1
|
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
|
2
|
Glading A. KRIT1 in vascular biology and beyond. Biosci Rep 2024; 44:BSR20231675. [PMID: 38980708 PMCID: PMC11263069 DOI: 10.1042/bsr20231675] [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: 02/10/2024] [Revised: 06/21/2024] [Accepted: 07/09/2024] [Indexed: 07/10/2024] Open
Abstract
KRIT1 is a 75 kDa scaffolding protein which regulates endothelial cell phenotype by limiting the response to inflammatory stimuli and maintaining a quiescent and stable endothelial barrier. Loss-of-function mutations in KRIT1 lead to the development of cerebral cavernous malformations (CCM), a disease marked by the formation of abnormal blood vessels which exhibit a loss of barrier function, increased endothelial proliferation, and altered gene expression. While many advances have been made in our understanding of how KRIT1, and the functionally related proteins CCM2 and PDCD10, contribute to the regulation of blood vessels and the vascular barrier, some important open questions remain. In addition, KRIT1 is widely expressed and KRIT1 and the other CCM proteins have been shown to play important roles in non-endothelial cell types and tissues, which may or may not be related to their role as pathogenic originators of CCM. In this review, we discuss some of the unsettled questions regarding the role of KRIT1 in vascular physiology and discuss recent advances that suggest this ubiquitously expressed protein may have a role beyond the endothelial cell.
Collapse
Affiliation(s)
- Angela J. Glading
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, U.S.A
| |
Collapse
|
3
|
Offenberger J, Chen B, Rossitto LA, Jin I, Conaboy L, Gallego-Gutierrez H, Nelsen B, Frias-Anaya E, Gonzalez DJ, Anagnostaras S, Lopez-Ramirez MA. Behavioral impairments are linked to neuroinflammation in mice with Cerebral Cavernous Malformation disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596485. [PMID: 38853989 PMCID: PMC11160801 DOI: 10.1101/2024.05.29.596485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background Cerebral Cavernous Malformations (CCMs) are neurovascular abnormalities in the central nervous system (CNS) caused by loss of function mutations in KRIT1 (CCM1), CCM2, or PDCD10 (CCM3) genes. One of the most common symptoms in CCM patients is associated with motor disability, weakness, seizures, stress, and anxiety, and the extent of the symptom or symptoms may be due to the location of the lesion within the CNS or whether multiple lesions are present. Previous studies have primarily focused on understanding the pathology of CCM using animal models. However, more research has yet to explore the potential impact of CCM lesions on behavioral deficits in animal models, including effects on short-term and long-term memory, motor coordination, and function. Methods We used the accelerating RotaRod test to assess motor and coordination deficits. We also used the open field test to assess locomotor activity and pathology-related behavior and Pavlovian fear conditioning to assess short-and long-term memory deficits. Our behavioral studies were complemented by proteomics, histology, immunofluorescence, and imaging techniques. We found that neuroinflammation is crucial in behavioral deficits in male and female mice with neurovascular CCM lesions (Slco1c1-iCreERT2; Pdcd10 fl/fl ; Pdcd10 BECKO ). Results Functional behavior tests in male and female Pdcd10 BECKO mice revealed that CCM lesions cause sudden motor coordination deficits associated with the manifestation of profound neuroinflammatory lesions. Our findings indicate that maturation of CCM lesions in Pdcd10 BECKO mice also experienced a significant change in short- and long-term memory compared to their littermate controls, Pdcd10 fl/fl mice. Proteomic experiments reveal that as CCM lesions mature, there is an increase in pathways associated with inflammation, coagulation, and angiogenesis, and a decrease in pathways associated with learning and plasticity. Therefore, our study shows that Pdcd10 BECKO mice display a wide range of behavioral deficits due to significant lesion formation in their central nervous system and that signaling pathways associated with neuroinflammation and learning impact behavioral outcomes. Conclusions Our study found that CCM animal models exhibited behavioral impairments such as decreased motor coordination and amnesia. These impairments were associated with the maturation of CCM lesions that displayed a neuroinflammatory pattern.
Collapse
Affiliation(s)
- Joseph Offenberger
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Bianca Chen
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Leigh-Ana Rossitto
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Irisa Jin
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Liam Conaboy
- Department of Psychology, University of California, San Diego, La Jolla, California, USA
| | | | - Bliss Nelsen
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Eduardo Frias-Anaya
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - David J. Gonzalez
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Stephan Anagnostaras
- Department of Psychology, University of California, San Diego, La Jolla, California, USA
- Program in Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Miguel Alejandro Lopez-Ramirez
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Lai CC, Nelsen B, Frias-Anaya E, Gallego-Gutierrez H, Orecchioni M, Herrera V, Ortiz E, Sun H, Mesarwi OA, Ley K, Gongol B, Lopez-Ramirez MA. Neuroinflammation Plays a Critical Role in Cerebral Cavernous Malformation Disease. Circ Res 2022; 131:909-925. [PMID: 36285625 PMCID: PMC9669201 DOI: 10.1161/circresaha.122.321129] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/11/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Cerebral cavernous malformations (CCMs) are neurovascular lesions caused by loss of function mutations in 1 of 3 genes, including KRIT1 (CCM1), CCM2, and PDCD10 (CCM3). CCMs affect ≈1 out of 200 children and adults, and no pharmacologic therapy is available. CCM lesion count, size, and aggressiveness vary widely among patients of similar ages with the same mutation or even within members of the same family. However, what determines the transition from quiescent lesions into mature and active (aggressive) CCM lesions is unknown. METHODS We use genetic, RNA-sequencing, histology, flow cytometry, and imaging techniques to report the interaction between CCM endothelium, astrocytes, leukocytes, microglia/macrophages, neutrophils (CCM endothelium, astrocytes, leukocytes, microglia/macrophages, neutrophils interaction) during the pathogenesis of CCMs in the brain tissue. RESULTS Expression profile of astrocytes in adult mouse brains using translated mRNAs obtained from the purification of EGFP (enhanced green fluorescent protein)-tagged ribosomes (Aldh1l1-EGFP/Rpl10a) in the presence or absence of CCM lesions (Slco1c1-iCreERT2;Pdcd10fl/fl; Pdcd10BECKO) identifies a novel gene signature for neuroinflammatory astrocytes. CCM-induced reactive astrocytes have a neuroinflammatory capacity by expressing genes involved in angiogenesis, chemotaxis, hypoxia signaling, and inflammation. RNA-sequencing analysis on RNA isolated from brain endothelial cells in chronic Pdcd10BECKO mice (CCM endothelium), identified crucial genes involved in recruiting inflammatory cells and thrombus formation through chemotaxis and coagulation pathways. In addition, CCM endothelium was associated with increased expression of Nlrp3 and Il1b. Pharmacological inhibition of NLRP3 (NOD [nucleotide-binding oligomerization domain]-' LRR [leucine-rich repeat]- and pyrin domain-containing protein 3) significantly decreased inflammasome activity as assessed by quantification of a fluorescent indicator of caspase-1 activity (FAM-FLICA [carboxyfluorescein-fluorochrome-labeled inhibitors of caspases] caspase-1) in brain endothelial cells from Pdcd10BECKO in chronic stage. Importantly, our results support the hypothesis of the crosstalk between astrocytes and CCM endothelium that can trigger recruitment of inflammatory cells arising from brain parenchyma (microglia) and the peripheral immune system (leukocytes) into mature active CCM lesions that propagate lesion growth, immunothrombosis, and bleedings. Unexpectedly, partial or total loss of brain endothelial NF-κB (nuclear factor κB) activity (using Ikkbfl/fl mice) in chronic Pdcd10BECKO mice does not prevent lesion genesis or neuroinflammation. Instead, this resulted in a trend increase in the number of lesions and immunothrombosis, suggesting that therapeutic approaches designed to target inflammation through endothelial NF-κB inhibition may contribute to detrimental side effects. CONCLUSIONS Our study reveals previously unknown links between neuroinflammatory astrocytes and inflamed CCM endothelium as contributors that trigger leukocyte recruitment and precipitate immunothrombosis in CCM lesions. However, therapeutic approaches targeting brain endothelial NF-κB activity may contribute to detrimental side effects.
Collapse
Affiliation(s)
| | - Bliss Nelsen
- Department of Medicine, University of California, San
Diego, La Jolla, California, USA
| | - Eduardo Frias-Anaya
- Department of Medicine, University of California, San
Diego, La Jolla, California, USA
| | | | - Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for
Immunology, La Jolla, California, USA
| | - Victoria Herrera
- Department of Medicine, University of California, San
Diego, La Jolla, California, USA
| | - Elan Ortiz
- Department of Medicine, University of California, San
Diego, La Jolla, California, USA
| | - Hao Sun
- Department of Medicine, University of California, San
Diego, La Jolla, California, USA
| | - Omar A. Mesarwi
- Department of Medicine, University of California, San
Diego, La Jolla, California, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for
Immunology, La Jolla, California, USA
| | - Brendan Gongol
- Department of Health Sciences, Victor Valley College,
Victorville, California, USA
- Institute for Integrative Genome Biology, 1207F Genomics
Building, University of California, Riverside, CA 92521, USA
| | - Miguel Alejandro Lopez-Ramirez
- Department of Medicine, University of California, San
Diego, La Jolla, California, USA
- Department of Pharmacology, University of California, San
Diego, La Jolla, California, USA
| |
Collapse
|
8
|
Dysregulated Hemostasis and Immunothrombosis in Cerebral Cavernous Malformations. Int J Mol Sci 2022; 23:ijms232012575. [PMID: 36293431 PMCID: PMC9604397 DOI: 10.3390/ijms232012575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Cerebral cavernous malformation (CCM) is a neurovascular disease that affects 0.5% of the general population. For a long time, CCM research focused on genetic mutations, endothelial junctions and proliferation, but recently, transcriptome and proteome studies have revealed that the hemostatic system and neuroinflammation play a crucial role in the development and severity of cavernomas, with some of these publications coming from our group. The aim of this review is to give an overview of the latest molecular insights into the interaction between CCM-deficient endothelial cells with blood components and the neurovascular unit. Specifically, we underscore how endothelial dysfunction can result in dysregulated hemostasis, bleeding, hypoxia and neurological symptoms. We conducted a thorough review of the literature and found a field that is increasingly poised to regard CCM as a hemostatic disease, which may have implications for therapy.
Collapse
|
9
|
Tu T, Peng Z, Ren J, Zhang H. Cerebral Cavernous Malformation: Immune and Inflammatory Perspectives. Front Immunol 2022; 13:922281. [PMID: 35844490 PMCID: PMC9280619 DOI: 10.3389/fimmu.2022.922281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/18/2022] [Indexed: 12/03/2022] Open
Abstract
Cerebral cavernous malformation (CCM) is a type of vascular anomaly that arises due to the dyshomeostasis of brain capillary networks. In the past two decades, many advances have been made in this research field. Notably, as a more reasonable current view, the CCM lesions should be attributed to the results of a great number of additional events related to the homeostasis disorder of the endothelial cell. Indeed, one of the most fascinating concerns in the research field is the inflammatory perturbation in the immune microenvironment, which would affect the disease progression as well as the patients’ outcomes. In this work, we focused on this topic, and underlined the immune-related factors’ contribution to the CCM pathologic progression.
Collapse
Affiliation(s)
- Tianqi Tu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenghong Peng
- Health Management Department, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jian Ren
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Hongqi Zhang,
| |
Collapse
|
10
|
Phillips CM, Stamatovic SM, Keep RF, Andjelkovic AV. Cerebral Cavernous Malformation Pathogenesis: Investigating Lesion Formation and Progression with Animal Models. Int J Mol Sci 2022; 23:5000. [PMID: 35563390 PMCID: PMC9105545 DOI: 10.3390/ijms23095000] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
Cerebral cavernous malformation (CCM) is a cerebromicrovascular disease that affects up to 0.5% of the population. Vessel dilation, decreased endothelial cell-cell contact, and loss of junctional complexes lead to loss of brain endothelial barrier integrity and hemorrhagic lesion formation. Leakage of hemorrhagic lesions results in patient symptoms and complications, including seizures, epilepsy, focal headaches, and hemorrhagic stroke. CCMs are classified as sporadic (sCCM) or familial (fCCM), associated with loss-of-function mutations in KRIT1/CCM1, CCM2, and PDCD10/CCM3. Identifying the CCM proteins has thrust the field forward by (1) revealing cellular processes and signaling pathways underlying fCCM pathogenesis, and (2) facilitating the development of animal models to study CCM protein function. CCM animal models range from various murine models to zebrafish models, with each model providing unique insights into CCM lesion development and progression. Additionally, these animal models serve as preclinical models to study therapeutic options for CCM treatment. This review briefly summarizes CCM disease pathology and the molecular functions of the CCM proteins, followed by an in-depth discussion of animal models used to study CCM pathogenesis and developing therapeutics.
Collapse
Affiliation(s)
- Chelsea M. Phillips
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| | - Svetlana M. Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
- Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| |
Collapse
|
11
|
Yau ACY, Globisch MA, Onyeogaziri FC, Conze LL, Smith R, Jauhiainen S, Corada M, Orsenigo F, Huang H, Herre M, Olsson AK, Malinverno M, Sundell V, Rezai Jahromi B, Niemelä M, Laakso A, Garlanda C, Mantovani A, Lampugnani MG, Dejana E, Magnusson PU. Inflammation and neutrophil extracellular traps in cerebral cavernous malformation. Cell Mol Life Sci 2022; 79:206. [PMID: 35333979 PMCID: PMC8949649 DOI: 10.1007/s00018-022-04224-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023]
Abstract
Cerebral Cavernous Malformation (CCM) is a brain vascular disease with various neurological symptoms. In this study, we describe the inflammatory profile in CCM and show for the first time the formation of neutrophil extracellular traps (NETs) in rodents and humans with CCM. Through RNA-seq analysis of cerebellum endothelial cells from wild-type mice and mice with an endothelial cell-specific ablation of the Ccm3 gene (Ccm3iECKO), we show that endothelial cells from Ccm3iECKO mice have an increased expression of inflammation-related genes. These genes encode proinflammatory cytokines and chemokines, as well as adhesion molecules, which promote recruitment of inflammatory and immune cells. Similarly, immunoassays showed elevated levels of these cytokines and chemokines in the cerebellum of the Ccm3iECKO mice. Consistently, both flow cytometry and immunofluorescence analysis showed infiltration of different subsets of leukocytes into the CCM lesions. Neutrophils, which are known to fight against infection through different strategies, including the formation of NETs, represented the leukocyte subset within the most pronounced increase in CCM. Here, we detected elevated levels of NETs in the blood and the deposition of NETs in the cerebral cavernomas of Ccm3iECKO mice. Degradation of NETs by DNase I treatment improved the vascular barrier. The deposition of NETs in the cavernomas of patients with CCM confirms the clinical relevance of NETs in CCM.
Collapse
Affiliation(s)
- Anthony C Y Yau
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Maria Ascencion Globisch
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Favour Chinyere Onyeogaziri
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Lei L Conze
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Ross Smith
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Suvi Jauhiainen
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Monica Corada
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Fabrizio Orsenigo
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Hua Huang
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Melanie Herre
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Matteo Malinverno
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Veronica Sundell
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden
| | - Behnam Rezai Jahromi
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Niemelä
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aki Laakso
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS Humanitas Research Hospital, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Maria Grazia Lampugnani
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy.,Mario Negri Institute for Pharmacological Research, 20157, Milan, Italy
| | - Elisabetta Dejana
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden.,Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Peetra U Magnusson
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Dag Hammarskjoldsv. 20, 751 85, Uppsala, Sweden.
| |
Collapse
|
12
|
Maderna C, Pisati F, Tripodo C, Dejana E, Malinverno M. A murine model of cerebral cavernous malformations with acute hemorrhage. iScience 2022; 25:103943. [PMID: 35265815 PMCID: PMC8898922 DOI: 10.1016/j.isci.2022.103943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 12/06/2021] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
Cavernomas are multi-lumen and blood-filled vascular malformations which form in the brain and the spinal cord. They lead to hemorrhage, epileptic seizures, neurological deficits, and paresthesia. An effective medical treatment is still lacking, and the available murine models for cavernomas have several limitations for preclinical studies. These include disease phenotypes that differ from human diseases, such as restriction of the lesions to the cerebellum, and absence of acute hemorrhage. Additional limitations of current murine models include rapid development of lesions, which are lethal before the first month of age. Here, we have characterized a murine model that recapitulates features of the human disease: lesions develop after weaning throughout the entire CNS, including the spinal cord, and undergo acute hemorrhage. This provides a preclinical model to develop new drugs for treatment of acute hemorrhage in the brain and spinal cord, as an unmet medical emergency for patients with cavernomas. Ccm3 deletion in endothelial progenitors drives cavernoma formation in a mouse model Mice develop acute hemorrhage and inflammation in brain and spinal cord The spleen has increased vascular density and altered hemopoiesis This model represents a useful tool for mechanistic studies and drug screening
Collapse
Affiliation(s)
- Claudio Maderna
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan 20139, Italy
| | - Federica Pisati
- Tumour and Microenvironment Histopathology Unit, FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Claudio Tripodo
- Tumour and Microenvironment Histopathology Unit, FIRC Institute of Molecular Oncology (IFOM), Milan, Italy.,Tumour Immunology Unit, University of Palermo, Palermo, Italy
| | - Elisabetta Dejana
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan 20139, Italy.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 752 37, Sweden
| | - Matteo Malinverno
- Vascular Biology Unit, The FIRC Institute of Molecular Oncology Foundation, Milan 20139, Italy
| |
Collapse
|
13
|
Castro C, Oyamada HAA, Cafasso MOSD, Lopes LM, Monteiro C, Sacramento PM, Alves-Leon SV, da Fontoura Galvão G, Hygino J, de Souza JPBM, Bento CAM. Elevated proportion of TLR2- and TLR4-expressing Th17-like cells and activated memory B cells was associated with clinical activity of cerebral cavernous malformations. J Neuroinflammation 2022; 19:28. [PMID: 35109870 PMCID: PMC8808981 DOI: 10.1186/s12974-022-02385-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Recent evidences have suggested the involvement of toll-like receptor (TLR)-4 in the pathogenesis of cerebral cavernous malformations (CCM). Elevated frequency of TLR+T-cells has been associated with neurological inflammatory disorders. As T-cells and B-cells are found in CCM lesions, the objective of the present study was to evaluate the cytokine profile of T-cells expressing TLR2 and TLR4, as well as B-cell subsets, in asymptomatic (CCMAsympt) and symptomatic (CCMSympt) patients. METHODS For our study, the cytokine profile from TLR2+ and TLR4+ T-cell and B-cell subsets in CCMAsympt and CCMSympt patients was investigated using flow cytometry and ELISA. T-cells were stimulated in vitro with anti-CD3/anti-CD28 beads or TLR2 (Pam3C) and TLR4 (LPS) ligands. RESULTS CCMSymptc patients presented a higher frequency of TLR4+(CD4+ and CD8+) T-cells and greater density of TLR4 expression on these cells. With regard to the cytokine profile, the percentage of TLR2+ and TLR4+ Th17 cells was higher in CCMSympt patients. In addition, an elevated proportion of TLR4+ Tc-1 cells, as well as Tc-17 and Th17.1 cells expressing TLR2 and TLR4, was observed in the symptomatic patients. By contrast, the percentage of TLR4+ IL-10+CD4+ T cells was higher in the CCMAsympt group. Both Pam3C and LPS were more able to elevate the frequency of IL-6+CD4+T cells and Th17.1 cells in CCMSympt cell cultures. Furthermore, in comparison with asymptomatic patients, purified T-cells from the CCMSympt group released higher levels of Th17-related cytokines in response to Pam3C and, mainly, LPS, as well as after activation via TCR/CD28. Concerning the B-cell subsets, a higher frequency of memory and memory activated B-cells was observed in CCMSympt patients. CONCLUSIONS Our findings reveal an increase in circulating Th17/Tc-17 cell subsets expressing functional TLR2 and, mainly, TLR4 molecules, associated with an increase in memory B-cell subsets in CCM patients with clinical activity of the disease.
Collapse
Affiliation(s)
- Camilla Castro
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hugo A A Oyamada
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Octávio S D Cafasso
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
| | - Lana M Lopes
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarice Monteiro
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila M Sacramento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Soniza Vieira Alves-Leon
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Translational Neuroscience Laboratory (LabNet), University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo da Fontoura Galvão
- Service of Neurosurgery, University Hospital of the Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Joana Hygino
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge Paes Barreto Marcondes de Souza
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Service of Neurosurgery, University Hospital of the Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Post-Graduate Program of Surgical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cleonice A M Bento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil.
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
14
|
Genetics and Vascular Biology of Brain Vascular Malformations. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00012-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
15
|
Snellings DA, Hong CC, Ren AA, Lopez-Ramirez MA, Girard R, Srinath A, Marchuk DA, Ginsberg MH, Awad IA, Kahn ML. Cerebral Cavernous Malformation: From Mechanism to Therapy. Circ Res 2021; 129:195-215. [PMID: 34166073 PMCID: PMC8922476 DOI: 10.1161/circresaha.121.318174] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cerebral cavernous malformations are acquired vascular anomalies that constitute a common cause of central nervous system hemorrhage and stroke. The past 2 decades have seen a remarkable increase in our understanding of the pathogenesis of this vascular disease. This new knowledge spans genetic causes of sporadic and familial forms of the disease, molecular signaling changes in vascular endothelial cells that underlie the disease, unexpectedly strong environmental effects on disease pathogenesis, and drivers of disease end points such as hemorrhage. These novel insights are the integrated product of human clinical studies, human genetic studies, studies in mouse and zebrafish genetic models, and basic molecular and cellular studies. This review addresses the genetic and molecular underpinnings of cerebral cavernous malformation disease, the mechanisms that lead to lesion hemorrhage, and emerging biomarkers and therapies for clinical treatment of cerebral cavernous malformation disease. It may also serve as an example for how focused basic and clinical investigation and emerging technologies can rapidly unravel a complex disease mechanism.
Collapse
Affiliation(s)
- Daniel A Snellings
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC (D.A.S., D.A.M.)
| | - Courtney C Hong
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia (C.C.H., A.A.R., M.L.K.)
| | - Aileen A Ren
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia (C.C.H., A.A.R., M.L.K.)
| | - Miguel A Lopez-Ramirez
- Department of Medicine (M.A.L.-R., M.H.G.), University of California, San Diego, La Jolla
- Department of Pharmacology (M.A.L.-R.), University of California, San Diego, La Jolla
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Abhinav Srinath
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC (D.A.S., D.A.M.)
| | - Mark H Ginsberg
- Department of Medicine (M.A.L.-R., M.H.G.), University of California, San Diego, La Jolla
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia (C.C.H., A.A.R., M.L.K.)
| |
Collapse
|
16
|
Rustenhoven J, Tanumihardja C, Kipnis J. Cerebrovascular Anomalies: Perspectives From Immunology and Cerebrospinal Fluid Flow. Circ Res 2021; 129:174-194. [PMID: 34166075 DOI: 10.1161/circresaha.121.318173] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Appropriate vascular function is essential for the maintenance of central nervous system homeostasis and is achieved through virtue of the blood-brain barrier; a specialized structure consisting of endothelial, mural, and astrocytic interactions. While appropriate blood-brain barrier function is typically achieved, the central nervous system vasculature is not infallible and cerebrovascular anomalies, a collective terminology for diverse vascular lesions, are present in meningeal and cerebral vasculature supplying and draining the brain. These conditions, including aneurysmal formation and rupture, arteriovenous malformations, dural arteriovenous fistulas, and cerebral cavernous malformations, and their associated neurological sequelae, are typically managed with neurosurgical or pharmacological approaches. However, increasing evidence implicates interacting roles for inflammatory responses and disrupted central nervous system fluid flow with respect to vascular perturbations. Here, we discuss cerebrovascular anomalies from an immunologic angle and fluid flow perspective. We describe immune contributions, both common and distinct, to the formation and progression of diverse cerebrovascular anomalies. Next, we summarize how cerebrovascular anomalies precipitate diverse neurological sequelae, including seizures, hydrocephalus, and cognitive effects and possible contributions through the recently identified lymphatic and glymphatic systems. Finally, we speculate on and provide testable hypotheses for novel nonsurgical therapeutic approaches for alleviating neurological impairments arising from cerebrovascular anomalies, with a particular emphasis on the normalization of fluid flow and alleviation of inflammation through manipulations of the lymphatic and glymphatic central nervous system clearance pathways.
Collapse
Affiliation(s)
- Justin Rustenhoven
- Center for Brain Immunology and Glia (J.R., J.K.), Washington University in St. Louis, St Louis, MO.,Department of Pathology and Immunology, School of Medicine (J.R., J.K.), Washington University in St. Louis, St Louis, MO
| | | | - Jonathan Kipnis
- Center for Brain Immunology and Glia (J.R., J.K.), Washington University in St. Louis, St Louis, MO.,Department of Pathology and Immunology, School of Medicine (J.R., J.K.), Washington University in St. Louis, St Louis, MO
| |
Collapse
|
17
|
Girard R, Li Y, Stadnik A, Shenkar R, Hobson N, Romanos S, Srinath A, Moore T, Lightle R, Shkoukani A, Akers A, Carroll T, Christoforidis GA, Koenig JI, Lee C, Piedad K, Greenberg SM, Kim H, Flemming KD, Ji Y, Awad IA. A Roadmap for Developing Plasma Diagnostic and Prognostic Biomarkers of Cerebral Cavernous Angioma With Symptomatic Hemorrhage (CASH). Neurosurgery 2021; 88:686-697. [PMID: 33469662 DOI: 10.1093/neuros/nyaa478] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/16/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Cerebral cavernous angioma (CA) is a capillary microangiopathy predisposing more than a million Americans to premature risk of brain hemorrhage. CA with recent symptomatic hemorrhage (SH), most likely to re-bleed with serious clinical sequelae, is the primary focus of therapeutic development. Signaling aberrations in CA include proliferative dysangiogenesis, blood-brain barrier hyperpermeability, inflammatory/immune processes, and anticoagulant vascular domain. Plasma levels of molecules reflecting these mechanisms and measures of vascular permeability and iron deposition on magnetic resonance imaging are biomarkers that have been correlated with CA hemorrhage. OBJECTIVE To optimize these biomarkers to accurately diagnose cavernous angioma with symptomatic hemorrhage (CASH), prognosticate the risk of future SH, and monitor cases after a bleed and in response to therapy. METHODS Additional candidate biomarkers, emerging from ongoing mechanistic and differential transcriptome studies, would further enhance the sensitivity and specificity of diagnosis and prediction of CASH. Integrative combinations of levels of plasma proteins and characteristic micro-ribonucleic acids may further strengthen biomarker associations. We will deploy advanced statistical and machine learning approaches for the integration of novel candidate biomarkers, rejecting noncorrelated candidates, and determining the best clustering and weighing of combined biomarker contributions. EXPECTED OUTCOMES With the expertise of leading CA researchers, this project anticipates the development of future blood tests for the diagnosis and prediction of CASH to clinically advance towards precision medicine. DISCUSSION The project tests a novel integrational approach of biomarker development in a mechanistically defined cerebrovascular disease with a relevant context of use, with an approach applicable to other neurological diseases with similar pathobiologic features.
Collapse
Affiliation(s)
- Romuald Girard
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Yan Li
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois.,Bioinformatics core, Center for Research Informatics, University of Chicago, Chicago, Illinois
| | - Agnieszka Stadnik
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Nicholas Hobson
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Thomas Moore
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Abdallah Shkoukani
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | | | - Timothy Carroll
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Gregory A Christoforidis
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - James I Koenig
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | | | - Kristina Piedad
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Helen Kim
- Department of Anesthesia & Perioperative Care, University of California at San Francisco, San Francisco, California
| | | | - Yuan Ji
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Surgery, Section of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| |
Collapse
|
18
|
Retta SF, Perrelli A, Trabalzini L, Finetti F. From Genes and Mechanisms to Molecular-Targeted Therapies: The Long Climb to the Cure of Cerebral Cavernous Malformation (CCM) Disease. Methods Mol Biol 2021; 2152:3-25. [PMID: 32524540 DOI: 10.1007/978-1-0716-0640-7_1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cerebral cavernous malformation (CCM) is a rare cerebrovascular disorder of genetic origin consisting of closely clustered, abnormally dilated and leaky capillaries (CCM lesions), which occur predominantly in the central nervous system. CCM lesions can be single or multiple and may result in severe clinical symptoms, including focal neurological deficits, seizures, and intracerebral hemorrhage. Early human genetic studies demonstrated that CCM disease is linked to three chromosomal loci and can be inherited as autosomal dominant condition with incomplete penetrance and highly variable expressivity, eventually leading to the identification of three disease genes, CCM1/KRIT1, CCM2, and CCM3/PDCD10, which encode for structurally unrelated intracellular proteins that lack catalytic domains. Biochemical, molecular, and cellular studies then showed that these proteins are involved in endothelial cell-cell junction and blood-brain barrier stability maintenance through the regulation of major cellular structures and mechanisms, including endothelial cell-cell and cell-matrix adhesion, actin cytoskeleton dynamics, autophagy, and endothelial-to-mesenchymal transition, suggesting that they act as pleiotropic regulators of cellular homeostasis, and opening novel therapeutic perspectives. Indeed, accumulated evidence in cellular and animal models has eventually revealed that the emerged pleiotropic functions of CCM proteins are mainly due to their ability to modulate redox-sensitive pathways and mechanisms involved in adaptive responses to oxidative stress and inflammation, thus contributing to the preservation of cellular homeostasis and stress defenses.In this introductory review, we present a general overview of 20 years of amazing progress in the identification of genetic culprits and molecular mechanisms underlying CCM disease pathogenesis, and the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Saverio Francesco Retta
- Department of Clinical and Biological Science, School of Medicine and Surgery, University of Torino, Orbassano (Torino), Italy. .,CCM Italia Research Network, Torino, Italy.
| | - Andrea Perrelli
- Department of Clinical and Biological Science, School of Medicine and Surgery, University of Torino, Orbassano (Torino), Italy.,CCM Italia Research Network, Torino, Italy
| | - Lorenza Trabalzini
- CCM Italia Research Network, Torino, Italy.,Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Federica Finetti
- CCM Italia Research Network, Torino, Italy.,Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| |
Collapse
|
19
|
Li W, Shenkar R, Detter MR, Moore T, Benavides C, Lightle R, Girard R, Hobson N, Cao Y, Li Y, Griffin E, Gallione C, Zabramski JM, Ginsberg MH, Marchuk DA, Awad IA. Propranolol inhibits cavernous vascular malformations by β1 adrenergic receptor antagonism in animal models. J Clin Invest 2021; 131:144893. [PMID: 33301422 PMCID: PMC7843213 DOI: 10.1172/jci144893] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/01/2020] [Indexed: 11/17/2022] Open
Abstract
Propranolol, a pleiotropic β-adrenergic blocker, has been anecdotally reported to reduce cerebral cavernous malformations (CCMs) in humans. However, propranolol has not been rigorously evaluated in animal models, nor has its mechanism of action in CCM been defined. We report that propranolol or its S(-) enantiomer dramatically reduced embryonic venous cavernomas in ccm2 mosaic zebrafish, whereas R-(+)-propranolol, lacking β antagonism, had no effect. Silencing of the β1, but not β2, adrenergic receptor mimicked the beneficial effects of propranolol in a zebrafish CCM model, as did the β1-selective antagonist metoprolol. Thus, propranolol ameliorated cavernous malformations by β1 adrenergic antagonism in zebrafish. Oral propranolol significantly reduced lesion burden in 2 chronic murine models of the exceptionally aggressive Pdcd10/Ccm3 form of CCM. Propranolol or other β1-selective antagonists may be beneficial in CCM disease.
Collapse
MESH Headings
- Adrenergic beta-1 Receptor Antagonists/adverse effects
- Adrenergic beta-1 Receptor Antagonists/pharmacology
- Animals
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Female
- G-Protein-Coupled Receptor Kinase 2/genetics
- G-Protein-Coupled Receptor Kinase 2/metabolism
- Hemangioma, Cavernous, Central Nervous System/chemically induced
- Hemangioma, Cavernous, Central Nervous System/drug therapy
- Hemangioma, Cavernous, Central Nervous System/genetics
- Hemangioma, Cavernous, Central Nervous System/metabolism
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Knockout
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/metabolism
- Propranolol/pharmacology
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Zebrafish
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Wenqing Li
- Department of Medicine, UCSD, San Diego, California, USA
| | - Robert Shenkar
- Department of Neurological Surgery, University of Chicago, Chicago, Illinois, USA
| | - Mathew R. Detter
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Thomas Moore
- Department of Neurological Surgery, University of Chicago, Chicago, Illinois, USA
| | - Christian Benavides
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Rhonda Lightle
- Department of Neurological Surgery, University of Chicago, Chicago, Illinois, USA
| | - Romuald Girard
- Department of Neurological Surgery, University of Chicago, Chicago, Illinois, USA
| | - Nicholas Hobson
- Department of Neurological Surgery, University of Chicago, Chicago, Illinois, USA
| | - Ying Cao
- Department of Neurological Surgery, University of Chicago, Chicago, Illinois, USA
| | - Yan Li
- Department of Neurological Surgery, University of Chicago, Chicago, Illinois, USA
- Bioinformatics Core, Center for Research Informatics, University of Chicago, Chicago, Illinois, USA
| | - Erin Griffin
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Carol Gallione
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Joseph M. Zabramski
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona, USA
| | | | - Douglas A. Marchuk
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Issam A. Awad
- Department of Neurological Surgery, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
20
|
Zhang D, Kinloch AJ, Srinath A, Shenkar R, Girard R, Lightle R, Moore T, Koskimäki J, Mohsin A, Carrión-Penagos J, Romanos S, Shen L, Clark MR, Shi C, Awad IA. Antibodies in cerebral cavernous malformations react with cytoskeleton autoantigens in the lesional milieu. J Autoimmun 2020; 113:102469. [PMID: 32362501 PMCID: PMC7483292 DOI: 10.1016/j.jaut.2020.102469] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 01/21/2023]
Abstract
Previous studies have reported robust inflammatory cell infiltration, synthesis of IgG, B-cell clonal expansion, deposition of immune complexes and complement within cerebral cavernous malformation (CCM) lesions. B-cell depletion has also been shown to reduce the maturation of CCM in murine models. We hypothesize that antigen(s) within the lesional milieu perpetuate the pathogenetic immune responses in CCMs. This study aims to identify those putative antigen(s) using monoclonal antibodies (mAbs) derived from plasma cells found in surgically removed human CCM lesions. We produced human mAbs from laser capture micro-dissected plasma cells from four CCM patients, and also germline-reverted versions. CCM mAbs were assayed using immunofluorescence on central nervous system (CNS) tissues and immunocytochemistry on human primary cell lines. Antigen characterization was performed using a combination of confocal microscopy, immunoprecipitation and mass spectrometry. Affinity was determined by enzyme-linked immunosorbent assay, and specificity by multi-color confocal microscopy and quantitative co-localization. CCM mAbs bound CNS tissue, especially endothelial cells and astrocytes. Non-muscle myosin heavy chain IIA (NMMHCIIA), vimentin and tubulin are three cytoskeleton proteins that were commonly targeted. Selection of cytoskeleton proteins by plasma cells was supported by a high frequency of immunoglobulin variable region somatic hypermutations, high affinity and selectivity of mAbs in their affinity matured forms, and profoundly reduced affinity and selectivity in the germline reverted forms. Antibodies produced by plasma cells in CCM lesions commonly target cytoplasmic and cytoskeletal autoantigens including NMMHCIIA, vimentin and tubulin that are abundant in endothelial cells and astrocytes. Binding to, and selection on autoantigen(s) in the lesional milieu likely perpetuates the pathogenetic immune response in CCMs. Blocking this in situ autoimmune response may yield a novel treatment for CCM.
Collapse
Affiliation(s)
- Dongdong Zhang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China; Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Andrew J Kinloch
- Department of Medicine, Section of Rheumatology, The University of Chicago, Gwen Knapp Center for Lupus and Immunology Research, 5841 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Abhinav Srinath
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Janne Koskimäki
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Azam Mohsin
- Department of Medicine, Section of Rheumatology, The University of Chicago, Gwen Knapp Center for Lupus and Immunology Research, 5841 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Julián Carrión-Penagos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Sharbel Romanos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Le Shen
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Marcus R Clark
- Department of Medicine, Section of Rheumatology, The University of Chicago, Gwen Knapp Center for Lupus and Immunology Research, 5841 S. Maryland Ave, Chicago, IL, 60637, United States
| | - Changbin Shi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5842 S. Maryland Ave, Chicago, IL, 60637, United States.
| |
Collapse
|
21
|
Detter MR, Shenkar R, Benavides CR, Neilson CA, Moore T, Lightle R, Hobson N, Shen L, Cao Y, Girard R, Zhang D, Griffin E, Gallione CJ, Awad IA, Marchuk DA. Novel Murine Models of Cerebral Cavernous Malformations. Angiogenesis 2020; 23:651-666. [PMID: 32710309 DOI: 10.1007/s10456-020-09736-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/06/2020] [Indexed: 12/21/2022]
Abstract
Cerebral cavernous malformations (CCMs) are ectatic capillary-venous malformations that develop in approximately 0.5% of the population. Patients with CCMs may develop headaches, focal neurologic deficits, seizures, and hemorrhages. While symptomatic CCMs, depending upon the anatomic location, can be surgically removed, there is currently no pharmaceutical therapy to treat CCMs. Several mouse models have been developed to better understand CCM pathogenesis and test therapeutics. The most common mouse models induce a large CCM burden that is anatomically restricted to the cerebellum and contributes to lethality in the early days of life. These inducible models thus have a relatively short period for drug administration. We developed an inducible CCM3 mouse model that develops CCMs after weaning and provides a longer period for potential therapeutic intervention. Using this new model, three recently proposed CCM therapies, fasudil, tempol, vitamin D3, and a combination of the three drugs, failed to substantially reduce CCM formation when treatment was administered for 5 weeks, from postnatal day 21 (P21) to P56. We next restricted Ccm3 deletion to the brain vasculature and provided greater time (121 days) for CCMs to develop chronic hemorrhage, recapitulating the human lesions. We also developed the first model of acute CCM hemorrhage by injecting mice harboring CCMs with lipopolysaccharide. These efficient models will enable future drug studies to more precisely target clinically relevant features of CCM disease: CCM formation, chronic hemorrhage, and acute hemorrhage.
Collapse
Affiliation(s)
- Matthew R Detter
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Christian R Benavides
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Catherine A Neilson
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Nicholas Hobson
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Le Shen
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Ying Cao
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Dongdong Zhang
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Erin Griffin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Carol J Gallione
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurosurgery, University of Chicago Medicine and Biological Sciences, Chicago, IL, 60637, USA
| | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, 27705, USA. .,James B Duke Professor, Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Box 3175, Durham, NC, 27710, USA.
| |
Collapse
|
22
|
Polster SP, Sharma A, Tanes C, Tang AT, Mericko P, Cao Y, Carrión-Penagos J, Girard R, Koskimäki J, Zhang D, Stadnik A, Romanos SG, Lyne SB, Shenkar R, Yan K, Lee C, Akers A, Morrison L, Robinson M, Zafar A, Bittinger K, Kim H, Gilbert JA, Kahn ML, Shen L, Awad IA. Permissive microbiome characterizes human subjects with a neurovascular disease cavernous angioma. Nat Commun 2020; 11:2659. [PMID: 32461638 PMCID: PMC7253448 DOI: 10.1038/s41467-020-16436-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 05/04/2020] [Indexed: 11/09/2022] Open
Abstract
Cavernous angiomas (CA) are common vascular anomalies causing brain hemorrhage. Based on mouse studies, roles of gram-negative bacteria and altered intestinal homeostasis have been implicated in CA pathogenesis, and pilot study had suggested potential microbiome differences between non-CA and CA individuals based on 16S rRNA gene sequencing. We here assess microbiome differences in a larger cohort of human subjects with and without CA, and among subjects with different clinical features, and conduct more definitive microbial analyses using metagenomic shotgun sequencing. Relative abundance of distinct bacterial species in CA patients is shown, consistent with postulated permissive microbiome driving CA lesion genesis via lipopolysaccharide signaling, in humans as in mice. Other microbiome differences are related to CA clinical behavior. Weighted combinations of microbiome signatures and plasma inflammatory biomarkers enhance associations with disease severity and hemorrhage. This is the first demonstration of a sensitive and specific diagnostic microbiome in a human neurovascular disease.
Collapse
Affiliation(s)
- Sean P Polster
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Anukriti Sharma
- Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA, 19146, USA
| | - Alan T Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Patricia Mericko
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Ying Cao
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Julián Carrión-Penagos
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Romuald Girard
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Janne Koskimäki
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Dongdong Zhang
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Agnieszka Stadnik
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Sharbel G Romanos
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Seán B Lyne
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Robert Shenkar
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
| | - Kimberly Yan
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Cornelia Lee
- Angioma Alliance, 520W 21st Street, Norfolk, VA, 23517, USA
| | - Amy Akers
- Angioma Alliance, 520W 21st Street, Norfolk, VA, 23517, USA
| | - Leslie Morrison
- Department of Neurology, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Myranda Robinson
- Department of Neurology, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Atif Zafar
- Department of Neurology, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, 2716 South Street, Philadelphia, PA, 19146, USA
| | - Helen Kim
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Jack A Gilbert
- Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA
- Department of Pediatrics, The University of California San Diego and Scripps Institution of Oceanography, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Le Shen
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA.
- Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA.
| | - Issam A Awad
- Section of Neurosurgery, Department of Surgery, The University of Chicago, 5841S. Maryland Avenue, Chicago, IL, 60637, USA.
| |
Collapse
|
23
|
Polster SP, Cao Y, Carroll T, Flemming K, Girard R, Hanley D, Hobson N, Kim H, Koenig J, Koskimäki J, Lane K, Majersik JJ, McBee N, Morrison L, Shenkar R, Stadnik A, Thompson RE, Zabramski J, Zeineddine HA, Awad IA. Trial Readiness in Cavernous Angiomas With Symptomatic Hemorrhage (CASH). Neurosurgery 2020; 84:954-964. [PMID: 29660039 DOI: 10.1093/neuros/nyy108] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/06/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Brain cavernous angiomas with symptomatic hemorrhage (CASH) are uncommon but exact a heavy burden of neurological disability from recurrent bleeding, for which there is no proven therapy. Candidate drugs to stabilize the CASH lesion and prevent rebleeding will ultimately require testing of safety and efficacy in multisite clinical trials. Much progress has been made in understanding the epidemiology of CASH, and novel biomarkers have been linked to the biological mechanisms and clinical activity in lesions. Yet, the ability to enroll and risk-stratify CASH subjects has never been assessed prospectively at multiple sites. Biomarkers and other outcomes have not been evaluated for their sensitivity and reliability, nor have they been harmonized across sites. OBJECTIVE To address knowledge gaps and establish a research network as infrastructure for future clinical trials, through the Trial Readiness grant mechanism, funded by National Institute of Neurological Disorders and Stroke/National Institutes of Health. METHODS This project includes an observational cohort study to assess (1) the feasibility of screening, enrollment rates, baseline disease categorization, and follow-up of CASH using common data elements at multiple sites, (2) the reliability of imaging biomarkers including quantitative susceptibility mapping and permeability measures that have been shown to correlate with lesion activity, and (3) the rates of recurrent hemorrhage and change in functional status and biomarker measurements during prospective follow-up. EXPECTED OUTCOMES We propose a harmonized multisite assessment of enrollment rates of CASH, baseline features relevant to stratification in clinical trials, and follow-up assessments of functional outcomes in relation to clinical bleeds. We introduce novel biomarkers of vascular leak and hemorrhage, with firm mechanistic foundations, which have been linked to clinical disease activity. We shall test their reliability and validity at multiple sites, and assess their changes over time, with and without clinical rebleeds, hence their fitness as outcome instruments in clinical trials. DISCUSSION The timing cannot be more opportune, with therapeutic targets identified, exceptional collaboration among researchers and the patient community, along with several drugs ready to benefit from development of a path to clinical testing using this network in the next 5 years.
Collapse
Affiliation(s)
- Sean P Polster
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Timothy Carroll
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Kelly Flemming
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Daniel Hanley
- Division of Brain Injury Outcomes, Department of Neurology, Johns Hopkins University Medical Institutions, Baltimore, Maryland
| | - Nicholas Hobson
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Helen Kim
- Center for Cerebrovascular Research, Department of Anesthesiology, University of California San Francisco, San Francisco, California
| | - James Koenig
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland
| | - Janne Koskimäki
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Karen Lane
- Division of Brain Injury Outcomes, Department of Neurology, Johns Hopkins University Medical Institutions, Baltimore, Maryland
| | | | - Nichol McBee
- Division of Brain Injury Outcomes, Department of Neurology, Johns Hopkins University Medical Institutions, Baltimore, Maryland
| | - Leslie Morrison
- Department of Neurology, University of New Mexico, Albuquerque, New Mexico
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Agnieszka Stadnik
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Richard E Thompson
- Division of Brain Injury Outcomes, Department of Neurology, Johns Hopkins University Medical Institutions, Baltimore, Maryland
| | - Joseph Zabramski
- Department of Neurological Surgery, The Barrow Neurological Institute, Phoenix, ArizonaAll except the first and final author are listed in alphabetic order
| | - Hussein A Zeineddine
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois
| |
Collapse
|
24
|
Abdelilah-Seyfried S, Tournier-Lasserve E, Derry WB. Blocking Signalopathic Events to Treat Cerebral Cavernous Malformations. Trends Mol Med 2020; 26:874-887. [PMID: 32692314 DOI: 10.1016/j.molmed.2020.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
Cerebral cavernous malformations (CCMs) are pathologies of the brain vasculature characterized by capillary-venous angiomas that result in recurrent cerebral hemorrhages. Familial forms are caused by a clonal loss of any of three CCM genes in endothelial cells, which causes the activation of a novel pathophysiological pathway involving mitogen-activated protein kinase and Krüppel-like transcription factor KLF2/4 signaling. Recent work has shown that cavernomas can undergo strong growth when CCM-deficient endothelial cells recruit wild-type neighbors through the secretion of cytokines. This suggests a treatment strategy based on targeting signalopathic events between CCM-deficient endothelial cells and their environment. Such approaches will have to consider recent evidence implicating 'third hits' from hypoxia-induced angiogenesis signaling or the microbiome in modulating the development of cerebral hemorrhages.
Collapse
Affiliation(s)
- Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, D-30625 Hannover, Germany.
| | - Elisabeth Tournier-Lasserve
- INSERM UMR-1141, NeuroDiderot, Université de Paris, Paris, France; AP-HP, Groupe hospitalier Saint-Louis, Lariboisière, Fernand-Widal, Service de génétique moléculaire neuro-vasculaire, Paris, France
| | - W Brent Derry
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8; Developmental and Cell Biology Program, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, Canada M5G 0A4
| |
Collapse
|
25
|
Symptomatic Brain Hemorrhages from Cavernous Angioma After Botulinum Toxin Injections, a Role of TLR/MEKK3 Mechanism? Case Report and Review of the Literature. World Neurosurg 2020; 136:7-11. [PMID: 31917316 DOI: 10.1016/j.wneu.2019.12.172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/29/2019] [Accepted: 12/30/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Cavernous angiomas (CAs) are vascular malformations that may result in stroke. CASE DESCRIPTION Herein, we evaluate a CA patient with chronic migraine who experienced 2 documented symptomatic hemorrhages after receiving respective high doses of botulinum toxin (Btx). CONCLUSIONS Recently, bacterial lipopolysaccharide has been reported to contribute to CA development through Toll-like receptor signaling, causing hemorrhagic angiogenic proliferation. Lipopolysaccharide and Btx share a common intracellular signaling pathway driving CA development and hemorrhage. Significance of these observations is demonstrated by previous works on plasma molecules showing prognostic associations with symptomatic hemorrhages in human CA, related to the same canonical pathways. Authors suggest careful tracking of the association of Btx and hemorrhage in CA patients.
Collapse
|
26
|
Shenkar R, Peiper A, Pardo H, Moore T, Lightle R, Girard R, Hobson N, Polster SP, Koskimäki J, Zhang D, Lyne SB, Cao Y, Chaudagar K, Saadat L, Gallione C, Pytel P, Liao JK, Marchuk D, Awad IA. Rho Kinase Inhibition Blunts Lesion Development and Hemorrhage in Murine Models of Aggressive Pdcd10/Ccm3 Disease. Stroke 2019; 50:738-744. [PMID: 30744543 DOI: 10.1161/strokeaha.118.024058] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background and Purpose- Previously, murine models Krit1 +/- Msh2 -/ - and Ccm2 +/ - Trp53 -/ - showed a reduction or no effect on cerebral cavernous malformation (CCM) burden and favorable effects on lesional hemorrhage by the robust Rock (Rho-associated protein kinase) inhibitor fasudil and by simvastatin (a weak pleiotropic inhibitor of Rock). Herein, we concurrently investigated treatment of the more aggressive Pdcd10/Ccm3 model with fasudil, simvastatin, and higher dose atorvastatin to determined effectiveness of Rock inhibition. Methods- The murine models, Pdcd10 +/ - Trp53 -/ - and Pdcd10 +/ - Msh2 -/ -, were contemporaneously treated from weaning to 5 months of age with fasudil (100 mg/kg per day in drinking water, n=9), simvastatin (40 mg/kg per day in chow, n=11), atorvastatin (80 mg/kg per day in chow, n=10), or with placebo (n=16). We assessed CCM volume in mouse brains by microcomputed tomography. Lesion burden was calculated as lesion volume normalized to total brain volume. We analyzed chronic hemorrhage in CCM lesions by quantitative intensity of Perls staining in brain sections. Results- The Pdcd10 +/ - Trp53 -/ - /Msh2 -/ - models showed a mean CCM lesion burden per mouse reduction from 0.0091 in placebos to 0.0042 ( P=0.027) by fasudil, and to 0.0047 ( P=0.025) by atorvastatin treatment, but was not changed significantly by simvastatin. Hemorrhage intensity per brain was commensurately decreased by Rock inhibition. Conclusions- These results support the exploration of proof of concept effect of high-dose atorvastatin on human CCM disease for potential therapeutic testing.
Collapse
Affiliation(s)
- Robert Shenkar
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Amy Peiper
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (A.P., H.P., C.G., D.M.)
| | - Heidy Pardo
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (A.P., H.P., C.G., D.M.)
| | - Thomas Moore
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Rhonda Lightle
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Romuald Girard
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Nicholas Hobson
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Sean P Polster
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Janne Koskimäki
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Dongdong Zhang
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Seán B Lyne
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Ying Cao
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Kiranj Chaudagar
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Laleh Saadat
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| | - Carol Gallione
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (A.P., H.P., C.G., D.M.)
| | - Peter Pytel
- Department of Pathology (P.P.), Biological Sciences Division, University of Chicago, IL
| | - James K Liao
- Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL
| | - Douglas Marchuk
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (A.P., H.P., C.G., D.M.)
| | - Issam A Awad
- From the Section of Neurosurgery (R.S., T.M., R.L., R.G., N.H., S.P.P., J.K., D.Z., S.B.L., Y.C., K.C., L.S., I.A.A.), Biological Sciences Division, University of Chicago, IL
| |
Collapse
|
27
|
McKerracher L, Shenkar R, Abbinanti M, Cao Y, Peiper A, Liao JK, Lightle R, Moore T, Hobson N, Gallione C, Ruschel J, Koskimäki J, Girard R, Rosen K, Marchuk DA, Awad IA. A Brain-Targeted Orally Available ROCK2 Inhibitor Benefits Mild and Aggressive Cavernous Angioma Disease. Transl Stroke Res 2019; 11:365-376. [PMID: 31446620 DOI: 10.1007/s12975-019-00725-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/01/2019] [Accepted: 08/13/2019] [Indexed: 12/01/2022]
Abstract
Cavernous angioma (CA) is a vascular pathology caused by loss of function in one of the 3 CA genes (CCM1, CCM2, and CCM3) that result in rho kinase (ROCK) activation. We investigated a novel ROCK2 selective inhibitor for the ability to reduce brain lesion formation, growth, and maturation. We used genetic methods to explore the use of a ROCK2-selective kinase inhibitor to reduce growth and hemorrhage of CAs. The role of ROCK2 in CA was investigated by crossing Rock1 or Rock2 hemizygous mice with Ccm1 or Ccm3 hemizygous mice, and we found reduced lesions in the Rock2 hemizygous mice. A ROCK2-selective inhibitor, BA-1049 was used to investigate efficacy in reducing CA lesions after oral administration to Ccm1+/- and Ccm3+/- mice that were bred into a mutator background. After assessing the dose range effective to target brain endothelial cells in an ischemic brain model, Ccm1+/- and Ccm3+/- transgenic mice were treated for 3 (Ccm3+/-) or 4 months (Ccm1+/-), concurrently, randomized to receive one of three doses of BA-1049 in drinking water, or placebo. Lesion volumes were assessed by micro-computed tomography. BA-1049 reduced activation of ROCK2 in Ccm3+/-Trp53-/- lesions. Ccm1+/-Msh2-/- (n=68) and Ccm3+/-Trp53-/- (n=71) mice treated with BA-1049 or placebo showed a significant dose-dependent reduction in lesion volume after treatment with BA-1049, and a reduction in hemorrhage (iron deposition) near lesions at all doses. These translational studies show that BA-1049 is a promising therapeutic agent for the treatment of CA, a disease with no current treatment except surgical removal of the brain lesions.
Collapse
Affiliation(s)
- Lisa McKerracher
- BioAxone BioSciences Inc., Cambridge, MA, USA.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | | | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Amy Peiper
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - James K Liao
- Section of Cardiology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Nicholas Hobson
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Carol Gallione
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | | | - Janne Koskimäki
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA
| | | | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL, USA.
| |
Collapse
|
28
|
Koskimäki J, Zhang D, Li Y, Saadat L, Moore T, Lightle R, Polster SP, Carrión-Penagos J, Lyne SB, Zeineddine HA, Shi C, Shenkar R, Romanos S, Avner K, Srinath A, Shen L, Detter MR, Snellings D, Cao Y, Lopez-Ramirez MA, Fonseca G, Tang AT, Faber P, Andrade J, Ginsberg M, Kahn ML, Marchuk DA, Girard R, Awad IA. Transcriptome clarifies mechanisms of lesion genesis versus progression in models of Ccm3 cerebral cavernous malformations. Acta Neuropathol Commun 2019; 7:132. [PMID: 31426861 PMCID: PMC6699077 DOI: 10.1186/s40478-019-0789-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023] Open
Abstract
Cerebral cavernous malformations (CCMs) are dilated capillaries causing epilepsy and stroke. Inheritance of a heterozygous mutation in CCM3/PDCD10 is responsible for the most aggressive familial form of the disease. Here we studied the differences and commonalities between the transcriptomes of microdissected lesional neurovascular units (NVUs) from acute and chronic in vivo Ccm3/Pdcd10ECKO mice, and cultured brain microvascular endothelial cells (BMECs) Ccm3/Pdcd10ECKO.We identified 2409 differentially expressed genes (DEGs) in acute and 2962 in chronic in vivo NVUs compared to microdissected brain capillaries, as well as 121 in in vitro BMECs with and without Ccm3/Pdcd10 loss (fold change ≥ |2.0|; p < 0.05, false discovery rate corrected). A functional clustered dendrogram generated using the Euclidean distance showed that the DEGs identified only in acute in vivo NVUs were clustered in cellular proliferation gene ontology functions. The DEGs only identified in chronic in vivo NVUs were clustered in inflammation and immune response, permeability, and adhesion functions. In addition, 1225 DEGs were only identified in the in vivo NVUs but not in vitro BMECs, and these clustered within neuronal and glial functions. One miRNA mmu-miR-3472a was differentially expressed (FC = - 5.98; p = 0.07, FDR corrected) in the serum of Ccm3/Pdcd10+/- when compared to wild type mice, and this was functionally related as a putative target to Cand2 (cullin associated and neddylation dissociated 2), a DEG in acute and chronic lesional NVUs and in vitro BMECs. Our results suggest that the acute model is characterized by cell proliferation, while the chronic model showed inflammatory, adhesion and permeability processes. In addition, we highlight the importance of extra-endothelial structures in CCM disease, and potential role of circulating miRNAs as biomarkers of disease, interacting with DEGs. The extensive DEGs library of each model will serve as a validation tool for potential mechanistic, biomarker, and therapeutic targets.
Collapse
Affiliation(s)
- Janne Koskimäki
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Dongdong Zhang
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Yan Li
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Laleh Saadat
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Sean P Polster
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Julián Carrión-Penagos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Seán B Lyne
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Hussein A Zeineddine
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Changbin Shi
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Kenneth Avner
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Le Shen
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Matthew R Detter
- The Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Daniel Snellings
- The Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | | | - Gregory Fonseca
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alan T Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Pieter Faber
- University of Chicago Genomics Facility, The University of Chicago, Chicago, IL, USA
| | - Jorge Andrade
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Mark Ginsberg
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas A Marchuk
- The Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA.
| |
Collapse
|
29
|
Awad IA, Polster SP. Cavernous angiomas: deconstructing a neurosurgical disease. J Neurosurg 2019; 131:1-13. [PMID: 31261134 PMCID: PMC6778695 DOI: 10.3171/2019.3.jns181724] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/15/2019] [Indexed: 01/08/2023]
Abstract
Cavernous angioma (CA) is also known as cavernoma, cavernous hemangioma, and cerebral cavernous malformation (CCM) (National Library of Medicine Medical Subject heading unique ID D006392). In its sporadic form, CA occurs as a solitary hemorrhagic vascular lesion or as clustered lesions associated with a developmental venous anomaly. In its autosomal dominant familial form (Online Mendelian Inheritance in Man #116860), CA is caused by a heterozygous germline loss-of-function mutation in one of three genes-CCM1/KRIT1, CCM2/Malcavernin, and CCM3/PDCD10-causing multifocal lesions throughout the brain and spinal cord.In this paper, the authors review the cardinal features of CA's disease pathology and clinical radiological features. They summarize key aspects of CA's natural history and broad elements of evidence-based management guidelines, including surgery. The authors also discuss evidence of similar genetic defects in sporadic and familial lesions, consequences of CCM gene loss in different tissues at various stages of development, and implications regarding the pathobiology of CAs.The concept of CA with symptomatic hemorrhage (CASH) is presented as well as its relevance to clinical care and research in the field. Pathobiological mechanisms related to CA include inflammation and immune-mediated processes, angiogenesis and vascular permeability, microbiome driven factors, and lesional anticoagulant domains. These mechanisms have motivated the development of imaging and plasma biomarkers of relevant disease behavior and promising therapeutic targets.The spectrum of discoveries about CA and their implications endorse CA as a paradigm for deconstructing a neurosurgical disease.
Collapse
|
30
|
Lyne SB, Girard R, Koskimäki J, Zeineddine HA, Zhang D, Cao Y, Li Y, Stadnik A, Moore T, Lightle R, Shi C, Shenkar R, Carrión-Penagos J, Polster SP, Romanos S, Akers A, Lopez-Ramirez M, Whitehead KJ, Kahn ML, Ginsberg MH, Marchuk DA, Awad IA. Biomarkers of cavernous angioma with symptomatic hemorrhage. JCI Insight 2019; 4:128577. [PMID: 31217347 PMCID: PMC6629090 DOI: 10.1172/jci.insight.128577] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/01/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUNDCerebral cavernous angiomas (CAs) with a symptomatic hemorrhage (CASH) have a high risk of recurrent hemorrhage and serious morbidity.METHODSEighteen plasma molecules with mechanistic roles in CA pathobiology were investigated in 114 patients and 12 healthy subjects. The diagnostic biomarker of a CASH in the prior year was derived as that minimizing the Akaike information criterion and validated using machine learning, and was compared with the prognostic CASH biomarker predicting bleeding in the subsequent year. Biomarkers were longitudinally followed in a subset of cases. The biomarkers were queried in the lesional neurovascular unit (NVU) transcriptome and in plasma miRNAs from CASH and non-CASH patients.RESULTSThe diagnostic CASH biomarker included a weighted combination of soluble CD14 (sCD14), VEGF, C-reactive protein (CRP), and IL-10 distinguishing CASH patients with 76% sensitivity and 80% specificity (P = 0.0003). The prognostic CASH biomarker (sCD14, VEGF, IL-1β, and sROBO-4) was confirmed to predict a bleed in the subsequent year with 83% sensitivity and 93% specificity (P = 0.001). Genes associated with diagnostic and prognostic CASH biomarkers were differentially expressed in CASH lesional NVUs. Thirteen plasma miRNAs were differentially expressed between CASH and non-CASH patients.CONCLUSIONShared and unique biomarkers of recent symptomatic hemorrhage and of future bleeding in CA are mechanistically linked to lesional transcriptome and miRNA. The biomarkers may be applied for risk stratification in clinical trials and developed as a tool in clinical practice.FUNDINGNIH, William and Judith Davis Fund in Neurovascular Surgery Research, Be Brave for Life Foundation, Safadi Translational Fellowship, Pritzker School of Medicine, and Sigrid Jusélius Foundation.
Collapse
Affiliation(s)
- Seán B. Lyne
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Romuald Girard
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Janne Koskimäki
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Hussein A. Zeineddine
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Dongdong Zhang
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Ying Cao
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Yan Li
- Center for Research Informatics, The University of Chicago, Chicago, Illinois, USA
| | - Agnieszka Stadnik
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Thomas Moore
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Rhonda Lightle
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Changbin Shi
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Robert Shenkar
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Julián Carrión-Penagos
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sean P. Polster
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sharbel Romanos
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Amy Akers
- Angioma Alliance, Norfolk, Virginia, USA
| | | | - Kevin J. Whitehead
- Division of Cardiology and Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Mark L. Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Douglas A. Marchuk
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, North Carolina, USA
| | - Issam A. Awad
- Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| |
Collapse
|
31
|
Zeineddine HA, Girard R, Saadat L, Shen L, Lightle R, Moore T, Cao Y, Hobson N, Shenkar R, Avner K, Chaudager K, Koskimäki J, Polster SP, Fam MD, Shi C, Lopez-Ramirez MA, Tang AT, Gallione C, Kahn ML, Ginsberg M, Marchuk DA, Awad IA. Phenotypic characterization of murine models of cerebral cavernous malformations. J Transl Med 2019; 99:319-330. [PMID: 29946133 PMCID: PMC6309944 DOI: 10.1038/s41374-018-0030-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 11/09/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are clusters of dilated capillaries that affect around 0.5% of the population. CCMs exist in two forms, sporadic and familial. Mutations in three documented genes, KRIT1(CCM1), CCM2, and PDCD10(CCM3), cause the autosomal dominant form of the disease, and somatic mutations in these same genes underlie lesion development in the brain. Murine models with constitutive or induced loss of respective genes have been applied to study disease pathobiology and therapeutic manipulations. We aimed to analyze the phenotypic characteristic of two main groups of models, the chronic heterozygous models with sensitizers promoting genetic instability, and the acute neonatal induced homozygous knockout model. Acute model mice harbored a higher lesion burden than chronic models, more localized in the hindbrain, and largely lacking iron deposition and inflammatory cell infiltrate. The chronic model mice showed a lower lesion burden localized throughout the brain, with significantly greater perilesional iron deposition, immune B- and T-cell infiltration, and less frequent junctional protein immunopositive endothelial cells. Lesional endothelial cells in both models expressed similar phosphorylated myosin light chain immunopositivity indicating Rho-associated protein kinase activity. These data suggest that acute models are better suited to study the initial formation of the lesion, while the chronic models better reflect lesion maturation, hemorrhage, and inflammatory response, relevant pathobiologic features of the human disease.
Collapse
Affiliation(s)
- Hussein A. Zeineddine
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Laleh Saadat
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Le Shen
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA,Department of Pathology, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Nick Hobson
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Kenneth Avner
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Kiranj Chaudager
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Janne Koskimäki
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Sean P. Polster
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Maged D. Fam
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | - Changbin Shi
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| | | | - Alan T. Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA USA
| | - Carol Gallione
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC USA
| | - Mark L. Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA USA
| | - Mark Ginsberg
- Department of Medicine, University of California, San Diego, CA USA
| | - Douglas A. Marchuk
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC USA
| | - Issam A. Awad
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL USA
| |
Collapse
|
32
|
Koskimäki J, Girard R, Li Y, Saadat L, Zeineddine HA, Lightle R, Moore T, Lyne S, Avner K, Shenkar R, Cao Y, Shi C, Polster SP, Zhang D, Carrión-Penagos J, Romanos S, Fonseca G, Lopez-Ramirez MA, Chapman EM, Popiel E, Tang AT, Akers A, Faber P, Andrade J, Ginsberg M, Derry WB, Kahn ML, Marchuk DA, Awad IA. Comprehensive transcriptome analysis of cerebral cavernous malformation across multiple species and genotypes. JCI Insight 2019; 4:126167. [PMID: 30728328 DOI: 10.1172/jci.insight.126167] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/03/2019] [Indexed: 12/18/2022] Open
Abstract
The purpose of this study was to determine important genes, functions, and networks contributing to the pathobiology of cerebral cavernous malformation (CCM) from transcriptomic analyses across 3 species and 2 disease genotypes. Sequencing of RNA from laser microdissected neurovascular units of 5 human surgically resected CCM lesions, mouse brain microvascular endothelial cells, Caenorhabditis elegans with induced Ccm gene loss, and their respective controls provided differentially expressed genes (DEGs). DEGs from mouse and C. elegans were annotated into human homologous genes. Cross-comparisons of DEGs between species and genotypes, as well as network and gene ontology (GO) enrichment analyses, were performed. Among hundreds of DEGs identified in each model, common genes and 1 GO term (GO:0051656, establishment of organelle localization) were commonly identified across the different species and genotypes. In addition, 24 GO functions were present in 4 of 5 models and were related to cell-to-cell adhesion, neutrophil-mediated immunity, ion transmembrane transporter activity, and responses to oxidative stress. We have provided a comprehensive transcriptome library of CCM disease across species and for the first time to our knowledge in Ccm1/Krit1 versus Ccm3/Pdcd10 genotypes. We have provided examples of how results can be used in hypothesis generation or mechanistic confirmatory studies.
Collapse
Affiliation(s)
- Janne Koskimäki
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Yan Li
- Center for Research Informatics, The University of Chicago, Chicago, Illinois, USA
| | - Laleh Saadat
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Hussein A Zeineddine
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Seán Lyne
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Kenneth Avner
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Changbin Shi
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sean P Polster
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Dongdong Zhang
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Julián Carrión-Penagos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | | | | | - Eric M Chapman
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Evelyn Popiel
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Alan T Tang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amy Akers
- Angioma Alliance, Norfolk, Virginia, USA
| | - Pieter Faber
- University of Chicago Genomics Facility, The University of Chicago, Chicago, Illinois, USA
| | - Jorge Andrade
- Center for Research Informatics, The University of Chicago, Chicago, Illinois, USA
| | - Mark Ginsberg
- Department of Medicine, UCSD, La Jolla, California, USA
| | - W Brent Derry
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Douglas A Marchuk
- The Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, North Carolina, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| |
Collapse
|
33
|
Girard R, Zeineddine HA, Fam MD, Mayampurath A, Cao Y, Shi C, Shenkar R, Polster SP, Jesselson M, Duggan R, Mikati AG, Christoforidis G, Andrade J, Whitehead KJ, Li DY, Awad IA. Plasma Biomarkers of Inflammation Reflect Seizures and Hemorrhagic Activity of Cerebral Cavernous Malformations. Transl Stroke Res 2017; 9:34-43. [PMID: 28819935 DOI: 10.1007/s12975-017-0561-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/01/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022]
Abstract
The clinical course of cerebral cavernous malformations (CCMs) is highly variable. Based on recent discoveries implicating angiogenic and inflammatory mechanisms, we hypothesized that serum biomarkers might reflect chronic or acute disease activity. This single-site prospective observational cohort study included 85 CCM patients, in whom 24 a priori chosen plasma biomarkers were quantified and analyzed in relation to established clinical and imaging parameters of disease categorization and severity. We subsequently validated the positive correlations in longitudinal follow-up of 49 subjects. Plasma levels of matrix metalloproteinase-2 and intercellular adhesion molecule 1 were significantly higher (P = 0.02 and P = 0.04, respectively, FDR corrected), and matrix metalloproteinase-9 was lower (P = 0.04, FDR corrected) in patients with seizure activity at any time in the past. Vascular endothelial growth factor and endoglin (both P = 0.04, FDR corrected) plasma levels were lower in patients who had suffered a symptomatic bleed in the prior 3 months. The hierarchical clustering analysis revealed a cluster of four plasma inflammatory cytokines (interleukin 2, interferon gamma, tumor necrosis factor alpha, and interleukin 1 beta) separating patients into what we designated "high" and "low" inflammatory states. The "high" inflammatory state was associated with seizure activity (P = 0.02) and more than one hemorrhagic event during a patient's lifetime (P = 0.04) and with a higher rate of new hemorrhage, lesion growth, or new lesion formation (P < 0.05) during prospective follow-up. Peripheral plasma biomarkers reflect seizure and recent hemorrhagic activity in CCM patients. In addition, four clustered inflammatory biomarkers correlate with cumulative disease aggressiveness and predict future clinical activity.
Collapse
Affiliation(s)
- Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Hussein A Zeineddine
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Maged D Fam
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Anoop Mayampurath
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Changbin Shi
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Sean P Polster
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Michael Jesselson
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Ryan Duggan
- Flow Cytometry Facility, The University of Chicago, Chicago, IL, USA
| | - Abdul-Ghani Mikati
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Gregory Christoforidis
- Section Neuroradiology, Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Jorge Andrade
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Kevin J Whitehead
- Division of Cardiology, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Dean Y Li
- Division of Cardiology, Department of Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA.
| |
Collapse
|
34
|
Zeineddine HA, Girard R, Cao Y, Hobson N, Fam MD, Stadnik A, Tan H, Shen J, Chaudagar K, Shenkar R, Thompson RE, McBee N, Hanley D, Carroll T, Christoforidis GA, Awad IA. Quantitative susceptibility mapping as a monitoring biomarker in cerebral cavernous malformations with recent hemorrhage. J Magn Reson Imaging 2017; 47:1133-1138. [PMID: 28791783 DOI: 10.1002/jmri.25831] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/24/2017] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Quantitative Susceptibility Mapping (QSM) MRI allows accurate assessment of iron content in cerebral cavernous malformations (CCM), and a threshold increase by 6% in QSM has been shown to reflect new symptomatic hemorrhage (SH) in previously stable lesions. PURPOSE/HYPOTHESIS It is unclear how lesional QSM evolves in CCMs after recent SH, and whether this could serve as a monitoring biomarker in clinical trials aimed at preventing rebleeding in these lesions. STUDY TYPE This is a prospective observational cohort study. POPULATION 16 CCM patients who experienced a SH within the past year, whose lesion was not resected or irradiated. FIELD STRENGTH/SEQUENCE The data acquisition was performed using QSM sequence implemented on a 3T MRI system ASSESSMENT: The lesional QSM assessments at baseline and yearly during 22 patient-years of follow-up were performed by a trained research staff including imaging scientists. STATISTICAL TESTS Biomarker changes were assessed in relation to clinical events. Clinical trial modeling was performed using two-tailed tests of time-averaged difference (assuming within-patient correlation of 0.8, power = 0.9 and alpha = 0.1) to detect 20%, 30% or 50% effects of intervention on clinical and biomarkers event rates during two years of follow-up. RESULTS The change in mean lesional QSM of index hemorrhagic lesions was +7.93% per patient-year in the whole cohort. There were 5 cases (31%) of recurrent SH or lesional growth, and twice as many instances (62%) with a threshold (6%) increase in QSM. There were no instances of SH hemorrhage or lesional growth without an associated threshold increase in QSM during the same epoch. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 4 J. Magn. Reson. Imaging 2018;47:1133-1138.
Collapse
Affiliation(s)
- Hussein A Zeineddine
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Romuald Girard
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Ying Cao
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Nicholas Hobson
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Maged D Fam
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Agnieszka Stadnik
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Huan Tan
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Jingjing Shen
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Kiranj Chaudagar
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Robert Shenkar
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Richard E Thompson
- Brain Injury Outcomes unit, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nichol McBee
- Brain Injury Outcomes unit, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel Hanley
- Brain Injury Outcomes unit, Johns Hopkins University, Baltimore, Maryland, USA
| | - Timothy Carroll
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Gregory A Christoforidis
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Issam A Awad
- Section of Neurosurgery, Department of Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| |
Collapse
|
35
|
Shenkar R, Shi C, Austin C, Moore T, Lightle R, Cao Y, Zhang L, Wu M, Zeineddine HA, Girard R, McDonald DA, Rorrer A, Gallione C, Pytel P, Liao JK, Marchuk DA, Awad IA. RhoA Kinase Inhibition With Fasudil Versus Simvastatin in Murine Models of Cerebral Cavernous Malformations. Stroke 2016; 48:187-194. [PMID: 27879448 DOI: 10.1161/strokeaha.116.015013] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/12/2016] [Accepted: 10/14/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND PURPOSE We sought to compare the effect of chronic treatment with commonly tolerated doses of Fasudil, a specific RhoA kinase (ROCK) inhibitor, and simvastatin (with pleiotropic effects including ROCK inhibition) on cerebral cavernous malformation (CCM) genesis and maturation in 2 models that recapitulate the human disease. METHODS Two heterozygous murine models, Ccm1+/-Msh2-/- and Ccm2+/-Trp53-/-, were treated from weaning to 4 to 5 months of age with Fasudil (100 mg/kg per day), simvastatin (40 mg/kg per day) or with placebo. Mouse brains were blindly assessed for CCM lesion burden, nonheme iron deposition (as a quantitative measure of chronic lesional hemorrhage), and ROCK activity. RESULTS Fasudil, but not simvastatin, significantly decreased mature CCM lesion burden in Ccm1+/-Msh2-/- mice, and in meta-analysis of both models combined, when compared with mice receiving placebo. Fasudil and simvastatin both significantly decreased the integrated iron density per mature lesion area in Ccm1+/-Msh2-/- mice, and in both models combined, compared with mice given placebo. ROCK activity in mature lesions of Ccm1+/-Msh2-/- mice was similar with both treatments. Fasudil, but not simvastatin, improved survival in Ccm1+/-Msh2-/- mice. Fasudil and simvastatin treatment did not affect survival or lesion development significantly in Ccm2+/-Trp53-/- mice alone, and Fasudil benefit seemed limited to males. CONCLUSIONS ROCK inhibitor Fasudil was more efficacious than simvastatin in improving survival and blunting the development of mature CCM lesions. Both drugs significantly decreased chronic hemorrhage in CCM lesions. These findings justify the development of ROCK inhibitors and the clinical testing of commonly used statin agents in CCM.
Collapse
Affiliation(s)
- Robert Shenkar
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Changbin Shi
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Cecilia Austin
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Thomas Moore
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Rhonda Lightle
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Ying Cao
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Lingjiao Zhang
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Meijing Wu
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Hussein A Zeineddine
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Romuald Girard
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - David A McDonald
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Autumn Rorrer
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Carol Gallione
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Peter Pytel
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - James K Liao
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Douglas A Marchuk
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk)
| | - Issam A Awad
- From the Section of Neurosurgery (R.S., C.S., C.A., T.M., R.L., Y.C., L.Z., M.W., H.A.Z., R.G., I.A.A.), Department of Pathology (P.P.), Section of Cardiology (J.K.L.), Biological Sciences Division, University of Chicago, IL; and the Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC (D.A. McDonald, A.R., C.G., D.A. Marchuk).
| |
Collapse
|
36
|
Retta SF, Glading AJ. Oxidative stress and inflammation in cerebral cavernous malformation disease pathogenesis: Two sides of the same coin. Int J Biochem Cell Biol 2016; 81:254-270. [PMID: 27639680 PMCID: PMC5155701 DOI: 10.1016/j.biocel.2016.09.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
Abstract
CCM proteins play pleiotropic roles in various redox-sensitive signaling pathways. CCM proteins modulate the crosstalk between redox signaling and autophagy that govern cell homeostasis and stress responses. Oxidative stress and inflammation are emerging as key focal determinants of CCM lesion formation, progression and severity. The pleiotropic functions of CCM proteins may prevent vascular dysfunctions triggered by local oxidative stress and inflammatory events. The distinct therapeutic compounds proposed so far for CCM disease share the ability to modulate redox signaling and autophagy.
Cerebral Cavernous Malformation (CCM) is a vascular disease of proven genetic origin, which may arise sporadically or is inherited as an autosomal dominant condition with incomplete penetrance and highly variable expressivity. CCM lesions exhibit a range of different phenotypes, including wide inter-individual differences in lesion number, size, and susceptibility to intracerebral hemorrhage (ICH). Lesions may remain asymptomatic or result in pathological conditions of various type and severity at any age, with symptoms ranging from recurrent headaches to severe neurological deficits, seizures, and stroke. To date there are no direct therapeutic approaches for CCM disease besides the surgical removal of accessible lesions. Novel pharmacological strategies are particularly needed to limit disease progression and severity and prevent de novo formation of CCM lesions in susceptible individuals. Useful insights into innovative approaches for CCM disease prevention and treatment are emerging from a growing understanding of the biological functions of the three known CCM proteins, CCM1/KRIT1, CCM2 and CCM3/PDCD10. In particular, accumulating evidence indicates that these proteins play major roles in distinct signaling pathways, including those involved in cellular responses to oxidative stress, inflammation and angiogenesis, pointing to pathophysiological mechanisms whereby the function of CCM proteins may be relevant in preventing vascular dysfunctions triggered by these events. Indeed, emerging findings demonstrate that the pleiotropic roles of CCM proteins reflect their critical capacity to modulate the fine-tuned crosstalk between redox signaling and autophagy that govern cell homeostasis and stress responses, providing a novel mechanistic scenario that reconciles both the multiple signaling pathways linked to CCM proteins and the distinct therapeutic approaches proposed so far. In addition, recent studies in CCM patient cohorts suggest that genetic susceptibility factors related to differences in vascular sensitivity to oxidative stress and inflammation contribute to inter-individual differences in CCM disease susceptibility and severity. This review discusses recent progress into the understanding of the molecular basis and mechanisms of CCM disease pathogenesis, with specific emphasis on the potential contribution of altered cell responses to oxidative stress and inflammatory events occurring locally in the microvascular environment, and consequent implications for the development of novel, safe, and effective preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Saverio Francesco Retta
- Department of Clinical and Biological Sciences, School of Medicine and Surgery, University of Torino, Regione Gonzole 10, 10043 Orbassano, Torino, Italy; CCM Italia Research Network(1).
| | - Angela J Glading
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, 14642 Rochester, NY, USA.
| |
Collapse
|