesville, VA
| | - Petr Tvrdik
- Department of Neuroscience, University of Virginia, Charlottesville, VA
- Department of Neurosurgery, University of Virginia Health System, Charlottesville, VA
| | - G Wilson Miller
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, VA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, VA
| |
Collapse
|
7
|
Liu D, Rodriguez M, Ross D, Ghahreman A. Concurrent multiple cerebral cavernous malformations and cauda equina paraganglioma: illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2024; 8:CASE24102. [PMID: 39102750 DOI: 10.3171/case24102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/15/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Cauda equina neuroendocrine tumors (CENETs), previously known as cauda equina paragangliomas, and multiple cerebral cavernous malformations (CCMs) are uncommon conditions affecting the central nervous system. To the authors' knowledge, they have not been reported in the same patient. OBSERVATIONS The authors present the case of a 45-year-old male with CENET and concurrent incidental MRI findings of multiple CCMs. Familial CCMs are associated with mutations in the KRIT1 (CCM1), MGC4607 (CCM2), and PDCD10 (CCM3) genes. Peripheral paragangliomas have been associated with mutations in succinate dehydrogenase (SDHx), RET (multiple endocrine neoplasia 2), VHL (von Hippel-Lindau syndrome), and NF1 (neurofibromatosis type 1) genes. Except for a single case, cauda equina paragangliomas have not been associated with any underlying genetic mutations. LESSONS It is unclear whether the co-occurrence of these two rare conditions in the same patient is coincidental or suggests a possible shared pathogenesis. https://thejns.org/doi/10.3171/CASE24102.
Collapse
Affiliation(s)
- Daniel Liu
- Departments of Neurosurgery, St George Hospital, Kogarah, New South Wales, Australia
| | - Michael Rodriguez
- Douglass Hanly Moir Pathology, Macquarie Park, New South Wales, Australia
- Macquarie University, Macquarie Park, New South Wales, Australia
| | - Dominic Ross
- Departments of Clinical Genetics, St George Hospital, Kogarah, New South Wales, Australia
| | - Ali Ghahreman
- Departments of Neurosurgery, St George Hospital, Kogarah, New South Wales, Australia
| |
Collapse
|
8
|
Min W, Qin L, Zhang H, López-Giráldez F, Jiang N, Kim Y, Mohan VK, Su M, Murray KN, Grutzendler J, Zhou JH. mTORC1 Signaling in Brain Endothelial Progenitors Contributes to CCM Pathogenesis. Circ Res 2024; 135:e94-e113. [PMID: 38957991 PMCID: PMC11293987 DOI: 10.1161/circresaha.123.324015] [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: 11/18/2023] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Cerebral vascular malformations (CCMs) are primarily found within the brain, where they result in increased risk for stroke, seizures, and focal neurological deficits. The unique feature of the brain vasculature is the blood-brain barrier formed by the brain neurovascular unit. Recent studies suggest that loss of CCM genes causes disruptions of blood-brain barrier integrity as the inciting events for CCM development. CCM lesions are proposed to be initially derived from a single clonal expansion of a subset of angiogenic venous capillary endothelial cells (ECs) and respective resident endothelial progenitor cells (EPCs). However, the critical signaling events in the subclass of brain ECs/EPCs for CCM lesion initiation and progression are unclear. METHODS Brain EC-specific CCM3-deficient (Pdcd10BECKO) mice were generated by crossing Pdcd10fl/fl mice with Mfsd2a-CreERT2 mice. Single-cell RNA-sequencing analyses were performed by the chromium single-cell platform (10× genomics). Cell clusters were annotated into EC subtypes based on visual inspection and GO analyses. Cerebral vessels were visualized by 2-photon in vivo imaging and tissue immunofluorescence analyses. Regulation of mTOR (mechanistic target of rapamycin) signaling by CCM3 and Cav1 (caveolin-1) was performed by cell biology and biochemical approaches. RESULTS Single-cell RNA-sequencing analyses from P10 Pdcd10BECKO mice harboring visible CCM lesions identified upregulated CCM lesion signature and mitotic EC clusters but decreased blood-brain barrier-associated EC clusters. However, a unique EPC cluster with high expression levels of stem cell markers enriched with mTOR signaling was identified from early stages of the P6 Pdcd10BECKO brain. Indeed, mTOR signaling was upregulated in both mouse and human CCM lesions. Genetic deficiency of Raptor (regulatory-associated protein of mTOR), but not of Rictor (rapamycin-insensitive companion of mTOR), prevented CCM lesion formation in the Pdcd10BECKO model. Importantly, the mTORC1 (mTOR complex 1) pharmacological inhibitor rapamycin suppressed EPC proliferation and ameliorated CCM pathogenesis in Pdcd10BECKO mice. Mechanistic studies suggested that Cav1/caveolae increased in CCM3-depleted EPC-mediated intracellular trafficking and complex formation of the mTORC1 signaling proteins. CONCLUSIONS CCM3 is critical for maintaining blood-brain barrier integrity and CCM3 loss-induced mTORC1 signaling in brain EPCs initiates and facilitates CCM pathogenesis.
Collapse
Affiliation(s)
- Wang Min
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Lingfeng Qin
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Haifeng Zhang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Francesc López-Giráldez
- Yale Center for Genomic Analysis, Department of Genetics, Yale University School of Medicine, New Haven, CT
| | - Ning Jiang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Yeaji Kim
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Varsha K. Mohan
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Minhong Su
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Katie N Murray
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT
| | - Jaime Grutzendler
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT
| | - Jenny Huanjiao Zhou
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
9
|
Ren J, Cui Z, Jiang C, Wang L, Guan Y, Ren Y, Zhang S, Tu T, Yu J, Li Y, Duan W, Guan J, Wang K, Zhang H, Xing D, Kahn ML, Zhang H, Hong T. GNA14 and GNAQ somatic mutations cause spinal and intracranial extra-axial cavernous hemangiomas. Am J Hum Genet 2024; 111:1370-1382. [PMID: 38917801 PMCID: PMC11267519 DOI: 10.1016/j.ajhg.2024.05.020] [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: 02/15/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
Extra-axial cavernous hemangiomas (ECHs) are complex vascular lesions mainly found in the spine and cavernous sinus. Their removal poses significant risk due to their vascularity and diffuse nature, and their genetic underpinnings remain incompletely understood. Our approach involved genetic analyses on 31 tissue samples of ECHs employing whole-exome sequencing and targeted deep sequencing. We explored downstream signaling pathways, gene expression changes, and resultant phenotypic shifts induced by these mutations, both in vitro and in vivo. In our cohort, 77.4% of samples had somatic missense variants in GNA14, GNAQ, or GJA4. Transcriptomic analysis highlighted significant pathway upregulation, with the GNAQ c.626A>G (p.Gln209Arg) mutation elevating PI3K-AKT-mTOR and angiogenesis-related pathways, while GNA14 c.614A>T (p.Gln205Leu) mutation led to MAPK and angiogenesis-related pathway upregulation. Using a mouse xenograft model, we observed enlarged vessels from these mutations. Additionally, we initiated rapamycin treatment in a 14-year-old individual harboring the GNAQ c.626A>G (p.Gln209Arg) variant, resulting in gradual regression of cutaneous cavernous hemangiomas and improved motor strength, with minimal side effects. Understanding these mutations and their pathways provides a foundation for developing therapies for ECHs resistant to current therapies. Indeed, the administration of rapamycin in an individual within this study highlights the promise of targeted treatments in treating these complex lesions.
Collapse
Affiliation(s)
- Jian Ren
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Ziwei Cui
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Chendan Jiang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Leiming Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yunqian Guan
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Yeqing Ren
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Shikun Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Tianqi Tu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Jiaxing Yu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Ye Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Wanru Duan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Jian Guan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Kai Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Hongdian Zhang
- Department of Neurosurgery, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China; Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China.
| | - Tao Hong
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China; Department of Neurosurgery, Xiongan Xuanwu Hospital, Xiong'an New Area, China.
| |
Collapse
|
10
|
Alcazar-Felix RJ, Shenkar R, Benavides CR, Bindal A, Srinath A, Li Y, Kinkade S, Terranova T, DeBose-Scarlett E, Lightle R, DeBiasse D, Almazroue H, Cruz DV, Romanos S, Jhaveri A, Koskimäki J, Hage S, Bennett C, Girard R, Marchuk DA, Awad IA. Except for Robust Outliers, Rapamycin Increases Lesion Burden in a Murine Model of Cerebral Cavernous Malformations. Transl Stroke Res 2024:10.1007/s12975-024-01270-9. [PMID: 38980519 DOI: 10.1007/s12975-024-01270-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
Cerebral cavernous malformation (CCM) is a hemorrhagic cerebrovascular disease where lesions develop in the setting of endothelial mutations of CCM genes, with many cases also harboring somatic PIK3CA gain of function (GOF) mutations. Rapamycin, an mTORC1 inhibitor, inhibited progression of murine CCM lesions driven by Ccm gene loss and Pik3ca GOF, but it remains unknown if rapamycin is beneficial in the absence of induction of Pik3ca GOF. We investigated the effect of rapamycin at three clinically relevant doses on lesion development in the Ccm3-/-PDGFb-icreERPositive murine model of familial CCM disease, without induction of Pik3ca GOF. Lesion burden, attrition, and acute and chronic hemorrhaging were compared between placebo and rapamycin-treated mice. Plasma miRNome was compared to identify potential biomarkers of rapamycin response. Outlier, exceptionally large CCM lesions (> 2 SD above the mean lesion burden) were exclusively observed in the placebo group. Rapamycin, across all dosages, may have prevented the emergence of large outlier lesions. Yet rapamycin also appeared to exacerbate mean lesion burden of surviving mice when outliers were excluded, increased attrition, and did not alter hemorrhage. miR-30c-2-3p, decreased in rapamycin-treated mouse plasma, has gene targets in PI3K/AKT and mTOR signaling. Progression of outlier lesions in a familial CCM model may have been halted by rapamycin treatment, at the potential expense of increased mean lesion burden and increased attrition. If confirmed, this can have implications for potential rapamycin treatment of familial CCM disease, where lesion development may not be driven by PIK3CA GOF. Further studies are necessary to determine specific pathways that mediate potential beneficial and detrimental effects of rapamycin treatment, and whether somatic PIK3CA mutations drive particularly aggressive lesions.
Collapse
Affiliation(s)
- Roberto J Alcazar-Felix
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Robert Shenkar
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Christian R Benavides
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Akash Bindal
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Abhinav Srinath
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Ying Li
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Serena Kinkade
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Tatiana Terranova
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Evon DeBose-Scarlett
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Rhonda Lightle
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Dorothy DeBiasse
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Hanadi Almazroue
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Diana Vera Cruz
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Sharbel Romanos
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Aditya Jhaveri
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Janne Koskimäki
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Stephanie Hage
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Carolyn Bennett
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Romuald Girard
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Issam A Awad
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
11
|
Li C, Huang S, Li Q, Zhuo L, Kang Y, Liu P, Huang W, Ma K, Lin X, Zhuang W, Chen D, Wang H, Yan L, Wang D, Lin Y, Kang D, Lin F. Plasma biomarkers in patients with familial cavernous malformation and their first-degree relatives. RESEARCH SQUARE 2024:rs.3.rs-4545797. [PMID: 39011103 PMCID: PMC11247933 DOI: 10.21203/rs.3.rs-4545797/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Background We aimed to explore the differences in plasma biomarker levels between patients with familial cerebral cavernous malformations (FCCM) and their healthy first-degree relatives (FDRs) and between FCCM patients with and without severe chronic disease aggressiveness (CDA). Methods Magnetic resonance imaging (MRI) scanning and genetic testing was performed in patients with multiple CCMs and their FDRs. Sixty-seven plasma biomarkers were tested using a customised multiplex bead immunoassay kit. Univariate and multivariate unconditional logistic regression analyses were conducted to determine the associations between plasma factors and the risk of developing FCCM and severe CDA. Receiver operating characteristic (ROC) curves were generated for each independent risk factor. Results Plasma factors of 37 patients with FCCM and 37 FDRs were examined. Low CD31 (P < 0.001) and BDNF levels (P = 0.013) were independent risk factors for FCCM. The best model was achieved by combining the results of CD31 and BDNF (AUC = 0.845, sensitivity 0.838, specificity 0.784, cutoff score - 4.295) to distinguish patients with FCCM from healthy FDRs. Low serpin E1/PAI-1 (P = 0.011) and high ROBO4 levels (P = 0.013) were independent risk factors for severe CDA in patients with FCCM. The best model was achieved by combining the results of E1/PAI-1 and ROBO4 levels (AUC = 0.913, sensitivity 1.000, specificity 0.760, cutoff score - 0.525) to identify patients with FCCM and severe CDA. Conclusions The plasma concentrations of CD31 and BDNF seem to be lower in patients with FCCM than in their healthy FDRs. Low serpin E1/PAI-1 and high ROBO4 concentrations may be correlated with high lesion burden and risk of recurrent bleeding.
Collapse
Affiliation(s)
- Chunwang Li
- First Affiliated Hospital of Fujian Medical University
| | - Shuna Huang
- First Affiliated Hospital of Fujian Medical University
| | - Qixuan Li
- First Affiliated Hospital of Fujian Medical University
| | - Lingyun Zhuo
- First Affiliated Hospital of Fujian Medical University
| | - Yaqing Kang
- First Affiliated Hospital of Fujian Medical University
| | - Penghui Liu
- First Affiliated Hospital of Fujian Medical University
| | - Weilin Huang
- First Affiliated Hospital of Fujian Medical University
| | - Ke Ma
- First Affiliated Hospital of Fujian Medical University
| | - Xinru Lin
- First Affiliated Hospital of Fujian Medical University
| | | | - Darong Chen
- First Affiliated Hospital of Fujian Medical University
| | - Huimin Wang
- First Affiliated Hospital of Fujian Medical University
| | - Lingjun Yan
- First Affiliated Hospital of Fujian Medical University
| | | | - Yuanxiang Lin
- First Affiliated Hospital of Fujian Medical University
| | - Dezhi Kang
- First Affiliated Hospital of Fujian Medical University
| | - Fuxin Lin
- First Affiliated Hospital of Fujian Medical University
| |
Collapse
|
12
|
Dao L, You Z, Lu L, Xu T, Sarkar AK, Zhu H, Liu M, Calandrelli R, Yoshida G, Lin P, Miao Y, Mierke S, Kalva S, Zhu H, Gu M, Vadivelu S, Zhong S, Huang LF, Guo Z. Modeling blood-brain barrier formation and cerebral cavernous malformations in human PSC-derived organoids. Cell Stem Cell 2024; 31:818-833.e11. [PMID: 38754427 PMCID: PMC11162335 DOI: 10.1016/j.stem.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 02/24/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
The human blood-brain barrier (hBBB) is a highly specialized structure that regulates passage across blood and central nervous system (CNS) compartments. Despite its critical physiological role, there are no reliable in vitro models that can mimic hBBB development and function. Here, we constructed hBBB assembloids from brain and blood vessel organoids derived from human pluripotent stem cells. We validated the acquisition of blood-brain barrier (BBB)-specific molecular, cellular, transcriptomic, and functional characteristics and uncovered an extensive neuro-vascular crosstalk with a spatial pattern within hBBB assembloids. When we used patient-derived hBBB assembloids to model cerebral cavernous malformations (CCMs), we found that these assembloids recapitulated the cavernoma anatomy and BBB breakdown observed in patients. Upon comparison of phenotypes and transcriptome between patient-derived hBBB assembloids and primary human cavernoma tissues, we uncovered CCM-related molecular and cellular alterations. Taken together, we report hBBB assembloids that mimic the core properties of the hBBB and identify a potentially underlying cause of CCMs.
Collapse
Affiliation(s)
- Lan Dao
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zhen You
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Lu Lu
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tianyang Xu
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Avijite Kumer Sarkar
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Hui Zhu
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Miao Liu
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Riccardo Calandrelli
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - George Yoshida
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Pei Lin
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yifei Miao
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sarah Mierke
- Divisions of Pediatric Neurosurgery and Interventional Neuroradiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Srijan Kalva
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Haining Zhu
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sudhakar Vadivelu
- Divisions of Pediatric Neurosurgery and Interventional Neuroradiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Sheng Zhong
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| | - L Frank Huang
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA.
| | - Ziyuan Guo
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
13
|
Chen C, Bao Y, Ju S, Jiang C, Zou X, Zhang X, Chen L. Single-cell and bulk RNA-seq unveils the immune infiltration landscape associated with cuproptosis in cerebral cavernous malformations. Biomark Res 2024; 12:57. [PMID: 38835051 DOI: 10.1186/s40364-024-00603-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Cerebral cavernous malformations (CCMs) are vascular abnormalities associated with deregulated angiogenesis. Their pathogenesis and optimal treatment remain unclear. This study aims to investigate the molecular signatures of cuproptosis, a newly identified type of cell death, associated with CCMs development. METHODS Bulk RNA sequencing (RNA-seq) from 15 CCM and 6 control samples were performed with consensus clustering and clustered to two subtypes based on expression levels of cuproptosis-related genes (CRGs). Differentially expressed genes and immune infiltration between subtypes were then identified. Machine learning algorithms including the least absolute shrinkage and selection operator and random forest were employed to screen for hub genes for CCMs associated with cuproptosis. Furthermore, Pathway enrichment and correlation analysis were used to explore the functions of hub genes and their association with immune phenotypes in CCMs. An external dataset was then employed for validation. Finally, employing the Cellchat algorithm on a single-cell RNA-seq dataset, we explored potential mechanisms underlying the participation of these hub genes in cell-cell communication in CCMs. RESULTS Our study revealed two distinct CCM subtypes with differential pattern of CRG expression and immune infiltration. Three hub genes (BTBD10, PFDN4, and CEMIP) were identified and validated, which may significantly associate with CCM pathogenesis. These genes were found to be significantly upregulated in CCM endothelial cells (ECs) and were validated through immunofluorescence and western blot analysis. Single-cell RNA-seq analysis revealed the cellular co-expression patterns of these hub genes, particularly highlighting the high expression of BTBD10 and PFDN4 in ECs. Additionally, a significant co-localization was also observed between BTBD10 and the pivotal cuproptosis gene FDX1 in Mki67+ tip cells, indicating the crucial role of cuproptosis for angiogenesis in CCMs. The study also explored the cell-cell communication between subcluster of ECs expressing these hub genes and immune cells, particularly M2 macrophages, suggesting a role for these interactions in CCM pathogenesis. CONCLUSION This study identifies molecular signatures linking cuproptosis to CCMs pathogenesis. Three hub genes-PFDN4, CEMIP, and BTBD10-may influence disease progression by modulating immunity. Further research is needed to understand their precise disease mechanisms and evaluate their potential as biomarkers or therapeutic targets for CCMs.
Collapse
Affiliation(s)
- Chengwei Chen
- Neurosurgical department of Huashan hospital and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- Tianqiao and Chrissy Chen Institute Clinical Translational Research Center, Shanghai, 200040, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yuting Bao
- Neurosurgical department of Huashan hospital and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- Tianqiao and Chrissy Chen Institute Clinical Translational Research Center, Shanghai, 200040, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Sihan Ju
- Neurosurgical department of Huashan hospital and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- Tianqiao and Chrissy Chen Institute Clinical Translational Research Center, Shanghai, 200040, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Conglin Jiang
- Neurosurgical department of Huashan hospital and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- Tianqiao and Chrissy Chen Institute Clinical Translational Research Center, Shanghai, 200040, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Xiang Zou
- Neurosurgical department of Huashan hospital and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- Tianqiao and Chrissy Chen Institute Clinical Translational Research Center, Shanghai, 200040, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Xin Zhang
- Neurosurgical department of Huashan hospital and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- Tianqiao and Chrissy Chen Institute Clinical Translational Research Center, Shanghai, 200040, China
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Liang Chen
- Neurosurgical department of Huashan hospital and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China.
- Tianqiao and Chrissy Chen Institute Clinical Translational Research Center, Shanghai, 200040, China.
- Research Unit of New Technologies of Micro-Endoscopy Combination in Skull Base Surgery (2018RU008), Chinese Academy of Medical Sciences, Beijing, China.
- National Center for Neurological Disorders, Shanghai, 200040, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China.
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China.
| |
Collapse
|
14
|
Chan KS, Daulton MR, Reddy VD, McComb EN, Lavine JA. Prevalence of Retinal Venous Malformations in Patients With Cerebral Cavernous or Arteriovenous Malformations. J Neuroophthalmol 2024; 44:226-231. [PMID: 37585271 PMCID: PMC10869638 DOI: 10.1097/wno.0000000000001974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
BACKGROUND Cerebral vascular malformations (CVMs) may result in hemorrhage, seizure, neurologic dysfunction, and death. CVMs include capillary telangiectasias, venous malformations, cavernous malformations, and arteriovenous malformations. Cavernous and arteriovenous malformations carry the highest risk of complications. Retinal venous malformations (RVMs) have been proposed as an associated finding. Our objective was to determine the prevalence of RVMs in patients with high-risk CVMs. METHODS We retrospectively reviewed patients diagnosed with cerebral cavernous or arteriovenous malformations (high-risk CVMs) who were evaluated by the ophthalmology service at Northwestern University between 2017 and 2020. Patients were stratified into 3 cohorts based on level of certainty: dilated funduscopic examination (DFE), DFE with any form of ocular imaging, and DFE with complete imaging of the macula. We recorded ophthalmic examination abnormalities, ocular imaging findings, and major CVM complications. RESULTS We evaluated 156 patients with high-risk CVMs who had undergone DFE. Ocular imaging of any type was performed in 56 patients, of whom 46 had complete imaging of the macula. Zero RVMs were identified in any cohort (95% confidence interval: 0%-1.9% for the entire cohort, 0%-5.4% for any ocular imaging cohort, and 0%-6.5% for the complete macular imaging cohort). Cerebral hemorrhage or seizure occurred in 15%-33% of patients. Associated visual field defects or cranial nerve palsies were found in 14%-20% of patients. CONCLUSIONS Zero RVMs were identified in patients with high-risk CVMs. However, neuro-ophthalmic findings were common. Therefore, we recommend neuroimaging for patients with RVMs and neuro-ophthalmic signs or symptoms. In asymptomatic patients with RVMs, a potential algorithm for neuroimaging is proposed.
Collapse
Affiliation(s)
- Kyle S Chan
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Melanie R Daulton
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Vishruth D Reddy
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Erin N McComb
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jeremy A Lavine
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
15
|
Alfa R, Considine T, Virani S, Pfeiffer M, Donato A, Dickerson D, Shuster D, Ellis J, Rushton K, Wei H, Gibson C. Clinical pharmacology and tolerability of REC-994, a redox-cycling nitroxide compound, in randomized phase 1 dose-finding studies. Pharmacol Res Perspect 2024; 12:e1200. [PMID: 38655895 PMCID: PMC11040693 DOI: 10.1002/prp2.1200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/28/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
Cerebral cavernous malformation (CCM) has variable clinical symptoms, including potentially fatal hemorrhagic stroke. Treatment options are very limited, presenting a large unmet need. REC-994 (also known as tempol), identified as a potential treatment through an unbiased drug discovery platform, is hypothesized to treat CCMs through a reduction in superoxide, a reactive oxygen species. We investigated the safety, tolerability, and pharmacokinetic profile of REC-994 in healthy volunteers. Single- and multiple-ascending dose (SAD and MAD, respectively) studies were conducted in adult volunteers (ages 18-55). SAD study participants received an oral dose of REC-994 or placebo. MAD study participants were randomized 3:1 to oral doses of REC-994 or matching placebo, once daily for 10 days. Thirty-two healthy volunteers participated in the SAD study and 52 in the MAD study. Systemic exposure increased in proportion to REC-994 dose after single doses of 50-800 mg and after 10 days of dosing over the 16-fold dose range of 50-800 mg. Median Tmax and mean t1/2 were independent of dose in both studies, and the solution formulation was more rapidly absorbed. REC-994 was well tolerated. Treatment-emergent adverse effects across both studies were mild and transient and resolved by the end of the study. REC-994 has a favorable safety profile and was well tolerated in single and multiple doses up to 800 mg with no dose-limiting adverse effects identified. Data support conducting a phase 2 clinical trial in patients with symptomatic CCM.
Collapse
Affiliation(s)
- Ron Alfa
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
- Present address:
NoetikSan FranciscoCaliforniaUSA
| | - Timothy Considine
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
- Present address:
Considine Comprehensive ConsultingSan DiegoCaliforniaUSA
| | | | - Matt Pfeiffer
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
| | - Anthony Donato
- Department of Internal MedicineUniversity of UtahSalt Lake CityUtahUSA
| | | | - Diana Shuster
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
- Present address:
CenExelSalt Lake CityUtahUSA
| | - Joel Ellis
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
- Present address:
Mariner BioPharmaSan JoseCaliforniaUSA
| | | | - Helen Wei
- Recursion Pharmaceuticals, Inc.Salt Lake CityUtahUSA
| | | |
Collapse
|
16
|
Frias-Anaya E, Gallego-Gutierrez H, Gongol B, Weinsheimer S, Lai CC, Orecchioni M, Sriram A, Bui CM, Nelsen B, Hale P, Pham A, Shenkar R, DeBiasse D, Lightle R, Girard R, Li Y, Srinath A, Daneman R, Nudleman E, Sun H, Guma M, Dubrac A, Mesarwi OA, Ley K, Kim H, Awad IA, Ginsberg MH, Lopez-Ramirez MA. Mild Hypoxia Accelerates Cerebral Cavernous Malformation Disease Through CX3CR1-CX3CL1 Signaling. Arterioscler Thromb Vasc Biol 2024; 44:1246-1264. [PMID: 38660801 PMCID: PMC11111348 DOI: 10.1161/atvbaha.123.320367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Heterogeneity in the severity of cerebral cavernous malformations (CCMs) disease, including brain bleedings and thrombosis that cause neurological disabilities in patients, suggests that environmental, genetic, or biological factors act as disease modifiers. Still, the underlying mechanisms are not entirely understood. Here, we report that mild hypoxia accelerates CCM disease by promoting angiogenesis, neuroinflammation, and vascular thrombosis in the brains of CCM mouse models. METHODS We used genetic studies, RNA sequencing, spatial transcriptome, micro-computed tomography, fluorescence-activated cell sorting, multiplex immunofluorescence, coculture studies, and imaging techniques to reveal that sustained mild hypoxia via the CX3CR1-CX3CL1 (CX3C motif chemokine receptor 1/chemokine [CX3C motif] ligand 1) signaling pathway influences cell-specific neuroinflammatory interactions, contributing to heterogeneity in CCM severity. RESULTS Histological and expression profiles of CCM neurovascular lesions (Slco1c1-iCreERT2;Pdcd10fl/fl; Pdcd10BECKO) in male and female mice found that sustained mild hypoxia (12% O2, 7 days) accelerates CCM disease. Our findings indicate that a small reduction in oxygen levels can significantly increase angiogenesis, neuroinflammation, and thrombosis in CCM disease by enhancing the interactions between endothelium, astrocytes, and immune cells. Our study indicates that the interactions between CX3CR1 and CX3CL1 are crucial in the maturation of CCM lesions and propensity to CCM immunothrombosis. In particular, this pathway regulates the recruitment and activation of microglia and other immune cells in CCM lesions, which leads to lesion growth and thrombosis. We found that human CX3CR1 variants are linked to lower lesion burden in familial CCMs, proving it is a genetic modifier in human disease and a potential marker for aggressiveness. Moreover, monoclonal blocking antibody against CX3CL1 or reducing 1 copy of the Cx3cr1 gene significantly reduces hypoxia-induced CCM immunothrombosis. CONCLUSIONS Our study reveals that interactions between CX3CR1 and CX3CL1 can modify CCM neuropathology when lesions are accelerated by environmental hypoxia. Moreover, a hypoxic environment or hypoxia signaling caused by CCM disease influences the balance between neuroinflammation and neuroprotection mediated by CX3CR1-CX3CL1 signaling. These results establish CX3CR1 as a genetic marker for patient stratification and a potential predictor of CCM aggressiveness.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Chemokine CX3CL1/metabolism
- Chemokine CX3CL1/genetics
- CX3C Chemokine Receptor 1/genetics
- CX3C Chemokine Receptor 1/metabolism
- Disease Models, Animal
- Hemangioma, Cavernous, Central Nervous System/genetics
- Hemangioma, Cavernous, Central Nervous System/metabolism
- Hemangioma, Cavernous, Central Nervous System/pathology
- Hypoxia/metabolism
- Hypoxia/complications
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic/metabolism
- Neuroinflammatory Diseases/metabolism
- Neuroinflammatory Diseases/pathology
- Neuroinflammatory Diseases/genetics
- Signal Transduction
Collapse
Affiliation(s)
- Eduardo Frias-Anaya
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Helios Gallego-Gutierrez
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Brendan Gongol
- Department of Health Sciences, Victor Valley College, Victorville, CA (B.G.)
- Institute for Integrative Genome Biology, 1207F Genomics Building, University of California, Riverside (B.G.)
| | - Shantel Weinsheimer
- Department of Anesthesia and Perioperative Care, Institute for Human Genetics, University of California, San Francisco (S.W., A.S., H.K.)
| | - Catherine Chinhchu Lai
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (M.O., K.L.)
| | - Aditya Sriram
- Department of Anesthesia and Perioperative Care, Institute for Human Genetics, University of California, San Francisco (S.W., A.S., H.K.)
| | - Cassandra M Bui
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Bliss Nelsen
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Preston Hale
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Angela Pham
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Dorothy DeBiasse
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Ying Li
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Richard Daneman
- Department of Pharmacology (R.D., M.A.L.-R.), University of California San Diego, La Jolla
| | - Eric Nudleman
- Department of Ophthalmology (E.N.), University of California San Diego, La Jolla
| | - Hao Sun
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Monica Guma
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montréal, Quebec, Canada. Département de Pathologie et Biologie Cellulaire, Université de Montréal, Quebec, Canada (A.D.)
| | - Omar A Mesarwi
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (M.O., K.L.)
| | - Helen Kim
- Department of Anesthesia and Perioperative Care, Institute for Human Genetics, University of California, San Francisco (S.W., A.S., H.K.)
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Mark H Ginsberg
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Miguel Alejandro Lopez-Ramirez
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
- Department of Pharmacology (R.D., M.A.L.-R.), University of California San Diego, La Jolla
| |
Collapse
|
17
|
Smith ER. Pediatric Cerebral Vascular Malformations : Current and Future Perspectives. J Korean Neurosurg Soc 2024; 67:326-332. [PMID: 38409785 PMCID: PMC11079569 DOI: 10.3340/jkns.2024.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/06/2024] [Accepted: 02/25/2024] [Indexed: 02/28/2024] Open
Abstract
Intracranial vascular malformations typically encountered by pediatric neurosurgeons include arteriovenous malformations, vein of Galen malformations and cavernous malformations. While these remain amongst some of the most challenging lesions faced by patients and caregivers, the past decade has produced marked advances in the understanding of the pathophysiology of these conditions, with concomitant innovations in treatment. This article will highlight present and future perspectives relevant to these diseases, with a focus on an emerging approach utilizing disease-specific mutations to develop a novel taxonomy for these conditions.
Collapse
Affiliation(s)
- Edward R. Smith
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Five steps to connect genetic risk variants to disease. Nature 2024:10.1038/d41586-024-00061-4. [PMID: 38570654 DOI: 10.1038/d41586-024-00061-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
|
19
|
Lorente-Herraiz L, Cuesta AM, Granado J, Recio-Poveda L, Botella LM, Albiñana V. Molecular and Cellular Characterization of Primary Endothelial Cells from a Familial Cavernomatosis Patient. Int J Mol Sci 2024; 25:3952. [PMID: 38612762 PMCID: PMC11012380 DOI: 10.3390/ijms25073952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Cerebral cavernous malformation (CCM) or familial cavernomatosis is a rare, autosomal dominant, inherited disease characterized by the presence of vascular malformations consisting of blood vessels with an abnormal structure in the form of clusters. Based on the altered gene (CCM1/Krit1, CCM2, CCM3) and its origin (spontaneous or familial), different types of this disease can be found. In this work we have isolated and cultivated primary endothelial cells (ECs) from peripheral blood of a type 1 CCM patient. Differential functional and gene expression profiles of these cells were analyzed and compared to primary ECs from a healthy donor. The mutation of the familial index case consisted of a heterozygous point mutation in the position +1 splicing consensus between exons 15 and 16, causing failure in RNA processing and in the final protein. Furthermore, gene expression analysis by quantitative PCR revealed a decreased expression of genes involved in intercellular junction formation, angiogenesis, and vascular homeostasis. Cell biology analysis showed that CCM1 ECs were impaired in angiogenesis and cell migration. Taken together, the results obtained suggest that the alterations found in CCM1 ECs are already present in the heterozygous condition, suffering from vascular impairment and somewhat predisposed to vascular damage.
Collapse
Affiliation(s)
- Laura Lorente-Herraiz
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
| | - Angel M. Cuesta
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Jaime Granado
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
| | - Lucía Recio-Poveda
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
| | - Luisa-María Botella
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
| | - Virginia Albiñana
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
| |
Collapse
|
20
|
Lin PK, Sun Z, Davis GE. Defining the Functional Influence of Endothelial Cell-Expressed Oncogenic Activating Mutations on Vascular Morphogenesis and Capillary Assembly. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:574-598. [PMID: 37838010 PMCID: PMC10988768 DOI: 10.1016/j.ajpath.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/02/2023] [Accepted: 08/15/2023] [Indexed: 10/16/2023]
Abstract
This study sought to define key molecules and signals controlling major steps in vascular morphogenesis, and how these signals regulate pericyte recruitment and pericyte-induced basement membrane deposition. The morphogenic impact of endothelial cell (EC) expression of activating mutants of Kirsten rat sarcoma virus (kRas), mitogen-activated protein kinase 1 (Mek1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), Akt serine/threonine kinase 1 (Akt1), Ras homolog enriched in brain (Rheb) Janus kinase 2 (Jak2), or signal transducer and activator of transcription 3 (Stat3) expression versus controls was evaluated, along with EC signaling events, pharmacologic inhibitor assays, and siRNA suppression experiments. Primary stimulators of EC lumen formation included kRas, Akt1, and Mek1, whereas PIK3CA and Akt1 stimulated a specialized type of cystic lumen formation. In contrast, the key drivers of EC sprouting behavior were Jak2, Stat3, Mek1, PIK3CA, and mammalian target of rapamycin (mTor). These conclusions are further supported by pharmacologic inhibitor and siRNA suppression experiments. EC expression of active Akt1, kRas, and PIK3CA led to markedly dysregulated lumen formation coupled to strongly inhibited pericyte recruitment and basement membrane deposition. For example, activated Akt1 expression in ECs excessively stimulated lumen formation, decreased EC sprouting behavior, and showed minimal pericyte recruitment with reduced mRNA expression of platelet-derived growth factor-BB, platelet-derived growth factor-DD, and endothelin-1, critical EC-derived factors known to stimulate pericyte invasion. The study identified key signals controlling fundamental steps in capillary morphogenesis and maturation and provided mechanistic details on why EC activating mutations induced a capillary deficiency state with abnormal lumens, impaired pericyte recruitment, and basement deposition: predisposing stimuli for the development of vascular malformations.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
21
|
Ge P, Tao C, Wang W, He Q, Liu C, Zheng Z, Mou S, Zhang B, Liu X, Zhang Q, Wang R, Li H, Zhang D, Zhao J. Circulating immune cell landscape and T-cell abnormalities in patients with moyamoya disease. Clin Transl Med 2024; 14:e1647. [PMID: 38566524 PMCID: PMC10988118 DOI: 10.1002/ctm2.1647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/23/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Moyamoya disease (MMD) stands as a prominent cause of stroke among children and adolescents in East Asian populations. Although a growing body of evidence suggests that dysregulated inflammation and autoimmune responses might contribute to the development of MMD, a comprehensive and detailed understanding of the alterations in circulating immune cells associated with MMD remains elusive. METHODS In this study, we employed a combination of single-cell RNA sequencing (scRNA-seq), mass cytometry and RNA-sequencing techniques to compare immune cell profiles in peripheral blood samples obtained from patients with MMD and age-matched healthy controls. RESULTS Our investigation unveiled immune dysfunction in MMD patients, primarily characterized by perturbations in T-cell (TC) subpopulations, including a reduction in effector TCs and an increase in regulatory TCs (Tregs). Additionally, we observed diminished natural killer cells and dendritic cells alongside heightened B cells and monocytes in MMD patients. Notably, within the MMD group, there was an augmented proportion of fragile Tregs, whereas the stable Treg fraction decreased. MMD was also linked to heightened immune activation, as evidenced by elevated expression levels of HLA-DR and p-STAT3. CONCLUSIONS Our findings offer a comprehensive view of the circulating immune cell landscape in MMD patients. Immune dysregulation in patients with MMD was characterized by alterations in T-cell populations, including a decrease in effector T-cells and an increase in regulatory T-cells (Tregs), suggest a potential role for disrupted circulating immunity in the aetiology of MMD.
Collapse
|
22
|
Smith ER. Cavernous Malformations of the Central Nervous System. N Engl J Med 2024; 390:1022-1028. [PMID: 38477989 DOI: 10.1056/nejmra2305116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Affiliation(s)
- Edward R Smith
- From the Department of Neurosurgery, Children's Hospital Boston, and Harvard Medical School - both in Boston
| |
Collapse
|
23
|
Yordanov TE, Keyser MS, Enriquez Martinez MA, Esposito T, Tefft JB, Morris EK, Labzin LI, Stehbens SJ, Rowan AE, Hogan BM, Chen CS, Lauko J, Lagendijk AK. Hyaluronic acid turnover controls the severity of cerebral cavernous malformations in bioengineered human micro-vessels. APL Bioeng 2024; 8:016108. [PMID: 38352162 PMCID: PMC10864035 DOI: 10.1063/5.0159330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Cerebral cavernous malformations (CCMs) are vascular lesions that predominantly form in blood vessels of the central nervous system upon loss of the CCM multimeric protein complex. The endothelial cells within CCM lesions are characterized by overactive MEKK3 kinase and KLF2/4 transcription factor signaling, leading to pathological changes such as increased endothelial cell spreading and reduced junctional integrity. Concomitant to aberrant endothelial cell signaling, non-autonomous signals from the extracellular matrix (ECM) have also been implicated in CCM lesion growth and these factors might explain why CCM lesions mainly develop in the central nervous system. Here, we adapted a three-dimensional microfluidic system to examine CCM1 deficient human micro-vessels in distinctive extracellular matrices. We validate that pathological hallmarks are maintained in this model. We further show that key genes responsible for homeostasis of hyaluronic acid, a major extracellular matrix component of the central nervous system, are dysregulated in CCM. Supplementing the matrix in our model with distinct forms of hyaluronic acid inhibits pathological cell spreading and rescues barrier function. Hyaluronic acid acts by dampening cell-matrix adhesion signaling in CCM, either downstream or in parallel of KLF2/4. This study provides a proof-of-principle that ECM embedded 3D microfluidic models are ideally suited to identify how changes in ECM structure and signaling impact vascular malformations.
Collapse
Affiliation(s)
- Teodor E. Yordanov
- Centre for Cell Biology and Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Mikaela S. Keyser
- Centre for Cell Biology and Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Marco A. Enriquez Martinez
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Juliann B. Tefft
- The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA
| | - Elysse K. Morris
- Centre for Cell Biology and Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | - Alan E. Rowan
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | - Jan Lauko
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | | |
Collapse
|
24
|
Wu G, Lin X, Jiang H, Liu Z. Association Between the miR-100 rs1834306 A>G Polymorphism and Susceptibility to Venous Malformation. Int J Gen Med 2024; 17:509-515. [PMID: 38356685 PMCID: PMC10864769 DOI: 10.2147/ijgm.s441542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
Background Venous malformation is related to genes and results in functional and morphologic anomalies. Genetic variations affecting the development of vessel endothelial cells are unclear. Therefore, this study aimed to investigate the potential value of the miR-100 rs1834306 A>G polymorphism as a marker of susceptibility to venous malformation. Methods In this case-control study in southern Chinese children, we collected blood samples from 1158 controls and 1113 patients with venous malformation. TaqMan genotyping of miR-100 rs1834306 A>G was performed by real-time fluorescent quantitative polymerase chain reaction. Results Multivariate logistic regression analysis showed that there was no significant association between the presence of the miR-100 rs1834306 A>G polymorphism and susceptibility to venous malformation by evaluating the values of pooled odds ratios and 95% confidence intervals. Similarly, among different sites, rs1834306 A>G was also not associated with venous malformation. Conclusion Our results suggest that the miR-100 rs1834306 A>G polymorphism is not associated with susceptibility to venous malformation in southern Chinese children. These results need to be further confirmed by investigating a more diverse ethnic population of patients with venous malformations.
Collapse
Affiliation(s)
- Guitao Wu
- Department of Interventional Radiology and Vascular Anomalies, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People’s Republic of China
| | - Xi Lin
- Department of Interventional Radiology and Vascular Anomalies, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People’s Republic of China
| | - Hua Jiang
- Department of Interventional Radiology and Vascular Anomalies, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People’s Republic of China
| | - Zhenyin Liu
- Department of Interventional Radiology and Vascular Anomalies, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510623, People’s Republic of China
| |
Collapse
|
25
|
Fisher DG, Sharifi KA, Shah IM, Gorick CM, Breza VR, Debski AC, Hoch MR, Cruz T, Samuels JD, Sheehan JP, Schlesinger D, Moore D, Lukens JR, Miller GW, Tvrdik P, Price RJ. Focused Ultrasound Blood-Brain Barrier Opening Arrests the Growth and Formation of Cerebral Cavernous Malformations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.577810. [PMID: 38352349 PMCID: PMC10862920 DOI: 10.1101/2024.01.31.577810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
BACKGROUND Cerebral cavernous malformations (CCM) are vascular lesions within the central nervous system, consisting of dilated and hemorrhage-prone capillaries. CCMs can cause debilitating neurological symptoms, and surgical excision or stereotactic radiosurgery are the only current treatment options. Meanwhile, transient blood-brain barrier opening (BBBO) with focused ultrasound (FUS) and microbubbles is now understood to exert potentially beneficial bioeffects, such as stimulation of neurogenesis and clearance of amyloid-β. Here, we tested whether FUS BBBO could be deployed therapeutically to control CCM formation and progression in a clinically-representative murine model. METHODS CCMs were induced in mice by postnatal, endothelial-specific Krit1 ablation. FUS was applied for BBBO with fixed peak-negative pressures (PNPs; 0.2-0.6 MPa) or passive cavitation detection-modulated PNPs. Magnetic resonance imaging (MRI) was used to target FUS treatments, evaluate safety, and measure longitudinal changes in CCM growth after BBBO. RESULTS FUS BBBO elicited gadolinium accumulation primarily at the perilesional boundaries of CCMs, rather than lesion cores. Passive cavitation detection and gadolinium contrast enhancement were comparable in CCM and wild-type mice, indicating that Krit1 ablation does not confer differential sensitivity to FUS BBBO. Acutely, CCMs exposed to FUS BBBO remained structurally stable, with no signs of hemorrhage. Longitudinal MRI revealed that FUS BBBO halted the growth of 94% of CCMs treated in the study. At 1 month, FUS BBBO-treated lesions lost, on average, 9% of their pre-sonication volume. In contrast, non-sonicated control lesions grew to 670% of their initial volume. Lesion control with FUS BBBO was accompanied by a marked reduction in the area and mesenchymal appearance of Krit mutant endothelium. Strikingly, in mice receiving multiple BBBO treatments with fixed PNPs, de novo CCM formation was significantly reduced by 81%. Mock treatment plans on MRIs of patients with surgically inaccessible lesions revealed their lesions are amenable to FUS BBBO with current clinical technology. CONCLUSIONS Our results establish FUS BBBO as a novel, non-invasive modality that can safely arrest murine CCM growth and prevent their de novo formation. As an incisionless, MR image-guided therapy with the ability to target eloquent brain locations, FUS BBBO offers an unparalleled potential to revolutionize the therapeutic experience and enhance the accessibility of treatments for CCM patients.
Collapse
Affiliation(s)
- Delaney G Fisher
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Khadijeh A Sharifi
- Department of Neuroscience, University of Virginia, Charlottesville, VA
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA
| | - Ishaan M Shah
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Catherine M Gorick
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Victoria R Breza
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Anna C Debski
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Matthew R Hoch
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Tanya Cruz
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Joshua D Samuels
- Department of Neuroscience, University of Virginia, Charlottesville, VA
| | - Jason P Sheehan
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA
| | - David Schlesinger
- Department of Radiation Oncology, University of Virginia Health System, Charlottesville, VA
| | - David Moore
- Focused Ultrasound Foundation, Charlottesville, VA
| | - John R Lukens
- Department of Neuroscience, University of Virginia, Charlottesville, VA
| | - G Wilson Miller
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, VA
| | - Petr Tvrdik
- Department of Neuroscience, University of Virginia, Charlottesville, VA
- Department of Neurological Surgery, University of Virginia Health System, Charlottesville, VA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, VA
| |
Collapse
|
26
|
Ayata C, Kim H, Morrison L, Liao JK, Gutierrez J, Lopez-Toledano M, Carrazana E, Rabinowicz AL, Awad IA. Role of Rho-Associated Kinase in the Pathophysiology of Cerebral Cavernous Malformations. Neurol Genet 2024; 10:e200121. [PMID: 38179414 PMCID: PMC10766084 DOI: 10.1212/nxg.0000000000200121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/20/2023] [Indexed: 01/06/2024]
Abstract
Cerebral cavernous malformations (CCMs) are vascular lesions characterized by a porous endothelium. The lack of a sufficient endothelial barrier can result in microbleeds and frank intracerebral hemorrhage. A primary mechanism for lesion development is a sequence variant in at least 1 of the 3 CCM genes (CCM1, CCM2, and CCM3), which influence various signaling pathways that lead to the CCM phenotype. A common downstream process associated with CCM gene loss of function involves overactivation of RhoA and its effector Rho-associated kinase (ROCK). In this study, we review RhoA/ROCK-related mechanisms involved in CCM pathophysiology as potential therapeutic targets. Literature searches were conducted in PubMed using combinations of search terms related to RhoA/ROCK and CCMs. In endothelial cells, CCM1, CCM2, and CCM3 proteins normally associate to form the CCM protein complex, which regulates the functions of a wide variety of protein targets (e.g., MAP3K3, SMURF1, SOK-1, and ICAP-1) that directly or indirectly increase RhoA/ROCK activity. Loss of CCM complex function and increased RhoA/ROCK activity can lead to the formation of stress fibers that contribute to endothelial junction instability. Other RhoA/ROCK-mediated pathophysiologic outcomes include a shift to a senescence-associated secretory phenotype (primarily mediated by ROCK2), which is characterized by endothelial cell migration, cell cycle arrest, extracellular matrix degradation, leukocyte chemotaxis, and inflammation. ROCK represents a potential therapeutic target, and direct (fasudil, NRL-1049) and indirect (statins) ROCK inhibitors have demonstrated various levels of efficacy in reducing lesion burden in preclinical models of CCM. Current (atorvastatin) and planned (NRL-1049) clinical studies will determine the efficacy of ROCK inhibitors for CCM in humans, for which no US Food and Drug Administration-approved or EU-approved pharmacologic treatment exists.
Collapse
Affiliation(s)
- Cenk Ayata
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Helen Kim
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Leslie Morrison
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - James K Liao
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Juan Gutierrez
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Miguel Lopez-Toledano
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Enrique Carrazana
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Adrian L Rabinowicz
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Issam A Awad
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| |
Collapse
|
27
|
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
|
28
|
Kahn ML. CCM Function in the Heart: Working From Outside-In Rather Than Inside-Out. JACC Basic Transl Sci 2024; 9:220-222. [PMID: 38510721 PMCID: PMC10950394 DOI: 10.1016/j.jacbts.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Affiliation(s)
- Mark L. Kahn
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
29
|
Schnitzler GR, Kang H, Fang S, Angom RS, Lee-Kim VS, Ma XR, Zhou R, Zeng T, Guo K, Taylor MS, Vellarikkal SK, Barry AE, Sias-Garcia O, Bloemendal A, Munson G, Guckelberger P, Nguyen TH, Bergman DT, Hinshaw S, Cheng N, Cleary B, Aragam K, Lander ES, Finucane HK, Mukhopadhyay D, Gupta RM, Engreitz JM. Convergence of coronary artery disease genes onto endothelial cell programs. Nature 2024; 626:799-807. [PMID: 38326615 PMCID: PMC10921916 DOI: 10.1038/s41586-024-07022-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/03/2024] [Indexed: 02/09/2024]
Abstract
Linking variants from genome-wide association studies (GWAS) to underlying mechanisms of disease remains a challenge1-3. For some diseases, a successful strategy has been to look for cases in which multiple GWAS loci contain genes that act in the same biological pathway1-6. However, our knowledge of which genes act in which pathways is incomplete, particularly for cell-type-specific pathways or understudied genes. Here we introduce a method to connect GWAS variants to functions. This method links variants to genes using epigenomics data, links genes to pathways de novo using Perturb-seq and integrates these data to identify convergence of GWAS loci onto pathways. We apply this approach to study the role of endothelial cells in genetic risk for coronary artery disease (CAD), and discover 43 CAD GWAS signals that converge on the cerebral cavernous malformation (CCM) signalling pathway. Two regulators of this pathway, CCM2 and TLNRD1, are each linked to a CAD risk variant, regulate other CAD risk genes and affect atheroprotective processes in endothelial cells. These results suggest a model whereby CAD risk is driven in part by the convergence of causal genes onto a particular transcriptional pathway in endothelial cells. They highlight shared genes between common and rare vascular diseases (CAD and CCM), and identify TLNRD1 as a new, previously uncharacterized member of the CCM signalling pathway. This approach will be widely useful for linking variants to functions for other common polygenic diseases.
Collapse
Affiliation(s)
- Gavin R Schnitzler
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA
- Divisions of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Helen Kang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
| | - Shi Fang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Divisions of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ramcharan S Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | - Vivian S Lee-Kim
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Divisions of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - X Rosa Ma
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
| | - Ronghao Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
| | - Tony Zeng
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
| | - Katherine Guo
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
| | - Martin S Taylor
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shamsudheen K Vellarikkal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Divisions of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Aurelie E Barry
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Divisions of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Oscar Sias-Garcia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Divisions of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Alex Bloemendal
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA
| | - Glen Munson
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Tung H Nguyen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Drew T Bergman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Stephen Hinshaw
- Department of Chemical and Systems Biology, ChEM-H, and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Nathan Cheng
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Brian Cleary
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Faculty of Computing and Data Sciences, Departments of Biology and Biomedical Engineering, Biological Design Center, and Program in Bioinformatics, Boston University, Boston, MA, USA
| | - Krishna Aragam
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Eric S Lander
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, MIT, Cambridge, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Hilary K Finucane
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | - Rajat M Gupta
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA.
- Divisions of Genetics and Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Jesse M Engreitz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute, Cambridge, MA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Basic Science and Engineering Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
30
|
Wang R, Lu D, Song R, Du L, Yang X, Wu ST, Wang X, Wong J, Xu Z, Zhao Q, Liu R, Zheng X. Epicardial CCM2 Promotes Cardiac Development and Repair Via its Regulation on Cytoskeletal Reorganization. JACC Basic Transl Sci 2024; 9:203-219. [PMID: 38510716 PMCID: PMC10950406 DOI: 10.1016/j.jacbts.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 03/22/2024]
Abstract
The epicardium provides epicardial-derived cells and molecular signals to support cardiac development and regeneration. Zebrafish and mouse studies have shown that ccm2, a cerebral cavernous malformation disease gene, is essential for cardiac development. Endocardial cell-specific deletion of Ccm2 in mice has previously established that Ccm2 is essential for maintenance of the cardiac jelly for cardiac development during early gestation. The current study aimed to explore the function of Ccm2 in epicardial cells for heart development and regeneration. Through genetic deletion of Ccm2 in epicardial cells, our in vivo and ex vivo experiments revealed that Ccm2 is required by epicardial cells to support heart development. Ccm2 regulates epicardial cell adhesion, cell polarity, cell spreading, and migration. Importantly, the loss of Ccm2 in epicardial cells delays cardiac function recovery and aggravates cardiac fibrosis following myocardial infarction. Molecularly, Ccm2 targets the production of cytoskeletal and matrix proteins to maintain epicardial cell function and behaviors. Epicardial Ccm2 plays a critical role in heart development and regeneration via its regulation of cytoskeleton reorganization.
Collapse
Affiliation(s)
- Rui Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Dongbo Lu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Renhua Song
- Epigenetics and RNA Biology Program, Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Luping Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xi Yang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shi-ting Wu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Justin Wong
- Epigenetics and RNA Biology Program, Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Zhelong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiang Zhao
- Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Renjing Liu
- Vascular Epigenetics Laboratory, Victor Chang Cardiac Research Institute and School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
31
|
Zhang J, Tang K, Liu Z, Zhang Z, Duan S, Wang H, Yang H, Yang D, Fan W. Tumor microenvironment-responsive degradable silica nanoparticles: design principles and precision theranostic applications. NANOSCALE HORIZONS 2024; 9:186-214. [PMID: 38164973 DOI: 10.1039/d3nh00388d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Silica nanoparticles have emerged as promising candidates in the field of nanomedicine due to their remarkable versatility and customizable properties. However, concerns about their potential toxicity in healthy tissues and organs have hindered their widespread clinical translation. To address this challenge, significant attention has been directed toward a specific subset of silica nanoparticles, namely degradable silica nanoparticles, primarily because of their excellent biocompatibility and responsive biodegradability. In this review, we provide a comprehensive understanding of degradable silica nanoparticles, categorizing them into two distinct groups: inorganic species-doped and organic moiety-doped silica nanoparticles based on their framework components. Next, the recent progress of tumor microenvironment (TME)-responsive degradable silica nanoparticles for precision theranostic applications is summarized in detail. Finally, current bottlenecks and future opportunities of theranostic nanomedicines based on degradable silica nanoparticles in clinical applications are also outlined and discussed. The aim of this comprehensive review is to shed light on the potential of degradable silica nanoparticles in addressing current challenges in nanomedicine, offering insights into their design, applications in tumor diagnosis and treatment, and paving the way for future advancements in clinical theranostic nanomedicines.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, P. R. China
| | - Kaiyuan Tang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Zilu Liu
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Zhijing Zhang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Shufan Duan
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Hui Wang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu 233030, P. R. China.
| | - Dongliang Yang
- Nanjing Polytechnic Institute, Nanjing 210048, P. R. China.
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing 211198, P. R. China.
| |
Collapse
|
32
|
Li Y, Girard R, Srinath A, Cruz DV, Ciszewski C, Chen C, Lightle R, Romanos S, Sone JY, Moore T, DeBiasse D, Stadnik A, Lee JJ, Shenkar R, Koskimäki J, Lopez-Ramirez MA, Marchuk DA, Ginsberg MH, Kahn ML, Shi C, Awad IA. Transcriptomic signatures of individual cell types in cerebral cavernous malformation. Cell Commun Signal 2024; 22:23. [PMID: 38195510 PMCID: PMC10775676 DOI: 10.1186/s12964-023-01301-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/30/2023] [Indexed: 01/11/2024] Open
Abstract
Cerebral cavernous malformation (CCM) is a hemorrhagic neurovascular disease with no currently available therapeutics. Prior evidence suggests that different cell types may play a role in CCM pathogenesis. The contribution of each cell type to the dysfunctional cellular crosstalk remains unclear. Herein, RNA-seq was performed on fluorescence-activated cell sorted endothelial cells (ECs), pericytes, and neuroglia from CCM lesions and non-lesional brain tissue controls. Differentially Expressed Gene (DEG), pathway and Ligand-Receptor (LR) analyses were performed to characterize the dysfunctional genes of respective cell types within CCMs. Common DEGs among all three cell types were related to inflammation and endothelial-to-mesenchymal transition (EndMT). DEG and pathway analyses supported a role of lesional ECs in dysregulated angiogenesis and increased permeability. VEGFA was particularly upregulated in pericytes. Further pathway and LR analyses identified vascular endothelial growth factor A/ vascular endothelial growth factor receptor 2 signaling in lesional ECs and pericytes that would result in increased angiogenesis. Moreover, lesional pericytes and neuroglia predominantly showed DEGs and pathways mediating the immune response. Further analyses of cell specific gene alterations in CCM endorsed potential contribution to EndMT, coagulation, and a hypoxic microenvironment. Taken together, these findings motivate mechanistic hypotheses regarding non-endothelial contributions to lesion pathobiology and may lead to novel therapeutic targets. Video Abstract.
Collapse
Affiliation(s)
- Ying Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Romuald Girard
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Abhinav Srinath
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Diana Vera Cruz
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Cezary Ciszewski
- Human Disease and Immune Discovery Core, The University of Chicago, Chicago, IL, USA
| | - Chang Chen
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Rhonda Lightle
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Sharbel Romanos
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Je Yeong Sone
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Thomas Moore
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Dorothy DeBiasse
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Agnieszka Stadnik
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Justine J Lee
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Robert Shenkar
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Janne Koskimäki
- Department of Neurosurgery, Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
- Department of Neurosurgery, Oulu University Hospital, Neurocenter, Oulu, Finland
| | - Miguel A Lopez-Ramirez
- Department of Medicine, University of California, La Jolla, San Diego, CA, USA
- Department of Pharmacology, University of California, La Jolla, San Diego, CA, USA
| | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Mark H Ginsberg
- Department of Medicine, University of California, La Jolla, San Diego, CA, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Changbin Shi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Issam A Awad
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA.
- Department of Neurological Surgery, University of Chicago Medicine, 5841 S Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA.
| |
Collapse
|
33
|
Kim Y, Jang Y, Shin YW, Jeon D, Yoo JS, Park DK, Lee HS, Ahn SJ, Park KI, Jung KH, Lee ST, Lee SK, Chu K. Gut microbiome diversity in a febrile seizure mouse model. ENCEPHALITIS 2024; 4:11-17. [PMID: 38195066 PMCID: PMC11007404 DOI: 10.47936/encephalitis.2023.00206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
Purpose Febrile seizures at a young age can provoke late-onset temporal lobe epilepsy. Since recent evidence has suggested that the gut microbiome affects central nervous system pathology across the blood-brain barrier, we hypothesized that febrile seizures alter the composition of the gut microbiome to provoke epilepsy. Methods Third-generation C57BL/6 mice were separated into two groups (n = 5 each), and hot air was applied to only one group to cause febrile seizures. After two weeks of heat challenge, the fecal pellets acquired from each group were analyzed. Results The gut microbiota of fecal pellets from each group revealed five taxa at the genus level and eight taxa at the species level that were significantly different in proportion between the groups. Conclusion Although there was no significant difference in the overall diversity of the gut microbiota between the two groups, the identified heterogeneity may imply the pathognomonic causative relevance of febrile seizures and the development of epilepsy.
Collapse
Affiliation(s)
- Yongmoo Kim
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Yoonhyuk Jang
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Yong-Won Shin
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
- Department of Neurosurgery, Seoul National University Hospital, Seoul, Korea
- Center for Hospital Medicine, Seoul National University Hospital, Seoul, Korea
| | | | - Jung-seok Yoo
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Dong-Kyu Park
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Han Sang Lee
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
- Center for Hospital Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seon-Jae Ahn
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
- Center for Hospital Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyung-Il Park
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
- Division of Neurology, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Keun-Hwa Jung
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Soon-Tae Lee
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Sang Kun Lee
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Kon Chu
- Laboratory for Neurotherapeutics, Department of Neurology, Comprehensive Epilepsy Center, Center for Medical Innovation, Biomedical Research Institute, Seoul National University College of Medicine and Hospital, Seoul, Korea
| |
Collapse
|
34
|
Lazzaroni F, Meessen JMTA, Sun Y, Lanfranconi S, Scola E, D'Alessandris QG, Tassi L, Carriero MR, Castori M, Marino S, Blanda A, Nicolis EB, Novelli D, Calabrese R, Agnelli NM, Bottazzi B, Leone R, Mazzola S, Besana S, Catozzi C, Nezi L, Lampugnani MG, Malinverno M, Grdseloff N, Rödel CJ, Rezai Jahromi B, Bolli N, Passamonti F, Magnusson PU, Abdelilah-Seyfried S, Dejana E, Latini R. Circulating biomarkers in familial cerebral cavernous malformation. EBioMedicine 2024; 99:104914. [PMID: 38113759 PMCID: PMC10767159 DOI: 10.1016/j.ebiom.2023.104914] [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: 08/02/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Cerebral Cavernous Malformation (CCM) is a rare cerebrovascular disease, characterized by the presence of multiple vascular malformations that may result in intracerebral hemorrhages (ICHs), seizure(s), or focal neurological deficits (FND). Familial CCM (fCCM) is due to loss of function mutations in one of the three independent genes KRIT1 (CCM1), Malcavernin (CCM2), or Programmed Cell death 10 (PDCD10/CCM3). The aim of this study was to identify plasma protein biomarkers of fCCM to assess the severity of the disease and predict its progression. METHODS Here, we have investigated plasma samples derived from n = 71 symptomatic fCCM patients (40 female/31 male) and n = 17 healthy donors (HD) (9 female/8 male) of the Phase 1/2 Treat_CCM trial, using multiplexed protein profiling approaches. FINDINGS Biomarkers as sCD14 (p = 0.00409), LBP (p = 0.02911), CXCL4 (p = 0.038), ICAM-1 (p = 0.02013), ANG2 (p = 0.026), CCL5 (p = 0.00403), THBS1 (p = 0.0043), CRP (p = 0.0092), and HDL (p = 0.027), were significantly different in fCCM compared to HDs. Of note, sENG (p = 0.011), THBS1 (p = 0.011) and CXCL4 (p = 0.011), were correlated to CCM genotype. sROBO4 (p = 0.014), TM (p = 0.026) and CRP (p = 0.040) were able to predict incident adverse clinical events, such as ICH, FND or seizure. GDF-15, FLT3L, CXCL9, FGF-21 and CDCP1, were identified as predictors of the formation of new MRI-detectable lesions over 2-year follow-up. Furthermore, the functional relevance of ang2, thbs1, robo4 and cdcp1 markers was validated by zebrafish pre-clinical model of fCCM. INTERPRETATION Overall, our study identifies a set of biochemical parameters to predict CCM progression, suggesting biological interpretations and potential therapeutic approaches to CCM disease. FUNDING Italian Medicines Agency, Associazione Italiana per la Ricerca sul Cancro (AIRC), ERC, Leducq Transatlantic Network of Excellence, Swedish Research Council.
Collapse
Affiliation(s)
- Francesca Lazzaroni
- Vascular Biology Unit, IFOM ETS-The AIRC Institute of Molecular Oncology, Milan, Italy; Hematology Department, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Jennifer M T A Meessen
- Department of Acute Brain and Cardiovascular Injury, Institute for Pharmacological Research Mario Negri IRCCS, Milan, Italy
| | - Ying Sun
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Silvia Lanfranconi
- Department of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Scola
- Department of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Neuroradiology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Quintino Giorgio D'Alessandris
- Department of Neurosurgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Neuroscience, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Laura Tassi
- Claudio Munari Epilepsy Surgery Centre, ASST Niguarda Hospital, Milan, Italy
| | - Maria Rita Carriero
- Cerebrovascular Disease Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Silvia Marino
- IRCCS Centro Neurolesi "Bonino Pulejo", Messina, Italy
| | - Adriana Blanda
- Department of Acute Brain and Cardiovascular Injury, Institute for Pharmacological Research Mario Negri IRCCS, Milan, Italy
| | - Enrico B Nicolis
- Department of Acute Brain and Cardiovascular Injury, Institute for Pharmacological Research Mario Negri IRCCS, Milan, Italy
| | - Deborah Novelli
- Department of Acute Brain and Cardiovascular Injury, Institute for Pharmacological Research Mario Negri IRCCS, Milan, Italy
| | - Roberta Calabrese
- Department of Acute Brain and Cardiovascular Injury, Institute for Pharmacological Research Mario Negri IRCCS, Milan, Italy
| | - Nicolò M Agnelli
- Department of Acute Brain and Cardiovascular Injury, Institute for Pharmacological Research Mario Negri IRCCS, Milan, Italy
| | | | | | - Selene Mazzola
- Laboratory Medicine, Desio Hospital, Università Milano Bicocca, Milan, Italy
| | - Silvia Besana
- Laboratory Medicine, Desio Hospital, Università Milano Bicocca, Milan, Italy
| | - Carlotta Catozzi
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Milano, Italy
| | - Luigi Nezi
- Department of Experimental Oncology, Istituto Europeo di Oncologia IRCCS, Milano, Italy
| | - Maria G Lampugnani
- Vascular Biology Unit, IFOM ETS-The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Acute Brain and Cardiovascular Injury, Institute for Pharmacological Research Mario Negri IRCCS, Milan, Italy
| | - Matteo Malinverno
- Vascular Biology Unit, IFOM ETS-The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Nastasja Grdseloff
- Department of Zoophysiology, Institute of Biochemistry and Biology, University of Potsdam, Germany
| | - Claudia J Rödel
- Department of Zoophysiology, Institute of Biochemistry and Biology, University of Potsdam, Germany
| | | | - Niccolò Bolli
- Hematology Department, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Francesco Passamonti
- Hematology Department, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Salim Abdelilah-Seyfried
- Department of Zoophysiology, Institute of Biochemistry and Biology, University of Potsdam, Germany
| | - Elisabetta Dejana
- Vascular Biology Unit, IFOM ETS-The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Roberto Latini
- Department of Acute Brain and Cardiovascular Injury, Institute for Pharmacological Research Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
35
|
Hage S, Kinkade S, Girard R, Flemming KD, Kim H, Torbey MT, Huang J, Huston J, Shu Y, Selwyn RG, Hart BL, Mabray MC, Feghali J, Sair HI, Narvid J, Lupo JM, Lee J, Stadnik A, Alcazar-Felix RJ, Shenkar R, Hobson N, DeBiasse D, Lane K, McBee NA, Treine K, Ostapkovich N, Wang Y, Thompson RE, Koenig JI, Carroll T, Hanley DF, Awad IA. Trial Readiness of Cavernous Malformations With Symptomatic Hemorrhage, Part II: Biomarkers and Trial Modeling. Stroke 2024; 55:31-39. [PMID: 38134265 PMCID: PMC10752356 DOI: 10.1161/strokeaha.123.044083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/12/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Quantitative susceptibility mapping (QSM) and dynamic contrast-enhanced quantitative perfusion (DCEQP) magnetic resonance imaging sequences assessing iron deposition and vascular permeability were previously correlated with new hemorrhage in cerebral cavernous malformations. We assessed their prospective changes in a multisite trial-readiness project. METHODS Patients with cavernous malformation and symptomatic hemorrhage (SH) in the prior year, without prior or planned lesion resection or irradiation were enrolled. Mean QSM and DCEQP of the SH lesion were acquired at baseline and at 1- and 2-year follow-ups. Sensitivity and specificity of biomarker changes were analyzed in relation to predefined criteria for recurrent SH or asymptomatic change. Sample size calculations for hypothesized therapeutic effects were conducted. RESULTS We logged 143 QSM and 130 DCEQP paired annual assessments. Annual QSM change was greater in cases with SH than in cases without SH (P=0.019). Annual QSM increase by ≥6% occurred in 7 of 7 cases (100%) with recurrent SH and in 7 of 10 cases (70%) with asymptomatic change during the same epoch and 3.82× more frequently than clinical events. DCEQP change had lower sensitivity for SH and asymptomatic change than QSM change and greater variance. A trial with the smallest sample size would detect a 30% difference in QSM annual change during 2 years of follow-up in 34 or 42 subjects (1 and 2 tailed, respectively); power, 0.8, α=0.05. CONCLUSIONS Assessment of QSM change is feasible and sensitive to recurrent bleeding in cavernous malformations. Evaluation of an intervention on QSM percent change may be used as a time-averaged difference between 2 arms using a repeated measures analysis. DCEQP change is associated with lesser sensitivity and higher variability than QSM. These results are the basis of an application for certification by the US Food and Drug Administration of QSM as a biomarker of drug effect on bleeding in cavernous malformations. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03652181.
Collapse
Affiliation(s)
- Stephanie Hage
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| | - Serena Kinkade
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| | | | - Helen Kim
- Department of Anesthesiology and Perioperative Care, Center for Cerebrovascular Research (H.K.), University of California, San Francisco
| | - Michel T Torbey
- Department of Neurology (M.T.T.), University of New Mexico, Albuquerque
| | | | - John Huston
- Radiology (J. Huston, Y.S.), Mayo Clinic, Rochester, MN
| | - Yunhong Shu
- Radiology (J. Huston, Y.S.), Mayo Clinic, Rochester, MN
| | - Reed G Selwyn
- Department of Diagnostic Radiology (R.G.S., B.L.H.), University of New Mexico, Albuquerque
| | - Blaine L Hart
- Department of Diagnostic Radiology (R.G.S., B.L.H.), University of New Mexico, Albuquerque
| | - Marc C Mabray
- Department of Radiology (M.C.M.), University of New Mexico, Albuquerque
| | | | - Haris I Sair
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD (H.I.S.)
| | - Jared Narvid
- Department of Radiology and Biomedical Imaging (J.N., J.M.L.), University of California, San Francisco
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging (J.N., J.M.L.), University of California, San Francisco
| | - Justine Lee
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| | - Agnieszka Stadnik
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| | - Roberto J Alcazar-Felix
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| | - Nicholas Hobson
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| | - Dorothy DeBiasse
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| | - Karen Lane
- Brain Injury Outcomes Unit, Department of Neurology (K.L., N.A.M., K.T., N.O., Y.W., R.E.T., D.F.H.), Johns Hopkins University Medical Institutions, Baltimore, MD
| | - Nichole A McBee
- Brain Injury Outcomes Unit, Department of Neurology (K.L., N.A.M., K.T., N.O., Y.W., R.E.T., D.F.H.), Johns Hopkins University Medical Institutions, Baltimore, MD
| | - Kevin Treine
- Brain Injury Outcomes Unit, Department of Neurology (K.L., N.A.M., K.T., N.O., Y.W., R.E.T., D.F.H.), Johns Hopkins University Medical Institutions, Baltimore, MD
| | - Noeleen Ostapkovich
- Brain Injury Outcomes Unit, Department of Neurology (K.L., N.A.M., K.T., N.O., Y.W., R.E.T., D.F.H.), Johns Hopkins University Medical Institutions, Baltimore, MD
| | - Ying Wang
- Brain Injury Outcomes Unit, Department of Neurology (K.L., N.A.M., K.T., N.O., Y.W., R.E.T., D.F.H.), Johns Hopkins University Medical Institutions, Baltimore, MD
| | - Richard E Thompson
- Brain Injury Outcomes Unit, Department of Neurology (K.L., N.A.M., K.T., N.O., Y.W., R.E.T., D.F.H.), Johns Hopkins University Medical Institutions, Baltimore, MD
| | - James I Koenig
- National Institute of Neurological Disorders and Stroke, Bethesda, MD (J.K.)
| | - Timothy Carroll
- Department of Diagnostic Radiology (T.C.), University of Chicago Medicine and Biological Sciences, IL
| | - Daniel F Hanley
- Brain Injury Outcomes Unit, Department of Neurology (K.L., N.A.M., K.T., N.O., Y.W., R.E.T., D.F.H.), Johns Hopkins University Medical Institutions, Baltimore, MD
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurological Surgery (S.H., S.K., R.G., J.L., A.S., R.J.A.-F., R.S., N.H., D.D., I.A.A.), University of Chicago Medicine and Biological Sciences, IL
| |
Collapse
|
36
|
Lee J, Lee H, Shin M, Park S. Cerebral Cavernous Malformation (CCM)-like Vessel Lesion in the Aged ANKS1A-deficient Brain. Exp Neurobiol 2023; 32:441-452. [PMID: 38196138 PMCID: PMC10789174 DOI: 10.5607/en23032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Abstract
In this study, we show that ANKS1A is specifically expressed in the brain endothelial cells of adult mice. ANKS1A deficiency in adult mice does not affect the differentiation, growth, or patterning of the cerebrovascular system; however, its absence significantly impacts the cerebrovascular system of the aged brain. In aged ANKS1A knock-out (KO) brains, vessel lesions exhibiting cerebral cavernous malformations (CCMs) are observed. In addition, CCM-like lesions show localized peripheral blood leakage into the brain. The CCM-like lesions reveal immune cells infiltrating the parenchyma. The CCM-like lesions also contain significantly fewer astrocyte endfeets and tight junctions, indicating that the integrity of the BBB has been partially compromised. CCM-like lesions display increased fibronectin expression in blood vessels, which is also confirmed in cultured endothelial cells deficient for ANKS1A. Therefore, we hypothesize that ANKS1A may play a role in maintaining or stabilizing healthy blood vessels in the brain during aging.
Collapse
Affiliation(s)
- Jiyeon Lee
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Haeryung Lee
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Miram Shin
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| | - Soochul Park
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea
| |
Collapse
|
37
|
Lin J, Weng X, Zheng J, Wu S, Bao Q, Peng F, Huang Y. Case report: Thrombolysis in patients with acute ischemic stroke and cerebral cavernous malformation. Front Neurol 2023; 14:1281412. [PMID: 38164201 PMCID: PMC10758226 DOI: 10.3389/fneur.2023.1281412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Background Cerebral cavernous malformation (CCM) is a rare disease associated with a latent risk of intracranial hemorrhage. However, due to limited evidence, the safety of recommending intravenous tissue plasminogen activators for patients with acute stroke and CCM remains uncertain. Methods Our study identified five patients with acute stroke and CCM treated between 2017 and 2023 across two hospitals. A comprehensive literature review was conducted, incorporating three similar case reports and two retrospective studies. Results Among 30 patients reviewed, three exhibited symptomatic intracranial hemorrhage, two of whom were women. Additionally, three patients presented with calcification in their CCM, with two experiencing symptomatic intracranial hemorrhage. Conclusion The observed incidence of symptomatic intracranial hemorrhage following intravenous tissue plasminogen activator administration appears to be elevated in patients with CCM. Therefore, before thrombolysis, a thorough evaluation of personalized risk-benefit ratios is crucial. Furthermore, conducting further research involving multiple centers and larger sample sizes is imperative to advance our understanding in this area, especially in identifying hemorrhage risk factors.
Collapse
Affiliation(s)
- Jie Lin
- Department of Neurology, Yueqing People’s Hospital, Wenzhou, China
| | - Xiongpeng Weng
- Department of Neurology, Affiliated Huangyan Hospital of Wenzhou Medical University, Taizhou First People's Hospital, Taizhou, China
| | - Jing Zheng
- Department of Neurology, Yueqing People’s Hospital, Wenzhou, China
| | - Saizhen Wu
- Department of Neurology, Yueqing People’s Hospital, Wenzhou, China
| | - Qiongqiong Bao
- Department of Neurology, Yueqing People’s Hospital, Wenzhou, China
| | - Feifei Peng
- Department of Neurology, Affiliated Huangyan Hospital of Wenzhou Medical University, Taizhou First People's Hospital, Taizhou, China
| | - Yanbin Huang
- Department of Neurology, Yueqing People’s Hospital, Wenzhou, China
| |
Collapse
|
38
|
Sirko S, Schichor C, Della Vecchia P, Metzger F, Sonsalla G, Simon T, Bürkle M, Kalpazidou S, Ninkovic J, Masserdotti G, Sauniere JF, Iacobelli V, Iacobelli S, Delbridge C, Hauck SM, Tonn JC, Götz M. Injury-specific factors in the cerebrospinal fluid regulate astrocyte plasticity in the human brain. Nat Med 2023; 29:3149-3161. [PMID: 38066208 PMCID: PMC10719094 DOI: 10.1038/s41591-023-02644-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 10/13/2023] [Indexed: 12/17/2023]
Abstract
The glial environment influences neurological disease progression, yet much of our knowledge still relies on preclinical animal studies, especially regarding astrocyte heterogeneity. In murine models of traumatic brain injury, beneficial functions of proliferating reactive astrocytes on disease outcome have been unraveled, but little is known regarding if and when they are present in human brain pathology. Here we examined a broad spectrum of pathologies with and without intracerebral hemorrhage and found a striking correlation between lesions involving blood-brain barrier rupture and astrocyte proliferation that was further corroborated in an assay probing for neural stem cell potential. Most importantly, proteomic analysis unraveled a crucial signaling pathway regulating this astrocyte plasticity with GALECTIN3 as a novel marker for proliferating astrocytes and the GALECTIN3-binding protein LGALS3BP as a functional hub mediating astrocyte proliferation and neurosphere formation. Taken together, this work identifies a therapeutically relevant astrocyte response and their molecular regulators in different pathologies affecting the human cerebral cortex.
Collapse
Affiliation(s)
- Swetlana Sirko
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.
- Institute of Stem Cell Research, Helmholtz Center München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany.
| | - Christian Schichor
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Patrizia Della Vecchia
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | | | - Giovanna Sonsalla
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Tatiana Simon
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Martina Bürkle
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Sofia Kalpazidou
- Chair of Cell Biology, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Jovica Ninkovic
- Institute of Stem Cell Research, Helmholtz Center München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
- Chair of Cell Biology, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- SYNERGY Excellence Cluster of Systems Neurology, LMU Munich, Munich, Germany
| | - Giacomo Masserdotti
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
- Institute of Stem Cell Research, Helmholtz Center München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | | | | | | | - Claire Delbridge
- Department of Neuropathology, Institute of Pathology, TUM School of Medicine, TU Munich, Munich, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Magdalena Götz
- Chair of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.
- Institute of Stem Cell Research, Helmholtz Center München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany.
- SYNERGY Excellence Cluster of Systems Neurology, LMU Munich, Munich, Germany.
| |
Collapse
|
39
|
Boubshait N, Alhassar H, Alsubaie N, Jan A, Asad F. A Case Report of Hemorrhagic Cavernoma Masquerading as a Thrombosed Aneurysm. Cureus 2023; 15:e50548. [PMID: 38226114 PMCID: PMC10788674 DOI: 10.7759/cureus.50548] [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] [Accepted: 12/14/2023] [Indexed: 01/17/2024] Open
Abstract
Cavernous malformations are vascular lesions characterized by dilated blood vessels with minimal intervening brain parenchyma. Although often asymptomatic, they can present with seizures, headaches, or neurological deficits. Accurate diagnosis relies on magnetic resonance imaging, with characteristic features such as a "popcorn" appearance. We present a case of a 45-year-old male with chronic headaches and seizures who underwent an extensive work-up. Initial magnetic resonance imaging suggested a thrombosed aneurysm, with subsequent cerebral angiography being unremarkable, supporting the final diagnosis of a cavernous malformation. Conservative management, initiated for asymptomatic lesions, led to effective seizure control and improved quality of life. This case underscores diagnostic complexities in neuroradiology, emphasizing the need for careful consideration of differentials when faced with unexpected imaging results. Clinicians must remain vigilant for alternative explanations, recognizing the dynamic nature of optimal strategies in neurovascular medicine.
Collapse
Affiliation(s)
| | - Hassan Alhassar
- General Practice, Imam Abdulrahman Bin Faisal University, Dammam, SAU
| | | | - Abdulla Jan
- Family Medicine, University of Jeddah, Jeddah, SAU
| | - Farah Asad
- Public Health, Primary Health Care Center, Hofuf, SAU
| |
Collapse
|
40
|
Glanz H, Damodharan SN, Smith-Simmer K, Helgager J, Puccetti D. Concurrent high-grade glioma with cavernous malformations and pathogenic variants in PDCD10 and SMARCA4. Childs Nerv Syst 2023; 39:3311-3315. [PMID: 37328659 DOI: 10.1007/s00381-023-06026-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/10/2023] [Indexed: 06/18/2023]
Abstract
The co-occurrence of multiple disease processes can pose diagnostic challenges. We report an unusual case of a patient found to have co-occurrences of an IDH1-mutant high-grade glioma along with cerebral cavernous malformations and pathogenic germline variants in PDCD10 and SMARCA4. Somatic testing was done on the tumor and identified a SMARCA4 and two TP53 variants. Within the literature, little is known about the association of high-grade gliomas with these germline variants. Such findings furthermore not only inform complex diagnoses, but have the potential to play a crucial role in the ongoing care of a patient.
Collapse
Affiliation(s)
- Hannah Glanz
- Department of Pediatrics, School of Medicine & Public Health, University of Wisconsin, Madison, WI, USA
| | - Sudarshawn N Damodharan
- Division of Pediatric Hematology, Oncology and Bone Marrow Transplant, Department of Pediatrics, School of Medicine & Public Health, University of Wisconsin, UW Carbone Cancer Center, 600 Highland Ave, Madison, WI, 53792, USA.
| | - Kelcy Smith-Simmer
- Master of Genetic Counselor Studies, Department of Pediatrics, School of Medicine & Public Health, University of Wisconsin, UW Carbone Cancer Center, Madison, WI, USA
| | - Jeffrey Helgager
- Department of Pathology, School of Medicine & Public Health, University of Wisconsin, Madison, WI, USA
| | - Diane Puccetti
- Division of Pediatric Hematology, Oncology and Bone Marrow Transplant, Department of Pediatrics, School of Medicine & Public Health, University of Wisconsin, UW Carbone Cancer Center, 600 Highland Ave, Madison, WI, 53792, USA
| |
Collapse
|
41
|
Li L, Ren AA, Gao S, Su YS, Yang J, Bockman J, Mericko-Ishizuka P, Griffin J, Shenkar R, Alcazar R, Moore T, Lightle R, DeBiasse D, Awad IA, Marchuk DA, Kahn ML, Burkhardt JK. mTORC1 Inhibitor Rapamycin Inhibits Growth of Cerebral Cavernous Malformation in Adult Mice. Stroke 2023; 54:2906-2917. [PMID: 37746705 PMCID: PMC10599232 DOI: 10.1161/strokeaha.123.044108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Cerebral cavernous malformations (CCMs) are vascular malformations that frequently cause stroke. CCMs arise due to loss of function in one of the genes that encode the CCM complex, a negative regulator of MEKK3-KLF2/4 signaling in vascular endothelial cells. Gain-of-function mutations in PIK3CA (encoding the enzymatic subunit of the PI3K (phosphoinositide 3-kinase) pathway associated with cell growth) synergize with CCM gene loss-of-function to generate rapidly growing lesions. METHODS We recently developed a model of CCM formation that closely reproduces key events in human CCM formation through inducible CCM loss-of-function and PIK3CA gain-of-function in mature mice. In the present study, we use this model to test the ability of rapamycin, a clinically approved inhibitor of the PI3K effector mTORC1, to treat rapidly growing CCMs. RESULTS We show that both intraperitoneal and oral administration of rapamycin arrests CCM growth, reduces perilesional iron deposition, and improves vascular perfusion within CCMs. CONCLUSIONS Our findings further establish this adult CCM model as a valuable preclinical model and support clinical testing of rapamycin to treat rapidly growing human CCMs.
Collapse
Affiliation(s)
- Lun Li
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
- Department of Neurosurgery, Perelman School of Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| | - Aileen A. Ren
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| | - Siqi Gao
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| | - Yourong S. Su
- Department of Neurosurgery, Perelman School of Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| | - Jisheng Yang
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| | - Jenna Bockman
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| | - Patricia Mericko-Ishizuka
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| | - Joanna Griffin
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA 60637
| | - Roberto Alcazar
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA 60637
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA 60637
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA 60637
| | - Dorothy DeBiasse
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA 60637
| | - Issam A. Awad
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA 60637
| | - Douglas A. Marchuk
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA 27708
| | - Mark L. Kahn
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| | - Jan-Karl Burkhardt
- Department of Neurosurgery, Perelman School of Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania, Philadelphia, Pennsylvania, USA 19104
| |
Collapse
|
42
|
Yao BH, Li D, Wang L, Wu Z. Extra-axial cavernous malformations of the foramen magnum: illustrative cases. JOURNAL OF NEUROSURGERY. CASE LESSONS 2023; 6:CASE23441. [PMID: 37871342 PMCID: PMC10599450 DOI: 10.3171/case23441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/14/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Extra-axial cavernous malformations involving the foramen magnum are rare, and preoperative diagnosis becomes difficult when they mimic meningiomas. OBSERVATIONS The authors present 2 cases of extra-axial cavernous malformations involving the foramen magnum. Surgical removal of the lesions was performed via far lateral craniotomy. The authors investigate the disease and elaborate the differential diagnosis. LESSONS The authors recommend that extra-axial cavernous malformations should be considered in the differential diagnosis of lesions in the foramen magnum region. Intraoperative frozen sections are helpful to the diagnosis, and resection warranted a favorable long-term outcome.
Collapse
Affiliation(s)
- Bo-Han Yao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Fengtai District, Beijing, China; and
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Da Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Fengtai District, Beijing, China; and
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Fengtai District, Beijing, China; and
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Fengtai District, Beijing, China; and
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
43
|
Gawade K, Raczynska KD. Imprinted small nucleolar RNAs: Missing link in development and disease? WILEY INTERDISCIPLINARY REVIEWS. RNA 2023:e1818. [PMID: 37722601 DOI: 10.1002/wrna.1818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/20/2023]
Abstract
The 14q32.2 (DLK1-DIO3) and 15q11-q13 (SNURF-SNRPN) imprinted gene loci harbor the largest known small nucleolar RNA clusters expressed from the respective maternal and paternal alleles. Recent studies have demonstrated significant roles for the 15q11-q13 located SNORD115-SNORD116 C/D box snoRNAs in Prader-Willi syndrome (PWS), a neurodevelopmental disorder. Even though the effect of SNORD116 deletion is apparent in the PWS phenotype, similar effects of a SNORD113-SNORD114 cluster deletion from the 14q32.2 locus in Kagami-Ogata syndrome (KOS14) and upregulation in Temple syndrome (TS14) remain to be explored. Moreover, apart from their probable involvement in neurodevelopmental disorders, snoRNAs from the SNORD113-SNORD114 cluster have been implicated in multiple biological processes, including pluripotency, development, cancers, and RNA modifications. Here we summarize the current understanding of the system to explore the possibility of a link between developmental disorders and C/D box snoRNA expression from the imprinted 14q32.2 locus. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development RNA Processing > Processing of Small RNAs.
Collapse
Affiliation(s)
- Kishor Gawade
- Laboratory of RNA Processing, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University in Poznan, Poznan, Poland
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Poznan, Poland
| | - Katarzyna D Raczynska
- Laboratory of RNA Processing, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University in Poznan, Poznan, Poland
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Poznan, Poland
| |
Collapse
|
44
|
Li Y, Srinath A, Alcazar-Felix RJ, Hage S, Bindal A, Lightle R, Shenkar R, Shi C, Girard R, Awad IA. Inflammatory Mechanisms in a Neurovascular Disease: Cerebral Cavernous Malformation. Brain Sci 2023; 13:1336. [PMID: 37759937 PMCID: PMC10526329 DOI: 10.3390/brainsci13091336] [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: 07/21/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Cerebral cavernous malformation (CCM) is a common cerebrovascular malformation causing intracranial hemorrhage, seizures, and focal neurologic deficits. A unique CCM lesional inflammatory microenvironment has been shown to influence the clinical course of the disease. This review addresses the inflammatory cell infiltrate in the CCM lesion and the role of a defined antigen-driven immune response in pathogenicity. We summarize immune mechanisms associated with the loss of the CCM gene and disease progression, including the potential role of immunothrombosis. We also review evidence of circulating inflammatory biomarkers associated with CCM disease and its clinical activity. We articulate future directions for this research, including the role of individual cell type contributions to the immune response in CCM, single cell transcriptomics of inflammatory cells, biomarker development, and therapeutic implications. The concepts are applicable for developing diagnostic and treatment strategies for CCM and for studying other neurovascular diseases.
Collapse
Affiliation(s)
- Ying Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (Y.L.); (C.S.)
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, Chicago, IL 60637, USA; (A.S.); (R.J.A.-F.); (S.H.); (A.B.); (R.L.); (R.S.); (R.G.)
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, Chicago, IL 60637, USA; (A.S.); (R.J.A.-F.); (S.H.); (A.B.); (R.L.); (R.S.); (R.G.)
| | - Roberto J. Alcazar-Felix
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, Chicago, IL 60637, USA; (A.S.); (R.J.A.-F.); (S.H.); (A.B.); (R.L.); (R.S.); (R.G.)
| | - Stephanie Hage
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, Chicago, IL 60637, USA; (A.S.); (R.J.A.-F.); (S.H.); (A.B.); (R.L.); (R.S.); (R.G.)
| | - Akash Bindal
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, Chicago, IL 60637, USA; (A.S.); (R.J.A.-F.); (S.H.); (A.B.); (R.L.); (R.S.); (R.G.)
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, Chicago, IL 60637, USA; (A.S.); (R.J.A.-F.); (S.H.); (A.B.); (R.L.); (R.S.); (R.G.)
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, Chicago, IL 60637, USA; (A.S.); (R.J.A.-F.); (S.H.); (A.B.); (R.L.); (R.S.); (R.G.)
| | - Changbin Shi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (Y.L.); (C.S.)
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, Chicago, IL 60637, USA; (A.S.); (R.J.A.-F.); (S.H.); (A.B.); (R.L.); (R.S.); (R.G.)
| | - Issam A. Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago, Chicago, IL 60637, USA; (A.S.); (R.J.A.-F.); (S.H.); (A.B.); (R.L.); (R.S.); (R.G.)
- Department of Neurological Surgery, University of Chicago Medicine, 5841 S Maryland, MC3026/Neurosurgery J341, Chicago, IL 60637, USA
| |
Collapse
|
45
|
Ren J, Huang Y, Ren Y, Tu T, Qiu B, Ai D, Bi Z, Bai X, Li F, Li JL, Chen XJ, Feng Z, Guo Z, Lei J, Tian A, Cui Z, Lindner V, Adams RH, Wang Y, Zhao F, Körbelin J, Sun W, Wang Y, Zhang H, Hong T, Ge WP. Somatic variants of MAP3K3 are sufficient to cause cerebral and spinal cord cavernous malformations. Brain 2023; 146:3634-3647. [PMID: 36995941 PMCID: PMC10473567 DOI: 10.1093/brain/awad104] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 03/07/2023] [Accepted: 03/12/2023] [Indexed: 03/31/2023] Open
Abstract
Cerebral cavernous malformations (CCMs) and spinal cord cavernous malformations (SCCMs) are common vascular abnormalities of the CNS that can lead to seizure, haemorrhage and other neurological deficits. Approximately 85% of patients present with sporadic (versus congenital) CCMs. Somatic mutations in MAP3K3 and PIK3CA were recently reported in patients with sporadic CCM, yet it remains unknown whether MAP3K3 mutation is sufficient to induce CCMs. Here we analysed whole-exome sequencing data for patients with CCM and found that ∼40% of them have a single, specific MAP3K3 mutation [c.1323C>G (p.Ile441Met)] but not any other known mutations in CCM-related genes. We developed a mouse model of CCM with MAP3K3I441M uniquely expressed in the endothelium of the CNS. We detected pathological phenotypes similar to those found in patients with MAP3K3I441M. The combination of in vivo imaging and genetic labelling revealed that CCMs were initiated with endothelial expansion followed by disruption of the blood-brain barrier. Experiments with our MAP3K3I441M mouse model demonstrated that CCM can be alleviated by treatment with rapamycin, the mTOR inhibitor. CCM pathogenesis has usually been attributed to acquisition of two or three distinct genetic mutations involving the genes CCM1/2/3 and/or PIK3CA. However, our results demonstrate that a single genetic hit is sufficient to cause CCMs.
Collapse
Affiliation(s)
- Jian Ren
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing 100053, China
| | - Yazi Huang
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Yeqing Ren
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing 100053, China
| | - Tianqi Tu
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing 100053, China
| | - Baoshan Qiu
- Chinese Institute for Brain Research, Beijing 102206, China
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Daosheng Ai
- Chinese Institute for Brain Research, Beijing 102206, China
- Academy for Advanced Interdisciplinary Studies (AAIS), Peking University, Beijing 100871, China
| | - Zhanying Bi
- Chinese Institute for Brain Research, Beijing 102206, China
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xue Bai
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Fengzhi Li
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Jun-Liszt Li
- Chinese Institute for Brain Research, Beijing 102206, China
- Academy for Advanced Interdisciplinary Studies (AAIS), Peking University, Beijing 100871, China
| | - Xing-jun Chen
- Chinese Institute for Brain Research, Beijing 102206, China
- Academy for Advanced Interdisciplinary Studies (AAIS), Peking University, Beijing 100871, China
| | - Ziyan Feng
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Zongpei Guo
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Jianfeng Lei
- Medical Imaging laboratory of Core Facility Center, Capital Medical University, Beijing 100054, China
| | - An Tian
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing 100053, China
| | - Ziwei Cui
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing 100053, China
| | - Volkhard Lindner
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, and Faculty of Medicine, University of Münster, D-48149 Münster, Germany
| | - Yibo Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Fei Zhao
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Wenzhi Sun
- Chinese Institute for Brain Research, Beijing 102206, China
- School of Basic Medical Sciences, Capital Medical University, Beijing 100054, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing 100053, China
| | - Tao Hong
- Department of Neurosurgery, Xuanwu Hospital, China International Neuroscience Institute, Capital Medical University, Beijing 100053, China
| | - Woo-ping Ge
- Chinese Institute for Brain Research, Beijing 102206, China
- Department of Neurosurgery, Xuanwu Hospital, Beijing Institute of Brain Disorders (BIBD), China International Neuroscience Institute, Capital Medical University, Beijing 100053, China
| |
Collapse
|
46
|
Sun Z, Lin PK, Yrigoin K, Kemp SS, Davis GE. Increased Matrix Metalloproteinase-1 Activation Enhances Disruption and Regression of k-RasV12-Expressing Arteriovenous Malformation-Like Vessels. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1319-1334. [PMID: 37328101 PMCID: PMC10477956 DOI: 10.1016/j.ajpath.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023]
Abstract
This study sought to identify potential mechanisms by which k-RasV12-expressing endothelial cell (EC) tubes demonstrate an increased propensity to regress compared with controls. Activated k-Ras mutations play a role in a variety of pathological conditions, including arteriovenous malformations, which are prone to bleed, causing serious hemorrhagic complications. ECs expressing active k-RasV12 demonstrate markedly excessive lumen formation with widened and shortened tubes accompanied by reduced pericyte recruitment and basement membrane deposition, leading to deficient capillary network assembly. The current study showed that active k-Ras-expressing ECs secreted greater amounts of MMP-1 proenzyme compared with control ECs, and readily converted it to increased active MMP-1 levels through the action of plasmin or plasma kallikrein (generated from their added zymogens). Active MMP-1 degraded three-dimensional collagen matrices, leading to more rapid and extensive regression of the active k-Ras-expressing EC tubes, in conjunction with matrix contraction, compared with control ECs. Under conditions where pericytes protect control EC tubes from plasminogen- and MMP-1-dependent tube regression, this failed to occur with k-RasV12 ECs, due to reduced pericyte interactions. In summary, k-RasV12-expressing EC vessels showed an increased propensity to regress in response to serine proteinases through accentuated levels of active MMP-1, a novel pathogenic mechanism that may underlie hemorrhagic events associated with arteriovenous malformation lesions.
Collapse
Affiliation(s)
- Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Ksenia Yrigoin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
47
|
Romanos SG, Srinath A, Li Y, Xie B, Chen C, Li Y, Moore T, Bi D, Sone JY, Lightle R, Hobson N, Zhang D, Koskimäki J, Shen L, McCurdy S, Lai CC, Stadnik A, Piedad K, Carrión-Penagos J, Shkoukani A, Snellings D, Shenkar R, Sulakhe D, Ji Y, Lopez-Ramirez MA, Kahn ML, Marchuk DA, Ginsberg MH, Girard R, Awad IA. Circulating Plasma miRNA Homologs in Mice and Humans Reflect Familial Cerebral Cavernous Malformation Disease. Transl Stroke Res 2023; 14:513-529. [PMID: 35715588 PMCID: PMC9758276 DOI: 10.1007/s12975-022-01050-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 01/16/2023]
Abstract
Patients with familial cerebral cavernous malformation (CCM) inherit germline loss of function mutations and are susceptible to progressive development of brain lesions and neurological sequelae during their lifetime. To date, no homologous circulating molecules have been identified that can reflect the presence of germ line pathogenetic CCM mutations, either in animal models or patients. We hypothesize that homologous differentially expressed (DE) plasma miRNAs can reflect the CCM germline mutation in preclinical murine models and patients. Herein, homologous DE plasma miRNAs with mechanistic putative gene targets within the transcriptome of preclinical and human CCM lesions were identified. Several of these gene targets were additionally found to be associated with CCM-enriched pathways identified using the Kyoto Encyclopedia of Genes and Genomes. DE miRNAs were also identified in familial-CCM patients who developed new brain lesions within the year following blood sample collection. The miRNome results were then validated in an independent cohort of human subjects with real-time-qPCR quantification, a technique facilitating plasma assays. Finally, a Bayesian-informed machine learning approach showed that a combination of plasma levels of miRNAs and circulating proteins improves the association with familial-CCM disease in human subjects to 95% accuracy. These findings act as an important proof of concept for the future development of translatable circulating biomarkers to be tested in preclinical studies and human trials aimed at monitoring and restoring gene function in CCM and other diseases.
Collapse
Affiliation(s)
- Sharbel G Romanos
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Abhinav Srinath
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Ying Li
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Bingqing Xie
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Chang Chen
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Bioinformatics Core, Biological Sciences Division, University of Chicago, Chicago, IL, USA
| | - Yan Li
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Bioinformatics Core, Biological Sciences Division, University of Chicago, Chicago, IL, USA
| | - Thomas Moore
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Dehua Bi
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Je Yeong Sone
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Rhonda Lightle
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Nick Hobson
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Dongdong Zhang
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Janne Koskimäki
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Le Shen
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Sara McCurdy
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Catherine Chinhchu Lai
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Agnieszka Stadnik
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Kristina Piedad
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Julián Carrión-Penagos
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Abdallah Shkoukani
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Daniel Snellings
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Robert Shenkar
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Dinanath Sulakhe
- Bioinformatics Core, Biological Sciences Division, University of Chicago, Chicago, IL, USA
| | - Yuan Ji
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Miguel A Lopez-Ramirez
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas A Marchuk
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Mark H Ginsberg
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Romuald Girard
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Issam A Awad
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA.
| |
Collapse
|
48
|
Hage S, Hagan M, Bi D, Stadnik A, Lee J, Romanos S, Srinath A, Shenkar R, Lee C, Horowitz PM, Girard R, Awad IA. Impact of socioeconomics and race on clinical follow-up and trial enrollment and adherence in cerebral cavernous malformation. J Stroke Cerebrovasc Dis 2023; 32:107167. [PMID: 37146402 PMCID: PMC10201538 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/07/2023] Open
Abstract
OBJECTIVES Cerebral cavernous malformation (CCM) affects more than a million Americans but advanced care for symptomatic lesions and access to research studies is largely limited to referral academic centers MATERIALS AND METHODS: A cohort of CCM patients screened for research studies at an accredited center of excellence for CCM was analyzed. Demographics, lesion location, history of hemorrhage, insurance type and area of deprivation index (ADI) were collected. Primary outcomes were clinical follow-up within a year from initial evaluation, and enrollment and adherence in clinical trials among eligible subjects RESULTS: A majority (52.8%) of CCM patients evaluated had a high socioeconomic status (SES) (ADI 1-3), and only 11.5% were African American. Patients who had a symptomatic bleed were more likely to follow-up (p=0.01), and those with brainstem lesion were more likely to enroll/adhere in a clinical trial (p=0.02). Rates of clinical follow-up were similar across different ADI groups, insurance coverage and race. Patients who were uninsured/self-paying, and African Americans were more likely to decline/drop from clinical trials (OR 2.4, 95% CI 0.46-10.20 and OR 2.2, 95% CI 0.33-10.75, respectively), but differences were not statistically significant CONCLUSIONS: Access of disadvantaged patients to center of excellence care and research remains limited despite geographic proximity to their community. Patients with lower SES and African Americans are as likely to follow-up clinically, but there were trends of differences in enrollment/adherence in clinical trials. Mitigation efforts should target systemic causes of low access to specialized care among uninsured and African American patients.
Collapse
Affiliation(s)
- Stephanie Hage
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Matthew Hagan
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Dehua Bi
- Department of Public Health Sciences, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Agnieszka Stadnik
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Justine Lee
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Cornelia Lee
- Alliance to Cure Cavernous Malformations, Charlottesville, Virginia, USA.
| | - Peleg M Horowitz
- Neurotrauma Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA.
| |
Collapse
|
49
|
Hage S, Kinkade S, Girard R, Flemming KD, Kim H, Torbey MT, Huang J, Huston J, Shu Y, Selwyn RG, Hart BL, Mabray MC, Feghali J, Sair HI, Narvid J, Lupo JM, Lee J, Stadnik A, Alcazar R, Shenkar R, Hobson N, DeBiasse D, Lane K, McBee N, Treine K, Ostapkovich N, Wang Y, Thompson RE, Mendoza-Puccini C, Koenig J, Carroll T, Hanley DF, Awad IA. Cavernous Angioma Symptomatic Hemorrhage (CASH) Trial Readiness II: Imaging Biomarkers and Trial Modeling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.01.23290854. [PMID: 37333396 PMCID: PMC10275015 DOI: 10.1101/2023.06.01.23290854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Quantitative susceptibility mapping (QSM) and dynamic contrast enhanced quantitative perfusion (DCEQP) MRI sequences assessing iron deposition and vascular permeability were previously correlated with new hemorrhage in cavernous angiomas. We assessed their prospective changes in cavernous angiomas with symptomatic hemorrhage (CASH) in a multisite trial readiness project ( clinicaltrials.gov NCT03652181 ). Methods Patients with CASH in the prior year, without prior or planned lesion resection or irradiation were enrolled. Mean QSM and DCEQP of CASH lesion were acquired at baseline, and at 1- and 2-year follow-ups. Sensitivity and specificity of biomarker changes were analyzed in relation to predefined lesional symptomatic hemorrhage (SH) or asymptomatic change (AC). Sample size calculations for hypothesized therapeutic effects were conducted. Results We logged 143 QSM and 130 DCEQP paired annual assessments. Annual QSM change was greater in cases with SH than in cases without SH (p= 0.019). Annual QSM increase by ≥ 6% occurred in 7 of 7 cases (100%) with recurrent SH and in 7 of 10 cases (70%) with AC during the same epoch, and 3.82 times more frequently than clinical events. DCEQP change had lower sensitivity for SH and AC than QSM change, and greater variance. A trial with smallest sample size would detect a 30% difference in QSM annual change in 34 or 42 subjects (one and two-tailed, respectively), power 0.8, alpha 0.05. Conclusions Assessment of QSM change is feasible and sensitive to recurrent bleeding in CASH. Evaluation of an intervention on QSM percent change may be used as a time-averaged difference between 2 arms using a repeated measures analysis. DCEQP change is associated with lesser sensitivity and higher variability than QSM. These results are the basis of an application for certification by the U.S. F.D.A. of QSM as a biomarker of drug effect in CASH.
Collapse
|
50
|
Ding J, Lee SJ, Vlahos L, Yuki K, Rada CC, van Unen V, Vuppalapaty M, Chen H, Sura A, McCormick AK, Tomaske M, Alwahabi S, Nguyen H, Nowatzke W, Kim L, Kelly L, Vollrath D, Califano A, Yeh WC, Li Y, Kuo CJ. Therapeutic blood-brain barrier modulation and stroke treatment by a bioengineered FZD 4-selective WNT surrogate in mice. Nat Commun 2023; 14:2947. [PMID: 37268690 PMCID: PMC10238527 DOI: 10.1038/s41467-023-37689-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/27/2023] [Indexed: 06/04/2023] Open
Abstract
Derangements of the blood-brain barrier (BBB) or blood-retinal barrier (BRB) occur in disorders ranging from stroke, cancer, diabetic retinopathy, and Alzheimer's disease. The Norrin/FZD4/TSPAN12 pathway activates WNT/β-catenin signaling, which is essential for BBB and BRB function. However, systemic pharmacologic FZD4 stimulation is hindered by obligate palmitoylation and insolubility of native WNTs and suboptimal properties of the FZD4-selective ligand Norrin. Here, we develop L6-F4-2, a non-lipidated, FZD4-specific surrogate which significantly improves subpicomolar affinity versus native Norrin. In Norrin knockout (NdpKO) mice, L6-F4-2 not only potently reverses neonatal retinal angiogenesis deficits, but also restores BRB and BBB function. In adult C57Bl/6J mice, post-stroke systemic delivery of L6-F4-2 strongly reduces BBB permeability, infarction, and edema, while improving neurologic score and capillary pericyte coverage. Our findings reveal systemic efficacy of a bioengineered FZD4-selective WNT surrogate during ischemic BBB dysfunction, with potential applicability to adult CNS disorders characterized by an aberrant blood-brain barrier.
Collapse
Affiliation(s)
- Jie Ding
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sung-Jin Lee
- Surrozen, Inc. South San Francisco, South San Francisco, CA, 94080, USA
| | - Lukas Vlahos
- Department of Systems Biology, Columbia University, Columbia, NY, 10032, USA
| | - Kanako Yuki
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Cara C Rada
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Vincent van Unen
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | - Hui Chen
- Surrozen, Inc. South San Francisco, South San Francisco, CA, 94080, USA
| | - Asmiti Sura
- Surrozen, Inc. South San Francisco, South San Francisco, CA, 94080, USA
| | - Aaron K McCormick
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Madeline Tomaske
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Samira Alwahabi
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Huy Nguyen
- Surrozen, Inc. South San Francisco, South San Francisco, CA, 94080, USA
| | - William Nowatzke
- Surrozen, Inc. South San Francisco, South San Francisco, CA, 94080, USA
| | - Lily Kim
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Lisa Kelly
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Douglas Vollrath
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University, Columbia, NY, 10032, USA
| | - Wen-Chen Yeh
- Surrozen, Inc. South San Francisco, South San Francisco, CA, 94080, USA
| | - Yang Li
- Surrozen, Inc. South San Francisco, South San Francisco, CA, 94080, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|