1
|
Chen Z, Zhou Q, Droux J, Liu YH, Glück C, Gezginer I, Wyss M, Yoshihara HAI, Kindler DR, Weber B, Wegener S, El Amki M, Razansky D. Transcranial Cortex-Wide Imaging of Murine Ischemic Perfusion With Large-Field Multifocal Illumination Microscopy. Stroke 2025; 56:170-182. [PMID: 39705394 DOI: 10.1161/strokeaha.124.047996] [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: 04/03/2024] [Revised: 10/20/2024] [Accepted: 11/07/2024] [Indexed: 12/22/2024]
Abstract
BACKGROUND Ischemic stroke is a common cause of death worldwide and a main cause of morbidity. Presently, laser speckle contrast imaging, x-ray computed tomography, and magnetic resonance imaging are the mainstay for stroke diagnosis and therapeutic monitoring in preclinical studies. These modalities are often limited in terms of their ability to map brain perfusion with sufficient spatial and temporal resolution, thus calling for development of new brain perfusion techniques featuring rapid imaging speed, cost-effectiveness, and ease of use. METHODS We report on a new preclinical high-resolution angiography technique for murine ischemic stroke imaging based on large-field high-speed multifocal illumination fluorescence microscopy. We subsequently showcase the proposed method by monitoring therapeutic effects of thrombolysis in stroke (n=6), further performing cross-strain comparison of perfusion dynamics (n=6) and monitoring the therapeutic effects of sensory stimulation-based treatment (n=11). RESULTS Quantitative readings of hemodynamic and structural changes in cerebral vascular network and pial vessels were attained with 14.4-µm spatial resolution at 80-Hz frame rate fully transcranially. The in vivo perfusion maps accurately delineated the ischemic core and penumbra, further exhibiting a strong correlation (86.1±4.5%) with ex vivo triphenyl tetrazolium chloride staining, significantly higher than for the conventional laser speckle contrast imaging method. Monitoring of therapeutic effects of thrombolysis confirmed that early recanalization could effectively save the penumbra while reducing the infarct area. Cross-strain comparison of perfusion dynamics affirmed that C57BL/6 mice feature a larger penumbra and smaller infarct core as compared with BALB/c mice, which have few or no collaterals. Sensory stimulation-based treatment could effectively enhance blood flow and abolish perfusion deficits in the ischemic core and penumbra regions. CONCLUSIONS A high-speed fluorescence-based angiography method for transcranial stroke imaging in mice is introduced, which is capable of localizing brain perfusion changes and accurately assessing the ischemic penumbra. Compared with the whole-brain x-ray computed tomography and magnetic resonance imaging methods, which are conventionally used for stroke diagnosis and therapeutic monitoring, the new approach is simple and cost-effective, further offering high resolution and speed for in vivo studies. It thus opens new venues for brain perfusion research under various disease conditions such as stroke, neurodegeneration, or epileptic seizures.
Collapse
Affiliation(s)
- Zhenyue Chen
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Department of Information Technology and Electrical Engineering, Institute for Biomedical Engineering, ETH Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., I.G., H.A.I.Y., D.R.K., D.R.)
- Institute of Precision Optical Engineering, School of Physics Science and Engineering, Tongji University, Shanghai, China (Z.C.)
| | - Quanyu Zhou
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Department of Information Technology and Electrical Engineering, Institute for Biomedical Engineering, ETH Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., I.G., H.A.I.Y., D.R.K., D.R.)
| | - Jeanne Droux
- Department of Neurology, University Hospital and University of Zurich, Switzerland (J.D., S.W., M.E.A.)
- Zurich Neuroscience Center, Switzerland (J.D., C.G., M.W., B.W., S.W., M.E.A., D.R.)
| | - Yu-Hang Liu
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Department of Information Technology and Electrical Engineering, Institute for Biomedical Engineering, ETH Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., I.G., H.A.I.Y., D.R.K., D.R.)
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Zurich Neuroscience Center, Switzerland (J.D., C.G., M.W., B.W., S.W., M.E.A., D.R.)
| | - Irmak Gezginer
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Department of Information Technology and Electrical Engineering, Institute for Biomedical Engineering, ETH Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., I.G., H.A.I.Y., D.R.K., D.R.)
| | - Matthias Wyss
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Zurich Neuroscience Center, Switzerland (J.D., C.G., M.W., B.W., S.W., M.E.A., D.R.)
| | - Hikari A I Yoshihara
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Department of Information Technology and Electrical Engineering, Institute for Biomedical Engineering, ETH Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., I.G., H.A.I.Y., D.R.K., D.R.)
| | - Diana Rita Kindler
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Department of Information Technology and Electrical Engineering, Institute for Biomedical Engineering, ETH Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., I.G., H.A.I.Y., D.R.K., D.R.)
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Zurich Neuroscience Center, Switzerland (J.D., C.G., M.W., B.W., S.W., M.E.A., D.R.)
| | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, Switzerland (J.D., S.W., M.E.A.)
- Zurich Neuroscience Center, Switzerland (J.D., C.G., M.W., B.W., S.W., M.E.A., D.R.)
| | - Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, Switzerland (J.D., S.W., M.E.A.)
- Zurich Neuroscience Center, Switzerland (J.D., C.G., M.W., B.W., S.W., M.E.A., D.R.)
| | - Daniel Razansky
- Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., C.G., I.G., M.W., H.A.I.Y., D.R.K., B.W., D.R.)
- Department of Information Technology and Electrical Engineering, Institute for Biomedical Engineering, ETH Zurich, Switzerland (Z.C., Q.Z., Y.-H.L., I.G., H.A.I.Y., D.R.K., D.R.)
- Zurich Neuroscience Center, Switzerland (J.D., C.G., M.W., B.W., S.W., M.E.A., D.R.)
| |
Collapse
|
2
|
Sugimoto K, Chung DY, Fischer P, Takizawa T, Qin T, Yaseen MA, Sakadžić S, Ayata C. Optogenetic Functional Activation Is Detrimental During Acute Ischemic Stroke in Mice. Stroke 2024; 55:2502-2509. [PMID: 39234742 PMCID: PMC11421960 DOI: 10.1161/strokeaha.124.048032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 07/03/2024] [Accepted: 08/12/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Functional activation of the focal ischemic brain has been reported to improve outcomes by augmenting collateral blood flow. However, functional activation also increases metabolic demand and might thereby worsen outcomes. Indeed, preclinical and clinical reports have been conflicting. Here, we tested the effect of functional activation during acute ischemic stroke using distal middle cerebral artery occlusion in anesthetized mice. METHODS Using transgenic mice expressing channelrhodopsin-2 in neurons, we delivered functional activation using physiological levels of transcranial optogenetic stimulation of the moderately ischemic cortex (ie, penumbra), identified using real-time full-field laser speckle perfusion imaging during a 1-hour distal microvascular clip of the middle cerebral artery. Neuronal activation was confirmed using evoked field potentials, and infarct volumes were measured in tissue slices 48 hours later. RESULTS Optogenetic stimulation of the penumbra was associated with more than 2-fold larger infarcts than stimulation of the contralateral homotopic region and the sham stimulation group (n=10, 7, and 9; 11.0±5.6 versus 5.1±4.3 versus 4.1±3.7 mm3; P=0.008, 1-way ANOVA). Identical stimulation in wild-type mice that do not express channelrhodopsin-2 did not have an effect. Optogenetic stimulation was associated with a small increase in penumbral perfusion that did not explain enlarged infarcts. CONCLUSIONS Our data suggest that increased neuronal activity during acute focal arterial occlusions can be detrimental, presumably due to increased metabolic demand, and may have implications for the clinical management of hyperacute stroke patients.
Collapse
Affiliation(s)
- Kazutaka Sugimoto
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurosurgery, Yamaguchi University School of Medicine, Japan
| | - David Y. Chung
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Paul Fischer
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurology with Experimental Neurology Charité-Universitätsmedizin Berlin and Berlin Institute of Health at Charité – Universitätsmedizin Berlin,BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin Germany
| | - Tsubasa Takizawa
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Tao Qin
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Mohammad A. Yaseen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Bioengineering Department, Northeastern University, Boston, MA, USA
| | - Sava Sakadžić
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Cenk Ayata
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA
| |
Collapse
|
3
|
Bhatti MS, Frostig RD. Astrocyte-neuron lactate shuttle plays a pivotal role in sensory-based neuroprotection in a rat model of permanent middle cerebral artery occlusion. Sci Rep 2023; 13:12799. [PMID: 37550353 PMCID: PMC10406860 DOI: 10.1038/s41598-023-39574-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/27/2023] [Indexed: 08/09/2023] Open
Abstract
We have previously demonstrated protection from impending cortical ischemic stroke is achievable by sensory stimulation of the ischemic area in an adult rat model of permanent middle cerebral artery occlusion (pMCAo). We have further demonstrated that a major underpinning mechanism that is necessary for such protection is the system of collaterals among cerebral arteries that results in reperfusion of the MCA ischemic territory. However, since such collateral flow is weak, it may be necessary but not sufficient for protection and therefore we sought other complementary mechanisms that contribute to sensory-based protection. We hypothesized that astrocytes-neuron lactate shuttle (ANLS) activation could be another potential underpinning mechanism that complements collateral flow in the protection process. Supporting our hypothesis, using functional imaging, pharmacological treatments, and postmortem histology, we showed that ANLS played a pivotal role in sensory stimulation-based protection of cortex and therefore serves as the other supporting mechanism underpinning the protection process.
Collapse
Affiliation(s)
- Mehwish S Bhatti
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, Irvine, CA, USA.
| | - Ron D Frostig
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, Irvine, CA, USA.
- Department of Biomedical Engineering, School of Engineering, University of California, Irvine, Irvine, CA, USA.
- Center for Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
4
|
Bhatti M, Frostig RD. Astrocyte-neuron lactate shuttle plays a pivotal role in sensory-based neuroprotection in a rat model of permanent middle cerebral artery occlusion. RESEARCH SQUARE 2023:rs.3.rs-2698138. [PMID: 37034797 PMCID: PMC10081351 DOI: 10.21203/rs.3.rs-2698138/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
We have previously demonstrated protection from impending cortical stroke is achievable by sensory stimulation of the ischemic area in an adult rat model of permanent middle cerebral artery occlusion (pMCAo). We have further demonstrated that a major underpinning mechanism that is necessary for such protection is the system of collaterals among cerebral arteries that results in reperfusion of the MCA ischemic territory. However, since such collateral flow is weak, it may be necessary but not sufficient for protection and therefore we were seeking other complementary mechanisms that contribute to sensory-based protection. We hypothesized that astrocytes-to-neuron shuttle (ANLS) is another potential underpinning mechanism that could complement collateral flow in the protection process. Supporting our hypothesis, using functional imaging, pharmacological treatments, and postmortem histology, we show that ANLS has a pivotal role in sensory-based protection of cortex and therefor serves as the other supporting mechanism underpinning the protection process.
Collapse
|
5
|
Li Y, Song R, Shen G, Huang L, Xiao D, Ma Q, Zhang L. MicroRNA-210 Downregulates TET2 (Ten-Eleven Translocation Methylcytosine Dioxygenase 2) and Contributes to Neuroinflammation in Ischemic Stroke of Adult Mice. Stroke 2023; 54:857-867. [PMID: 36734233 PMCID: PMC10151037 DOI: 10.1161/strokeaha.122.041651] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 12/16/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Stroke is a leading cause of morbidity and mortality worldwide. Neuroinflammation plays a key role in acute brain injury of ischemic stroke. MicroRNA-210 (miR210) is the master hypoxamir and regulates microglial activation and inflammation in a variety of diseases. In this study, we uncovered the mechanism of miR210 in orchestrating ischemic stroke-induced neuroinflammation through repression of TET2 (ten-eleven translocation methylcytosine dioxygenase 2) in the adult mouse brain. METHODS Ischemic stroke was induced in adult WT (wild type) or miR210 KO (miR210 deficient) mice by transient intraluminal middle cerebral artery occlusion. Injection of TET2 silencing RNA or miR210 complementary locked nucleic acid oligonucleotides, or miR210 KO mice were used to validate miR210-TET2 axis and its role in ischemic brain injury. Furthermore, the effect of TET2 overexpression on miR210-stimulated proinflammatory cytokines was examined in BV2 microglia. Post assays included magnetic resonance imaging scan for brain infarct size; neurobehavioral tests, reverse transcription-quantitative polymerase chain reaction, and Western blot for miR210; and TET2 levels, flow cytometry, and ELISA for neuroinflammation in the brain after stroke or microglia in vitro. RESULTS miR210 injection significantly reduced TET2 protein abundance in the brain, while miR210 complementary locked nucleic acid oligonucleotides or miR210 KO preserved TET2 regardless of ischemic brain injury. TET2 knockdown reversed the protective effects of miR210 inhibition or miR210 KO on ischemic stroke-induced brain infarct size and neurobehavioral deficits. Moreover, flow cytometry and ELISA assays showed that TET2 knockdown also significantly dampened the anti-inflammatory effect of miR210 inhibition on microglial activation and IL (interleukin)-6 release after stroke. In addition, overexpression of TET2 in BV2 microglia counteracted miR210-induced increase in cytokines. CONCLUSIONS miR210 inhibition reduced ischemic stroke-induced neuroinflammatory response via repression of TET2 in the adult mouse brain, suggesting that miR210 is a potential treatment target for acute brain injury after ischemic stroke.
Collapse
Affiliation(s)
- Yong Li
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Guofang Shen
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lei Huang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - DaLiao Xiao
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Qingyi Ma
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
6
|
Bonnin P, Kubis N, Charriaut-Marlangue C. Collateral Supply in Preclinical Cerebral Stroke Models. Transl Stroke Res 2021; 13:512-527. [PMID: 34797519 PMCID: PMC9232412 DOI: 10.1007/s12975-021-00969-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 02/01/2023]
Abstract
Enhancing the collateral blood supply during the acute phase of cerebral ischemia may limit both the extension of the core infarct, by rescuing the penumbra area, and the degree of disability. Many imaging techniques have been applied to rodents in preclinical studies, to evaluate the magnitude of collateral blood flow and the time course of responses during the early phase of ischemic stroke. The collateral supply follows several different routes at the base of the brain (the circle of Willis) and its surface (leptomeningeal or pial arteries), corresponding to the proximal and distal collateral pathways, respectively. In this review, we describe and illustrate the cerebral collateral systems and their modifications following pre-Willis or post-Willis occlusion in rodents. We also review the potential pharmaceutical agents for stimulating the collateral blood supply tested to date. The time taken to establish a collateral blood flow supply through the leptomeningeal anastomoses differs between young and adult animals and between different species and genetic backgrounds. Caution is required when transposing preclinical findings to humans, and clinical trials must be performed to check the added value of pharmacological agents for stimulating the collateral blood supply at appropriate time points. However, collateral recruitment appears to be a rapid, beneficial, endogenous mechanism that can be stimulated shortly after artery occlusion. It should be considered a treatment target for use in addition to recanalization strategies.
Collapse
Affiliation(s)
- Philippe Bonnin
- APHP, Physiologie Clinique - Explorations Fonctionnelles, Hôpital Lariboisiere, Université de Paris, 2 rue Ambroise Paré, F-75010, Paris, France. .,INSERM U1148, LVTS, Hôpital Bichat, Université de Paris, F-75018, Paris, France.
| | - Nathalie Kubis
- APHP, Physiologie Clinique - Explorations Fonctionnelles, Hôpital Lariboisiere, Université de Paris, 2 rue Ambroise Paré, F-75010, Paris, France.,INSERM U1148, LVTS, Hôpital Bichat, Université de Paris, F-75018, Paris, France
| | | |
Collapse
|
7
|
Gamma frequency activation of inhibitory neurons in the acute phase after stroke attenuates vascular and behavioral dysfunction. Cell Rep 2021; 34:108696. [PMID: 33535035 DOI: 10.1016/j.celrep.2021.108696] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/06/2020] [Accepted: 01/06/2021] [Indexed: 11/24/2022] Open
Abstract
Alterations in gamma oscillations occur in several neurological disorders, and the entrainment of gamma oscillations has been recently proposed as a treatment for neurodegenerative disease. Optogenetic stimulation enhances recovery in models of stroke when applied weeks after injury; however, the benefits of acute brain stimulation have not been investigated. Here, we report beneficial effects of gamma-frequency modulation in the acute phase, within 1 h, after stroke. Transgenic VGAT-ChR2 mice are subject to awake photothrombotic stroke in an area encompassing the forelimb sensory and motor cortex. Optogenetic stimulation at 40 Hz in the peri-infarct zone recovers neuronal activity 24 h after stroke in motor and parietal association areas, as well as blood flow over the first week after stroke. Stimulation significantly reduces lesion volume and improves motor function. Our results suggest that acute-phase modulation of cortical oscillatory dynamics may serve as a target for neuroprotection against stroke.
Collapse
|
8
|
Yanagisawa T, Zhang H, Suzuki T, Kamio Y, Takizawa T, Morais A, Chung DY, Qin T, Murayama Y, Faber JE, Patel AB, Ayata C. Sex and Genetic Background Effects on the Outcome of Experimental Intracranial Aneurysms. Stroke 2020; 51:3083-3094. [PMID: 32912097 DOI: 10.1161/strokeaha.120.029651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Intracranial aneurysm formation and rupture risk are, in part, determined by genetic factors and sex. To examine their role, we compared 3 mouse strains commonly used in cerebrovascular studies in a model of intracranial aneurysm formation and rupture. METHODS Intracranial aneurysms were induced in male CD1 (Crl:CD1[ICR]), male and female C57 (C57BL/6NCrl), and male 129Sv (129S2/SvPasCrl or 129S1/SvImJ) mice by stereotaxic injection of elastase at the skull base, combined with systemic deoxycorticosterone acetate-salt hypertension. Neurological deficits and mortality were recorded. Aneurysms and subarachnoid hemorrhage grades were quantified postmortem, either after spontaneous mortality or at 7 to 21 days if the animals survived. In separate cohorts, we examined proinflammatory mediators by quantitative reverse transcriptase-polymerase chain reaction, arterial blood pressure via the femoral artery, and the circle of Willis by intravascular latex casting. RESULTS We found striking differences in aneurysm formation, rupture, and postrupture survival rates among the groups. 129Sv mice showed the highest rates of aneurysm rupture (80%), followed by C57 female (36%), C57 male (27%), and CD1 (21%). The risk of aneurysm rupture and the presence of unruptured aneurysms significantly differed among all 3 strains, as well as between male and female C57. The same hierarchy was observed upon Kaplan-Meier analysis of both overall survival and deficit-free survival. Subarachnoid hemorrhage grades were also more severe in 129Sv. CD1 mice showed the highest resistance to aneurysm rupture and the mildest outcomes. Higher mean blood pressures and the major phenotypic difference in the circle of Willis anatomy in 129Sv provided an explanation for the higher incidence of and more severe aneurysm ruptures. TNFα (tumor necrosis factor-alpha), IL-1β (interleukin-1-beta), and CCL2 (chemokine C-C motif ligand 2) expressions did not differ among the groups. CONCLUSIONS The outcome of elastase-induced intracranial aneurysm formation and rupture in mice depends on genetic background and shows sexual dimorphism.
Collapse
Affiliation(s)
- Takeshi Yanagisawa
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.).,Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston. (T.Y., A.B.P.).,Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan (T.Y., Y.M.)
| | - Hua Zhang
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill. (H.Z., J.E.F.)
| | - Tomoaki Suzuki
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.)
| | - Yoshinobu Kamio
- Department of Neurosurgery, Hamamatsu University School of Medicine, Japan (Y.K.)
| | - Tsubasa Takizawa
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.)
| | - Andreia Morais
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.).,National Institute of Translational Neuroscience, Biomedical Science Institute, Federal University of Rio de Janeiro, Brazil (A.M.)
| | - David Y Chung
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.).,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston. (D.Y.C., C.A.)
| | - Tao Qin
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.)
| | - Yuichi Murayama
- Department of Neurosurgery, Jikei University School of Medicine, Tokyo, Japan (T.Y., Y.M.)
| | - James E Faber
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill. (H.Z., J.E.F.).,Department of Neurobiology Curriculum, McAllister Heart Institute, University of North Carolina, Chapel Hill. (J.E.F.)
| | - Aman B Patel
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston. (T.Y., A.B.P.)
| | - Cenk Ayata
- Neurovascular Research Laboratory, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown (T.Y., T.S., T.T., D.Y.C., T.Q., A.M., C.A.).,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston. (D.Y.C., C.A.)
| |
Collapse
|
9
|
Sugimoto K, Chung DY, Böhm M, Fischer P, Takizawa T, Aykan SA, Qin T, Yanagisawa T, Harriott A, Oka F, Yaseen MA, Sakadžić S, Ayata C. Peri-Infarct Hot-Zones Have Higher Susceptibility to Optogenetic Functional Activation-Induced Spreading Depolarizations. Stroke 2020; 51:2526-2535. [PMID: 32640946 PMCID: PMC7387208 DOI: 10.1161/strokeaha.120.029618] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Spreading depolarizations (SDs) are recurrent and ostensibly spontaneous depolarization waves that may contribute to infarct progression after stroke. Somatosensory activation of the metastable peri-infarct tissue triggers peri-infarct SDs at a high rate. METHODS We directly measured the functional activation threshold to trigger SDs in peri-infarct hot zones using optogenetic stimulation after distal middle cerebral artery occlusion in Thy1-ChR2-YFP mice. RESULTS Optogenetic activation of peri-infarct tissue triggered SDs at a strikingly high rate (64%) compared with contralateral homotopic cortex (8%; P=0.004). Laser speckle perfusion imaging identified a residual blood flow of 31±2% of baseline marking the metastable tissue with a propensity to develop SDs. CONCLUSIONS Our data reveal a spatially distinct increase in SD susceptibility in peri-infarct tissue where physiological levels of functional activation are capable of triggering SDs. Given the potentially deleterious effects of peri-infarct SDs, the effect of sensory overstimulation in hyperacute stroke should be examined more carefully.
Collapse
Affiliation(s)
- Kazutaka Sugimoto
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurosurgery, Yamaguchi University School of Medicine, Japan
| | - David Y. Chung
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Maximilian Böhm
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Paul Fischer
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Tsubasa Takizawa
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Sanem Aslihan Aykan
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Tao Qin
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Takeshi Yanagisawa
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Andrea Harriott
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA
| | - Fumiaki Oka
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurosurgery, Yamaguchi University School of Medicine, Japan
| | - Mohammad A. Yaseen
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Sava Sakadžić
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Cenk Ayata
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, USA
| |
Collapse
|
10
|
Zhu J, Hancock AM, Qi L, Telkmann K, Shahbaba B, Chen Z, Frostig RD. Spatiotemporal dynamics of pial collateral blood flow following permanent middle cerebral artery occlusion in a rat model of sensory-based protection: a Doppler optical coherence tomography study. NEUROPHOTONICS 2019; 6:045012. [PMID: 31824979 PMCID: PMC6903432 DOI: 10.1117/1.nph.6.4.045012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/19/2019] [Indexed: 05/05/2023]
Abstract
There is a growing recognition regarding the importance of pial collateral flow in the protection from impending ischemic stroke both in preclinical and clinical studies. Collateral flow is also a major player in sensory stimulation-based protection from impending ischemic stroke. Doppler optical coherence tomography has been employed to image spatiotemporal patterns of collateral flow within the dorsal branches of the middle cerebral artery (MCA) as it provides a powerful tool for quantitative in vivo flow parameters imaging (velocity, flux, direction of flow, and radius of imaged branches). It was employed prior to and following dorsal permanent MCA occlusion (pMCAo) in rat models of treatment by protective sensory stimulation, untreated controls, or sham surgery controls. Unexpectedly, following pMCAo in the majority of subjects, some MCA branches continued to show anterograde blood flow patterns over time despite severing of the MCA. Further, in the presence of protective sensory stimulation, the anterograde velocity and flux were stronger and lasted longer than in retrograde flow branches, even within different branches of single subjects, but stimulated retrograde branches showed stronger flow parameters at 24 h. Our study suggests that the spatiotemporal patterns of collateral-based dorsal MCA flow are dynamic and provide a detailed description on the differential effects of protective sensory stimulation.
Collapse
Affiliation(s)
- Jiang Zhu
- University of California Irvine, Beckman Laser Institute, Irvine, California, United States
| | - Aneeka M. Hancock
- University of California Irvine, Department of Neurobiology and Behavior, Irvine, California, United States
| | - Li Qi
- University of California Irvine, Beckman Laser Institute, Irvine, California, United States
| | - Klaus Telkmann
- University of California Irvine, Department of Statistics, Irvine, California, United States
| | - Babak Shahbaba
- University of California Irvine, Department of Statistics, Irvine, California, United States
| | - Zhongping Chen
- University of California Irvine, Beckman Laser Institute, Irvine, California, United States
- University of California Irvine, Department of Biomedical Engineering, Irvine, California, United States
| | - Ron D. Frostig
- University of California Irvine, Department of Neurobiology and Behavior, Irvine, California, United States
- University of California Irvine, Department of Biomedical Engineering, Irvine, California, United States
- University of California Irvine, Center for the Neurobiology of Learning and Memory, Irvine, California, United States
| |
Collapse
|
11
|
Baron JC. Protecting the ischaemic penumbra as an adjunct to thrombectomy for acute stroke. Nat Rev Neurol 2019; 14:325-337. [PMID: 29674752 DOI: 10.1038/s41582-018-0002-2] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
After ischaemic stroke, brain damage can be curtailed by rescuing the 'ischaemic penumbra' - that is, the severely hypoperfused, at-risk but not yet infarcted tissue. Current evidence-based treatments involve restoration of blood flow so as to salvage the penumbra before it evolves into irreversibly damaged tissue, termed the 'core'. Intravenous thrombolysis (IVT) can salvage the penumbra if given within 4.5 h after stroke onset; however, the early recanalization rate is only ~30%. Direct removal of the occluding clot by mechanical thrombectomy considerably improves outcomes over IVT alone, but despite early recanalization in > 80% of cases, ~50% of patients who receive this treatment do not enjoy functional independence, usually because the core is already too large at the time of recanalization. Novel therapies aiming to 'freeze' the penumbra - that is, prevent core growth until recanalization is complete - hold potential as adjuncts to mechanical thrombectomy. This Review focuses on nonpharmacological approaches that aim to restore the physiological balance between oxygen delivery to and oxygen demand of the penumbra. Particular emphasis is placed on normobaric oxygen therapy, hypothermia and sensory stimulation. Preclinical evidence and early pilot clinical trials are critically reviewed, and future directions, including clinical translation and trial design issues, are discussed.
Collapse
Affiliation(s)
- Jean-Claude Baron
- Department of Neurology, Hôpital Sainte-Anne, Université Paris 5, INSERM U894, Paris, France.
| |
Collapse
|
12
|
Murmu RP, Fordsmann JC, Cai C, Brazhe A, Thomsen KJ, Lauritzen M. Sensory Stimulation-Induced Astrocytic Calcium Signaling in Electrically Silent Ischemic Penumbra. Front Aging Neurosci 2019; 11:223. [PMID: 31496947 PMCID: PMC6712371 DOI: 10.3389/fnagi.2019.00223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/07/2019] [Indexed: 11/14/2022] Open
Abstract
Middle cerebral artery occlusion (MCAO) induces ischemia characterized by a densely ischemic focus, and a less densely ischemic penumbral zone in which neurons and astrocytes display age-dependent dynamic variations in spontaneous Ca2+ activities. However, it is unknown whether penumbral nerve cells respond to sensory stimulation early after stroke onset, which is critical for understanding stimulation-induced stroke therapy. In this study, we investigated the ischemic penumbra’s capacity to respond to somatosensory input. We examined adult (3- to 4-month-old) and old (18- to 24-month-old) male mice at 2–4 h after MCAO, using two-photon microscopy to record somatosensory stimulation-induced neuronal and astrocytic Ca2+ signals in the ischemic penumbra. In both adult and old mice, MCAO abolished spontaneous and stimulation-induced electrical activity in the penumbra, and strongly reduced stimulation-induced Ca2+ responses in neuronal somas (35–82%) and neuropil (92–100%) in the penumbra. In comparison, after stroke, stimulation-induced astrocytic Ca2+ responses in the penumbra were only moderately reduced (by 54–62%) in adult mice, and were even better preserved (reduced by 31–38%) in old mice. Our results suggest that somatosensory stimulation evokes astrocytic Ca2+ activity in the ischemic penumbra. We hypothesize that the relatively preserved excitability of astrocytes, most prominent in aged mice, may modulate protection from ischemic infarcts during early somatosensory activation of an ischemic cortical area. Future neuroprotective efforts in stroke may target spontaneous or stimulation-induced activity of astrocytes in the ischemic penumbra.
Collapse
Affiliation(s)
- Reena P Murmu
- Translational Neurobiology Group, Department of Neuroscience, Panum Institute, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Jonas C Fordsmann
- Translational Neurobiology Group, Department of Neuroscience, Panum Institute, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Changsi Cai
- Translational Neurobiology Group, Department of Neuroscience, Panum Institute, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Alexey Brazhe
- Faculty of Biology, Moscow State University, Moscow, Russia
| | - Kirsten J Thomsen
- Translational Neurobiology Group, Department of Neuroscience, Panum Institute, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark
| | - Martin Lauritzen
- Translational Neurobiology Group, Department of Neuroscience, Panum Institute, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark
| |
Collapse
|
13
|
von Bornstädt D, Gertz K, Lagumersindez Denis N, Seners P, Baron JC, Endres M. Sensory stimulation in acute stroke therapy. J Cereb Blood Flow Metab 2018; 38:1682-1689. [PMID: 30073883 PMCID: PMC6168904 DOI: 10.1177/0271678x18791073] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/14/2018] [Accepted: 06/17/2018] [Indexed: 02/06/2023]
Abstract
The beneficial effects of cortical activation for functional recovery after ischemic stroke have been well described. However, little is known about the role of early sensory stimulation, i.e. stimulation during first 6 h after stroke onset even during acute treatment. In recent years, various preclinical studies reported significant effects of acute sensory stimulation that range from entire neuroprotection to increased infarct volumes by 30-50%. Systematic knowledge about the effect of acute sensory stimulation on stroke outcome is highly relevant as stroke patients are subject to uncontrolled sensory stimulation during transport, acute treatment, and critical care. This article discusses the current stage of knowledge about acute sensory stimulation and provides directions for future experimental and clinical trials.
Collapse
Affiliation(s)
- Daniel von Bornstädt
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Center for Stroke Research Berlin, Berlin, Germany
| | - Karen Gertz
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Berlin, Germany
| | - Nielsen Lagumersindez Denis
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Berlin, Germany
| | - Pierre Seners
- Department of Neurology, Hôpital Sainte-Anne, University Paris Descartes, INSERM U894, France
| | - Jean-Claude Baron
- Department of Neurology, Hôpital Sainte-Anne, University Paris Descartes, INSERM U894, France
| | - Matthias Endres
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research) Partner Site, Berlin, Germany
| |
Collapse
|