1
|
Shapeti A, Barrasa-Fano J, Abdel Fattah AR, de Jong J, Sanz-Herrera JA, Pezet M, Assou S, de Vet E, Elahi SA, Ranga A, Faurobert E, Van Oosterwyck H. Force-mediated recruitment and reprogramming of healthy endothelial cells drive vascular lesion growth. Nat Commun 2024; 15:8660. [PMID: 39370485 PMCID: PMC11456588 DOI: 10.1038/s41467-024-52866-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
Force-driven cellular interactions are crucial for cancer cell invasion but remain underexplored in vascular abnormalities. Cerebral cavernous malformations (CCM), a vascular abnormality characterized by leaky vessels, involves CCM mutant cells recruiting wild-type endothelial cells to form and expand mosaic lesions. The mechanisms behind this recruitment remain poorly understood. Here, we use an in-vitro model of angiogenic invasion with traction force microscopy to reveal that hyper-angiogenic Ccm2-silenced endothelial cells enhance angiogenic invasion of neighboring wild-type cells through force and extracellular matrix-guided mechanisms. We demonstrate that mechanically hyperactive CCM2-silenced tips guide wild-type cells by transmitting pulling forces and by creating paths in the matrix, in a ROCKs-dependent manner. This is associated with reinforcement of β1 integrin and actin cytoskeleton in wild-type cells. Further, wild-type cells are reprogrammed into stalk cells and activate matrisome and DNA replication programs, thereby initiating proliferation. Our findings reveal how CCM2 mutants hijack wild-type cell functions to fuel lesion growth, providing insight into the etiology of vascular malformations. By integrating biophysical and molecular techniques, we offer tools for studying cell mechanics in tissue heterogeneity and disease progression.
Collapse
Affiliation(s)
- Apeksha Shapeti
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium.
| | - Jorge Barrasa-Fano
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
| | - Abdel Rahman Abdel Fattah
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
- CeMM The Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Janne de Jong
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
| | - José Antonio Sanz-Herrera
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBIS), Seville, Spain
| | - Mylène Pezet
- Univ. Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Said Assou
- IRMB, University of Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Emilie de Vet
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
| | - Seyed Ali Elahi
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
- KU Leuven, Department of Movement Sciences, Human Movement Biomechanics Research Group, Leuven, Belgium
| | - Adrian Ranga
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium
| | - Eva Faurobert
- Univ. Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, Grenoble, France.
| | - Hans Van Oosterwyck
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Belgium.
- KU Leuven, Prometheus, Division of Skeletal Tissue Engineering, Leuven, Belgium.
| |
Collapse
|
2
|
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
|
3
|
Alcazar-Felix RJ, Shenkar R, Benavides CR, Bindal A, Srinath A, Li Y, Kinkade S, Terranova T, DeBose-Scarlett E, Lightle R, DeBiasse D, Almazroue H, Cruz DV, Romanos S, Jhaveri A, Koskimäki J, Hage S, Bennett C, Girard R, Marchuk DA, Awad IA. Except for Robust Outliers, Rapamycin Increases Lesion Burden in a Murine Model of Cerebral Cavernous Malformations. Transl Stroke Res 2024:10.1007/s12975-024-01270-9. [PMID: 38980519 DOI: 10.1007/s12975-024-01270-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
Cerebral cavernous malformation (CCM) is a hemorrhagic cerebrovascular disease where lesions develop in the setting of endothelial mutations of CCM genes, with many cases also harboring somatic PIK3CA gain of function (GOF) mutations. Rapamycin, an mTORC1 inhibitor, inhibited progression of murine CCM lesions driven by Ccm gene loss and Pik3ca GOF, but it remains unknown if rapamycin is beneficial in the absence of induction of Pik3ca GOF. We investigated the effect of rapamycin at three clinically relevant doses on lesion development in the Ccm3-/-PDGFb-icreERPositive murine model of familial CCM disease, without induction of Pik3ca GOF. Lesion burden, attrition, and acute and chronic hemorrhaging were compared between placebo and rapamycin-treated mice. Plasma miRNome was compared to identify potential biomarkers of rapamycin response. Outlier, exceptionally large CCM lesions (> 2 SD above the mean lesion burden) were exclusively observed in the placebo group. Rapamycin, across all dosages, may have prevented the emergence of large outlier lesions. Yet rapamycin also appeared to exacerbate mean lesion burden of surviving mice when outliers were excluded, increased attrition, and did not alter hemorrhage. miR-30c-2-3p, decreased in rapamycin-treated mouse plasma, has gene targets in PI3K/AKT and mTOR signaling. Progression of outlier lesions in a familial CCM model may have been halted by rapamycin treatment, at the potential expense of increased mean lesion burden and increased attrition. If confirmed, this can have implications for potential rapamycin treatment of familial CCM disease, where lesion development may not be driven by PIK3CA GOF. Further studies are necessary to determine specific pathways that mediate potential beneficial and detrimental effects of rapamycin treatment, and whether somatic PIK3CA mutations drive particularly aggressive lesions.
Collapse
Affiliation(s)
- Roberto J Alcazar-Felix
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Robert Shenkar
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Christian R Benavides
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Akash Bindal
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Abhinav Srinath
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Ying Li
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Serena Kinkade
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Tatiana Terranova
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Evon DeBose-Scarlett
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Rhonda Lightle
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Dorothy DeBiasse
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Hanadi Almazroue
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Diana Vera Cruz
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Sharbel Romanos
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Aditya Jhaveri
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Janne Koskimäki
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Stephanie Hage
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Carolyn Bennett
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Romuald Girard
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, 27710, USA
| | - Issam A Awad
- Department of Neurological Surgery, Pritzker School of Medicine and Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
4
|
Frias-Anaya E, Gallego-Gutierrez H, Gongol B, Weinsheimer S, Lai CC, Orecchioni M, Sriram A, Bui CM, Nelsen B, Hale P, Pham A, Shenkar R, DeBiasse D, Lightle R, Girard R, Li Y, Srinath A, Daneman R, Nudleman E, Sun H, Guma M, Dubrac A, Mesarwi OA, Ley K, Kim H, Awad IA, Ginsberg MH, Lopez-Ramirez MA. Mild Hypoxia Accelerates Cerebral Cavernous Malformation Disease Through CX3CR1-CX3CL1 Signaling. Arterioscler Thromb Vasc Biol 2024; 44:1246-1264. [PMID: 38660801 PMCID: PMC11111348 DOI: 10.1161/atvbaha.123.320367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/05/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Heterogeneity in the severity of cerebral cavernous malformations (CCMs) disease, including brain bleedings and thrombosis that cause neurological disabilities in patients, suggests that environmental, genetic, or biological factors act as disease modifiers. Still, the underlying mechanisms are not entirely understood. Here, we report that mild hypoxia accelerates CCM disease by promoting angiogenesis, neuroinflammation, and vascular thrombosis in the brains of CCM mouse models. METHODS We used genetic studies, RNA sequencing, spatial transcriptome, micro-computed tomography, fluorescence-activated cell sorting, multiplex immunofluorescence, coculture studies, and imaging techniques to reveal that sustained mild hypoxia via the CX3CR1-CX3CL1 (CX3C motif chemokine receptor 1/chemokine [CX3C motif] ligand 1) signaling pathway influences cell-specific neuroinflammatory interactions, contributing to heterogeneity in CCM severity. RESULTS Histological and expression profiles of CCM neurovascular lesions (Slco1c1-iCreERT2;Pdcd10fl/fl; Pdcd10BECKO) in male and female mice found that sustained mild hypoxia (12% O2, 7 days) accelerates CCM disease. Our findings indicate that a small reduction in oxygen levels can significantly increase angiogenesis, neuroinflammation, and thrombosis in CCM disease by enhancing the interactions between endothelium, astrocytes, and immune cells. Our study indicates that the interactions between CX3CR1 and CX3CL1 are crucial in the maturation of CCM lesions and propensity to CCM immunothrombosis. In particular, this pathway regulates the recruitment and activation of microglia and other immune cells in CCM lesions, which leads to lesion growth and thrombosis. We found that human CX3CR1 variants are linked to lower lesion burden in familial CCMs, proving it is a genetic modifier in human disease and a potential marker for aggressiveness. Moreover, monoclonal blocking antibody against CX3CL1 or reducing 1 copy of the Cx3cr1 gene significantly reduces hypoxia-induced CCM immunothrombosis. CONCLUSIONS Our study reveals that interactions between CX3CR1 and CX3CL1 can modify CCM neuropathology when lesions are accelerated by environmental hypoxia. Moreover, a hypoxic environment or hypoxia signaling caused by CCM disease influences the balance between neuroinflammation and neuroprotection mediated by CX3CR1-CX3CL1 signaling. These results establish CX3CR1 as a genetic marker for patient stratification and a potential predictor of CCM aggressiveness.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Chemokine CX3CL1/metabolism
- Chemokine CX3CL1/genetics
- CX3C Chemokine Receptor 1/genetics
- CX3C Chemokine Receptor 1/metabolism
- Disease Models, Animal
- Hemangioma, Cavernous, Central Nervous System/genetics
- Hemangioma, Cavernous, Central Nervous System/metabolism
- Hemangioma, Cavernous, Central Nervous System/pathology
- Hypoxia/metabolism
- Hypoxia/complications
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic/metabolism
- Neuroinflammatory Diseases/metabolism
- Neuroinflammatory Diseases/pathology
- Neuroinflammatory Diseases/genetics
- Signal Transduction
Collapse
Affiliation(s)
- Eduardo Frias-Anaya
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Helios Gallego-Gutierrez
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Brendan Gongol
- Department of Health Sciences, Victor Valley College, Victorville, CA (B.G.)
- Institute for Integrative Genome Biology, 1207F Genomics Building, University of California, Riverside (B.G.)
| | - Shantel Weinsheimer
- Department of Anesthesia and Perioperative Care, Institute for Human Genetics, University of California, San Francisco (S.W., A.S., H.K.)
| | - Catherine Chinhchu Lai
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Marco Orecchioni
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (M.O., K.L.)
| | - Aditya Sriram
- Department of Anesthesia and Perioperative Care, Institute for Human Genetics, University of California, San Francisco (S.W., A.S., H.K.)
| | - Cassandra M Bui
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Bliss Nelsen
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Preston Hale
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Angela Pham
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Dorothy DeBiasse
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Ying Li
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Richard Daneman
- Department of Pharmacology (R.D., M.A.L.-R.), University of California San Diego, La Jolla
| | - Eric Nudleman
- Department of Ophthalmology (E.N.), University of California San Diego, La Jolla
| | - Hao Sun
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Monica Guma
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montréal, Quebec, Canada. Département de Pathologie et Biologie Cellulaire, Université de Montréal, Quebec, Canada (A.D.)
| | - Omar A Mesarwi
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Immunology, CA (M.O., K.L.)
| | - Helen Kim
- Department of Anesthesia and Perioperative Care, Institute for Human Genetics, University of California, San Francisco (S.W., A.S., H.K.)
| | - Issam A Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, IL (R.S., D.D., R.L., R.G., Y.L., A.S., I.A.A.)
| | - Mark H Ginsberg
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
| | - Miguel Alejandro Lopez-Ramirez
- Department of Medicine (E.F.-A., H.G.-G., C.C.L., C.M.B., B.N., P.H., A.P., H.S., M.G., O.A.M., M.H.G., M.A.L.-R.), University of California San Diego, La Jolla
- Department of Pharmacology (R.D., M.A.L.-R.), University of California San Diego, La Jolla
| |
Collapse
|
5
|
Xu C, Li S, Cai Y, Lu J, Teng Y, Yang X, Wang J. Generation of Slco1a4-CreERT2-tdTomato Knock-in Mice for Specific Cerebrovascular Endothelial Cell Targeting. Int J Mol Sci 2024; 25:4666. [PMID: 38731886 PMCID: PMC11083393 DOI: 10.3390/ijms25094666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
The cerebrovascular endothelial cells with distinct characteristics line cerebrovascular blood vessels and are the fundamental structure of the blood-brain barrier, which is important for the development and homeostatic maintenance of the central nervous system. Cre-LoxP system-based spatial gene manipulation in mice is critical for investigating the physiological functions of key factors or signaling pathways in cerebrovascular endothelial cells. However, there is a lack of Cre recombinase mouse lines that specifically target cerebrovascular endothelial cells. Here, using a publicly available single-cell RNAseq database, we screened the solute carrier organic anion transporter family member 1a4 (Slco1a4) as a candidate marker of cerebrovascular endothelial cells. Then, we generated an inducible Cre mouse line in which a CreERT2-T2A-tdTomato cassette was placed after the initiation codon ATG of the Slco1a4 locus. We found that tdTomato, which can indicate the endogenous Slco1a4 expression, was expressed in almost all cerebrovascular endothelial cells but not in any other non-endothelial cell types in the brain, including neurons, astrocytes, oligodendrocytes, pericytes, smooth muscle cells, and microglial cells, as well as in other organs. Consistently, when crossing the ROSA26LSL-EYFP Cre reporter mouse, EYFP also specifically labeled almost all cerebrovascular endothelial cells upon tamoxifen induction. Overall, we generated a new inducible Cre line that specifically targets cerebrovascular endothelial cells.
Collapse
Affiliation(s)
- Chengfang Xu
- Beijing Institute of Lifeomics, Beijing 102206, China; (C.X.); (Y.C.); (Y.T.)
| | - Shounian Li
- Beijing Institute of Lifeomics, Beijing 102206, China; (C.X.); (Y.C.); (Y.T.)
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Yunting Cai
- Beijing Institute of Lifeomics, Beijing 102206, China; (C.X.); (Y.C.); (Y.T.)
| | - Jinjin Lu
- Beijing Institute of Lifeomics, Beijing 102206, China; (C.X.); (Y.C.); (Y.T.)
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yan Teng
- Beijing Institute of Lifeomics, Beijing 102206, China; (C.X.); (Y.C.); (Y.T.)
| | - Xiao Yang
- Beijing Institute of Lifeomics, Beijing 102206, China; (C.X.); (Y.C.); (Y.T.)
| | - Jun Wang
- Beijing Institute of Lifeomics, Beijing 102206, China; (C.X.); (Y.C.); (Y.T.)
| |
Collapse
|
6
|
Li Y, Girard R, Srinath A, Cruz DV, Ciszewski C, Chen C, Lightle R, Romanos S, Sone JY, Moore T, DeBiasse D, Stadnik A, Lee JJ, Shenkar R, Koskimäki J, Lopez-Ramirez MA, Marchuk DA, Ginsberg MH, Kahn ML, Shi C, Awad IA. Transcriptomic signatures of individual cell types in cerebral cavernous malformation. Cell Commun Signal 2024; 22:23. [PMID: 38195510 PMCID: PMC10775676 DOI: 10.1186/s12964-023-01301-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/30/2023] [Indexed: 01/11/2024] Open
Abstract
Cerebral cavernous malformation (CCM) is a hemorrhagic neurovascular disease with no currently available therapeutics. Prior evidence suggests that different cell types may play a role in CCM pathogenesis. The contribution of each cell type to the dysfunctional cellular crosstalk remains unclear. Herein, RNA-seq was performed on fluorescence-activated cell sorted endothelial cells (ECs), pericytes, and neuroglia from CCM lesions and non-lesional brain tissue controls. Differentially Expressed Gene (DEG), pathway and Ligand-Receptor (LR) analyses were performed to characterize the dysfunctional genes of respective cell types within CCMs. Common DEGs among all three cell types were related to inflammation and endothelial-to-mesenchymal transition (EndMT). DEG and pathway analyses supported a role of lesional ECs in dysregulated angiogenesis and increased permeability. VEGFA was particularly upregulated in pericytes. Further pathway and LR analyses identified vascular endothelial growth factor A/ vascular endothelial growth factor receptor 2 signaling in lesional ECs and pericytes that would result in increased angiogenesis. Moreover, lesional pericytes and neuroglia predominantly showed DEGs and pathways mediating the immune response. Further analyses of cell specific gene alterations in CCM endorsed potential contribution to EndMT, coagulation, and a hypoxic microenvironment. Taken together, these findings motivate mechanistic hypotheses regarding non-endothelial contributions to lesion pathobiology and may lead to novel therapeutic targets. Video Abstract.
Collapse
Affiliation(s)
- Ying Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Romuald Girard
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Abhinav Srinath
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Diana Vera Cruz
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Cezary Ciszewski
- Human Disease and Immune Discovery Core, The University of Chicago, Chicago, IL, USA
| | - Chang Chen
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Rhonda Lightle
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Sharbel Romanos
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Je Yeong Sone
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Thomas Moore
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Dorothy DeBiasse
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Agnieszka Stadnik
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Justine J Lee
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Robert Shenkar
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA
| | - Janne Koskimäki
- Department of Neurosurgery, Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
- Department of Neurosurgery, Oulu University Hospital, Neurocenter, Oulu, Finland
| | - Miguel A Lopez-Ramirez
- Department of Medicine, University of California, La Jolla, San Diego, CA, USA
- Department of Pharmacology, University of California, La Jolla, San Diego, CA, USA
| | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Mark H Ginsberg
- Department of Medicine, University of California, La Jolla, San Diego, CA, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Changbin Shi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Issam A Awad
- Department of Neurological Surgery, Neurovascular Surgery Program, The University of Chicago, Chicago, IL, USA.
- Department of Neurological Surgery, University of Chicago Medicine, 5841 S Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA.
| |
Collapse
|
7
|
Phillips CM, Johnson AM, Stamatovic SM, Keep RF, Andjelkovic AV. 20 kDa isoform of connexin-43 augments spatial reorganization of the brain endothelial junctional complex and lesion leakage in cerebral cavernous malformation type-3. Neurobiol Dis 2023; 186:106277. [PMID: 37652184 DOI: 10.1016/j.nbd.2023.106277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/14/2023] [Accepted: 08/29/2023] [Indexed: 09/02/2023] Open
Abstract
Cerebral cavernous malformation type-3 (CCM3) is a type of brain vascular malformation caused by mutations in programmed cell death protein-10 (PDCD10). It is characterized by early life occurrence of hemorrhagic stroke and profound blood-brain barrier defects. The pathogenic mechanisms responsible for microvascular hyperpermeability and lesion progression in CCM3 are still largely unknown. The current study examined brain endothelial barrier structural defects formed in the absence of CCM3 in vivo and in vitro that may lead to CCM3 lesion leakage. We found significant upregulation of a 20 kDa isoform of connexin 43 (GJA1-20 k) in brain endothelial cells (BEC) in both non-leaky and leaky lesions, as well as in an in vitro CCM3 knockdown model (CCM3KD-BEC). Morphological, biochemical, FRET, and FRAP analyses of CCM3KD-BEC found GJA1-20 k regulates full-length GJA1 biogenesis, prompting uncontrolled gap junction growth. Furthermore, by binding to a tight junction scaffolding protein, ZO-1, GJA1-20 k interferes with Cx43/ZO-1 interactions and gap junction/tight junction crosstalk, promoting ZO-1 dissociation from tight junction complexes and diminishing claudin-5/ZO-1 interaction. As a consequence, the tight junction complex is destabilized, allowing "replacement" of tight junctions with gap junctions leading to increased brain endothelial barrier permeability. Modifying cellular levels of GJA1-20 k rescued brain endothelial barrier integrity re-establishing the spatial organization of gap and tight junctional complexes. This study highlights generation of potential defects at the CCM3-affected brain endothelial barrier which may underlie prolonged vascular leakiness.
Collapse
Affiliation(s)
- Chelsea M Phillips
- Neuroscience Graduate program, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA; Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Anuska V Andjelkovic
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Dudley AC, Griffioen AW. Pathological angiogenesis: mechanisms and therapeutic strategies. Angiogenesis 2023; 26:313-347. [PMID: 37060495 PMCID: PMC10105163 DOI: 10.1007/s10456-023-09876-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/26/2023] [Indexed: 04/16/2023]
Abstract
In multicellular organisms, angiogenesis, the formation of new blood vessels from pre-existing ones, is an essential process for growth and development. Different mechanisms such as vasculogenesis, sprouting, intussusceptive, and coalescent angiogenesis, as well as vessel co-option, vasculogenic mimicry and lymphangiogenesis, underlie the formation of new vasculature. In many pathological conditions, such as cancer, atherosclerosis, arthritis, psoriasis, endometriosis, obesity and SARS-CoV-2(COVID-19), developmental angiogenic processes are recapitulated, but are often done so without the normal feedback mechanisms that regulate the ordinary spatial and temporal patterns of blood vessel formation. Thus, pathological angiogenesis presents new challenges yet new opportunities for the design of vascular-directed therapies. Here, we provide an overview of recent insights into blood vessel development and highlight novel therapeutic strategies that promote or inhibit the process of angiogenesis to stabilize, reverse, or even halt disease progression. In our review, we will also explore several additional aspects (the angiogenic switch, hypoxia, angiocrine signals, endothelial plasticity, vessel normalization, and endothelial cell anergy) that operate in parallel to canonical angiogenesis mechanisms and speculate how these processes may also be targeted with anti-angiogenic or vascular-directed therapies.
Collapse
Affiliation(s)
- Andrew C Dudley
- Department of Microbiology, Immunology and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA.
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Romanos SG, Srinath A, Li Y, Xie B, Chen C, Li Y, Moore T, Bi D, Sone JY, Lightle R, Hobson N, Zhang D, Koskimäki J, Shen L, McCurdy S, Lai CC, Stadnik A, Piedad K, Carrión-Penagos J, Shkoukani A, Snellings D, Shenkar R, Sulakhe D, Ji Y, Lopez-Ramirez MA, Kahn ML, Marchuk DA, Ginsberg MH, Girard R, Awad IA. Circulating Plasma miRNA Homologs in Mice and Humans Reflect Familial Cerebral Cavernous Malformation Disease. Transl Stroke Res 2023; 14:513-529. [PMID: 35715588 PMCID: PMC9758276 DOI: 10.1007/s12975-022-01050-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 01/16/2023]
Abstract
Patients with familial cerebral cavernous malformation (CCM) inherit germline loss of function mutations and are susceptible to progressive development of brain lesions and neurological sequelae during their lifetime. To date, no homologous circulating molecules have been identified that can reflect the presence of germ line pathogenetic CCM mutations, either in animal models or patients. We hypothesize that homologous differentially expressed (DE) plasma miRNAs can reflect the CCM germline mutation in preclinical murine models and patients. Herein, homologous DE plasma miRNAs with mechanistic putative gene targets within the transcriptome of preclinical and human CCM lesions were identified. Several of these gene targets were additionally found to be associated with CCM-enriched pathways identified using the Kyoto Encyclopedia of Genes and Genomes. DE miRNAs were also identified in familial-CCM patients who developed new brain lesions within the year following blood sample collection. The miRNome results were then validated in an independent cohort of human subjects with real-time-qPCR quantification, a technique facilitating plasma assays. Finally, a Bayesian-informed machine learning approach showed that a combination of plasma levels of miRNAs and circulating proteins improves the association with familial-CCM disease in human subjects to 95% accuracy. These findings act as an important proof of concept for the future development of translatable circulating biomarkers to be tested in preclinical studies and human trials aimed at monitoring and restoring gene function in CCM and other diseases.
Collapse
Affiliation(s)
- Sharbel G Romanos
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Abhinav Srinath
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Ying Li
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Bingqing Xie
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Chang Chen
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Bioinformatics Core, Biological Sciences Division, University of Chicago, Chicago, IL, USA
| | - Yan Li
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Bioinformatics Core, Biological Sciences Division, University of Chicago, Chicago, IL, USA
| | - Thomas Moore
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Dehua Bi
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Je Yeong Sone
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Rhonda Lightle
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Nick Hobson
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Dongdong Zhang
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Janne Koskimäki
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Le Shen
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Sara McCurdy
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Catherine Chinhchu Lai
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Agnieszka Stadnik
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Kristina Piedad
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Julián Carrión-Penagos
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Abdallah Shkoukani
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Daniel Snellings
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Robert Shenkar
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Dinanath Sulakhe
- Bioinformatics Core, Biological Sciences Division, University of Chicago, Chicago, IL, USA
| | - Yuan Ji
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Miguel A Lopez-Ramirez
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas A Marchuk
- Molecular Genetics and Microbiology Department, Duke University Medical Center, Durham, NC, USA
| | - Mark H Ginsberg
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Romuald Girard
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA
| | - Issam A Awad
- Department of Neurological Surgery, Neurovascular Surgery Program, University of Chicago Medicine and Biological Sciences, 5841 S. Maryland, MC3026/Neurosurgery J341, Chicago, IL, 60637, USA.
| |
Collapse
|
10
|
Fisher DG, Sharifi KA, Ulutas EZ, Kumar JS, Kalani MYS, Miller GW, Price RJ, Tvrdik P. Magnetic Resonance Imaging of Mouse Cerebral Cavernomas Reveal Differential Lesion Progression and Variable Permeability to Gadolinium. Arterioscler Thromb Vasc Biol 2023; 43:958-970. [PMID: 37078284 PMCID: PMC10257814 DOI: 10.1161/atvbaha.122.318938] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/04/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND Cerebral cavernous malformations, also known as cavernous angiomas, are blood vessel abnormalities comprised of clusters of grossly enlarged and hemorrhage-prone capillaries. The prevalence in the general population, including asymptomatic cases, is estimated to be 0.5%. Some patients develop severe symptoms, including seizures and focal neurological deficits, whereas others remain asymptomatic. The causes of this remarkable presentation heterogeneity within a primarily monogenic disease remain poorly understood. METHODS We established a chronic mouse model of cerebral cavernous malformations, induced by postnatal ablation of Krit1 with Pdgfb-CreERT2, and examined lesion progression in these mice with T2-weighted 7T magnetic resonance imaging (MRI). We also established a modified protocol for dynamic contrast-enhanced MRI and produced quantitative maps of gadolinium tracer gadobenate dimeglumine. After terminal imaging, brain slices were stained with antibodies against microglia, astrocytes, and endothelial cells. RESULTS These mice develop cerebral cavernous malformations lesions gradually over 4 to 5 months of age throughout the brain. Precise volumetric analysis of individual lesions revealed nonmonotonous behavior, with some lesions temporarily growing smaller. However, the cumulative lesional volume invariably increased over time and after about 2 months followed a power trend. Using dynamic contrast-enhanced MRI, we produced quantitative maps of gadolinium in the lesions, indicating a high degree of heterogeneity in lesional permeability. MRI properties of the lesions were correlated with cellular markers for endothelial cells, astrocytes, and microglia. Multivariate comparisons of MRI properties of the lesions with cellular markers for endothelial and glial cells revealed that increased cell density surrounding lesions correlates with stability, whereas denser vasculature within and surrounding the lesions may correlate with high permeability. CONCLUSIONS Our results lay a foundation for better understanding individual lesion properties and provide a comprehensive preclinical platform for testing new drug and gene therapies for controlling cerebral cavernous malformations.
Collapse
Affiliation(s)
- Delaney G. Fisher
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Khadijeh A. Sharifi
- Department of Neuroscience, University of Virginia, Charlottesville, VA
- Department of Neurosurgery, University of Virginia Health System, Charlottesville, VA
| | - E. Zeynep Ulutas
- Department of Neuroscience, Georgia Institute of Technology, Atlanta, GA
| | - Jeyan S. Kumar
- 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
| | - Petr Tvrdik
- Department of Neuroscience, University of Virginia, Charlottesville, VA
- Department of Neurosurgery, University of Virginia Health System, Charlottesville, VA
| |
Collapse
|
11
|
Grdseloff N, Boulday G, Rödel CJ, Otten C, Vannier DR, Cardoso C, Faurobert E, Dogra D, Tournier-Lasserve E, Abdelilah-Seyfried S. Impaired retinoic acid signaling in cerebral cavernous malformations. Sci Rep 2023; 13:5572. [PMID: 37019926 PMCID: PMC10076292 DOI: 10.1038/s41598-023-31905-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/20/2023] [Indexed: 04/07/2023] Open
Abstract
The capillary-venous pathology cerebral cavernous malformation (CCM) is caused by loss of CCM1/Krev interaction trapped protein 1 (KRIT1), CCM2/MGC4607, or CCM3/PDCD10 in some endothelial cells. Mutations of CCM genes within the brain vasculature can lead to recurrent cerebral hemorrhages. Pharmacological treatment options are urgently needed when lesions are located in deeply-seated and in-operable regions of the central nervous system. Previous pharmacological suppression screens in disease models of CCM led to the discovery that treatment with retinoic acid improved CCM phenotypes. This finding raised a need to investigate the involvement of retinoic acid in CCM and test whether it has a curative effect in preclinical mouse models. Here, we show that components of the retinoic acid synthesis and degradation pathway are transcriptionally misregulated across disease models of CCM. We complemented this analysis by pharmacologically modifying retinoic acid levels in zebrafish and human endothelial cell models of CCM, and in acute and chronic mouse models of CCM. Our pharmacological intervention studies in CCM2-depleted human umbilical vein endothelial cells (HUVECs) and krit1 mutant zebrafish showed positive effects when retinoic acid levels were increased. However, therapeutic approaches to prevent the development of vascular lesions in adult chronic murine models of CCM were drug regiment-sensitive, possibly due to adverse developmental effects of this hormone. A treatment with high doses of retinoic acid even worsened CCM lesions in an adult chronic murine model of CCM. This study provides evidence that retinoic acid signaling is impaired in the CCM pathophysiology and suggests that modification of retinoic acid levels can alleviate CCM phenotypes.
Collapse
Affiliation(s)
- Nastasja Grdseloff
- Institute of Biochemistry and Biology, Department of Zoophysiology, University of Potsdam, Karl-Liebknecht-Strasse 24-25, 14476, Potsdam, Germany
| | - Gwenola Boulday
- InsermNeuroDiderot, Université Paris Cité, 75019, Paris, France
| | - Claudia J Rödel
- Institute of Biochemistry and Biology, Department of Zoophysiology, University of Potsdam, Karl-Liebknecht-Strasse 24-25, 14476, Potsdam, Germany
| | - Cécile Otten
- Institute of Biochemistry and Biology, Department of Zoophysiology, University of Potsdam, Karl-Liebknecht-Strasse 24-25, 14476, Potsdam, Germany
- Institut Ruđer Bošković, Bijenička cesta 54, 10000, Zagreb, Croatia
| | - Daphné Raphaelle Vannier
- Institute for Advanced Biosciences, INSERM 1209 CNRS, University Grenoble Alpes, 5309, Grenoble, France
| | - Cécile Cardoso
- InsermNeuroDiderot, Université Paris Cité, 75019, Paris, France
| | - Eva Faurobert
- Institute for Advanced Biosciences, INSERM 1209 CNRS, University Grenoble Alpes, 5309, Grenoble, France
| | - Deepika Dogra
- Institute of Biochemistry and Biology, Department of Zoophysiology, University of Potsdam, Karl-Liebknecht-Strasse 24-25, 14476, Potsdam, Germany
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Elisabeth Tournier-Lasserve
- InsermNeuroDiderot, Université Paris Cité, 75019, Paris, France
- Service de Génétique Neurovasculaire, AP-HP, Hôpital Saint-Louis, 75010, Paris, France
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Department of Zoophysiology, University of Potsdam, Karl-Liebknecht-Strasse 24-25, 14476, Potsdam, Germany.
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
12
|
Yang X, Dai Z, Gao C, Yin Y, Shi C, Liu R, Zhuge Q, Huang Y, Zhou B, Han Z, Zheng X. Cerebral cavernous malformation development in chronic mouse models driven by dual recombinases induced gene deletion in brain endothelial cells. J Cereb Blood Flow Metab 2022; 42:2230-2244. [PMID: 35686705 PMCID: PMC9669998 DOI: 10.1177/0271678x221105995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cerebral cavernous malformation (CCM) is a brain vascular disease which can cause stroke, cerebral hemorrhage and neurological deficits in affected individuals. Loss-of-function mutations in three genes (CCM1, CCM2 and CCM3) cause CCM disease. Multiple mouse models for CCM disease have been developed although each of them are associated with various limitations. Here, we employed the Dre-Cre dual recombinase system to specifically delete Ccm genes in brain endothelial cells. In this new series of CCM mouse models, robust CCM lesions now develop in the cerebrum. The survival curve and lesion burden analysis revealed that Ccm2 deletion causes modest CCM lesions with a median life expectance of ∼10 months and Ccm3 gene deletion leads to the most severe CCM lesions with median life expectance of ∼2 months. The extended lifespan of these mutant mice enables their utility in behavioral analyses of neurologic deficits in adult mice, and allow the development of methods to quantify lesion burden in mice over time and also permit longitudinal drug testing in live animals.
Collapse
Affiliation(s)
- Xi Yang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zifeng Dai
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Caixia Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yongqiang Yin
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Changbin Shi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Renjing Liu
- Vascular Epigenetics Laboratory, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue Huang
- China National Clinical Research Centre for Neurological Disorders, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Zhiming Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
13
|
McCurdy S, Lin J, Shenkar R, Moore T, Lightle R, Faurobert E, Lopez-Ramirez MA, Awad I, Ginsberg MH. β1 integrin monoclonal antibody treatment ameliorates cerebral cavernous malformations. FASEB J 2022; 36:e22629. [PMID: 36349990 PMCID: PMC9674378 DOI: 10.1096/fj.202200907rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/24/2022] [Accepted: 10/15/2022] [Indexed: 11/10/2022]
Abstract
β1 integrins are important in blood vessel formation and function, finely tuning the adhesion of endothelial cells to each other and to the extracellular matrix. The role of integrins in the vascular disease, cerebral cavernous malformation (CCM) has yet to be explored in vivo. Endothelial loss of the gene KRIT1 leads to brain microvascular defects, resulting in debilitating and often fatal consequences. We tested administration of a monoclonal antibody that enforces the active β1 integrin conformation, (clone 9EG7), on a murine neonatal CCM mouse model, Krit1flox/flox ;Pdgfb-iCreERT2 (Krit1ECKO ), and on KRIT1-silenced human umbilical vein endothelial cells (HUVECs). In addition, endothelial deletion of the master regulator of integrin activation, Talin 1 (Tln1), in Krit1ECKO mice was performed to assess the effect of completely blocking endothelial integrin activation on CCM. Treatment with 9EG7 reduced lesion burden in the Krit1ECKO model and was accompanied by a strong reduction in the phosphorylation of the ROCK substrate, myosin light chain (pMLC), in both retina and brain endothelial cells. Treatment of KRIT1-silenced HUVECs with 9EG7 in vitro stabilized cell-cell junctions. Overnight treatment of HUVECs with 9EG7 resulted in significantly reduced total surface expression of β1 integrin, which was associated with reduced pMLC levels, supporting our in vivo findings. Genetic blockade of integrin activation by Tln1ECKO enhanced bleeding and did not reduce CCM lesion burden in Krit1ECKO mice. In sum, targeting β1 integrin with an activated-specific antibody reduces acute murine CCM lesion development, which we found to be associated with suppression of endothelial ROCK activity.
Collapse
Affiliation(s)
- Sara McCurdy
- Department of Medicine, University of California San Diego, LA Jolla CA
| | - Jenny Lin
- Department of Medicine, University of California San Diego, LA Jolla CA
| | - Robert Shenkar
- Department of Neurological Surgery, University of Chicago, Chicago IL
| | - Thomas Moore
- Department of Neurological Surgery, University of Chicago, Chicago IL
| | - Rhonda Lightle
- Department of Neurological Surgery, University of Chicago, Chicago IL
| | - Eva Faurobert
- Univ. Grenoble Alpes, CNRS 5309, Inserm 1209, Institute for Advanced Biosciences, Grenoble, France
| | | | - Issam Awad
- Department of Neurological Surgery, University of Chicago, Chicago IL
| | - Mark H. Ginsberg
- Department of Medicine, University of California San Diego, LA Jolla CA
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Abstract
While inhibiting pathological angiogenesis has been long associated with the field of oncology, recent advances in angiogenesis research have impacted the progress of disease treatment for additional non-malignant diseases or chronic conditions in the fields of ophthalmology, cardiology, and gynecology. Moreover, stimulators of angiogenesis find application in ischemic diseases, while inhibitors of angiogenesis are being used to limit blood vessel formation, but in judicious ways that modify or "reprogram" the vasculature as a reinforcement for immunotherapy. We have noticed an increasing impact, as evidenced by increases in the total number of citations, in the literature surrounding the angiogenesis field suggesting that targeting angiogenesis per se is well established as a tractable approach for therapy in diverse conditions.
Collapse
Affiliation(s)
- Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, & The Emily Couric Cancer Center, Charlottesville, VA, 22908, USA
| |
Collapse
|
17
|
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
|
18
|
Zhang J, Abou-Fadel J, Renteria M, Belkin O, Chen B, Zhu Y, Dammann P, Rigamonti D. Cerebral cavernous malformations do not fall in the spectrum of PIK3CA-related overgrowth. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2022-328901. [PMID: 35477890 DOI: 10.1136/jnnp-2022-328901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/23/2022] [Indexed: 11/04/2022]
Abstract
Somatic gain-of-function (GOF) mutations in phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), the catalytic subunit of phosphoinositide 3-kinase (PI3K), have been recently discovered in cerebral cavernous malformations (CCMs), raising the possibility that the activation of PI3K pathways is a possible universal regulator of vascular morphogenesis. However, there have been contradicting data presented among various groups and studies. To enhance the current understanding of vascular anomalies, it is essential to explore this possible relationship between altered PI3K signalling pathways and its influence on the pathogenesis of CCMs. GOF PIK3CA-mutants have been linked to overgrowth syndromes, allowing this group of disorders, resulting from somatic activating mutations in PIK3CA, to be collectively named as PIK3CA-related overgrowth spectrum disorders. This paper reviews and attempts to conceptualise the relationships and differences among clinical presentations, genotypic and phenotypic correlations and possible coexistence of PIK3CA and CCM mutations/phenotypes in CCM lesions. Finally, we present a model reflecting our hypothetical understanding of CCM pathogenesis based on a systematic review and conceptualisation of data obtained from other studies.
Collapse
Affiliation(s)
- Jun Zhang
- Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Johnathan Abou-Fadel
- Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Mellisa Renteria
- Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Ofek Belkin
- Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Bixia Chen
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany
| | - Yuan Zhu
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany
| | - Philipp Dammann
- Department of Neurosurgery, University of Duisburg-Essen, Essen, Germany
| | | |
Collapse
|
19
|
Magid-Bernstein J, Girard R, Polster S, Srinath A, Romanos S, Awad IA, Sansing LH. Cerebral Hemorrhage: Pathophysiology, Treatment, and Future Directions. Circ Res 2022; 130:1204-1229. [PMID: 35420918 PMCID: PMC10032582 DOI: 10.1161/circresaha.121.319949] [Citation(s) in RCA: 168] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke with high morbidity and mortality. This review article focuses on the epidemiology, cause, mechanisms of injury, current treatment strategies, and future research directions of ICH. Incidence of hemorrhagic stroke has increased worldwide over the past 40 years, with shifts in the cause over time as hypertension management has improved and anticoagulant use has increased. Preclinical and clinical trials have elucidated the underlying ICH cause and mechanisms of injury from ICH including the complex interaction between edema, inflammation, iron-induced injury, and oxidative stress. Several trials have investigated optimal medical and surgical management of ICH without clear improvement in survival and functional outcomes. Ongoing research into novel approaches for ICH management provide hope for reducing the devastating effect of this disease in the future. Areas of promise in ICH therapy include prognostic biomarkers and primary prevention based on disease pathobiology, ultra-early hemostatic therapy, minimally invasive surgery, and perihematomal protection against inflammatory brain injury.
Collapse
Affiliation(s)
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sean Polster
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Issam A. Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
20
|
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
|
21
|
TMEM60 Promotes the Proliferation and Migration and Inhibits the Apoptosis of Glioma through Modulating AKT Signaling. JOURNAL OF ONCOLOGY 2022; 2022:9913700. [PMID: 35027926 PMCID: PMC8749377 DOI: 10.1155/2022/9913700] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/16/2021] [Accepted: 10/26/2021] [Indexed: 02/03/2023]
Abstract
Glioma is a highly fatal malignancy with aggressive proliferation, migration, and invasion metastasis due to aberrant genetic regulation. This work aimed to determine the function of transmembrane protein 60 (TMEM60) during glioma development. The level of TMEM60 in glioma tissues and normal tissues and its correlation with glioma prognosis were checked in The Cancer Genome Atlas (TCGA) database. The levels of TMEM60 in glioma cell lines and normal astrocytes were determined by quantitative real-time PCR and western blotting assay. TMEM60 knockdown and overexpression were conducted, followed by detection of cell viability, migration, invasion, and apoptosis. CCK-8 and colony formation assay were adopted to detect cell viability proliferation. Transwell assay was performed to measure cell migration and invasion. Cell apoptosis was evaluated by flow cytometry. The alternation of key proteins in the PI3K/Akt signaling pathway was measured by western blotting. TMEM60 expression was significantly higher in glioma tissues than that in the healthy control and was correlated with poor overall survival of patients. The protein and mRNA levels of TMEM60 were both elevated in glioma cell lines in comparison with the normal cell lines. Elevated level of TMEM60 led to enhanced proliferation, migration, and invasion and suppressed cell apoptosis. TMEM60 promoted the activation of PI3K/Akt signaling. Our data suggested that TMEM60 plays an oncogenic role in glioma progression via activating the PI3K/Akt signaling pathway.
Collapse
|
22
|
FUNDC1 activates the mitochondrial unfolded protein response to preserve mitochondrial quality control in cardiac ischemia/reperfusion injury. Cell Signal 2022; 92:110249. [DOI: 10.1016/j.cellsig.2022.110249] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 12/28/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022]
|
23
|
Wang J, Wang X, Du W, Xue Z, Huang W, Guan Z, Wang H. BI-1 ameliorates myocardial injury by activating the mitochondrial unfolded protein response and FUNDC1-related mitophagy in cardiorenal syndrome type 3. Cell Signal 2021; 91:110218. [PMID: 34921980 DOI: 10.1016/j.cellsig.2021.110218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 01/25/2023]
Abstract
It has been suggested that mitochondrial dysfunction underlies the myocardial injury seen following cardiorenal syndrome type 3 (CRS-3). Both mitophagy and the mitochondrial unfolded protein response (UPRmt) are protective programs that preserve mitochondrial homeostasis. Here, we explored whether Bax inhibitor-1 (BI-1) overexpression attenuates CRS-3-related myocardial injury through activation of mitophagy and the UPRmt in cardiomyocytes. Following CRS-3 induction via renal ischemia-reperfusion injury, BI-1 transgenic (BI1TG) mice showed greater preservation of myocardial integrity and relaxation function and less cardiomyocyte apoptosis than wild-type (WT) mice. Moreover, BI-1 overexpression attenuated CRS-3-mediated myocardial inflammation, as indicated by decreased MCP-1 and IL-6 expression and normalized ATP production in cardiomyocytes. After CRS-3 induction, mitophagy was inhibited in cardiomyocytes from WT mice, as indicated by both decreased Fundc1 transcription and mt-Keima fluorescence, and modest activation of the UPRmt, denoted by a slight increase in Atf6 mRNA levels. By contrast, activation of mitophagy and marked UPRmt upregulation were observed in cardiac tissue from BI1TG mice. shRNA-mediated silencing of Fundc1 or Atf6 greatly impaired mitochondrial metabolism and survival in cultured cardiomyocytes overexpressing BI-1. Thus, upregulation of BI-1 expression aimed at activating mitophagy and the UPRmt may represent a useful therapeutic approach for the treatment of CRS-3.
Collapse
Affiliation(s)
- Jin Wang
- Department of Vascular Medicine, Peking University Shougang Hospital, Beijing 100144, China.
| | - Xiaohua Wang
- National Clinical Research Center for Geriatric Diseases, People's Liberation Army General Hospital, Beijing, China
| | - Wenjuan Du
- Laboratory of Radiation Injury Treatment, Medical Innovation Research Division, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhe Xue
- Department of Orthopedics, Peking University Shougang Hospital, Beijing 100144, China
| | - Wei Huang
- Department of Vascular Medicine, Peking University Shougang Hospital, Beijing 100144, China
| | - Zhenpeng Guan
- Department of Orthopedics, Peking University Shougang Hospital, Beijing 100144, China
| | - Hongyu Wang
- Department of Vascular Medicine, Peking University Shougang Hospital, Beijing 100144, China.
| |
Collapse
|
24
|
Zhang H, Qu X, Han L, Di X. Mst2 Overexpression Inhibits Thyroid Carcinoma Growth and Metastasis by Disrupting Mitochondrial Fitness and Endoplasmic Reticulum Homeostasis. JOURNAL OF ONCOLOGY 2021; 2021:1262291. [PMID: 34557228 PMCID: PMC8455210 DOI: 10.1155/2021/1262291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/27/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022]
Abstract
Although the incidence of thyroid carcinoma has increased over the past several decades, it has an excellent prognosis and overall 5-year survival, with a stable mortality rate, except in cases with advanced stages or rare malignant tumor types. Biomarkers have emerged as effective targets of molecular therapy against thyroid carcinoma due to their rapid and convenient detection; however, there has been little clinical application. Macrophage stimulating 2 (Mst2) is a proapoptotic protein with implications in carcinogenesis and metastasis. We found that Mst2 overexpression-induced endoplasmic reticulum (ER) stress in MDA-T32 thyroid carcinoma cells, accompanied by elevated caspase-12 activity, increased apoptotic rate, and reduced cell viability. In addition, Mst2 overexpression contributed to mitochondrial damage, as evidenced by increased mitochondrial oxidative stress and activated the mitochondrial apoptotic pathway. Inhibition of the JNK pathway abolished these effects. These results show Mst2 to be a novel tumor suppressor that induces mitochondrial dysfunction and ER stress via the JNK pathway. Thus, Mst2 could potentially serve as a biomarker for developing targeted therapy against thyroid carcinoma.
Collapse
Affiliation(s)
- Haichao Zhang
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Xin Qu
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Lu Han
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Xu Di
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| |
Collapse
|
25
|
Cheng D, Zheng J, Hu F, Lv W, Lu C. Abnormal Mitochondria-Endoplasmic Reticulum Communication Promotes Myocardial Infarction. Front Physiol 2021; 12:717187. [PMID: 34413791 PMCID: PMC8369510 DOI: 10.3389/fphys.2021.717187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/08/2021] [Indexed: 01/06/2023] Open
Abstract
Myocardial infarction is characterized by cardiomyocyte death, and can be exacerbated by mitochondrial damage and endoplasmic reticulum injury. In the present study, we investigated whether communication between mitochondria and the endoplasmic reticulum contributes to cardiomyocyte death after myocardial infarction. Our data demonstrated that hypoxia treatment (mimicking myocardial infarction) promoted cardiomyocyte death by inducing the c-Jun N-terminal kinase (JNK) pathway. The activation of JNK under hypoxic conditions was dependent on overproduction of mitochondrial reactive oxygen species (mtROS) in cardiomyocytes, and mitochondrial division was identified as the upstream inducer of mtROS overproduction. Silencing mitochondrial division activators, such as B cell receptor associated protein 31 (BAP31) and mitochondrial fission 1 (Fis1), repressed mitochondrial division, thereby inhibiting mtROS overproduction and preventing JNK-induced cardiomyocyte death under hypoxic conditions. These data revealed that a novel death-inducing mechanism involving the BAP31/Fis1/mtROS/JNK axis promotes hypoxia-induced cardiomyocyte damage. Considering that BAP31 is localized within the endoplasmic reticulum and Fis1 is localized in mitochondria, abnormal mitochondria-endoplasmic reticulum communication may be a useful therapeutic target after myocardial infarction.
Collapse
Affiliation(s)
- Degang Cheng
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Jia Zheng
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Fang Hu
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Wei Lv
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Chengzhi Lu
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
26
|
LATS2 Deletion Attenuates Myocardial Ischemia-Reperfusion Injury by Promoting Mitochondrial Biogenesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1058872. [PMID: 34457109 PMCID: PMC8390173 DOI: 10.1155/2021/1058872] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/25/2021] [Accepted: 08/07/2021] [Indexed: 02/03/2023]
Abstract
Reperfusion therapy is the most effective treatment for acute myocardial infarction, but it can damage cardiomyocytes through a mechanism known as myocardial ischemia/reperfusion injury (MIRI). In this study, we investigated whether the large tumor suppressor kinase 2 (LATS2) contributes to the development of myocardial MIRI by disrupting mitochondrial biogenesis. Our in vitro data demonstrate that cardiomyocyte viability was reduced and apoptosis was increased in response to hypoxia/reoxygenation (H/R) injury. However, suppression of LATS2 by shRNA sustained cardiomyocyte viability by maintaining mitochondrial function. Compared to H/R-treated control cardiomyocytes, cardiomyocytes transfected with LATS2 shRNA exhibited increased mitochondrial respiration, improved mitochondrial ATP generation, and more stable mitochondrial membrane potential. LATS2 suppression increased cardiomyocyte viability and mitochondrial biogenesis in a manner dependent on PGC1α, a key regulator of mitochondrial metabolism. These results identify LATS2 as a new inducer of mitochondrial damage and myocardial MIRI and suggest that approaches targeting LATS2 or mitochondrial biogenesis may be beneficial in the clinical management of cardiac MIRI.
Collapse
|
27
|
Zhang J, Zhang F, Wang Y. Mitofusin-2 Enhances Mitochondrial Contact With the Endoplasmic Reticulum and Promotes Diabetic Cardiomyopathy. Front Physiol 2021; 12:707634. [PMID: 34305656 PMCID: PMC8298037 DOI: 10.3389/fphys.2021.707634] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/02/2021] [Indexed: 12/21/2022] Open
Abstract
Diabetic cardiomyopathy has been associated with mitochondrial damage. Mitochondria–endoplasmic reticulum (ER) contact is an important determinant of mitochondrial function and ER homeostasis. We therefore investigated whether hyperglycemia can damage the mitochondria by increasing their contact with the ER in cardiomyocytes. We found that hyperglycemia induced mitochondria–ER contact in cardiomyocytes, as evidenced by the increased MMM1, MDM34, and BAP31 expressions. Interestingly, the silencing of Mfn2 reduced the cooperation between the mitochondria and the ER in cardiomyocytes. Mfn2 silencing improved cardiomyocyte viability and function under hyperglycemic conditions. Additionally, the silencing of Mfn2 markedly attenuated the release of calcium from the ER to the mitochondria, thereby preserving mitochondrial metabolism in cardiomyocytes under hyperglycemic conditions. Mfn2 silencing reduced mitochondrial reactive oxygen species production, which reduced mitochondria-dependent apoptosis in hyperglycemia-treated cardiomyocytes. Finally, Mfn2 silencing attenuated ER stress in cardiomyocytes subjected to high-glucose stress. These results demonstrate that Mfn2 promotes mitochondria–ER contact in hyperglycemia-treated cardiomyocytes. The silencing of Mfn2 sustained mitochondrial function, suppressed mitochondrial calcium overload, prevented mitochondrial apoptosis, and reduced ER stress, thereby enhancing cardiomyocyte survival under hyperglycemic conditions.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Cardiology, Tianjin First Central Hospital, Tianjing, China
| | - Feng Zhang
- Department of Cardiology, Tianjin First Central Hospital, Tianjing, China
| | - Yanou Wang
- Health Management Department, Tianjin First Central Hospital, Tianjing, China
| |
Collapse
|
28
|
Lopez-Ramirez MA, Lai CC, Soliman SI, Hale P, Pham A, Estrada EJ, McCurdy S, Girard R, Verma R, Moore T, Lightle R, Hobson N, Shenkar R, Poulsen O, Haddad GG, Daneman R, Gongol B, Sun H, Lagarrigue F, Awad IA, Ginsberg MH. Astrocytes propel neurovascular dysfunction during cerebral cavernous malformation lesion formation. J Clin Invest 2021; 131:139570. [PMID: 34043589 PMCID: PMC8245174 DOI: 10.1172/jci139570] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/24/2021] [Indexed: 12/13/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are common neurovascular lesions caused by loss-of-function mutations in 1 of 3 genes, including KRIT1 (CCM1), CCM2, and PDCD10 (CCM3), and generally regarded as an endothelial cell-autonomous disease. Here we reported that proliferative astrocytes played a critical role in CCM pathogenesis by serving as a major source of VEGF during CCM lesion formation. An increase in astrocyte VEGF synthesis is driven by endothelial nitric oxide (NO) generated as a consequence of KLF2- and KLF4-dependent elevation of eNOS in CCM endothelium. The increased brain endothelial production of NO stabilized HIF-1α in astrocytes, resulting in increased VEGF production and expression of a "hypoxic" program under normoxic conditions. We showed that the upregulation of cyclooxygenase-2 (COX-2), a direct HIF-1α target gene and a known component of the hypoxic program, contributed to the development of CCM lesions because the administration of a COX-2 inhibitor significantly prevented the progression of CCM lesions. Thus, non-cell-autonomous crosstalk between CCM endothelium and astrocytes propels vascular lesion development, and components of the hypoxic program represent potential therapeutic targets for CCMs.
Collapse
MESH Headings
- Animals
- Apoptosis Regulatory Proteins/deficiency
- Apoptosis Regulatory Proteins/genetics
- Astrocytes/pathology
- Astrocytes/physiology
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Disease Models, Animal
- Disease Progression
- Endothelial Cells/metabolism
- Hemangioma, Cavernous, Central Nervous System/etiology
- Hemangioma, Cavernous, Central Nervous System/pathology
- Hemangioma, Cavernous, Central Nervous System/physiopathology
- Human Umbilical Vein Endothelial Cells
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Mice
- Mice, Knockout
- Models, Neurological
- Mutation
- Nitric Oxide/biosynthesis
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Vascular Endothelial Growth Factor A/biosynthesis
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | | | - Thomas Moore
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Nicholas Hobson
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | | | - Gabriel G. Haddad
- Department of Pediatrics, and
- Department of Neuroscience, Division of Respiratory Medicine, University of California, San Diego, La Jolla, California, USA
- Rady Children’s Hospital, San Diego, California, USA
| | - Richard Daneman
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | | | | | | | - Issam A. Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, The University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | | |
Collapse
|
29
|
Ji H, Wu D, Kimberlee O, Li R, Qian G. Molecular Perspectives of Mitophagy in Myocardial Stress: Pathophysiology and Therapeutic Targets. Front Physiol 2021; 12:700585. [PMID: 34276422 PMCID: PMC8279814 DOI: 10.3389/fphys.2021.700585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/27/2021] [Indexed: 01/15/2023] Open
Abstract
A variety of complex risk factors and pathological mechanisms contribute to myocardial stress, which ultimately promotes the development of cardiovascular diseases, including acute cardiac insufficiency, myocardial ischemia, myocardial infarction, high-glycemic myocardial injury, and acute alcoholic cardiotoxicity. Myocardial stress is characterized by abnormal metabolism, excessive reactive oxygen species production, an insufficient energy supply, endoplasmic reticulum stress, mitochondrial damage, and apoptosis. Mitochondria, the main organelles contributing to the energy supply of cardiomyocytes, are key determinants of cell survival and death. Mitophagy is important for cardiomyocyte function and metabolism because it removes damaged and aged mitochondria in a timely manner, thereby maintaining the proper number of normal mitochondria. In this review, we first introduce the general characteristics and regulatory mechanisms of mitophagy. We then describe the three classic mitophagy regulatory pathways and their involvement in myocardial stress. Finally, we discuss the two completely opposite effects of mitophagy on the fate of cardiomyocytes. Our summary of the molecular pathways underlying mitophagy in myocardial stress may provide therapeutic targets for myocardial protection interventions.
Collapse
Affiliation(s)
- Haizhe Ji
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China.,Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dan Wu
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| | - O'Maley Kimberlee
- School of Public Health, University of California, Berkeley, Berkeley, CA, United States
| | - Ruibing Li
- Department of Clinical Laboratory Medicine, The First Medical Center, Medical School of Chinese People's Liberation Army, Beijing, China
| | - Geng Qian
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
30
|
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
|
31
|
Protective Effect of Optic Atrophy 1 on Cardiomyocyte Oxidative Stress: Roles of Mitophagy, Mitochondrial Fission, and MAPK/ERK Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3726885. [PMID: 34211623 PMCID: PMC8205577 DOI: 10.1155/2021/3726885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/08/2021] [Accepted: 05/21/2021] [Indexed: 12/14/2022]
Abstract
Myocardial infarction is associated with oxidative stress and mitochondrial damage. However, the regulatory mechanisms underlying cardiomyocyte oxidative stress during myocardial infarction are not fully understood. In the present study, we explored the cardioprotective action of optic atrophy 1- (Opa1-) mediated mitochondrial autophagy (mitophagy) in oxidative stress-challenged cardiomyocytes, with a focus on mitochondrial homeostasis and the MAPK/ERK pathway. Our results demonstrated that overexpression of Opa1 in cultured rat H9C2 cardiomyocytes, a procedure that stimulates mitophagy, attenuates oxidative stress and increases cellular antioxidant capacity. Activation of Opa1-mediated mitophagy suppressed cardiomyocyte apoptosis by downregulating Bax, caspase-9, and caspase-12 and upregulating Bcl-2 and c-IAP. Using mitochondrial tracker staining and a reactive oxygen species indicator, our assays showed that Opa1-mediated mitophagy attenuated mitochondrial fission and reduced ROS production in cardiomyocytes. In addition, we found that inhibition of the MAPK/ERK pathway abolished the antioxidant action of Opa1-mediated mitophagy in these cells. Taken together, our data demonstrate that Opa1-mediated mitophagy protects cardiomyocytes against oxidative stress damage through inhibition of mitochondrial fission and activation of MAPK/ERK signaling. These findings reveal a critical role for Opa1 in the modulation of cardiomyocyte redox balance and suggest a potential target for the treatment of myocardial infarction.
Collapse
|
32
|
Abstract
Vascular endothelial cells are highly plastic and show great phenotypic heterogeneity. In recent years, emerging technologies have identified a range of novel endothelial phenotypes and functions. In this Special Issue of Angiogenesis, we present a series of papers from leading experts in the field, highlighting the heterogeneity and plasticity of endothelial cells in health and disease.
Collapse
Affiliation(s)
- Coert Margadant
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Location VUmc, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The use of genetic models has facilitated the study of the origins and mechanisms of vascular disease. Mouse models have been developed to specifically target endothelial cell populations, with the goal of pinpointing when and where causative mutations wreck their devastating effects. Together, these approaches have propelled the development of therapies by providing an in-vivo platform to evaluate diagnoses and treatment options. This review summarizes the most widely used mouse models that have facilitated the study of vascular disease, with a focus on mouse models of vascular malformations and the road ahead. RECENT FINDINGS Over the past 3 decades, the vascular biology scientific community has been steadily generating a powerful toolkit of useful mouse lines that can be used to tightly regulate gene ablation, or to express transgenic genes, in the murine endothelium. Some of these models inducibly (constitutively) alter gene expression across all endothelial cells, or within distinct subsets, by expressing either Cre recombinase (or inducible versions such as CreERT), or the tetracycline controlled transactivator protein tTA (or rtTA). This now relatively standard technology has been used to gain cutting edge insights into vascular disorders, by allowing in-vivo modeling of key molecular pathways identified as dysregulated across the vast spectrum of vascular anomalies, malformations and dysplasias. However, as sequencing of human patient samples expands, the number of interesting candidate molecular culprits keeps increasing. Consequently, there is now a pressing need to create new genetic mouse models to test hypotheses and to query mechanisms underlying vascular disease. SUMMARY The current review assesses the collection of mouse driver lines that have been instrumental is identifying genes required for blood vessel formation, remodeling, maintenance/quiescence and disease. In addition, the usefulness of these driver lines is underscored here by cataloguing mouse lines developed to experimentally assess the role of key candidate genes in vascular malformations. Despite this solid and steady progress, numerous new candidate vascular malformation genes have recently been identified for which no mouse model yet exists.
Collapse
Affiliation(s)
- Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
34
|
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
|